Skip to main content
PEDIATRÍA INTEGRAL - Revista de formación continuada dirigida al pediatra y profesionales interesados de otras especialidades médicas

Syncope

A. Ortigado Matamala
Topics on
Continuous Training


A. Ortigado Matamala

Head of the Pediatric Service. Pediatric Cardiology. Guadalajara University Hospital. Professor of Medicine at the University of Alcalá

Abstract

Syncope is defined as the sudden and self-limited loss of consciousness and postural tone resulting from a transient global cerebral hypoperfusion, sometimes preceded by a prodrome and always followed by a spontaneous complete recovery. Syncope is a common pediatric problem, although syncope is almost always benign, in a few cases it may be a clue to the presence of an underlying cardiovascular problem and may predict a risk of sudden death. A detailed history of the event, a comprehensive physical examination and an electrocardiogram will help in the differential diagnosis and must show the key features for identifying high-risk patients and exclude life threatening disorder.

 

Resumen

El síncope se define como la pérdida súbita y transitoria de la conciencia y del tono postural por una hipoperfusión cerebral transitoria, alguna vez, precedida por unos pródromos y, siempre, seguida de una recuperación completa y espontánea. El síncope es un problema pediátrico frecuente, afecta al 15-25% de los niños y, especialmente a los adolescentes, con un pico de incidencia entre los 15 y 19 años de edad, y con predominio en el sexo femenino. La etiología es variada, con múltiples causas y, aunque casi siempre es benigno, en unos pocos casos puede ser el aviso de una enfermedad cardiovascular subyacente con riesgo de muerte súbita. Una historia detallada del evento, una exploración física y un electrocardiograma, deben orientarnos en el diagnóstico diferencial y pueden mostrarnos los puntos clave para identificar pacientes de alto riesgo y enfermedades que amenacen la vida.

 

Key words: Syncope; Vasovagal syncope; Cardiac syncope; Transient loss of consciousness; Sudden death; Pediatrics.

Palabras clave: Síncope; Síncope vasovagal; Síncope cardiaco; Pérdida transitoria del conocimiento; Muerte súbita; Pediatría.

 

 

Pediatr Integral 2021; XXV (8): 339 – 405

 


 

Syncope

Introduction

Syncope is the sudden loss of consciousness and postural tone, of brief duration and with spontaneous recovery.

Syncope is a sudden loss of consciousness and postural tone, due to a transient cerebral hypoperfusion, characterized by a rapid onset, a fleeting duration and a spontaneous and complete recovery(1). Syncope is a prevalent pathology in pediatric emergencies, generally benign, but generating great concern to the patient and his family. One of the challenges that syncope poses for the pediatrician is to identify the cases with underlying and potentially fatal cardiac pathology(2).

When we evaluate episodes of this nature, we must avoid terms such as: “dizziness”, “fainting”, “attack” or “crisis”, because they are ambiguous. We should also know how to differentiate a syncope from vertigo or balance disturbances, characteristic of cerebellar or vestibular dysfunction.

We must also know how to differentiate syncope from other situations with transient loss of consciousness (TLoC) which are not due to a transient cerebral hypoperfusion, such as: neurological causes (epilepsy, migraine, head trauma, transient cerebrovascular accident…), metabolic causes (hypoglycemia), poisonings (medications, drugs of abuse, carbon monoxide…) or those of psychogenic origin(3,4).

Although it seems a clear and well-defined topic, there are still open issues in the medical literature that raise the need for a consensus. There are two important international clinical practice guidelines for the diagnosis and management of syncope; on the one hand, the 2017 American guideline of the “American College of Cardiology/American Heart Association/Heart Rhythm Society” (ACC/AHA/HRS), and on the other hand, the 2018 fourth edition of the European guideline “European Society of Cardiology/European Heart Rhythm Association” (ESC/EHRA(5,6). Interestingly, there are certain differences between the American and the European guidelines. Another issue to highlight is that, although both guidelines are very thorough, both hardly expound on the subject of syncope in the pediatric age, where there are specific forms, such as sobbing spasms, with its two variants, pale and cyanotic.

It is also important not to confuse syncope with presyncope. The term “presyncope” refers to the clinical situation with the symptoms previous to syncope, that is, the vegetative symptoms including: dizziness, paleness, cold sweating and blurred vision, but without losing consciousness. Therefore, presyncope would be a failed episode of syncope.

If the cerebral anoxia in a syncope extends for more than 15 seconds, a convulsive syncope may occur, characterized by generalized tonic spasms, mandibular trismus, opisthotonos, myoclonic jerks and sphincter relaxation(7). Convulsive syncope, although infrequent (5% of syncope episodes), can pose one of the main challenges for the pediatrician in its differential diagnosis with an epileptic disorder(3,4).

Epidemiology

Syncope is common in pediatrics, especially in adolescence, being benign in most cases.

Syncope is a frequent clinical situation in Pediatrics, affecting 15-25% of children and adolescents, with a peak of maximum incidence at 18 years of age (30-50%). Syncope is more common in females, especially between the ages of 15 and 19 years(4). Syncope is uncommon below 6 years of age, except if there is an underlying neurological (epilepsy) or cardiac pathology (arrhythmia), or sobbing spasms. Syncope is the reason for attending the pediatric emergency service in 0.5-3 cases per 1,000.

Most episodes of syncope in pediatrics are benign, 75% correspond to reflex or neurally mediated syncope, where among them, vasovagal syncope, also known as neurocardiogenic syncope, stands out(10,11). Cardiac syncope accounts for 5-10%, and its prognosis underscores its great clinical interest. Recurrence rate varies between 33-51% in patients who have been followed for 5 years(9).

Etiopathogenesis

Syncope can have a varied etiology, where vasovagal syncope is the most common, however cardiac syncope can represent a potentially fatal underlying disease.

The causes of syncope are multiple, with various underlying pathogenic mechanisms, and not all of them completely clarified.

Low blood pressure and cerebral hypoperfusion are the main causes of syncope. Systemic blood pressure is determined by cardiac output and peripheral vascular resistance, and a decline in one of these two factors can cause syncope.

There are three causes of low vascular resistance:

1. Reflex response abnormality with vasodilation (vasodepressor-type reflex syncope).

2. Functional failure of the autonomic nervous system.

3. Structural failure of the autonomic nervous system.

Autonomic nervous system failure can be primary or secondary, for instance, drug-induced.

The four causes of low cardiac output are:

1. Reflex bradycardia (cardioinhibitory-type reflex syncope).

2. Structural heart diseases and arrhythmias, especially ventricular tachycardias.

3. Inadequate venous return: hypovolemia or venous accumulation.

4. Chronotropism and inotropism anomaly due to autonomic nervous system dysfunction.

It must be borne in mind that these different primary mechanisms can interfere and interact with each other(5).

To facilitate the understanding and clinical management of syncope, it is advisable to classify syncope based on its etiopathogenesis, which can be summarized into 3 large groups (Table I):

1. Reflex or neurally mediated syncope.

2. Cardiac syncope

3. Non-cardiac syncope.

Reflex or neurally mediated syncope

Reflex syncope is the most common one (75%). Diverse factors such as erect position of the body and venous pool in the lower extremities condition a sudden decrease in cardiac preload, which for instance is accentuated in prolonged standing. There is a paradoxical reflex response in syncope. The relative hypovolemia due to a decrease in blood pressure causes an initial release of catecholamines to improve chronotropism and inotropism. However, cardiac contractions in relatively empty ventricles stimulate the left ventricular mechanoreceptors and via the afferent vagal pathway (unmyelinated C fibers) reach the brainstem. Subsequently, the efferent vagal response is twofold, causing bradycardia (cardioinhibitory response) and vasodilation (vasodepressor response).

Nevertheless, the mechanisms involved are broader and require further clarification. Syncope triggered by an intense emotion, the sight of blood or trauma from afar, suggest the involvement of brain structures. There are studies that indicate the existence of low levels of serotonin and high levels of beta-endorphins in patients with this type of syncope(10).

Another relevant component that has been related to neurally mediated syncope is endothelial function(11,12). Nitric oxide (NO) is a powerful vasodilator in our body, generated by endothelial cells through the nitric oxide synthase (NOS) enzyme. In children and adolescents with this type of syncope, higher levels of NO and NOS have been detected than in the control group(13).

Vagal or neurocardiogenic syncope is the most frequent and representative form of this type of syncope, where, in addition to the mechanisms already described, there is usually a precipitating factor that would act as a trigger, such as: emotional distress (anxiety, fear, pain) or physical stress (heat, prolonged standing, fatigue, dehydration, fasting, intercurrent illness…).

Sometimes syncope occurs in specific circumstances, such as during urination, especially in men (whilst standing) and in the first void of the morning (fasting and after sitting up from a previous lying down position). Other situational syncope episodes are related to defecation, ingestion of cold drinks, physical exertion, hot baths, coughing, intense laughter, blowing or playing wind instruments (Valsalva maneuver).

Another clinical picture within this group is carotid sinus syndrome. The carotid sinus is a regulator of blood pressure through its baroreceptors. In case of hypersensitivity, performing a stimulus such as a carotid sinus massage can lead to syncope(14).

In Pediatrics, there is a specific entity of reflex syncope called sobbing spasms with its two variants, the cyanotic and the pale type. Cyanotic-type sobbing spasm occurs between 6 months and 5 years of age, peaking at 2 years, in which the syncope begins with a loud cry (“tantrum”) followed by an apnea pause. The pale-type sobbing spasm appears between 12 and 24 months of age, and the syncope occurs suddenly after a stimulus (pain or fright) and without prior crying. The prognosis of these clinical entities is good, 100% will disappear with age. 25% of the cases of pale-type sobbing spasms will develop vasovagal syncope in adolescence(3).

Another entity to take into account is postural orthostatic tachycardia syndrome (POTS), frequent in adolescents, especially in females. This entity consists of an intolerance to standing that occurs in the first 10 minutes after modifying the posture to standing up, with an acceleration of the heart rate greater than 30 bpm (maximum rate of: 130 bpm for 6-12 years, 125 bpm for 13-18 years, and more than 120 bpm in adults) in the absence of hypotension. Dizziness, weakness, blurred vision, and syncope may manifest along with tachycardia. POTS would be an exaggeration of a compensatory physiological mechanism. Although its pathophysiology is not fully clear, it has been related to various factors such as: hypovolemia, iron deficiency, hyperadrenergic state, dysfunction of local regulation of vascular tension, endothelial dysfunction, autoimmune dysfunction (antibodies against cholinesterase receptors) or mast cell activation(15).

Cardiac syncope

Although syncope of cardiac origin is rare (5-10%), it is of great significance due to its morbidity and mortality. Syncope in these cases may be the first manifestation of a life-threatening heart disease(16).

Globally, they can be classified into three large groups: the first one being situations of obstruction to the ventricular outflow tract; in second place, myocardial dysfunction and, finally, arrhythmias.

Left ventricular outflow tract obstructions include aortic stenosis and obstructive hypertrophic cardiomyopathy, whereas in the case of the right ventricle, severe pulmonary stenosis and primary pulmonary hypertension stand out(3).

Myocardial dysfunction can be primary (e.g., familial cardiomyopathies) or secondary (e.g., viral myocarditis). Ischemic cardiomyopathy due to coronary lesion is uncommon in Pediatrics, but, as a congenital pathology, we must bear in mind the Anomalous origin of the Left Coronary Artery from the Pulmonary Artery (ALCAPA) syndrome and, as an acquired pathology, Kawasaki disease(17).

Among the bradyarrhythmia that can cause syncope, carotid sinus disease is an etiological condition, and, above all, complete atrioventricular block, which can be congenital (child of mother with systemic lupus erythematosus) or acquired (post-cardiac surgery, inflammatory/infectious diseases of the heart)(4).

Tachyarrhythmias are also a cause of syncope. Supraventricular tachycardias (e.g., Wolff-Parkinson-White syndrome) are usually better tolerated in Pediatrics, but ventricular tachycardias have a worse prognosis, being potentially fatal. Special mention should be made to channelopathies, such as long QT syndrome, short QT syndrome, Brugada syndrome or catecholaminergic polymorphic ventricular tachycardia that, having structurally normal hearts, are associated with severe ventricular tachyarrhythmias that can culminate in sudden death. Physicians must recall that the QT interval can be lengthened, not only congenitally (17 subtypes of long QT syndrome have already been described), but also acquired by electrolyte disturbances (hypokalemia) and especially by the administration of drugs (certain antiarrhythmics, psychotropics, macrolides or antihistamines)(18). It is important to be aware of these medications, especially in patients with congenital long QT syndrome. The lists of drugs that prolong the QT interval, grouped into three categories according to the level of evidence, are available on the website of the Arizona Center for Education and Research in Therapeutics (AZCERT) (www.crediblemeds.org). These lists are periodically updated taking into consideration notifications from regulatory agencies and available clinical evidence. One only has to register for free on the website to receive these updates(19).

Non-cardiac syncope

There are other entities that, although they do not meet the requirements of syncope, must be taken into consideration, especially for the differential diagnosis with other forms of transient loss of consciousness (TLoC).

In this group, epilepsy should be highlighted, due to the challenge it poses for the physician so as not to confuse an epileptic seizure with a syncopal seizure (Table II).

Other neurological abnormalities that can simulate a syncopal episode are migraines (basilar migraine), transient cerebrovascular accidents, cataplegic seizures in narcolepsy, “vestibular syncope” (Tumarkin otolithic seizures or drop attacks) or acute elevations of cerebrospinal fluid pressure (hydrocephalus)(3).

Among the “psychogenic syncope” entities, various disorders of a psychiatric nature must be taken into account, such as hysterical crises, panic attacks, conversion reactions or major depression. In case of hyperventilation, hypocapnia occurs which, if not compensated, derives in acute respiratory alkalosis, with dizziness, confusion, peripheral and perioral paresthesias, cramps and finally syncope(4).

Hypoglycemia can mimic syncopal manifestations due to the adrenergic response associated with: anxiety, tachycardia, sweating, paleness, tremor, weakness, and sleepiness.

We should not forget the possibility of ingestion of toxins, medications or drugs of abuse in patients with syncope, especially in adolescents. Among medications, there are those that lengthen the QT segment of the electrocardiogram, drugs that decrease blood volume (antihypertensives and diuretics), and psychotropic drugs(10).

Diagnosis

The diagnosis of syncope is based on a detailed medical history, a full physical examination, and an electrocardiogram.

The diagnosis of syncope is based on three basic pillars:

1. Medical history.

2. Physical examination.

3. Electrocardiogram (EKG).

Medical history

The medical history is the cornerstone and essential tool in the diagnosis of syncope, with a detailed recall of all the information, both from the patient and from possible witnesses. The clinical history must include the following information(5):

• Personal and family history. It is important to know the patient’s history and possible diseases of interest (cardiac diseases, epilepsy, diabetes…), as well as family´s (cardiomyopathies, cardiac arrhythmias, cases of sudden death, vasovagal syncope in parents…). In adolescent girls, assess the possibility of pregnancy.

• Predisposing factors. Look for data on the circumstances prior to syncope, which may be predisposing factors, such as: possible fasting, medication intake, the environment where it took place (indoor or outdoor premises, temperature, crowded spaces), activity carried out (rest, sport, bath, urination…) or position of the patient (standing, decubitus or sitting).

• Triggering or precipitating factors. The report of the episode should consider possible syncope triggers that may have acted as a trigger for the loss of consciousness (postural change, panic, fear, trauma…).

• In congenital long QT syndromes, syncope may be associated with severe ventricular arrhythmia (“Torsade de pointes”) induced by a specific trigger. Type 1 (LQTS1) is related to physical exercise (especially swimming), type 2 (LQTS2) to emotional stress or a sudden intense auditory stimulus, and type 3 (LQTS3) with sleep (bradycardia).

• Initial clinical manifestations (prodrome). Next, the initial symptoms that precede the loss of consciousness (paleness, sweating, nausea, vomiting, abdominal pain, chest pain, palpitations, weakness, dizziness, blurred vision…) should be described.

• Description of the syncope. The characteristics of the episode of transient loss of consciousness (TLoC), its duration, body attitude, mucocutaneous color and description of possible abnormal movements (convulsive crisis?) are important.

• Later recovery. Confirm normality, later symptoms or existence of a post-critical state (drowsiness, confusion, neurological focus…).

Physical examination

The physical examination should be complete, paying special attention to cardiovascular signs (palpation of central arterial pulses, heart rate or auscultation of heart murmurs) and neurological signs (level of consciousness, balance, vestibular function, neurological focus…). Vital signs should be measured, especially heart rate and blood pressure.

An important issue to assess in the physical examination is changes with orthostatism, requesting the patient to move from the decubitus position to standing and after 3 minutes checking if symptoms appear and measuring the heart rate and blood pressure. A drop of more than 20 mmHg in systolic blood pressure, or 10 mmHg in diastolic blood pressure, or an acceleration in the heart rate greater than 30 bpm, suggests a neurally mediated mechanism of the syncope(4).

One method to evaluate autonomic function is through the Valsalva maneuver, which is more profitable in adult patients than in Pediatrics. The patient performs a forced expiration for 15 seconds with the glottis closed (mouth and nose closed). Initially, blood pressure rises slightly due to filling of the left ventricle, but soon decreases due to the abrupt reduction in venous return, cardiac output decreases, and heart rate increases to compensate (baroreceptor reflex). This hypotension in healthy individuals will cause a compensatory automatic sympathetic response: it increases systemic vascular resistance and heart rate. At the end of the maneuver, when the patient releases all the trapped air and breathes normally, the intrathoracic overpressure disappears, a drop in blood pressure occurs, and the heart rate normalizes. In situational syndrome, for example, when playing a wind instrument or lifting excessive weight, normal heart rate response is associated with pronounced hypotension due to lack of the compensatory automatic response(6).

Another of the diagnostic tests is the carotid sinus massage, which reproduces the syncope with a reflex mechanism (bradycardia and hypotension), however, this maneuver is not indicated in Pediatrics.

Complementary tests

In addition to measuring blood pressure in decubitus and standing position, a capillary blood glucose should also be checked.

There is a broad consensus on the recommendation to perform a 12-lead electrocardiogram (EKG) in all syncope episodes, although the Canadian guidelines indicate it in an atypical syndrome or if associated risk factors(7,8). In the EKG the heart rate and rhythm should be checked, arrhythmias should be ruled out, and the presence of sinus rhythm confirmed.

Even in an EKG with sinus rhythm, the following warning signs, indicative of possible cardiac pathology in syncope, should be ruled out:

• Wolff-Parkinson-White (WPW) syndrome pattern: short PR interval (from the beginning of the P wave to the beginning of the QRS complex), wide QRS complex and delta wave (initial filling of the QRS complex, positive or negative).

• QT interval: the corrected QT (QTc) must be obtained using the Bazett formula, because the QT interval varies with heart rate (Fig. 1), preferably measured in leads II and V5. Remember that the QT interval goes from the beginning of the Q wave to the end of the T wave (that of the tangent of the T wave), not including the U wave, if identified.

Figura1

Figure 1. Calculation of the QTc (Bazzet’s formula).

• Long QT syndrome (LQTS): QTc prolongation, greater than 450 msec in men and greater than 460 msec in women.

• Short QT syndrome (SQTC): shortening of the QTc, less than 330 msec.

• Brugada syndrome pattern: downward elevation of the ST segment greater than or equal to 2 mm (“shark fin”) in more than one right lead (V1-V2), followed by negative T waves (type I pattern).

• Hypertrophic cardiomyopathy: left ventricular enlargement with changes in repolarization, pathological wide and deep Q waves, unevenness of the ST segment, negative T waves on the lower and left side.

• Arrhythmogenic cardiomyopathy: presence of epsilon waves which, although infrequent, are specific (electrical potentials of low amplitude between the end of the QRS complex and the beginning of the ST segment), inverted T waves in the right precordial (V1, V2, V3) in individuals older than 14 years of age and in the absence of complete right bundle branch block.

The tilt-table test or upright tilt testing allows the provocation of syncope symptoms in predisposed people and under a controlled situation with monitoring of blood pressure, heart rate, respiratory rate and peripheral oxygen saturation. Tilt table performance is suboptimal. The indication for this test would be limited to patients with recurrent syncope and reflex syncope suspicion, but in whom the clinical evaluation has failed to specify it. Therefore, the tilt table test is useful for the diagnosis of reflex syncope, in autonomic failure that occurs with orthostatic hypotension, in postural orthostatic tachycardia syndrome (POTS), and it could also be useful in differentiating a vasovagal syncope from a psychiatric pseudosyncope(6).

Electroencephalogram (EEG) is not indicated as a routine test in syncope and would be limited to those cases with suspected underlying neurological etiology, specifically, epilepsy. An epileptic seizure with loss of consciousness can simulate a convulsive syncope and, therefore, it is important to consider it in the differential diagnosis (Table II). Neuroimaging tests, such as Computerized Axial Tomography (CT) or Magnetic Resonance Imaging (MRI), would also be limited in their indication, for the same reason.

Blood tests, except for capillary blood glucose, are not necessary in every syncope, hence full blood count (e.g., hypovolemia or infection) or biochemistry (suspected metabolic abnormality) must be taken only if justified by clinical criteria. In certain cases, the possibility of requesting urine toxicology tests or a pregnancy test should be taken into account.

If the patient is referred to the pediatric cardiology clinic, the study can be completed with echocardiography, ambulatory EKG-Holter and ergometry. In specific cases, other studies may also be indicated such as: cardiac MRI, subcutaneous implantable Holter, electrophysiological study and cardiac catheterization.

Treatment

Treatment is based on three main pillars: reassuring, educating and preventing.

Most syncope episodes are neurally mediated and it is, therefore, important to inform the patient and family about the benign nature of the process, avoiding or minimizing predisposing and triggering factors for syncope episodes (avoidance of prolonged standing, stressful, hot environments or agglomerations). Thus, it is important to help them to learn how to detect prodromal symptoms and guide them with instructions to carry out maneuvers to interrupt the episode (supine position, physical counter-pressure maneuvers)(20).

The most common counterpressure maneuvers are the following three(6):

1. In a standing position cross the legs, making maximum tension of the muscles of the legs, abdomen and buttocks, in an intense and sustained manner.

2. Contraction of hands, squeezing a rubber ball or a soft object with the greatest possible force, in an intense and sustained way.

3. Tense the arms, with the hands remaining intertwined and correctly grasped, producing an intense and sustained isometric contraction, with the arms in the horizontal plane and pulling outwards with the elbows (centrifugal force).

As a general rule, maintaining good hydration (fluid intake 30-50 mL/kg/day, 1.5-2.5 L/day) and a controlled increase in salt intake (2-5 g/day ) are recommended(7).

The use of pharmacological treatment is controversial and limited to specific cases (fludrocortisone, beta-blockers, alpha1-agonists).

Role of the Primary Care pediatrician

The Primary Care pediatrician is essential in syncope, because it is usually the doctor responsible for its initial assessment and the one who decides its clinical management.

Syncope is a frequent cause of consultation in Primary Care and, the pediatrician, the doctor who attends it. Most syncope episodes are benign, and a detailed medical history, a complete physical examination, and an EKG are sufficient to reach a diagnosis. This is the case of neurally mediated syncope, specifically, vasovagal syncope, which does not require further study or referral to a specialist, but does need to inform the patient and family of the benign nature of the condition, and of its possible prevention or how to act in the event of another episode(21).

One of the challenges for the pediatrician in a patient with syncope is to detect the possibility of an underlying pathology, especially of cardiac origin, which requires referral to the hospital emergency department or to the cardiology clinic (Fig. 2).

Figura1

Figure 2. Syncope clinical management algorithm. PAT: pediatric assessment triangle.

 

Bibliography

The asterisks show the interest of the article in the opinion of the author.

1.** Zavala R, Metais B, Tuckfield L, DelVecchio M, Aronoff S. Pediatric syncope: a systematic review. Pediatr Emer Care. 2020; 36: 442-5.

2.*** Koene RJ, Adkinsson WO, Benditt DG. Syncope ant the risk of sudden cardiac death: evaluation, management and prevention. J Arrhythm. 2017; 5: 33-44.

3.*** Kanjwal K, Calkins H. Syncope in children and adolescents. Cardiol Clin. 2015; 33: 397409.

4.** Anderson JB, Willis M, Lancaster H, Leonard K, Thomas C. The evaluation and management of pediatric Syncope. Pediatr Neurol. 2016; 55: 6-13.

5.** Shen W-K, Sheldon RS, Benditt DG, Cohen MI, Forman DE, Goldberger ZD, et al. 2017 ACC/AHA/HRS Guideline for the evaluation and management of patients with syncope. A report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation. 2017; 136: e60-122.

6.*** Brignole M, Moya A, de Lange FJ, Deharo JC, Elliott PM, Fanciulli A, et al. ESC Scientific Document Group. 2018 ESC Guidelines for the diagnosis and management of syncope. Eur Heart J. 2018; 39: 1883-948.

7.*** Moodley M. Clinical approach to syncope in children. Semin Pediatr Neurol. 2013; 20: 12-7.

8.*** Sanatani S, Chau V, Fournier A, Dixon A, Blondin R, Sheldon R. Canadian Cardiovascular Society and Canadian Pediatric Cardiology Association Position Statement on the Approach to Syncope in the Pediatric Patient. Can J Cardiol. 2017; 33: 189-198.

9.** Zhang Q, Du J, Wang Z, Du Z, Wang L, Tang C. The diagnostic protocol in children and adolescents with syncope: a multi-center prospective study. Acta Paediatr. 2009; 98: 879-84.

10.** Puñal JE, Rodríguez A, Gómez C, Martinón-Torres F, Castro-Gago M, Martinón JM. Síncope en el adolescente. Orientación diagnóstica y terapéutica. An Pediatr. 2005; 63: 330-9.

11.** Dehghan B, Sabri MR, Javanmard SH, Ahmadi AR, Mansourian M. Neurally mediated syncope: is it really an endothelial dysfunction? Anatol J Cardiol. 2016; 16: 701-6.

12.** Santini L, Capria A, Brusca V, Violo A, Smurra F, Scarf I. et al. An increased endothelial-independent vasodilation is the hall mark of the neurally mediated syncope. Clin Cardiol. 2012; 35: 107-10.

13.** Liao Y, Chen S, Liu X, Zhang Q, Ai Y, Wang Y, et al. Flow-mediated vasodilation and endothelium function in children with postural orthostatic tachycardia syndrome. Am J Cardiol. 2010; 106: 378-82.

14.** Moya-i-Mitjans A, Rivas-Gándana N, Sarrias-Mercè A, Pérez-Rodón J, Roca-Luque I. Síncope. Rev Esp Cardiol. 2012; 65: 755-65.

15.** Chen G, Du J, Jin J, Huang Y. Postural tachycardia syndrome in children and adolescents: pathologhysiology anc clinical management. Font Pediatr. 2020; 8: 34. Doi:10.3389/fped.2020.00474.

16.** Hammond BH, Zahka KG, aziz PF. Suden cardiac death: a pediatrician´s role. Pediatr Rev. 2019; 40: 456-67.

17.** Lee TM, Hsu DT, Kantor P, Towbin JA, Ware SM, Colan SD, et al. Pediatric cardiomyopathies. Circ Res. 2017; 121: 855-73.

18.** Wallace E, Howard L, Liu M, O´Brien T, Ward D, Shen S, et al. Long QT syndrome: genetics and future perspective. Pediatr Cardiol. 2019; 40: 1419-30.

19.** Web de Arizona Center for Education and Research on Therapeutics. Accessed september 6th 2021. Available at: www.azcert.org o www. qtdrugs.org.

20.** Strickberger SA, Benson W, Biaggioni I, Callans DJ, Cohen MI, Ellenbogen KA, et al. AHA/ACCF scientific statement on the evaluation of syncope. From the American Heart Association Councils on clinical cardiology, cardiovascular nursing, cardiovascular disease in the young, and stroke, and the quality of care and outcomes research interdisciplinary working group; and the American College of cardiology Foundation in collaboration with the Heart Rhythm Society. J Am Coll Cardiol. 2006; 47: 473-84.

21.** Rivera DR, Cartón AJ. Síncope. En: Guerrero-Fernández J, Cartón A, Barreda A, Menéndez J, Ruiz J, ed. Manuel de Diagnóstico y Terapéutica en Pediatría. Madrid: Editorial Médica Panamericana S.A.; 2018. p. 325-30.

Recommended bibliography

- Brignole M, Moya A, de Lange FJ, Deharo JC, Elliott PM, Fanciulli A, et al. ESC Scientific Document Group. 2018 ESC Guidelines for the diagnosis and management of syncope. Eur Heart J. 2018; 39: 1883-948.

Clinical practice international guide where all the scientific evidence on syncope is collected and evaluated. This article is important to clarify concepts and definitions, as well as to assist the physician in the evaluation, management, and treatment of these patients.

- Kanjwal K, Calkins H. Syncope in children and adolescents. Cardiol Clin. 2015; 33: 397409.

Article that focuses on syncope in the pediatric patient, describing its peculiarities and providing a global vision of the subject.

- Moodley M. Clinical approach to syncope in children. Semin Pediatr Neurol. 2013; 20: 12-7.

Short article in length, but helpful for the pediatrician by providing a practical approach in the clinical management of syncope in Pediatrics.

- Koene RJ, Adkinsson WO, Benditt DG. Syncope ant the risk of sudden cardiac death: evaluation, management and prevention.J Arrhythm. 2017; 5: 33-44.

This article highlights the importance of syncope as a clinical presentation of sudden death in different situations and diseases.

- Sanatani S, Chau V, Fournier A, Dixon A, Blondin R, Sheldon R. Canadian Cardiovascular Society and Canadian PediatricCardiology Association Position Statement on the Approach to Syncope in the Pediatric Patient. Can J Cardiol. 2017; 33: 189-198.

This article presents a very thorough guide of the approach to syncope and it is specific for pediatric patients.

 

Clinical case

 

A school teacher observes and reports how a healthy 9-year-old boy, whilst playing with his friends in the schoolyard, right after standing up, suddenly loses consciousness, subsequently lying on the floor without abnormal movements. After a minute he regains consciousness, he is scared and only refers that prior to losing consciousness he noticed pain in his chest. His parents are notified and, although the child is apparently asymptomatic, they attend their pediatrician´s clinic at the Health Care Center. The pediatrician already knows the patient, as he is a thin child (weight in 3rd centile), with lack of appetite and gets tired easily. For this reason, his pediatrician had already requested a complete blood test a few weeks before. There are no data of interest in the personal and family history.

On physical examination the patient is in good general condition, blood pressure 100/65 mmHg, oxygen saturation 98%, a new heart murmur is detected, a systolic murmur 2/6 in the tricuspid focus, the rest of the examination being normal. An EKG is performed (Fig. 3).

Figura1

Figure 3. 12-lead EKG. Note that the voltage setting in the frontal and precordial leads changes (10 mm/mV vs. 5 mm/mV).

 

Follow-up of childhood cancer in Primary Care. How to detect late effects

J. Huerta Aragonés, C. Mata Fernández
Topics on
Continuous Training


J. Huerta Aragonés, C. Mata Fernández

Pediatric and Adolescent Hematology and Oncology Section. Pediatrics Service. Gregorio Marañón General University Hospital. Maternal and child Hospital. School of Medicine. Complutense University of Madrid. Gregorio Marañón Health Research Institute (IiSGM). CIBEREHD

Abstract

Over the last several decades there has been a significant increase in the survival of most pediatric tumors, which is undoubtedly a success, but generates a new scenario with an increase in late side effects and premature mortality compared to the general population. Therefore, it is highly important to develop models for follow-up care of childhood cancer survivors, which allow fluid and efficient communication between healthcare professionals, individualized management for each patient, implementation of healthy lifestyle habits and early diagnosis protocols for relapses and late sequelae. Following this approach, adherence to follow-up will be increased and the severity of chronic toxicity will be reduced.

There are national and international consensus guidelines of increasing quality, but with a high degree of heterogeneity between them. In this article reference will be made to these follow-up guidelines, highlighting the specific late side effects on each organ system, their prompt recognition and recommendations for longterm management.

 

Resumen

En las últimas décadas, se ha experimentado un notable incremento en la supervivencia de los tumores diagnosticados en edad pediátrica, lo cual supone indudablemente un éxito de la medicina, pero genera un nuevo escenario, con un aumento de los efectos secundarios tardíos y de la mortalidad prematura respecto a la población general. Es de vital importancia, por tanto, el desarrollo de modelos de seguimiento del paciente superviviente, que permitan una comunicación fluida y eficiente entre profesionales sanitarios, un manejo individualizado para cada paciente, la implementación de hábitos de vida saludables y de protocolos de diagnóstico precoz, de recaídas y de secuelas tardías. De esta forma, se aumentará la adherencia al seguimiento y se reducirá la gravedad de la toxicidad crónica.
Existen guías de consenso nacionales e internacionales, cada vez de mayor calidad, pero con una elevada heterogeneidad entre ellas. A lo largo de la siguiente revisión, se hará referencia a estas guías de seguimiento, haciendo especial hincapié en los efectos secundarios tardíos por aparatos, su diagnóstico precoz y recomendaciones de manejo a largo plazo.

 

Key words: Supervivientes de cáncer infantil; Efectos secundarios tardíos; Toxicidad a largo plazo; Segundas neoplasias.

Palabras clave: Childhood cancer survivors; Late side effects; Long-term toxicity; Second malignancies.

 

 

Pediatr Integral 2021; XXV (7): 372 – 385

 


 

Follow-up of childhood cancer in Primary Care. How to detect late effects

Introduction

Cancer survival rates in the pediatric age have notably increased in recent decades, where prevention as well as early diagnosis of late sequelae strategies have become essential.

The number of childhood cancer survivors has increased significantly in the last 40 years, increasing from 58% survival between 1975-1977 to 83% between 2008-2014(1). In Spain, a 23% increase in survival was observed (from 54 to 77%, from 1980-1984 to 2000-2004), with a 50% decrease in the risk of death in that period (https://www.uv.es/rnti/cifrasCancer.html). In Europe, 1 in 500-600 children will develop a malignancy before the age of 15 and it is estimated that, in the next few years, one in 450 young adults will be a childhood cancer survivor in Europe(2). In contrast to this great advance, there is a high prevalence of morbidity in the form of late side effects. The Children’s Oncology Group (COG) in a childhood cancer survival study observed the existence of at least one chronic problem in the second decade of life in almost 60% of survivors and more than 30% of severe chronic sequelae 30 years after diagnosis(3). The St. Jude Lifetime Cohort Study published a study in which it was observed that at 45 years of age, 95.2% of the survivors reported at least one chronic health condition, being it serious, disabling or life threatening in 80% of the cases (twice that of the general population at that age)(4).

Cancer treatments predispose recipients to excess morbidity and premature mortality compared to the general population. The risk is directly proportional to the intensity of the treatment to achieve the cure of the disease, being more burdensome in the case of multimodal therapy and in patients who have received several treatments due to relapse. Age is a determining factor, the younger the more toxic effects on linear growth, skeletal maturation, intellectual functionality, sexual development and organic functionality.

Although these are worrisome data, the impact of these late effects can be modified through early detection and adequate management of the pathologies, as well as, prevented in part through a modification of lifestyle habits(1). Likewise, it has become a generalized trend of current protocols to seek, in cases where a good long-term survival result has been achieved, to reduce acute and chronic toxicity without detriment to survival (e.g., acute lymphoblastic leukemia, nephroblastoma, Hodgkin lymphoma, certain brain tumors…), in particular, reducing the use of radiotherapy and adjusting the intensity of treatment to the individual risk(5,6).

Reducing late effects and early detection of them, if they exist, has become a goal for these patients, in order to improve their quality of life. Follow-up and recommendations must be individualized according to the personal characteristics of each survivor, type of tumor, treatment received, age, comorbidities, lifestyle, and existence of dysmorphic syndromes or predisposition to cancer(1) (Fig. 1). Side effects increase with age, but can manifest throughout life, hence, an adequate monitoring based on recommendations and consensus guidelines, from pediatric age to adulthood is essential.

Figura

Figure 1. Lifestyle recommendations and health promotion.

Follow-up Models of Childhood Cancer Survivors

There is not a unique model for the monitoring of long-term side effects, and the superiority of one over the others has not been demonstrated by studies of adequate duration and methodology. Table I contains a comparison between the different models, their advantages and disadvantages(1).

Figura

In general, the most suitable option would be the one that best suits the individual characteristics of the patient. In our environment, joint work between healthcare professionals from the hospital and Primary Care setting is essential, as well as a progressive and structured transition to the adult care model. Information transfer is crucial, especially: previous medical history, dates of diagnosis and end of treatment, diagnostic tests at diagnosis and during follow-up, multimodal treatments received and their doses, complications related to them or to the disease itself, relapses and follow-up plan (Table II).

Figura

Producing detailed medical reports for the physicians who will provide ongoing care for survivors in Primary Care or during their adulthood is paramount and often one of the deficits of hospital care. In this sense, a good number of Pediatric and Adolescent Oncology and Hematology services are making efforts to implement specific follow-up consultations for the survivor and transition to adult care, paying special attention to the elaboration of “survivor’s passport”. There are tools promoted by the International Society of European Pediatric Oncology (SIOPe) (http://www.survivorshippassport.org/) and through European projects such as the PanCareSurPass project (Horizon 2020-EU.3.1.5 Framework Program, https://cordis.europa.eu/project/id/899999/es).

Follow-up recommendations

There are consensus guidelines for the follow-up of survivors from different international groups that, although heterogeneous and variable in the recommendations, result very useful.

In recent years, national and international guidelines have been developed for the monitoring of long-term side effects in survivors. In the United States, the Children’s Oncology Group (COG) periodically publishes the “Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent, and Young Adult Cancers” (latest version: 5.0, October 2018). It is a guide based on the individual risk, tools for diagnosis and management of side effects. The current version is accessible through the website www.survivorshipguidelines.org(7). In Europe, three large groups have developed similar guidelines: the UKCCSG (United Kingdom Children’s Cancer Study Group) “Therapy Based Long-Term Follow Up Practice Statement”(8), the “Long Term Follow Up of Survivors of Childhood Cancer, A National Clinical Guideline” (Scottish Intercollegiate Guidelines Network, SIGN)(9) and the “Guidelines for follow-up after childhood cancer more than 5 years after diagnosis” (Late Effects Taskforce of the Dutch Childhood Oncology Group, DCOG LATER)(10). Each of these recommendations uses a different methodology and, at times, the recommendations are variable. With the aim of homogenizing them, the International Late Effects of Childhood Cancer Guideline Harmonization Group (IGHG) was created, however, given the complexity of the issue, for now, it has only prepared documents for the screening of breast cancer, cardiomyopathy, early ovarian failure, male gonadal toxicity, thyroid cancer and ototoxicity (www.ighg.org/guidelines/). The Spanish Society of Pediatric Hematology and Oncology (SEHOP) published the guide “Late effects in survivors of childhood cancer” in 2012, which constitutes a tool of indisputable value(11). The need to develop consensus guidelines is obvious, but the long-term cost-effectiveness of these recommendations remains unknown(1).

Late effects on organs and body systems

There are guidelines for side effects tailored to each specific treatment that, due to their breadth, far exceed the length of this article(7,8). Thus, we will focus on side effects by organs or systems. They are shown in table III in a very summarized manner.

Figura

Cardiology sequelae

Patients at greatest risk are those receiving high-dose anthracyclines, chest or mediastinal radiation therapy, or both; however, other conventional and targeted therapies also require vigilance.

Cardiovascular complications are one of the main problems for survivors, with an increased risk of heart failure compared to the general population (15 times higher) and premature death from cardiac causes (7 times higher)(12). Anthracyclines, regardless of the type, produce direct and indirect toxicity (free radicals) on the myocyte. There is no safe dose if long-term normal cardiovascular status is the goal. Chronic toxicity manifests itself months or even many years later, consisting of dose-dependent non-ischemic degenerative dilated cardiomyopathy (more frequent than restrictive) due to irreversible myocardial damage, as well as arterial hypertension, ischemic heart disease, and heart failure. Serious arrhythmias, tachycardia, ventricular fibrillation and even second- and third-degree blocks may appear. The incidence of conventional cardiotoxicity is 7.5-10% 30 years after finishing treatment(12,13). Female sex, earlier age, higher cumulative dose (> 250 mg/m2), and concomitant cardiothoracic irradiation are associated with an increased risk of cardiotoxicity and greater severity. There are also other classic chemotherapy agents (cyclophosphamide, cytarabine, cisplatin, ifosfamide, paclitaxel, 5-fluorouracil…) that produce cardiac toxicity by other mechanisms(14). In recent years, other emerging therapies must also be considered, such as proteasome, HER2, VEGF and tyrosine kinase (TK) inhibitors, as well as immunotherapy, including CAR-T cell therapy and immune checkpoint inhibitors(15) (Table IV).

Figura

The development of cardioprotective molecules has been a desire over the last years. Dexrazoxane has been postulated as a strong candidate, as it improves the cardiotoxicity observed with anthracyclines. Its use has been limited by the suspicion that it could interfere with the efficacy of anthracyclines, as well as by a possible contribution to the development of secondary neoplasms. None of these aspects have been proven in recent studies, including a meta-analysis with a sample of 4,639 children treated with anthracyclines for different neoplasms (Shaikh F et al., J Natl Cancer Inst. 2016). The American Heart Association and the American Academy of Pediatrics recommend its use as a cardioprotective agent in protocols that use anthracyclines (the Children’s Oncology Group -COG- recommends it since 2015 in those protocols that involve doses ≥ 150 mg/m2 or cardiothoracic radiotherapy). In our setting, it is not routinely used in the clinical practice. The use of liposomal forms of anthracyclines and analogues (epirubicin, idarubicin, mitoxantrone) could improve long-term toxicity(14) and long-term (6-96 hours) dosing regimens are recommended instead of boluses. Drugs such as enalapril or phosphocreatine (Cheuk DKL et al. Cochrane Database Syst Rev. 2016), beta-blockers, statins or anti-aldosteronic agents have not been shown to have a protective effect.

The mediastinal/thoracic radiotherapy produces acute inflammation on the cardiomyocytes and generates hypercoagulability. Oxidative stress leads to chronic inflammation and subsequent fibrosis (Velásquez CA et al. Rev Colombiana Cardiol 2018). Risk factors include: high cumulative doses (> 30-35 Gy), high daily doses (> 2 Gy/session), age <50 years at the time of therapy, previous history of heart disease or cardiovascular risk factors, and concomitant cardiotoxic chemotherapy treatment(7). Toxicity manifests as: ventricular dysfunction, endothelial damage (increased vascular risk, coronary artery disease, degenerative aortic vascular disease or supra-aortic trunk disease, stroke), dilated or restrictive cardiomyopathy, chronic/constrictive pericarditis, degenerative valvular disease and arrhythmias.

In summary, high-risk patients are those who have received ≥ 250 mg/m2 of anthracyclines, > 35 Gy of chest radiation or ≥ 100 mg/m2 of anthracyclines + ≥ 15 Gy of irradiation(12). Surveillance in high-risk patients should begin no later than 2 years after the end of cardiotoxic treatment and should be repeated 5 years after diagnosis, continuing every 5 years or sooner if necessary.

The semiology to be monitored will be dyspnea, chest pain, palpitations, intolerance to exercise and activities of daily life. Routine cardiopulmonary auscultation is recommended, as well as ruling out the presence of carotid murmurs. Echocardiography is the main imaging test for diagnosis and follow-up, with magnetic resonance imaging being an interesting supplementation in selected cases (not as a screening technique)(16). The electrocardiogram is a tool for the diagnosis of arrhythmias and cardiomyopathy, being necessary, on occasions, the performance of stress tests and Holter tests(7).

Regarding biomarkers, atrial natriuretic peptide, especially its pro-B NT fraction, could be a predictor of subsequent dysfunction if elevated during treatment. In asymptomatic patients, it should be interpreted with caution and always in combination with other elements. They are not recommended as the sole follow-up strategy in high-risk patients(12).

Regular exercise is recommended for survivors with normal ventricular function. The American College of Sports Medicine recommends 30-40 minutes of aerobic exercise, five times a week, along with 2 days of higher intensity training. In case of exercise-related symptoms, these should be promptly communicated. Other cardiovascular risk factors (hypertension, diabetes, dyslipidemia and obesity) should be monitored, and tobacco use avoided. During pregnancy, these patients should be closely followed, especially during the first trimester(12).

Finally, chronic fatigue syndrome should be considered, as it has a variable prevalence depending on the series (10-80%), although it is generally considered to affect 30% of children, adolescents and young adults. It consists of a subjective experience of persistent fatigue and severe tiredness, years after diagnosis, unexplained by organic causes, and that does not improve with rest. It is difficult to differentiate from depression. Risk factors include: having received radiotherapy (pulmonary therapy is the most clearly related), psychological stress, relapses, comorbidity of other side effects, female sex, unemployment and affective loneliness. It is inversely correlated with time progression since treatment and with an adequate social and labor integration. There are tools for its diagnosis (PROMIS pediatric fatigue measures, PedsQL MFS). Management is carried out through behavioral therapy (physical exercise, time management, social and occupational integration…), as the efficacy of pharmacological therapies has not been demonstrated(17).

Respiratory sequelae

Pulmonary toxicity is a common late complication, which can have a great impact on the quality of life of survivors, hence the importance in having a high index of suspicion.

Chronic respiratory tract damage can be related to primary or metastatic disease, infectious complications during treatment, and multimodal therapies themselves. The presentation can be acute or chronic, as summarized in Table V.

Figura

The cumulative incidence of pulmonary complications increases with the time elapsed since diagnosis(16), in particular, pulmonary fibrosis and chronic pneumonitis. Radiation therapy can cause oxidative damage to the vascular endothelium and affect the normal development of the chest wall, especially beyond 20 Gy(8,16). In patients receiving a hematopoietic stem cell transplant there is a special vulnerability, due to the conditioning regimens, complications derived from graft versus host disease (GVHD) and respiratory infections associated with the procedure.

During follow-up, watchful vigilance should be kept for guiding symptoms such as persistent dry cough, dyspnea, or exercise intolerance. Lung auscultation should be performed during visits, with baseline O2 saturation and chest X-ray, if symptoms are present. In these cases, the study of lung function is essential, specially assessing restrictive patterns (forced vital capacity, diffusion capacity). Many patients have asymptomatic or oligosymptomatic lung function abnormalities, which should be followed for possible progressive deterioration. The avoidance of tobacco use and its passive inhalation should be advised(11).

Neuropsychological sequelae

Many patients, particularly in high-risk groups, present late neurological, sensory, or cognitive toxicity that impacts their cognitive function and psychosocial integration in adult life.

Neurocognitive sequelae are observed in up to 40-45% of survivors, mainly in those who have suffered a primary or secondary tumor involvement of the central nervous system and in those who have received radiotherapy at that level (CNS tumors, acute lymphoblastic leukemia in previous protocols). Sometimes, there is a functional deficit (visual abnormalities, cranial nerve deficit, hemiparesis, ataxia…) derived from the growth of the tumor itself or from its resection surgery. In the long term, the manifestations can be sensory-motor (aphasia, paresis, focal deficits, peripheral neuropathy…) or neurocognitive (memory, attention, learning, speed of processing and execution of tasks, decreased school performance…). Radiation therapy is especially harmful in children younger than 3-5 years of age. A higher dose is associated with a higher risk of neurocognitive sequelae, in relation to leukoencephalopathy and vascular microangiopathic lesion(11).

At the peripheral nervous system level, sequelae can be observed in patients treated with cisplatin doses > 300 mg/m2 and with vinca alkaloids (especially vincristine, particularly in malnourished children). Cisplatin is often associated with sensory neuropathy. Vincristine rarely causes chronic neuropathy, but if present, it can be multiple (sensorimotor, autonomic, cranial nerves). In these cases, neurophysiological studies are required, as well as offering adequate rehabilitation support, occupational therapy and analgesia if the neuropathy is painful(9).

A follow-up assessment of intellectual, visual, sensory perception, memory, language and learning capacity is recommended, assessing academic performance and behavior in the family and social environment. When stagnation or deficits are observed in any of these areas, early intervention is deemed necessary. Imaging tests (CT angiography, MR angiography) may be required to complete the global assessment, as well as an assessment by Neuropediatrics and Psychology(10).

At a psychological level, there is up to 80% more probability of presenting psychological limitations that affect the quality of life and two times more emotional stress when a comparison with the patient’s siblings is established. Not all tumors present the same pattern of psychological sequelae. Brain tumor survivors display a higher rate of depression, somatization, fatigue, and drowsiness; and those of leukemia, more stress, depression and anxiety during adolescence, compared to other tumors and the general population(8).

Brain irradiation is associated with psychological stress, somatization, a greater feeling of fatigue, poorer physical and mental performance, sleep disruption, and poorer school and social integration. Patients treated with intensive chemotherapy, especially with alkylating agents, present with more stress, anxiety, depression and somatization, especially with alkylating treatments. Risk behaviors associated with psychological dysfunction, high smoking rate and alcohol consumption (in relation to worse socioeconomic status) have been described. In adults, a high rate of suicidal ideation and suicide attempts has been described, with brain tumor survivors having the highest incidence (10.4%)(7). No association has been found with age at diagnosis, time since diagnosis, type of treatment, recurrence or second tumors. Low educational level, low income, unemployment and emotional loneliness have been described as risk factors. In this regard, close monitoring of the social, psychological and emotional situation is relevant.

Sequelae on the senses

Sensory sequelae, especially vision and hearing, can limit the social or work capacity of survivors, so these must be carefully monitored.

The most frequent ocular abnormality is the detection of cataracts. It is related to the use of high and prolonged doses of steroids, radiotherapy at this level (ocular-orbit, cranial, total body, being dose dependent and, especially, if ≥ 2 Gy directly on the lens), busulfan or intrathecal chemotherapy(16). Xerophthalmia and lacrimal duct atrophy can occur and, more rarely: orbital hypoplasia, enophthalmos, keratitis, telangiectasia, retinopathy, maculopathy, optic chiasm neuropathy, papillary damage and glaucoma. The risk increases in case of radiotherapy ≥ 30 Gy on the eye-orbit, in case of treatment with 131-I for thyroid cancer, actinomycin-D or doxorubicin combined with radiotherapy, GVHD, frequent ocular exposure to sunlight and if there is comorbidity (diabetes mellitus, arterial hypertension). Other visual disturbances that may develop are: hypersensitivity to light, blurred vision, diplopia, nyctalopia (night blindness) and alterations in ocular refraction. The risk lasts for more than 20 years beyond the end of treatment(7).

Annual ophthalmological examination is recommended in all survivors, and in those with a history of treatment with corticosteroids or radiotherapy to the eyeball-orbit, orbital tumors, GVHD, treatment with radioactive iodine or abnormal diaphoresis. The use of approved glasses with UV protection is recommended(7).

When it comes to hearing, the most common long-term side effect is ototoxicity. Risk factors include having received high doses of cisplatin or carboplatin, radiation therapy over the head or ear (≥ 30 Gy) and having received therapies such as: aminoglycosides, furosemide, NSAIDs or iron chelators (deferasirox). The usual manifestation is sensorineural deafness, sometimes with tinnitus, and less frequently vertigo, transmission hearing loss and otosclerosis(7). Hearing evaluation is recommended at the end of treatment in patients at risk and then annually for those aged < 6 years (auditory evoked potentials), every 2 years between 6-12 years of age (tonal audiometry of 1,000-8,000 Hz, being best high frequency > 8,000 Hz) and, subsequently, every 5 years in those aged > 12 years. In patients with a ventriculoperitoneal bypass valve, a hearing assessment is recommended at the end of treatment and then every 5 years, even in the absence of other risk factors(7). Avoiding loud noises is recommended.

Smell can be affected in the form of anosmia or chronic rhinitis/sinusitis, usually in patients who have received radiotherapy or surgical resection of a nasal tumor.

Oral and dental sequelae

Patients undergoing chemotherapy before 5 years of age, especially with high-dose cyclophosphamide, are of special risk. One of the main problems of the oral cavity is xerostomia, in general, related to craniofacial radiotherapy, which can improve with frequent intake of liquids, sugar-free candies and artificial saliva tablets. Treatment predisposes to an increased risk of cavities, dental infections, and even speech or sleep disorders. Other dental problems are frequently described (enamel hypoplasia, tooth decay, tooth loss, microdontia, hypodontia, malocclusion). Careful and regular dental hygiene is essential, as well as an assessment by stomatology or dentistry in case of presenting problems. Occasionally, abnormalities in craniofacial development and trismus develop secondary to alterations in the temporomandibular joint(9).

Radiation therapy also increases the risk of developing second neoplasms. Occasionally, the oral cavity may be the site of GVHD involvement in hematopoietic stem cell transplant recipients. An adequate follow-up is important to detect these abnormalities early enough.

Endocrine sequelae

Half of childhood cancer survivors will present at least one late endocrinological disorder in their lifetime, with central nervous system tumors being the most frequently related(7). In the case of craniopharyngioma, abnormality of at least one hormonal axis takes place in 75% of patients. Risk factors are: direct damage from tumor growth itself, that derived from resection or biopsy surgeries, and radiotherapy ≥ 30 Gy. Cranial radiotherapy produces more disturbances in this axis the younger the age of the patient. The most common sequela is short stature due to GH deficiency, particularly with radiation therapy doses > 18 Gy. In adult life, the deficiency of this hormone is associated with metabolic problems such as: increased body fat, altered serum lipid profile, and decreased exercise capacity, bone mineral density and insulin sensitivity. Short stature may, however, be due to other causes (hypothyroidism, precocious puberty, growth impairment due to spinal or long bone radiation, and prolonged corticosteroid therapy)(7). Occasionally, a “catch up” growth occurs reaching the target height with adequate replacement treatment, but it does not always happen(16).

On other occasions, endocrine late effects manifest as central adrenal insufficiency due to ACTH deficiency (very rare, due to direct tumor or surgical damage or radiotherapy dose ≥ 30-50 Gy), central hypothyroidism due to TSH deficiency (cranial RT ≥ 30 Gy), hypogonadotropic hypogonadism due to LHRH, LH or FSH deficiency (second axis affected in order of frequency), prolactin deficiency or central diabetes insipidus due to ADH deficiency (infundibulum involvement by craniopharyngioma, germ cell tumor or optic glioma). The term panhypopituitarism is coined when deficiency of ≥ 3 hormones is present.

Precocious puberty, consisting of early pubertal development, rapid bone maturation, and final height shorter than target, can occur. It is related to radiotherapy dose > 18 Gy, younger age at exposition, and female sex. An annual physical examination is recommended, with anthropometric data and pubertal development monitoring, as well as bone age in case of suspicion. Hyperprolactinemia is observed in some patients, generally related to high-dose irradiation to the pituitary gland or development of second pituitary tumors (adenomas) after treatment. In case of suspicion, serum prolactin will be determined and pituitary MRI performed, as well as referral to Endocrinology(7).

At the thyroid level, hypothyroidism is usually associated with cervical, mediastinal, or spinal radiation therapy. It usually occurs in the first 5 years post-treatment, if exposed to doses > 30 Gy (especially ≥ 45 Gy). Other risk factors are: treatment with therapeutic metaiodobenzylguanidine (MIBG) (despite prophylaxis with oral iodine), anti-GD2 (neuroblastoma), tyrosine kinase inhibitors and secondary to thyroidectomy or ablative treatment with radioactive 131I. Rarely, hyperthyroidism, autoimmune thyroiditis, and thyroid nodules (benign and malignant) may be seen. Hypoparathyroidism secondary to thyroid resection surgery may occur, which requires follow-up and treatment(7).

At the gonadal level, toxicity derived from the use of high-dose alkylating agents (busulfan, melphalan, carmustine, lomustine, cyclophosphamide, ifosfamide, thiotepa, procarbazine, dacarbazine, platinum derivatives) can be observed, especially in combination with radiotherapy on the gonads. Girls are at higher risk if they receive the treatments during or after puberty. Ovarian failure occurs in up to 50% of patients who receive radiation therapy to the gonads (≥ 15 Gy), particularly in association with alkylating agents. Infertility has been described from doses ≥ 5 Gy on the ovaries. Likewise, surgery can produce an iatrogenic menopause (bilateral oophorectomy) or an advance of it (unilateral oophorectomy). In this case, hypergonadotropic hypogonadism would occur, which consequently can lead to an increased risk of premature osteoporosis and cardiovascular problems. Abdominal radiation therapy that involves the uterus increases the risk of intrauterine growth delay and preterm delivery. Annual clinical review of sexual maturation development, menstruation characteristics, hormonal determination (FSH, LH, estrogens) is recommended and, in case of suspected ovarian failure, a bone density scan (DEXA scan) and referral to Endocrinology should be performed(7,11).

In any case, patients whose treatment has placed them at risk for ovarian failure, may have premature menopause with a shortened fertility span, so it is the role of the Primary Care physician to warn of this risk and advise, if the woman is capable and willing to have offspring, to conceive no later than the first years of the fourth decade of life.

In men, germ cells (Sertoli) are sensitive to both chemotherapy and radiotherapy (at doses as low as 2-3 Gy), leading to azoospermia that can be irreversible in the case of radiotherapy or mostly temporary in the case of chemotherapy. Leydig cells are less radiosensitive, but can suffer toxicity from 24 Gy(16). In cases of male hypogonadism due to Leydig cell failure, referral should be made to Endocrinology so as to assess testosterone supplementation in order to avoid the symptoms that its lack generates(9).

In order to anticipate these problems, fertility preservation techniques may be performed in girls (ovarian cortex in prepubertal/postpubertal girls and oocyte vitrification in postpubertal women) and in boys (cryopreservation of semen in postpubertal men and in a few centers, cryopreservation of tissue testicular in prepubertal). The development of consensus guidelines is essential in our setting, since the preservation rate is still low, but efforts are being made to improve this situation(18). Likewise, there are surgical techniques for transposition of the testes or ovaries prior to the administration of scrotal, pelvic or inguinal radiotherapy that would avoid direct irradiation, which is the main risk factor for hypogonadism in these patients.

Regarding follow-up, height (percentile), weight, BMI and pubertal stage should be checked every 6 months until completion of sexual development and final height. Menstrual onset (prepubertal) or re-onset (pubertal), menopausal symptoms (hot flashes, dyspareunia) should be inquired, considering the risk of premature menopause. Referral to Endocrinology should be made if there is failure to thrive, delayed pubertal development, or risk of hypogonadism. It is important to advise on fertility, premature menopause, reproductive advice and individualized risks.

Finally, in the long term, some of the most relevant endocrine and metabolic sequelae are type 2 diabetes mellitus and metabolic syndrome (obesity, arterial hypertension, glucose intolerance – increased insulin resistance and dyslipidemia), which count as main risk factors along with abdominal radiotherapy, total body irradiation, prolonged corticosteroid treatment, inadequate eating habits with excessive caloric intake, sedentary lifestyle and a family history of type 2 diabetes mellitus. Consequently, there is an increase in cardiovascular risk with an increase in morbidity and mortality. Implementation of early dietary measures, increased physical activity, blood pressure and dyslipidemia control are recommended. A tight control of body mass index, blood pressure, lipid profile, blood glucose and Hb A1C will be carried out in clinic(19).

Gastrointestinal sequelae

Gastrointestinal complications can be multiple and heterogeneous depending on the therapy received, the location of the primary tumor and whether there is a history of surgery with organic lesion.

The most relevant gastrointestinal complications are shown in Table VI.

Figura

Tumor resection surgery and radiotherapy can have visceral obstructive sequelae. In the latter case, it can cause fibrosis and ischemia due to vascular injury. It can appear as a chronic manifestation in case of intestinal GVHD. Likewise, the specific sequelae derived from surgeries with wide resections should be considered (gastrectomy, pancreaticoduodenectomy, cholecystectomy, splenectomy, intestinal resection with discharge ostomy…). Guiding symptoms such as: dysphagia, heartburn, dyspepsia, reflux, nausea, vomiting, chronic diarrhea, constipation, or progressive weight loss should be monitored. In the cases with a short intestine, hypovitaminosis and lack of absorption of other nutrients ought to be monitored(7).

Chronic hepatotoxicity can appear after a long latency period, being secondary to some chemotherapeutic agents, radiotherapy, obesity, viral hepatitis or iron overload secondary to transfusions. The consumption of hepatotoxic agents, such as alcohol and other drugs of abuse, should be avoided.

Nephrourinary sequelae

Delayed kidney damage in the form of chronic kidney failure is one of the most serious complications in survivors.

The main risk factors for kidney damage at the glomerular and proximal tubular level are abdominal radiation and treatment with high doses of ifosfamide (> 16 g/m2), cisplatin (> 450 mg/m2) and, to a lesser extent, carboplatin. Other factors are the use of nephrotoxic drugs (aminoglycosides, amphotericin B, foscarnet, tacrolimus, and cyclosporine A). It should also be considered that some patients may have a single kidney, due to nephrectomy of their primary tumor (nephroblastoma) or due to metastatic renal involvement(20). Consequently, chronic renal failure, proximal tubulopathy, or Fanconi syndrome may develop(7). Some survivors of Wilms’ tumors are at particular risk, as in addition to having a single kidney, they may have an underlying syndrome (Denys-Drash) and will have received chemotherapy and radiotherapy. High blood pressure is common (30%) during follow-up. In hematopoietic stem cell transplant recipients, nephrotoxicity derived from immunosuppressants, as well as nephritis secondary to BK virus, may occur.

Bladder damage is usually associated with the use of cyclophosphamide or ifosfamide, as well as viral infections (BK virus, adenovirus, cytomegalovirus)(9). There may be lesions of the lower urinary tract, secondary to surgery of solid tumors and radiotherapy/brachytherapy. Secondarily, bladder incontinence (neurogenic bladder), bladder fibrosis, bladder cancer, sexual dysfunction, chronic pelvic pain, bladder floor disorders, fistulas and female vaginal dryness, can also occur(16).

Annual BP control is recommended, with urea, creatinine and electrolytes to assess renal function, venous blood gas, and urine electrolytes. If the post-treatment study is normal, it should be repeated after 5 years. In case of HT, proteinuria or signs of tubulopathy, refer to Nephrology(7).

Musculoskeletal sequelae

Healthy lifestyle habits are important, including regular physical exercise and sufficient amounts of vitamin D and calcium in the diet, as well as the early detection of musculoskeletal problems.

Cranio-spinal radiation, prolonged use of corticosteroids or methotrexate, nutritional alterations and a sedentary lifestyle lead to high risk of this type of sequelae. Soft tissue fibrosis and hypotrophy can occur, as well as a decrease in bone mass with osteopenia/osteoporosis derived from the treatments, prolonged bedridden and radiotherapy on musculoskeletal groups. Other long-term complications are: osteonecrosis (avascular necrosis), exostoses, pathological fractures, bone undergrowth or asymmetric growth, dysmetria and, on some occasions, the sequelae derived from amputations and prosthetic malfunctions(7). With regards to bone mineral density, there is a remarkable margin of recovery throughout adolescence provided a healthy lifestyle, regular physical activity (especially, weight-bearing exercises), controlled sun exposure and optimization of the calcium intake in the diet. Sometimes vitamin D supplements are required. Avoidance of alcohol, tobacco, excessive caffeine, and obesity are recommended(9).

Gynecological sequelae

Breast self-examination and screening based on the individual risk are essential to anticipate breast tumors in adulthood.

In the case of adolescent women, it is important to make the patient aware of the importance of breast self-examination (monthly), as well as a regular gynecological examination. The risk of secondary breast cancer is higher if the patient received chest wall or mediastinal radiation therapy, family predisposition to breast cancer (BRCA1/2 mutations) or history of Li-Fraumeni syndrome (TP53). In these high-risk cases, annual gynecological follow-up is recommended from puberty to age 25, then every 6 months. Annual mammograms should be performed from the age of 25 (other groups recommend it from the age of 30)(10) or from 8 years after treatment (whichever occurs last). Breast MRI is recommended concomitantly with the same timelines as mammograms. In case of mammary hypoplasia, an annual follow-up should take place, referring the patient to Plastic Surgery if breast reconstruction is required, once the pubertal development is completed(7).

Vaccination in the oncohematological patient

Revaccination of the oncohematological patient is of vital importance in the months and years following treatment, in a scheduled manner according to national guidelines and recommendations.

In general, routine vaccinations are suspended during treatment, which added to the fact that some patients have an incomplete vaccination schedule due to their young age at diagnosis, the loss of vaccine immunity acquired by the treatments received and the immunodeficiency maintained by some therapies (rituximab, hematopoietic stem cell transplantation, immunosuppressants), makes them a particularly high infectious risk population at the end of treatment. Given the length of this review, the reader is referred to the periodic updates of the Vaccine Advisory Committee of the AEP (https://vacunasaep.org/documentos/manual/cap-14 y -16, updated as of August 2021).

Risk of premature mortality

There is an increased risk of premature death in survivors, thus, health promotion (Fig. 1) and early diagnosis of complications, as well as second tumors, are of vital importance.

The premature mortality rate of survivors is increased compared to the general population, being higher in the first years of follow-up (cumulative mortality: 6.5% at 10 years, 11.9% at 20 years, and 18.1% at 30 years of diagnosis). The risk is higher in women, brain tumors and Ewing’s sarcoma(7). The main cause is the recurrence of the original disease (67%), which decreases in importance with the passage of time. As the years go by, the toxicity associated with the treatment takes on a more important role in the reduction of life expectancy (late mortality). A 7 times higher risk of dying from cardiovascular events has been described (especially in women, kidney tumors and Hodgkin lymphoma), almost 9 times the risk of dying from lung or respiratory problems (especially patients with AML or neuroblastoma) and more than 2 times from other medical reasons. After 20-30 years of treatment, mortality secondary to second neoplasms is the main cause of death (15 times higher than in the healthy population). Therefore, follow-up strictly restricted to 5 years post-treatment is insufficient in patients who have overcome a tumor in the pediatric age(7-9).

Risk of second neoplasms

Up to 5-20 times higher risk of suffering second tumors histologically different from the initial ones compared to the general population has been described. In general, they are observed at least 2 years after the primary tumor, and within the first 10 years of follow-up, although they can have a latency of up to 30 years(21). The cumulative incidence after 20 years is 3-10%, and after 30 years 5-30%, depending on the series. The primary tumors with the highest incidence of second neoplasms are Hodgkin lymphoma and soft tissue sarcomas. The most common secondary tumors are breast, thyroid, AML, and sarcomas(7). The main risk factors are(6,7,16):

Chemotherapy. Alkylating agents are associated with myelodysplastic syndromes, which may or may not precede secondary acute myeloblastic leukemias. Their latency is usually 5-7 years and they are accompanied by cytogenetic abnormalities (monosomies and partial deletions of chromosomes 5 and 7). Epipodophyllotoxins (etoposide) are also associated with AML, but this usually develops earlier (2-3 years after completion of treatment). There is a greater risk with doses ≥ 2,000 mg/m2 and associated abnormalities of the MLL gene (which induces 11q23 translocations). Anthracyclines can also cause this type of leukemia, 2-3 years after treatment. The prognosis is unfavorable. Annual full blood count is recommended during long-term follow-up, at least up to 10 years after exposure to chemotherapy. The latency for the development of solid tumors is somewhat longer, on average 14 years. Etoposide increases the risk of secondary Hodgkin lymphoma, neuroblastoma, nephroblastoma, and rhabdomyosarcoma, among others.

Radiotherapy. It is a relevant risk factor, especially when high doses are received at an early age. It is frequently used in CNS tumors, solid tumors and, historically, in Hodgkin lymphoma and the conditioning with total body irradiation of some transplants. The risk increases as time passes, with a maximum 10-15 years after treatment (although it remains until 30 years later). The types mainly observed are: breast tumors, meningiomas (cranial), bone and soft tissue tumors, thyroid cancer, non-melanoma skin cancer and bladder cancer.

Hematopoietic stem cell transplantation. It can increase the risk of second neoplasms by up to 8 times compared to the general population (up to 60 times in recipients below the age of 10 years). Its origin is multifactorial (chemotherapy, conditioning radiotherapy and GVHD). Immunosuppressants are associated with the development of post-transplant lymphoproliferative syndrome.

Cancer predisposition syndromes (Li-Fraumeni, DICER1, NF1-2, telomeropathies…). In recent years, hand in hand with advances in genetics, more are becoming diagnosed. This has allowed the identification of alterations in the germ line or mosaicisms that predispose to one or more tumors, increasing their importance in pediatric Hematology-Oncology. Given the length of the article, we refer the reader to review the bibliography in case of interest(22).

Sufficient information and adequate education about the personalized risks of each patient, promoting self-observation and reducing, as far as possible, risk factors (tobacco, alcohol, sun exposure) is important. Symptoms or signs that may be related to the primary tumor and that suggest a relapse should also be monitored. In this sense, to make an early diagnosis it is essential to have a high index of suspicion, to recognize the risk groups and the “red flags” (warning signs) in each patient(23,24).

The role of the Primary Care pediatrician

As previously discussed in this review, there is not a sole model for pediatric cancer survivor follow-up, and it is essential to adapt it to the individual characteristics and risks of each case, as well as to the real conditions of our healthcare system. In this sense, we have a Pediatric Primary Care network excellently prepared for the promotion of health (Fig. 1) and accompaniment of the patient during their growth. At the hospital level, progress is being made in the development of specialized units to follow-up this type of patients and in their transition to adult care, so we have a mixed care and follow-up model in the routine practice.

Primary Care professionals are a fundamental pillar of multidisciplinary teams, acting as a first line for the diagnosis of sequelae, relapses and second tumors, as well as promoting healthy lifestyle habits. An adequate transmission of the personal history, personalized risks and follow-up plan by hospital health care providers is necessary, as well as a fluid communication between both areas, which will result in a health benefit to our patients. Similarly, the patient and his family should be integrated in the responsibility of long-term care, avoidance of risk behaviors and perception of alarm signs or symptoms, which will prevent loss of adherence to follow-up or low perception of its importance in the long term.

Bibliography

The asterisks show the interest of the article in the opinion of the authors.

1.** Song A, Fish JD. Caring for survivors of childhood cancer: it takes a village. Current Opinion in Pediatrics. 2018; 30: 864-73.

2. Klassen AF, Anthony SJ, Khan A, Sung L, Klaassen R. Identifying determinants of quality of life of children with cancer and childhood cancer survivors: a systematic review. Support Care Cancer. 2011; 19: 1275-87.

3. Shad A, Myers SN, Hennessy K. Late effects in cancer survivors: “the shared care model”. Curr Oncol Rep. 2012; 14: 182-90.

4.** Bhakta N, Liu Q, Ness KK, Baassiri M, Eissa H, Yeo F, et al. The cumulative burden of surviving childhood cancer: an initial report from the St Jude Lifetime Cohort Study (SJLIFE). The Lancet. 2017;390(10112):2569-82.

5. Essig S, Li Q, Chen Y, Hitzler J, Leisenring W, Greenberg M, et al. Risk of late effects of treatment in children newly diagnosed with standard-risk acute lymphoblastic leukaemia: a report from the Childhood Cancer Survivor Study cohort. The Lancet Oncology. 2014; 15: 841-51.

6.** Armstrong GT, Chen Y, Yasui Y, Leisenring W, Gibson TM, Mertens AC, et al. Reduction in Late Mortality among 5-Year Survivors of Childhood Cancer. N Engl J Med. 2016; 374: 833-42.

7.*** Children’s Oncology Group. Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent, and Young Adult Cancers (Internet). 2018. Disponible en: http://www.survivorshipguidelines.org/pdf/2018/COG_LTFU_Guidelines_v5.pdf.

8.*** Skinner R, Wallace W, Levitt GA. Therapy based long term follow up: practice statement: United Kingdom Children’s Cancer Study Group (Late Effects Group) (Internet). Disponible en: https://www.cclg.org.uk/write/MediaUploads/Member%20area/Treatment%20guidelines/LTFU-full.pdf.

9.*** Scottish Intercollegiate Guidelines Network (SIGN), Scotland, Healhcare Improvement Scotland. Long Term Follow Up of Survivors of Childhood Cancer, A National Clinical Guideline (Internet). Disponible en: https://www.sign.ac.uk/media/1070/sign132.pdf.

10.*** Late Effects Taskforce of the Dutch Childhood Oncology Group (DCOG LATER). Guidelines for follow-up after childhood cancer more than 5 years after diagnosis (Internet). Disponible en: https://www.skion.nl/workspace/uploads/vertaling-richtlijn-LATER-versie-final-okt-2014_2.pdf.

11.*** Grupo de trabajo sobre efectos secundarios a largo plazo y segundos tumores de la Sociedad Española de Hematología y Oncología Pediátricas. “Efectos tardíos en supervivientes al cáncer en la infancia”. Cevagraf. 2012.

12.*** Armenian SH, Hudson MM, Mulder RL, Chen MH, Constine LS, Dwyer M, et al. Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. The Lancet Oncology. 2015; 16: e123-36.

13. Chow EJ, Leger KJ, Bhatt NS, Mulrooney DA, Ross CJ, Aggarwal S, et al. Paediatric cardio-oncology: epidemiology, screening, prevention, and treatment. Cardiovascular Research. 2019; 115: 922-34.

14. Bansal N, Amdani S, Lipshultz ER, Lipshultz SE. Chemotherapy-induced cardiotoxicity in children. Expert Opinion on Drug Metabolism & Toxicology. 2017; 13: 817-32.

15. Jin Y, Xu Z, Yan H, He Q, Yang X, Luo P. A Comprehensive Review of Clinical Cardiotoxicity Incidence of FDA-Approved Small-Molecule Kinase Inhibitors. Front Pharmacol. 2020; 11: 891.

16.*** Mendoza Sánchez MC. Seguimiento en Atención Primaria del niño oncológico. Cómo detectar las secuelas tardías. Pediatr Integral. 2016; XX(7): 475-84.

17. Christen S, Roser K, Mulder RL, Ilic A, Lie HC, Loonen JJ, et al. Recommendations for the surveillance of cancer-related fatigue in childhood, adolescent, and young adult cancer survivors: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. J Cancer Surviv. 2020; 14: 923-38.

18.** Garrido-Colino C, Lassaletta A, Vázquez MÁ, Echevarría A, Gutiérrez I, Andión M, et al. Situación de la preservación de fertilidad en pacientes con cáncer en nuestro medio: grado de conocimiento, información e implicación de los profesionales. Anales de Pediatría. 2017; 87: 3-8.

19. Friedman DN, Tonorezos ES, Cohen P. Diabetes and Metabolic Syndrome in Survivors of Childhood Cancer. Horm Res Paediatr. 2019; 91: 118-27.

20. Dekkers IA, Blijdorp K, Cransberg K, Pluijm SM, Pieters R, Neggers SJ, et al. Long-Term Nephrotoxicity in Adult Survivors of Childhood Cancer. CJASN. 2013; 8: 922-9.

21.** Choi DK, Helenowski I, Hijiya N. Secondary malignancies in pediatric cancer survivors: Perspectives and review of the literature: Secondary malignancies in pediatric cancer survivors. Int J Cancer. 2014; 135: 1764-73.

22.** Ripperger T, Bielack SS, Borkhardt A, Brecht IB, Burkhardt B, Calaminus G, et al. Childhood cancer predisposition syndromes-A concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology. Am J Med Genet. 2017; 173: 1017-37.

23.** Huerta Aragonés J. Oncología para el pediatra de Atención Primaria (I): signos y síntomas sugerentes de patología neoplásica. Form Act Pediatr Aten Prim. 2014; 1: 4-15.

24.** Huerta Aragonés J. Oncología para el pediatra de Atención Primaria (II): formas de presentación de las diferentes neoplasias infantiles. Form Act Pediatr Aten Prim. 2014; 7: 67-74.

Recommended bibliography

- Children’s Oncology Group. Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent, and Young Adult Cancers (Internet). 2018. Available at:http://www.survivorshipguidelines.org/pdf/2018/COG_LTFU_Guidelines_v5.pdf.

These guidelines are a mainstay in the follow-up, diagnosis and management of late effects in survivors of childhood cancer, with a dilated experience (almost 20 years). They are currently in their 5th version, being updated approximately every 5 years.

- Scottish Intercollegiate Guidelines Network (SIGN), Scotland, Healthcare Improvement Scotland. Long Term Follow Up of Survivors of Childhood Cancer, A National Clinical Guideline. [Internet]. Available in: https://www.sign.ac.uk/media/1070/sign132.pdf.

High quality consensus guidelines, from the year 2013, organized by body organs and systems.

- Skinner R, Wallace W, Levitt GA. Therapy based long term follow up: practice statement: United Kingdom Children’s Cancer Study Group (Late Effects Group) (Internet). Available at: https://www.cclg.org.uk/write/MediaUploads/Member%20area/Treatment%20guidelines/LTFU-full.pdf.

These consensus guidelines focus on both the long-term side effects in the various body organs and those specific to the chemotherapy or radiotherapy treatment received, providing an interesting point of view.

- Grupo de trabajo sobre efectos secundarios a largo plazo y segundos tumores de la Sociedad Española de Hematología y Oncología Pediátricas. “Efectos tardíos en supervivientes al cáncer en la infancia”. Cevagraf. 2012. (Working group on long-term side effects and second tumors of the Spanish Society of Pediatric Hematology and Oncology. “Late effects in survivors of childhood cancer”. Cevagraf. 2012.)

Interesting publication by the SEHOP Working Group on Side Effects, in the form of a textbook. Excellent description of each topic that surpasses in exposure and understanding that obtained, sometimes, in more schematic guides.

- Armenian SH, Hudson MM, Mulder RL, Chen MH, Constine LS, Dwyer M, et al. Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. The Lancet Oncology. 2015; 16: e123-36.

Very good article that summarizes the recommendations for cardiac monitoring and surveillance in patients at risk, with special emphasis on risk groups and long-term problems. It is the result of the cooperative work between the Children’s Oncology Group and several European groups, which make up the International Late Effects of Childhood Cancer Guideline Harmonization Group (IGHG).

- Ripperger T, Bielack SS, Borkhardt A, Brecht IB, Burkhardt B, Calaminus G, et al. Childhood cancer predisposition syndromes-A concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology. Am J Med Genet. 2017; 173: 1017-37.

Review of the most relevant cancer predisposition syndromes, with special interest in the risks of pediatric tumor development and specific follow-up recommendations.

 

Clinical case

 

3-year-old boy attended the emergency room due to progressive onset of severe respiratory difficulty. Physical examination revealed marked hypoventilation of the left hemithorax, with increased work of breathing at all levels and decreased oxygen saturation (92%). In the initial chest X-ray, a large hemithorax mass with massive ipsilateral pleural effusion was observed. A chest CT was performed confirming a large mass in the left hemithorax, with doubtful pleural dependence, as well as a severe pleural effusion. After stabilization of the patient and drainage of the pleural effusion, the diagnostic studies were completed in the following days. Thoracic ultrasound-guided percutaneous biopsy of the mass concluded the histological diagnosis of a primitive neuroectodermal tumor of the chest wall. No pulmonary metastases were identified.

The patient received treatment according to the protocol in force at that time (Ewing SEOP-2001, treatment group 3). She received induction chemotherapy with 6 VIDE cycles (vincristine, ifosfamide, doxorubicin and etoposide), after which a resection of the tumor followed with reconstruction of the thoracic wall. The response to chemotherapy showed a poor response (80% necrosis), with free surgical margins. A VAC consolidation cycle (vincristine, D-actinomycin, and cyclophosphamide) was administered and, subsequently, an autologous hematopoietic stem cell transplant conditioned with melphalan and etoposide performed. The cumulative dose of cytostatics is as follows: vincristine: 10.5 mg/m2; ifosfamide: 54 g/m2; doxorubicin: 360 mg/m2; etoposide: 4.5 g/m2; cyclophosphamide: 1.5 g/m2; actinomycin-D: 1.5 mg/m2; and melphalan: 140 mg/m2. Finally, 8 weeks after transplantation, radiotherapy with tomotherapy modality was performed with 48 Gy on the primary tumor and 15 Gy on the ipsilateral lung.

She attends her follow-up consultations in a timely manner according to the prevailing recommendations in her treatment protocol, with no relapse being observed after 10 years of follow-up. However, at 9 years of age, a lesser growth of the left rib cage is observed, conditioning a significant scoliosis, which requires intervention. Likewise, at 8 years of follow-up, data of mild dilated cardiomyopathy were observed, verified by echocardiography and by magnetic resonance imaging, with mild systolic ventricular dysfunction, for which treatment with enalapril was started.

 

 

How to suspect cancer in Primary Care

V. Losa Frías*, M. Herrera López**, I. Cabello García***, P.I. Navas Alonso****
Topics on
Continuous Training


V. Losa Frías, M. Herrera López, I. Cabello García, P.I. Navas Alonso

*Fuensalida Primary Care Center. Toledo. **Pediatric Service. Virgen de la Salud Hospital. Toledo. ***La Puebla de Montalbán Primary Care Center. Toledo. ****Pedro Fuente Primary Care Center. Bargas. Toledo

Abstract

Cancer in childhood has a low incidence and its clinical presentation is often nonspecific simulating common pathologies of benign course, therefore making its diagnosis highly challenging. The main symptoms and signs of suspected childhood cancer in Primary Care are: pallor, masses (in the head, neck and other locations), lymphadenopathy, abnormal movement, bruising and signs of bleeding, fatigue, headache, visual abnormalities, pain and musculoskeletal symptoms. The Primary Care pediatrician must recognize when an apparently benign symptomatology may be the beginning of a severe pathology, identifying those warning signs that require immediate assessment. A detailed medical history, a complete physical examination and a clinical follow-up are essential. The objective is to reduce the time from the onset of symptoms to the final diagnosis, so as to allow an early diagnosis of childhood cancer.

 

Key words: Lactante; Niño; Neoplasias; Atención Primaria; Diagnóstico precoz de cáncer.

Palabras clave: Infant; Child; Neoplasms; Primary health care; Early detection of cancer.

 

 

Pediatr Integral 2021; XXV (6): 283 – 295

 


 

How to suspect cancer in 
Primary Care

Introduction

Childhood cancer has a low incidence and often manifests nonspecifically, simulating other frequent and benign processes. These particularities make it difficult to diagnose cancer in Primary Care. However, an early suspicion, together with a rapid referral of the patient to a specialized center, may have implications at the prognostic and therapeutic level, as well as in the emotional impact secondary to the diagnosis on the patient and his family.

Epidemiology

Childhood cancer is after accidents, the second leading cause of death beyond the first year of life.

The standardized annual incidence of childhood cancer in Spain is 159 new cases per year per million children aged 0 to 14 years, which represents 1,100 new cases of childhood cancer per year, an incidence similar to that of the rest of Europe(1). It is estimated that a Primary Care pediatrician following 1,500 patients in his clinic, will see a new case of cancer every 5 years. Overall 5-year survival after the diagnosis is around 79%(1). Despite recent advances, childhood cancer is the second leading cause of death from the first year of life through adolescence. In the year 2018, 192 children under 14 years of age died in Spain from cancer, which means 4 children died per week due to this cause(2). The most frequent neoplasms from birth to 14 years of age are: leukemias (28%), central nervous system (CNS) tumors (23%) and lymphomas (12%), with a distribution pattern by sex and age similar to that of the rest of Europe, whilst between 15 and 19 years of age they are: bone tumors (24%), lymphomas (21%) and CNS tumors (15%). The most frequent diagnoses according to the age group are shown in Table I(1).

Patients at risk

Medical history is the most effective tool in identifying cancer predisposition syndromes.

There is a hereditary basis in 8-10% of all neoplasms. Within this percentage, cancer predisposition syndromes (CPS) are included, conforming a heterogeneous group of genetic conditions and immunodeficiencies, which predispose to a greater risk of cancer (Table II).

Most of these syndromes are rare and show variable expressiveness within the family members. It is important to identify these patients, as they can benefit from prevention and early detection measures, as well as the possibility of genetic counseling. During history-taking, CPS may be suspected should there be(3):

• Several cases of cancer within the family, usually of the same type.

• Multi-generational disease cases, presenting at an earlier age than in the general population.

• Presence of tumors in association with developmental defects: generalized or asymmetric body overgrowth, dysmorphic features, congenital malformations or mental retardation.

• Presence of bilateral or multifocal tumors.

Individuals with more than one primary tumor.

• Presence of rare, benign tumors or cysts associated with CPS.

Warning signs and symptoms

Apparently benign symptomatology, but with atypical presentation or torpid course, may be the beginning of a neoplastic process.

Childhood cancer can manifest in its initial stages, with symptoms similar to frequent and benign processes(4). The purpose is to recognize when this apparently benign symptomatology may be the beginning of a serious pathology, as well as to identify those findings (red flags) that, in combination with the rest of the data from history and physical examination, should alert us of the possibility of cancer (Table III).

For this, it is necessary to listen and pay special attention to parents(5), who in general are the best observers of their children’s symptoms, and also to adolescents(6); taking a complete medical history including personal and family history, and a thorough physical examination.

Qualitative studies highlight the importance of behavioral and affective changes detected by parents, that motivate the first visits to Primary Care prior to the diagnosis of cancer, sometimes in the absence of other signs of alarm(5,7). Such is the case, that the National Institute for Clinical Excellence (NICE) recommends considering the persistent parental concern regarding their children’s symptoms as a reason for study or referral(8). In Primary Care, the following signs and symptoms have been reported to increase the possibility of cancer: paleness, head and neck masses, abdominal masses, lymphadenopathy, motor abnormalities, bruises and other signs of bleeding, asthenia, headache, visual anomalies, pain and musculoskeletal symptoms. However, except for abdominal masses, the positive predictive value of these symptoms is low, given the low frequency of childhood cancer. Even so, given the severity of the diagnosis, the presence of the aforementioned symptoms, fundamentally when it occurs without a clear cause and leads to an increase in the number of consultations (3 or more in a period of 3 months), should alert of the possibility of a neoplastic process(9). In this sense, the Pan American Health Organization has published an evaluation strategy, a cancer probability classification and attitude based on the findings of the history and examination shown in Algorithm 1(10).

Headache and other neurological signs and symptoms

Primary CNS tumors are the second most common neoplasia in childhood after leukemias(1), as well as the second leading cause of death from childhood cancer(2). Their symptoms are due to the invasion and compression of the adjacent nervous tissue, as well as the increase in intracranial pressure due to mass effect or obstructive hydrocephalus (Fig. 1), manifesting a very heterogeneous clinical presentation.

Figure 1. Sagittal MRI showing a mass in the posterior fossa compatible with medulloblastoma. The 13-year-old patient presented with headache and papilledema.

Wilne et al.(11) analyzed 74 articles (n = 4,171), identifying up to a total of 56 signs and symptoms at the diagnosis of a CNS tumor, which depended on age, location and history of neurofibromatosis (NF). For intracranial tumors, excluding NF, the most frequent symptoms were: headache, nausea and vomiting, abnormality of gait and coordination, and papilledema. In intracranial tumors associated with NF these were: decreased visual acuity, exophthalmos, optic atrophy and strabismus. In intracranial tumors in children under 4 years they were: macrocephaly, nausea and vomiting, irritability, lethargy and ataxia. And in spinal cord tumors they were: back pain, gait and coordination abnormality, spinal deformity, focal weakness and sphincter alterations. Given this clinical variability, they subsequently studied the evolution of symptoms in a retrospective cohort (n = 139), describing a progressive increase in the number of symptoms. Thus, half of the patients went from one symptom at the beginning of the process to six at the diagnosis of the tumor(12). In Primary Care, we must be alert to patients with non-resolving symptoms or in which new ones are associated, especially: visual, motor, endocrine or behavioral, as well as signs of intracranial hypertension(12-14). In this line, Ansell et al.(15) described the reasons for consultation in Primary Care, from birth to the diagnosis of a CNS tumor in a series of patients, comparing it with a control group. They observed how the cases consulted three times more often for a sign or symptom suggestive of a CNS tumor, reaching seven times more when they associated two or more symptoms.

The Children’s Brain Tumour Research Centre group has developed an evidence-based clinical guide(16) (Table IV), as well as the awareness strategy “HeadSmart: be brain tumours aware” (https://www.headsmart.org.uk/), which has shown positive results in reducing the time from the onset of symptoms to diagnosis (median 14 to 6.7 weeks), as well as the time from the first consultation to an imaging test (3.3 to 1.4 weeks)(17).

Fever and constitutional symptoms

Fever is one of the most frequent reasons for consultation in Pediatrics, being, in most cases, of infectious etiology. Only 6% of the cases of fever of unknown origin correspond to neoplasms(18). This fever can be of tumor origin (Ewing’s sarcoma, neuroblastoma, Hodking’s lymphoma…) or due to infections secondary to the alteration of the immune system due to cancer, as can occur in leukemias.

Leukemia is the most common pediatric tumor(1). Clarke et al.(19) analyzed the symptoms at the diagnosis of leukemia in childhood and adolescence in 33 studies (n = 3,084), and identified a total of 95 signs and symptoms, of which five were present in more than half of the patients: hepatomegaly, splenomegaly, paleness, fever and bruising. In addition, between a third and a half of the patients presented: recurrent infections, asthenia, pain in the extremities, hepatosplenomegaly, hematomas / petechiae, lymphadenopathies, bleeding tendency and skin rash. These findings highlight the importance of performing a complete physical examination in children with common symptoms, such as fever, but with a torpid or persistent course, paying special attention to abdominal palpation, lymphadenopathy search and careful skin examination. Despite the fact that leukemia is the most frequent pediatric cancer, there is currently no evidence of the predictive value of clinical data at the individual level, or of their combination. NICE guidelines recommend that pediatric patients with fever of unclear cause or in combination with unjustified clinical data such as: paleness, asthenia, lymphadenopathies, splenomegaly, osteoarticular pain, bruising, night sweats or weight loss, should be evaluated with a full blood count and peripheral blood smear within 48 hours; in the case of associating petechiae or unexplained hepatosplenomegaly, immediate referral is recommended(8).

Lymphadenopathies

Lymph nodes are dynamic structures that change in size during children´s growth, usually in response to infections. During childhood, palpation of small lymph nodes in cervical, axillary, or inguinal regions is normal. An increase in size above 1 cm in cervical and axillary nodes, 1.5 cm in inguinal and 0.5 cm in epitrochlear nodes, as well as stone-like consistency, irregular surface, existence of skin ulceration or fixation is considered pathological(20) (Fig. 2).

Figure 2. Mediastinal widening in a 12-year-old patient diagnosed with Hodgkin’s lymphoma. Clinically, she had multiple adherent, non-tender and larger than 2 cm laterocervical and supraclavicular lymphadenopathies.

Lymphadenopathies are generalized, when they extend beyond more than 2 non-contiguous ganglion chains, and localized, when they appear in a single region. According to the course in terms of length of time, we distinguish between acute (less than three weeks) and subacute / chronic (more than three weeks / months)(21). In the history, we should inquire for: the age of the patient, the form of onset, the time of progression and the speed of growth, as well as the presence of recent or recurrent infections, contact with sick people, associated symptoms, previous antibiotic treatments, similar episodes, vaccination status, medications, contact with animals or recent travels. A complete physical examination should be performed, looking for signs of systemic disease and paying special attention to the presence of skin lesions, paleness, signs of bleeding, oropharyngeal or conjunctival lesions, hepatosplenomegaly, and abdominal masses. Adenopathies will be evaluated based on their size, location, tenderness, consistency, mobility, local inflammatory signs, and presence of skin fistulas. We must systematically palpate all accessible ganglion chains: occipital, retroauricular, preauricular, parotid, tonsillar, submandibular, submental, anterior and posterior neck, supraclavicular, infraclavicular, axillary, epitrochlear, inguinal and popliteal. Warning signs in the evaluation of adenopathies are: size greater than 3 cm, rapid growth in the absence of inflammatory signs, hard consistency, fixation to deep planes, supraclavicular, axillary, generalized or confluent location, as well as constitutional symptoms, the presence of abdominal masses, hard hepatosplenomegaly, signs of respiratory distress, paleness, jaundice or bleeding(8,20-22).

Mediastinal mass

Mediastinal masses in childhood are rare and, in most cases, are malignant. The most frequent location is the anterior mediastinum and the most frequent etiology is lymphoma(23,24). A thorough clinical evaluation and a high index of suspicion are important for an early diagnosis. Upon clinical suspicion, immediate referral to a hospital center to complete the study is recommended(8,23).

From an anatomical point of view, the mediastinum is divided into three compartments: anterior, middle, and posterior. The location of the mass will guide the diagnosis (Table V).

The most frequent neoplasms according to their location are: acute lymphoblastic leukemia and T lymphoma (Fig. 3) in the anterior mediastinum; Hodgkin lymphoma in the middle mediastinum; and neurogenic tumors (neuroblastoma and ganglioneuroma) in the posterior mediastinum. Posterior mediastinal masses (neuroblastoma) are more common in infants and young children, while anterior mediastinal masses (leukemias, lymphomas) are more common in older children and adolescents(24).

Figure 3. Anterior mediastinal mass in a 12-year-old boy with T-leukemia. He reported a 3 day course of asthenia and bilateral eyelid edema, as well as 48 hours of bruising, ecchymosis and petechiae in face, neck and upper arm.

A high percentage of patients are asymptomatic at diagnosis. In symptomatic cases, the symptoms are secondary to compression of adjacent structures, so the symptoms depend on the location of the mass, its size, and the rate of growth. Compression of the airway is the most frequent symptom, giving rise to nonspecific symptoms, such as: stridor, non-productive cough, wheezing, recurrent respiratory infections, chest pain and respiratory distress that often simulate frequent respiratory diseases, such as asthma or laryngitis. Esophageal compression leads to dysphagia. Compression of the spinal cord (common in neuroblastoma) results in band or radicular back pain that increases with Valsalva, gait weakness, loss of strength and sensory and sphincter alterations. Compression of the superior vena cava (characteristic of leukemias and T lymphomas) manifests with: facial plethora, headache, blurred vision, cough, chest pain, orthopnea that increases with Valsalva, hypotension and heart failure. Compression of the phrenic leads to hemidiaphragmatic elevation. Finally, injury to the sympathetic pathway (especially neuroblastomas) can cause Horner syndrome (ptosis, miosis, and enophthalmos). And, in turn, we can find systemic symptoms secondary to the tumor process itself. Compression of the phrenic leads to hemidiaphragmatic elevation. Finally, injury of the sympathetic pathway (especially neuroblastomas) can cause Horner syndrome (ptosis, miosis, and enophthalmos). In addition, we can find systemic symptoms secondary to the tumor process itself.

We must bear in mind that treatment prior to diagnosis with systemic corticosteroids in patients with hematological malignancies may have diagnostic and prognostic implications, and precipitate serious complications, such as tumor lysis syndrome. For this reason, in patients with atypical respiratory symptoms or laryngitis in older children, performing a chest X-ray prior to starting corticosteroid treatment is recommended(23,25).

Abdominal mass

The finding of an abdominal mass is one of the most frequent forms of presentation of neoplasms in childhood. Although they may be of benign etiology, all patients with an abdominal mass must be evaluated with the suspicion of malignancy and referred to a specialized center within 48 hours(8).

An abdominal mass is often asymptomatic, and is usually detected accidentally by the parents or in a routine examination. In the history, we will take into account the patient’s age (Table VI), the associated symptoms, their intensity and duration, taking into account which rapidly evolving symptoms are suggestive of malignancy(26,27).

The most frequent presentation symptoms are: pain, organ dysfunction due to mass effect (intestinal or urinary obstruction), hematuria (nephroblastoma) and systemic symptoms (night sweats, fever, asthenia, weight loss or bone pain…). The most frequent etiology involves: genitourinary congenital malformations in children under one year of age; neuroblastoma (Fig. 4) and nephroblastoma (Fig. 5) between one and five years; and non-Hodgkin lymphoma (NHL) in older children and adolescents.

Figure 4. Neuroblastoma in a 7-year-old patient who reported weeks of rib pain associated with asthenia and anorexia in the most recent days.

Figura 5. Right Wilms tumor. The 2-year-old patient consulted because the parents palpated the abdominal mass while dressing her.

In this age group, Burkitt-type NHL presents as: a rapidly growing abdominal mass that associates abdominal distention and pain, obstructive symptoms, intussusception, and metabolic alterations secondary to tumor lysis(26). In girls and adolescents, we will take into account ovarian tumors and pregnancy. In the personal history, factors such as prematurity and low birth weight (hepatoblastoma), should also be assessed.

Physical examination should be performed with the patient relaxed and calm. It must be meticulous, checking vital signs, including blood pressure(27). On inspection, we will look for irregularities on the abdominal surface. On palpation, we must take into account that, in healthy patients, especially infants, some structures may be palpable, such as: liver, spleen, kidneys, abdominal aorta, sigmoid colon, feces and / or spine(28). It is important to establish the location, size, shape, and contour of the mass, its adherence to deep planes, as well as the presence of tenderness. Depending on the location it should be considered that: palpable masses in the right upper quadrant are usually of hepatic, renal or adrenal origin; those in the upper left quadrant often depend on the spleen and may be secondary to metastatic infiltration; and those located in the hypogastrium, are usually secondary to ovarian tumors or intestinal lymphomas. In the patient with suspected neuroblastoma, a complete neurological examination is necessary due to the possibility of invasion of the medullary canal. Other examination signs that can guide the diagnosis are: aniridia, hemihypertrophy and genitourinary malformations (nephroblastoma); subcutaneous nodules, periorbital ecchymoses, proptosis, intractable watery diarrhea, Horner syndrome or opsoclonus-myoclonus syndrome (neuroblastoma); precocious puberty, feminization or virilization (hepatic, gonadal, adrenal masses or germ tumors); and Cushing phenotype (neoplasms of the adrenal cortex).

Soft tissue and skin masses

Soft tissue sarcomas (STS) are a heterogeneous group of tumors that originate from primitive mesenchymal cells. They are subdivided into rhabdomyosarcoma (RBM) and non-rhabdomyosarcoma sarcomas. RBM is the most frequent, it has its origin in the primitive mesenchymal cells involved in the development of skeletal muscle, and it presents its maximum incidence around the age of five years and in adolescence. It is subdivided into: embryonal, alveolar and pleomorphic. On the other hand, non-rhabdomyosarcoma sarcomas are more common in older children and adolescents.

STS can appear in any anatomical location, the most frequent being: the genitourinary region, head and neck, and extremities. The clinical presentation depends on the location, size and adjacent structures. An unexplained tumor in any location with any of the following characteristics is suspicious of STS: diameter greater than 2 cm, fixation to deep planes, increased consistency, progressive growth, and presence of regional lymphadenopathies(4). In the head, orbital locations usually develop proptosis and a differential diagnosis must be made with benign pathologies, such as orbital cellulitis. Parameningeal locations can lead to nasal, sinus, or ear obstruction, persistent mucopurulent discharge, or cranial nerve involvement. In the genitourinary region, it can present as: hematuria, voiding syndrome, constipation, pelvic mass or increased testicular size. The botryoid variety is a subtype of embryonic RMS characterized by multiple polypoid projections that form clusters of gelatinous and friable consistency, that develop under the mucosal surface of body orifices, such as vagina and nose.

When a soft tissue tumor is suspected, NICE guidelines recommend performing an ultrasound within 48 hours(8).

Musculoskeletal symptoms and signs

Musculoskeletal pain is a frequent reason for consultation in Primary Care. Its etiology varies with age, the most frequent causes being traumatic. The differential diagnosis will include overuse syndromes and osteochondroses. Less frequently, but highly important as their diagnosis delay can increase morbidity, are neoplasms and osteoarticular infections. Among the most frequent neoplasms that present with bone and / or joint pain, primary bone tumors, neuroblastomas, NHL and leukemias, can be found.

Patients with primary bone tumors, such as osteosarcoma or Ewing’s sarcoma, frequently present with localized, persistent, asymmetric, progressive bone pain that responds poorly to common analgesics and may wake the child up at night. They can associate a palpable indurated mass fixed to deep planes and of rapid and progressive growth and, occasionally, a pathological fracture can occur. Generalized musculoskeletal pain manifests as: lower limb pain, back pain, arthralgia or arthritis. The tumors that produce it are leukemias, especially lymphoblastic leukemia and bone or medullary metastases of tumors, such as Ewing’s sarcoma or neuroblastoma.

Several authors(29-31) have highlighted the relevance of incorporating leukemias and bone tumors in the differential diagnosis of patients with suspected osteomyelitis or rheumatological diseases. Leukemias that present with joint symptoms (generally in the form of asymmetric oligoarthritis) are less frequently associated with typical leukemia symptoms, such as: constitutional syndrome, hepatosplenomegaly or cytopenia, which makes diagnosis difficult(29). In the evaluation of patients with musculoskeletal symptoms, the association of leukopenia (less than 4 x 109 / L), platelets in the lower limit of normality (150-250 x 109 / L) and a history of nocturnal pain, represents a sensitivity of 100% and specificity of 85% in the diagnosis of leukemia(30). The importance of an accurate diagnosis prior to the start of steroid treatment should be noted, given the possibility of masking a hematological neoplasm or triggering a tumor lysis syndrome. For this reason, some authors suggest performing a bone marrow study prior to initiating treatment with steroids, in those patients with suspected rheumatological disease and atypical data(31).

A complete medical history that includes: characterization of the pain (onset, location, duration, intensity, number of affected joints); presence of other accompanying symptoms (inflammation, increased temperature, joint instability); precipitating factors (previous infection or trauma); and associated systemic symptoms, as well as a thorough physical examination, will guide the diagnosis. In a patient with bone and / or joint pain with suspected cancer, an X-ray as well as a full blood count and peripheral blood smear shall be performed within 48 hours. In case of a pathological X-ray, the patient should be referred to a specialized center within 48 hours. When leukemia is suspected (cytopenia of two or more cell lines and / or blasts), referral will be immediate(8).

Eye disorders

Retinoblastoma is the most common ocular neoplasm, representing 3% of childhood tumors(1). It is usually diagnosed between the first and third year of life, and 95% of them before the age of 5 years(1). 30% are bilateral and 40% hereditary. Leukocoria is the presenting sign in more than half of the cases, and it appears as a consequence of the presence of a mass located behind the crystalline lens. In the differential diagnosis of leukocoria, in addition to retinoblastoma, congenital cataracts (history of infection in pregnancy, such as toxoplasmosis, should be investigated) and Coats disease (retinal telangiectasia with deposition of intraretinal or subretinal exudates that affects younger children). Other symptoms and signs that should alert us include: strabismus, loss of visual acuity, eye pain or proptosis. In addition to retinoblastoma, other tumors that can manifest as proptosis include: neuroblastoma, rhabdomyosarcoma, lymphoma, and histiocytosis. The successful management of retinoblastoma depends on the ability to detect the disease while it still remains intraocular(32). Hence, it is very important to test for the red reflex in all newborns and in each programmed child health visit. An abnormal result of the red reflex examination is an indication for preferential referral (in less than two weeks) to the ophthalmologist(8). Patients with a family history of retinoblastoma must be referred from birth for close ophthalmological follow-up(3).

Another ocular manifestation of cancer is the paraneoplastic opsoclonus-myoclonus syndrome, which is associated with neuroblastoma in 50% of cases. It is characterized by multidirectional, involuntary and chaotic rapid eye movements, persistent during sleep, myoclonus, ataxia, and behavioral disturbances.

Time lapse to childhood cancer diagnosis

Decreasing the time to diagnosis has prognostic implications in some childhood tumors.

The most effective strategy to reduce the morbidity and mortality of childhood cancer is to focus, both on the reduction of the time to diagnosis (TD), understood as the time elapsed from the onset of symptoms to the diagnosis of cancer(33) (Fig. 6), as in the early initiation of treatment based on scientific evidence.

Figure 6. Time to diagnosis in childhood cancer.
Modified from Lethaby et al
(33).

Therefore, the recognition of alarm symptoms by families and Primary Care professionals, as well as easy access to the latter, is a priority. Screenings in childhood are usually not helpful, unless the child has a high risk of cancer associated with hereditary disorders. Among factors that have been related to TD, the following stand out: the patient’s age (the older, the greater the TD); the type of tumor (CNS, bone, germ cells and retinoblastomas have longer TD compared to leukemia and kidney tumors); and tumor biology(33-35). The relationship of TD with survival is complex. Thus, some authors(35,36) indicate that it is precisely the biology of the tumor, the factor that most influences TD and survival. In any case, there is extensive literature supporting a positive correlation between improved survival and early diagnosis in tumors such as retinoblastoma(35), being this correlation more ambiguous for CNS and other solid tumors, probably in relation to high-grade tumors having a more abrupt onset of prediagnostic symptoms and, therefore, a shorter TD(37). In the group of adolescents and young adults, although females more frequently consult physicians, they have more prolonged TD. Bone tumors and lymphomas have longer TD, probably due to the non-specific clinical presentation of these types of cancer(6).

It should be noted how, in the COVID-19 pandemic situation, several publications have warned of the increase in TD of childhood cancer in neighboring countries(38,39).

Role of the Primary Care pediatrician

The Primary Care pediatrician shares the responsibility of reducing TD, identifying those patients suspected of cancer and making an early referral to specialized care. This decrease in TD may have a prognostic role for some tumors and, in addition, contributes to the reduction of anxiety and stress experienced by patients and their families during the difficult period prior to the diagnosis of childhood cancer(5).

Bibliography

The asterisks show the interest of the article in the opinion of the authors.

1. Pardo Romaguera E, Muñoz López A, Valero Poveda S, Porta Cebolla S, Fernández-Delgado R, Barreda Reines MS PBR. Cáncer infantil en España. Estadísticas 1980-2017. Registro Español de Tumores Infantiles (RETI-SEHOP). Valencia: Universitat de Valéncia, 2018 (Preliminar edition, CD-Rom).

2. Ministerio de Sanidad, Servicios Sociales e Igualdad. Portal Estadístico (sede Web). Accessed february 28, 2021. Available at: https://pestadistico.inteligenciadegestion.mscbs.es/publicoSNS/S.

3.*** Coury SA, Schneider KA, Schienda J, Tan WH. Recognizing and managing children with a pediatric cancer predisposition syndrome: A guide for the pediatrician. Pediatr Ann. 2018; 47: e204-16.

4.*** Fragkandrea I, Nixon JA, Panagopoulou P. Signs and symptoms of childhood cancer: a guide for early recognition. Am Fam Physician. 2013; 88: 185-92.

5.*** Dixon-Woods M, Findlay M, Young B, Cox H, Heney D. Parents’ accounts of obtaining a diagnosis of childhood cancer. Lancet. 2001; 357: 670-4.

6.** Herbert A, Lyratzopoulos G, Whelan J, Taylor RM, Barber J, Gibson F, et al. Diagnostic timeliness in adolescents and young adults with cancer: a cross-sectional analysis of the BRIGHTLIGHT cohort. Lancet Child Adolesc Health. 2018; 2: 180-90.

7.*** Clarke RT, Jones CHD, Mitchell CD, Thompson MJ. Shouting from the roof tops: a qualitative study of how children with leukaemia are diagnosed in primary care. BMJ Open. 2014; 4: e004640. doi:10.1136/bmjopen-2013-004640.

8.** NICE. Suspected cancer recognition and referral: symptoms and findings. Updated january 29, 2021. Accessed april 1st, 2021. Available at: https://www.nice.org.uk/guidance/ng12.

9.** Dommett RM, Redaniel T, Stevens MCG, Martín RM, Hamilton W. Risk of childhood cancer with symptoms in primary care: a population-based case-control study. Br J Gen Pract. 2013; 63: e:22-9.

10.** Pan American Health Organization. Early diagnosis of childhood cancer. Washington, DC; OPS. 2014.

11.** Wilne S, Collier J, Kennedy C, Koller K, Grundy R, Walker D. Presentation of childhood CNS tumours: a systematic review and meta-analysis. Lancet Oncol. 2007; 8: 685-95.

12. Wilne S, Collier J, Kennedy C, Jenkins A, Grout J, MacKie S, et al. Progression from first symptom to diagnosis in childhood brain tumours. Eur J Pediatr. 2012; 171: 87-93.

13. Chu TPC, Shah A, Walker D, Coleman MP. Where are the opportunities for an earlier diagnosis of primary intracranial tumours in children and young adults? Eur J Paediatr Neurol. 2017; 21: 388-95.

14. Shanmugavadivel D, Liu JF, Murphy L, Wilne S, Walker D. Accelerating diagnosis for childhood brain tumours: An analysis of the HeadSmart UK population data. Arch Dis Child. 2020; 105: 355-62.

15. Ansell P, Johnston T, Simpson J, Crouch S, Roman E, Picton S. Brain tumor signs and symptoms: Analysis of primary health care records from the UKCCS. Pediatrics. 2010; 125: 112-9.

16.*** Wilne S, Koller K, Collier J, Kennedy C, Grundy R, Walker D. The diagnosis of brain tumours in children: A guideline to assist healthcare professionals in the assessment of children who may have a brain tumour. Arch Dis Child. 2010; 95: 534-9.

17.*** Walker D, Wilne S, Grundy R, Kennedy C, Neil, Dickson A, et al. A new clinical guideline from the Royal College of Paediatrics and Child Health with a national awareness campaign accelerates brain tumor diagnosis in UK children – “headSmart: Be Brain Tumour Aware.” Neuro Oncol. 2016; 18: 445-54.

18. Chow A, Robinson JL. Fever of unknown origin in children: a systematic review. World J Pediatr. 2011; 7: 5-10.

19. Clarke RT, Van Den Bruel A, Bankhead C, Mitchell CD, Phillips B, Thompson MJ. Clinical presentation of childhood leukaemia: A systematic review and meta-analysis. Arch Dis Child. 2016; 101: 894-901.

20.** Del Rosal Rabes T, Baquero Artigao F. Adenitis cervical. Pediatr Integral. 2018; XXII(7): 307-15.

21. Nield LS, Kamat D. Lymphadenopathy in Children: When and How to Evaluate. Clin Pediatr (Phila). 2004; 43: 25-33.

22. Chiappini E, Camaioni A, Benazzo M, Biondi A, Bottero S, De Masi S, et al. Development of an algorithm for the management of cervical lymphadenopathy in children: Consensus of the Italian Society of Preventive and Social Pediatrics, jointly with the Italian Society of Pediatric Infectious Diseases and the Italian Society of Pedi. Expert Rev Anti Infect Ther. 2015; 13: 1557-67.

23.** Green K, Behjati S, Cheng D. Fifteen-minute consultation: Obvious and not-so-obvious mediastinal masses. Arch Dis Child Educ Pract Ed. 2019; 104: 298-303.

24. Chen C, Wu K, Chao Y, Weng D, Chang J-S, Lin C-H. Clinical manifestation of pediatric mediastinal tumors, a single center experience. Medicine. 2019; 98: 32.

25.** Saraswatula A, McShane D, Tideswell D, Burke GAA, Williams DM, Nicholson JC, et al. Mediastinal masses masquerading as common respiratory conditions of childhood: A case series. Eur J Pediatr. 2009; 168: 1395-9.

26.** Uzunova L, Bailie H, Murray MJ. Fifteen-minute consultation: A general paediatrician’s guide to oncological abdominal masses. Arch Dis Child Educ Pract Ed. 2019; 104: 129-34.

27. Potisek NM, Antoon JW. Abdominal masses. Pediatr Rev. 2017; 38: 101-3.

28. Steuber PC. Clinical assessment of the child with suspected cancer. UpToDate; 2021. p. 1-24.

29. Brix N, Rosthøj S, Herlin T, Hasle H. Arthritis as presenting manifestation of acute lymphoblastic leukaemia in children. Arch Dis Child. 2015; 100: 821-25.

30. Jones OY, Spencer CH, Bowyer SL, Dent PB, Beth S, Rabinovich CE. A Multicenter Case-Control Study on Predictive Factors Distinguishing Childhood Leukemia From Juvenile Rheumatoid Arthritis. 2006; 117: e840-4.

31. Sen ES, Moppett JP, Ramanan AV. Are you missing leukaemia? Arch Dis Child. 2015; 100: 811-2.

32. Parrilla Vallejo M, Perea Pérez R, Relimpio López I, Montero de Espinosa I, Rodríguez de la Rúa E, Terrón León JA, et al. Retinoblastoma: The importance of early diagnosis. Arch Soc Esp Oftalmol. 2018; 93: 423-30.

33.** Lethaby CD, Picton S, Kinsey SE, Phillips R, Van Laar M, Feltbower RG. A systematic review of time to diagnosis in children and young adults with cancer. Arch Dis Child. 2013; 98: 349-55.

34. Barr RD. “Delays” in diagnosis: A misleading concept, yet providing opportunities for advancing clinical care. J Pediatr Hematol Oncol. 2014; 36: 169-72.

35.*** Brasme JF, Morfouace M, Grill J, Martinot A, Amalberti R, Bons-Letouzey C, et al. Delays in diagnosis of paediatric cancers: A systematic review and comparison with expert testimony in lawsuits. Lancet Oncol 2012; 13: e445-59.

36. Brasme JF, Chalumeau M, Oberlin O, Valteau-Couanet D, Gaspar N. Time to diagnosis of Ewing tumors in children and adolescents is not associated with metastasis or survival: A prospective multicenter study of 436 patients. J Clin Oncol. 2014; 32: 1935-40.

37.** Ferrari A, Lo Vullo S, Giardiello D, Veneroni L, Magni C, Clerici CA, et al. The Sooner the Better? How Symptom Interval Correlates With Outcome in Children and Adolescents With Solid Tumors: Regression Tree Analysis of the findings of a prospective study. Pediatr Blood Cancer. 2016; 63: 479-85.

38. Soares Martins QC, Gomes de Morais Fernandes FC, Pereira Santos VE, Guerra Azevedo I, Góes de Carvalho Nascimento, LS, Dos Santos Xavier CC, et al. Factors associated with the detection of childhood and adolescent cancer in primary health care: A prospective cross-sectional study. J Multidiscip Healthc. 2020; 13: 329-37.

39. Chiaravalli S, Ferrari A, Sironi G, Gattuso G, Bergamaschi L, Puma N, et al. A collateral effect of the COVID-19 pandemic: Delayed diagnosis in pediatric solid tumors. Pediatr Blood Cancer. 2020; 67: 2-3.

Recommended bibliography

- Fragkandrea I, Nixon JA, Panagopoulou P. Signs and Symptoms of childhood cancer: A guide for early recognition. Am Fam Physician. 2013; 88: 185-92.

Article focused on the early diagnosis of childhood cancer in Primary Care, by focusing on those warning signs that should alert us of the possibility of neoplastic processes.

- Clarke RT, Jones CHD, Mitchell CD, Thompson MJ. Shouting from the roof tops: a qualitative study of how children with leukaemia are diagnosed in primary care. BMJ Open. 2014; 4: e004640. doi: 10.1136 / bmjopen-2013-004640.

Highly recommended. Qualitative methodology article that explores the presentation of childhood leukemia and the factors that influence its suspicion from the point of view of parents and Primary Care physicians.

- Wilne S, Koller K, Collier J, Kennedy C, Grundy R, Walker D. The diagnosis of brain tumors in children: a guideline to assist healthcare professionals in the assessment of children who may have a brain tumor. Arch Dis Child. 2010; 95: 534-9.

A must-read. Clinical guide for the suspicion and early diagnosis of tumors of the central nervous system.

- Walker D, Wilne S, Grundy R, Kennedy C, Neil, Dickson A, et al. A new clinical guideline from the Royal College of Paediatrics and Child Health with a national awareness campaign accelerates brain tumor diagnosis in UK children – “headSmart: Be Brain Tumor Aware.” Neuro Oncol. 2016; 18: 445-54.

Very interesting. Strategy launched in 2011 in the United Kingdom, aimed at healthcare professionals and the general public, with the aim of reducing the time interval from the onset of symptoms to the diagnosis of a CNS tumor in pediatric patients.

 

 

Clinical case

 

7-year-old patient who presents with a 3-day course of frontal, evening headache, as well as progressive 2-week loss of vision in the left eye.

Personal history

Normal course pregnancy. Vaginal delivery. Normal neonatal period. Normal psychomotor development. Up-to-date vaccination schedule.

Family background

Mother: 32 years old, healthy, gestations: 1/miscarriages: 0/life births: 1. Father: 38 years old, healthy.

Physical examination

HR: 90 bpm. RR: 18 rpm. BP: 100/60 mmHg. Sat: 97%. Height: 122 cm (50th centile). Weight: 24 kg (50th centile). Good general condition. Well hydrated and perfused. No rashes or petechiae. No significant lymphadenopathies. Cardio-pulmonary auscultation: normal. Abdomen: soft, non-tender, no masses or megalies. Neurological: Glasgow 15, with normal cranial nerves, tone, strength, sensitivity, tendon reflexes and gait. No dysmetria or dysdiadochokinesia. Ophthalmological examination: left eye with decreased visual acuity, signs of papillary atrophy, as well as left temporal hemianopsia.

Complementary tests

Normal full blood count, biochemistry and coagulation. A brain MRI was performed which evidenced a midline, suprasellar, multicystic, heterogeneous mass measuring 40 x 36 x 39 mm (Fig. 7), with intense and heterogeneous enhancement following contrast administration (Fig. 8). Normal ventricular system, without signs of hydrocephalus.

Figure 7. Heterogeneous midline, suprasellar, multicystic mass measuring 40 x 36 x 39 mm.

Figure 8. Midline mass with intense and heterogeneous enhancement following contrast administration.

Diagnosis

Craniopharyngioma (adamantinomatous pattern).

 

 

 

 

Anemia. Classification and diagnosis

B. Rosich del Cacho*, Y. Mozo del Castillo**
Topics on
Continuous Training



B. Rosich del Cacho*, Y. Mozo del Castillo**

*Consultant physician, Pediatrics Service, Hospital Universitario Joan XXIII, Tarragona. **Consultant physician, Pediatric Hemato-Oncology and Hematopoietic Stem Cell Transplantation Service, Hospital Universitario La Paz, Madrid

Abstract

Anemia is defined as a reduction in hemoglobin concentration below normal levels for age, gender and ethnicity. It is the result of an imbalance between the production and destruction of red blood cells, which characterizes or accompanies various conditions. It is the most common hematological abnormality in childhood, the main cause of which is iron deficiency. Clinical manifestations are often nonspecific. Diagnosis begins with a full blood count, peripheral blood smear and biochemical parameters of hemolysis and iron metabolism. The overall diagnostic approach of the child with anemia is here reviewed, and an algorithm is proposed based on basic hematological data. Finally, a brief list of references is provided.

 

Key words: Anemia; Child; Infant; Classification; Diagnosis.

Palabras clave: Palabras clave: Anemia; Niño; Lactante; Clasificación; Diagnóstico.

 

 

Pediatr Integral 2021; XXV (5): 214 – 221

 


 

Anemia. Classification and diagnosis

Introduction

Anemia is defined as a reduction in the concentration of hemoglobin or hematocrit, the normal levels of which depend on age, sex, and ethnicity. Iron deficiency anemia is the most prevalent hematologic disorder of childhood.

In this article, the general concepts of anemia in childhood and its diagnostic approach are reviewed. Iron deficiency anemia (the most common cause of anemia in the pediatric age) and hemolytic anemia are specifically discussed in other articles.

Definition(1,2)

The word anemia is of Greek origin, meaning “without blood.” It is defined as the reduction in the concentration of hemoglobin (Hb), erythrocyte mass or hematocrit in peripheral blood below 2 standard deviations (-2 SD) for the age, sex and ethnicity of the patient (Table I).

Hemoglobin (Hb): complex protein made up of heme groups containing iron and a protein portion, globin. The concentration of this erythrocyte pigment is presented in grams (g) per 100 ml (dl) of whole blood.

Hematocrit (Hct): fraction of the volume of erythrocyte mass with respect to total blood volume. It is expressed as a percentage (%).

Epidemiology(2-4,7)

Anemia is the most common hematological disorder in childhood. In 2008, the World Health Organization (WHO) published the results of a survey of 192 member states, establishing the following Hb thresholds according to age group:

• 11 g/dl in children 0.5-4.99 years-old (preschool age).

11.5 g/dl between 5-11.99 years of age.

• 12 g/dl between ages 12-14.99 years.

The global prevalence of anemia was 47.4% ([95% CI] 45.7-49.1) in preschool-age children and 25.4% ([95% CI] 19.9-30.9) in school-age children. The prevalence in preschool children varied widely by country, led by countries in South America and Africa. This is because iron deficiency accounts for 50% of this prevalence and it is closely linked to nutritional deficiencies, therefore, to the social and development conditions in these countries.

In addition to acknowledging iron deficiency as the most common cause of anemia in pediatric age worldwide, we must take into account the factors and causes of this disorder that influence its prevalence:

Age: Hb and Hct counts vary throughout childhood (Table I), as well as the causes of anemia differ according to the age of the patients:

Birth-3 months: Hb achieves maximum concentrations (16.5-18.5 g/dl) in the newborn and drops to 9-10 g/dl between 6-9 weeks of life, as a consequence of increased oxygenation of tissues and a drastic decrease in erythropoiesis; in what is referred to as “physiological anemia of infancy”. Any anemia in this age group that differs from the characteristics of physiological anemia (Hb <9 g/dl, anemia prior to 1 month of age, or signs of hemolysis) will require further study.

3-6 months: iron deficiency is rare during this period, and hemoglobinopathies must be ruled out.

6 months-adolescence: there are differences in Hb counts according to age and sex (Table I). During this entire stage, the main cause of anemia is iron deficiency.

Sex: starting at puberty, testosterone secretion induces an increase in erythrocyte mass, which is why the normal level of Hb is higher in men than in women. On the other hand, some hereditary anemias are X-linked, and hence, being more frequent in men (eg, glucose-6-phosphate dehydrogenase (G6PDH) deficiency and sideroblastic anemia).

Race and ethnicity: normal Hb levels are observed with approximately 0.5 g/dl less in black children compared to those observed in Caucasians or Asians. HbS and HbC are more common in black and Hispanic populations. Furthermore, within the same country there are areas with a higher prevalence of hemoglobinopathies, endocellular parasites, such as malaria and infestation with intestinal parasites that impact on the prevalence of anemia. Thus, thalassemic syndromes are more prevalent on the Mediterranean coast, a large part of Africa, the Middle East, the Indian subcontinent, and Southeast Asia; conversely, G6PDH deficiency is observed, predominantly, in malaria endemic areas, since it seems to be a protective factor against this infection (a higher prevalence is found among: Kurdish Jews, Sardinian, Nigerian, African-American, Filipino and Greeks).

Height above sea level: the higher above the sea level, the higher the Hb count, since lower oxygen content in the air results in a stimulus for hematopoiesis.

Pathophysiology(2,5)

Anemia is the result of the imbalance between production and loss of red blood cells. The redistribution of blood, the stimulation of erythropoiesis and the decrease in the affinity of Hb for O2 are compensatory mechanisms.

Erythropoiesis mainly takes place in the bone marrow during postnatal and adult life (in the fetal period and up to 6 months of extrauterine life, the endodermal sinus also participates, where it begins at 3-4 weeks of gestation, and later on in the liver). Various regulatory factors (being blood oxygen saturation the main one) act on the peritubular cells of the kidneys involved in the synthesis of erythropoietin (EPO), a hormone that acts on the hematopoietic precursors of the bone marrow, which finally give rise to mature red blood cells. During this complex process of differentiation and maturation leading to the production of the mature erythrocyte, the participation of different molecules, growth factors (G and GM-CSF), trace elements (such as iron, essential for the elaboration of the heme group of Hb, copper and zinc) and cytokines (IL 1, 3, 4, 6, 9 and 11).

Mature erythrocytes are shaped like a biconcave disc, they are filled with Hb in the inside and are devoid of mitochondria or other organelles. Hb is composed of 4 globin subunits and heme groups, and it is involved in the exchange of oxygen and carbon dioxide throughout the body.

After erythrocytes have been in circulation for a long period (half-life 120 days), they are taken up and destroyed by the reticuloendothelial system of the spleen. To maintain normal Hb levels, there must be a balance between the continued loss of senescent red cells and erythropoiesis in the bone marrow. Therefore, anemia is the result of the imbalance between production (decreased) and destruction or loss of red blood cells (increased).

There are different compensation mechanisms as an adaptive response to the situation of anemia:

Redistribution of blood flow: this guarantees the oxygenation of vital organs (brain and myocardium), with the consequent vasoconstriction of less needy areas, such as skin and kidney.

Stimulation of erythropoiesis: this is mediated by an increase in EPO synthesis, the main trigger of which is tissue hypoxia. This mechanism is only effective if the bone marrow is able to respond with increased production of red blood cells and the consequent increase in reticulocytes in peripheral blood.

Increased ability of Hb to deliver oxygen to tissues: through an increase in the concentration of 2,3-diphosphoglycerate, which decreases the affinity of Hb for O2 and favors oxygenation of the tissues.

Understanding all of the above will help to understand the clinical manifestations and the pathophysiological classification of anemia, which are explained in the following sections.

Clinical manifestations(4,6)

Anemia can have both nonspecific clinical manifestations as well as guiding signs and symptoms for the etiological diagnosis.

The clinical picture of the anemic syndrome has common manifestations determined by: adaptation mechanisms, age of onset, underlying disease and type of onset (acute or chronic):

Pale skin and mucous membranes: direct consequence of the decrease in Hb and the accompanying peripheral vasoconstriction. Sometimes pallor may not be evident until the Hb level falls below 8 g/dl and can be difficult to identify depending on the pigmentation of the skin.

Cardiocirculatory symptoms and signs (palpitations, tachycardia, systolic murmur, exertional dyspnea and tachypnea): in general, they are due to the onset of compensatory mechanisms due to the decrease in blood volume, and become more evident the greater the degree of anemia and the speed of its establishment.

General symptoms (headache, irritability, mood swings, asthenia, anorexia): due to tissue hypoxia. In chronic anemia, there may also be an impact on diverse organs, leading to: neurological dysfunction (impaired psychomotor development, learning difficulty), delayed puberty, osteopenia, cardiological abnormalities (left ventricular hypertrophy that can lead to heart failure), etc.

Accompanying symptoms: derived from the causes and pathogenic mechanisms involved, such as:

– Hemolytic anemia: jaundice, choluria, abdominal pain, splenomegaly (due to the role of the spleen in the destruction of erythrocytes) or hepatosplenomegaly (because of extramedullary erythropoiesis), gallstones and bone abnormalities (due to extramedullary erythropoiesis).

– Deficiency anemia: trophic disorders of the skin and mucous membranes.

– Anemia of central origin (medullary): bleeding and infections if associated with thrombopenia and leukopenia.

Diagnosis(2,7-9)

Adequate history-taking, physical examination, full blood count with red cell indices, reticulocytes, peripheral blood smear and biochemistry, are cost-effective tools for the diagnostic approach of anemia.

Medical history

An adequate medical history is the starting point for the etiological diagnosis of anemia. In addition to noticing the age, gender, ethnicity and geographical origin of the patient, the following should be investigated:

Symptoms (see previous section): beginning and speed of onset, tolerance, history of bleeding (digestive, menstrual, etc.), symptoms suggestive of hemolysis, etc.

Neonatal history: gestational age, blood group, history of hospital admission for jaundice/anemia, neonatal screening results for endocrine-metabolic diseases (sickle cell anemia is included in Spain).

Underlying pathology: previous episodes of anemia and treatment received, presence of coagulopathy, concomitant diseases (infectious and/or inflammatory), malabsorptive problems (eg, celiac disease).

Family history: the former existence of: anemia, jaundice, gallstones, splenomegaly or the need for cholecystectomy in family members, can guide the diagnosis of hereditary hemolytic anemia.

Diet: to be queried in order to consider possible nutritional deficiencies (iron, vitamin B12 and folic acid). It is important to document the type of lactation (breastfeeding/artificial formula), the amount and the possibility of supplementation/fortification. The presence of pica can guide towards a deficit of nutrients. To consider that the intake of certain foods (eg: fava beans) can trigger hemolytic crises in G6PDH deficiency.

Exposure to drugs/toxins: medications (antibiotics, anti-inflammatory drugs, anticonvulsants), herbs, homeopathic products, drinking water containing nitrates, oxidants or products with lead.

Physical examination

Special attention should be paid to: skin, eyes, mouth, face, chest, hands and abdomen. Skin paleness is a specific but poorly sensitive finding; and so is tachycardia, as a manifestation of severity. Jaundice and hepatosplenomegaly, characteristic of hemolysis, are also specific data, but with relatively low sensitivity.

Table II shows certain physical signs which can guide towards a specific cause of anemia.

Laboratory evaluations

Work-up tests should begin with a full blood count with red cell indices and a thorough examination of the peripheral blood smear. In addition, first-level studies require: reticulocyte count, basic biochemistry and study of iron metabolism.

In any study of anemia, we must take into account the following parameters of the full blood count, which will aid in the classification of anemia and consequently in its etiological diagnosis:

Mean corpuscular volume (MCV): this is the average size (fl) of the red blood cells. Depending on this value, the morphological classification of anemia will be carried out (see next section).

Red cell distribution width (RDW): this informs of the coexistence of red cell populations of different sizes. This parameter helps to distinguish between iron deficiency and thalassemia, since in the former it is usually high, due to the different distribution of Hb depending on the iron availability at each moment; whilst in thalassemia, RDW is usually normal (although it may be elevated), because the Hb distribution is uniform.

Reticulocyte Production Index (RPI): reticulocytes report on the regenerative capacity of the bone marrow, so this parameter will allow a pathophysiological classification of anemia (see next section). Normal values of RPI range between 2 and 3. The correct interpretation of the reticulocyte number requires the adjustment of the crude number (%) according to the real number of red cells of each patient, using the formula shown in figure 1.

 

Figura

Figura

Figure 1. Formula for adjusting the reticulocyte count.

Peripheral blood smear examination: this must be performed by an experienced hematologist. The size and morphology of the red blood cells can be essential to identify disorders such as: sickle cell disease (sickle cells), spherocytosis (spherocytes), hemoglobinopathies (target cells), hemolysis (Heinz bodies), etc. Additionally, the presence of other cytopenias or leukocytosis with immature forms can point toward certain etiologies (infections, medullary aplasia/hypoplasia, infiltration of the bone marrow due to leukemia/lymphoma, etc.).

Iron metabolism: ferritin is the most useful parameter to measure iron stores; values below 15mcg/l are indicative of iron deficiency. However, its usefulness is limited by its role as an acute phase reactant, increasing with inflammation/infection and tissue destruction.

According to the previous findings, we will have to expand the investigations with second level studies (Diagnostic algorithms, algorithms 1 and 2):

– If reticulocytosis and data suggestive of hemolysis are present, the existence of hemolytic anemia should be ruled out and the following should be requested: haptoglobin, Coombs test to determine autoimmunity, and if negative, hemoglobin electrophoresis to rule out hemoglobinopathy (considering that HbF is predominant within the first months of life, and its decrease is slower in sickle cell disease, where it persists in variable amounts) and/or quantification of enzymes (G6PDH, pyruvate-kinase) to study enzymopathies, and/or membrane studies (osmotic fragility, cytometry) for membranopathies.

– If macrocytosis (elevated MCV) is found: there may be a deficiency cause (vitamin B12, folic acid) or hypothyroidism (request TSH and FT4), but it could also be a central maturational disorder (bone marrow aspirate/biopsy must be performed).

– If there is reticulocytopenia and parvovirus B19 infection has been ruled out, a bone marrow aspirate/biopsy must be carried out to assess anemia of central origin.

Classification of anemia(2,7-9)

Morphological and pathophysiological classifications of anemia are complementary and required for the etiological diagnosis approach.

Following the results obtained in the first-level investigations, the morphological and pathophysiological classifications of anemia will be carried out, which will lead to the determination of the etiology after conducting specific additional work-up (Table III and Algorithms 1 and 2).

Morphological classification: based on the MCV (Table I).

Microcytic: MCV 2.5 percentile for age, sex, and ethnicity. In childhood, these are iron deficiency anemia and thalassemia par excellence.

Normocytic: Normal MCV (between 2.5 and 97.5 percentiles) for age, sex and ethnicity. This can be due to: blood loss, chronic disorders, infections or it can be the initial stage of micro or macrocytic anemia.

Macrocytic: MCV 97.5 percentile for age, sex, and ethnicity. In childhood the most common ones are due to vitamin B12 and/or folic deficiency or exposure to certain drugs (eg, anticonvulsants and immunosuppressants). Macrocytosis can also be found in acute regenerative anemia, due to the presence of reticulocytosis, and in maturation disorders of the bone marrow (myelodysplasia and aplasia).

Pathophysiological classification: based on the medullary regenerative capacity, determined by the RPI.

Regenerative: there is an elevated reticulocyte response (reticulocytes >3% or RPI 3). Examples of this include: hemolytic anemia and anemia secondary to hemorrhage.

A/hyporegenerative: normal or low reticulocyte response for the degree of anemia (reticulocytes <1-1.5% or RPI <2). It translates the existence of a hypo/inactive bone marrow as a result of various reasons: deficiency of substrates (iron, folic acid, vitamin B12), tumor medullary infiltration, infections, deposit diseases (Nieman-Pick, Gaucher…) or congenital or acquired aplasia.

Role of the Primary Care pediatrician

• From the Primary Care setting, full blood count with erythrocyte indices, reticulocytes, biochemistry with hemolysis parameters and iron profile, can be requested, which provide the diagnostic approach of most anemia cases in the pediatric age.

• In our environment, the anemia cases that the Primary Care pediatrician can most frequently find are: iron deficiency anemia, physiological anemia of infancy, β-heterozygous thalassemia or thalassemic trait, anemia of chronic disorders, anemia due to hemorrhage and, currently also, sickle cell anemia.

• In general terms, any anemia of the following characteristics: combined with another cytopenia, hemoglobinopathies, hemolytic anemia, aregenerative anemia, those that do not respond to treatment, and whenever there is any other warning sign, is susceptible to referral to specialized care.

Bibliography

The asterisks show the interest of the article in the opinion of the authors.

1. Glader B. Anemia. In: Kliegman RM, Behrman RE, Jenson HB, Stanton BF, ed. Nelson. Textbook of Pediatrics. 18th ed., Barcelona. Elsevier España; 2009. p. 2003-6.

2. Hernández Merino A. Anemias en la infancia y adolescencia; clasificación y diagnóstico. Pediatr Integral. 2016; XX(5): 287-96.

3. Word Health Organization. Worldwide prevalence of anemia 1993-2005: WHO global database on anemia. Geneva: World Health Organization; 2008.

4.** Allali S, Brousse V, Sacri AS, Chalumeau M, de Montalembert M. Anemia in children: prevalence, causes, diagnostic work-up, and long-term consequences. Expert Rev Hematol. 2017; 10: 1023-8.

5.** Arrizabalaga B, González FA, Remacha. Eritropatología. Edición Ambos Marketing Services. Barcelona. 2017.

6.** Prudencio García-Paje M. Aproximación diagnóstica al paciente con anemia. In: Madero L, Lassaletta A, Sevilla J, ed. Hematología y Oncología Pediátricas. 3ª edición, Madrid. Ergon; 2015. p. 81-6.

7.*** Sandoval C. Approach to the child with anemia (Literature review: September 2020. Last update: June 20, 2019). Available at: www.uptodate.com.

8.** San Román Pacheco S, Mozo del Castillo Y. Síndrome anémico. In: Guerrero-Fdez. J, Cartón Sánchez A, Barreda Bonis A, Menéndez Suso J, Ruiz Domínguez J, ed. Manual de Diagnóstico y Terapéutica en Pediatría. 6ª edición, Madrid. Editorial Panamericana; 2017. p. 1117-30.

9. Buttarello M. Laboratory diagnosis of anemia: are the old and new red cell parameters useful in classification and treatment, how? Int J Lab Hematol. 2016; 38: 123-32.

10. Sevilla Navarro J. Abordaje de la anemia microcítica; nuevas herramientas diagnósticas. In: AEPap, ed. 7º Curso de Actualización en Pediatría 2010. Madrid: Exlibris Ediciones; 2010. p. 239-9.

Recommended bibliography

– Allali S, Brousse V, Sacri AS, Chalumeau M, de Montalembert M. Anemia in children: prevalence, causes, diagnostic work-up, and long-term consequences. Expert Rev Hematol. 2017; 10: 1023-8.

This article summarizes in a practical way, the causes of anemia in Pediatrics and the investigations necessary for its diagnosis, highlighting the comorbidity that this pathology implies in the long term.

– Arrizabalaga B, González FA, Remacha. Eritropatología. Edición Ambos Marketing Services. Barcelona. 2017.

General hematology chapter, suitable for the study of hematopoiesis and the pathophysiology of anemia.

– Prudencio García-Paje M. Aproximación diagnóstica al paciente con anemia. In: Madero L, Lassaletta A, Sevilla J, ed. Hematología y Oncología Pediátricas. 3ª edición, Madrid. Ergon; 2015. p. 81-6.

Chapter where a general presentation is made on anemia in Pediatrics, useful to make a global approach to the subject. In the same book, there are other specific chapters describing the different types of anemia in greater depth.

– Sandoval C. Approach to the child with anemia (Literature review: September 2020. Last update: June 20, 2019). Available at: www.uptodate.com.

This frequently updated source of evidence-based medicine offers a comprehensive review of childhood anemia, presented in a clear educational structure.

– San Román Pacheco S, Mozo del Castillo Y. Síndrome anémico. In: Guerrero-Fdez. J, Cartón Sánchez A, Barreda Bonis A, Menéndez Suso J, Ruiz Domínguez J, ed. Manual de Diagnóstico y Terapéutica en Pediatría. 6ª edición, Madrid. Editorial Panamericana; 2017. p. 1117-30.

Presentation of the topic of anemia in childhood, with practical tables and algorithms.

 

Clinical case

 

Presentation

A 7-year-old girl is seen in clinic because of progressive paleness and a one-month clinical picture of asthenia, initially attributed by the family to various infectious processes of probable viral etiology (gastroenteritis, upper respiratory tract catarrh and otitis), without any of them requiring admission or antibiotic therapy. Her physical activity remains intact. She has not had: fever, sweating, anorexia, weight loss or musculoskeletal pain. She had taken mebendazole in the month prior to the consultation as treatment for oxyuriasis, and ibuprofen occasionally. Her pediatrician had requested a test where Hb of 8 g/dl stands out. Deficiency and/or parainfectious etiology was suspected, and iron therapy was prescribed. She attends the clinic today after two weeks of starting treatment for clinical and analytical control.

Personal history

Up-to-date vaccination, according to her region´s (autonomous community) immunization schedule.

No known allergies.

No medical-surgical history of interest.

Pets: a dog who has lived at home for 10 years, correctly vaccinated.

She lives in an urban area. No recent trips.

Exercise: swimming.

Family background

Parents and a 10-year-old brother, all healthy.

Maternal grandmother: deficiency anemia.

No other background of interest.

Physical examination

Weight: 20 kg. Temperature: 36.5°C. Respiratory rate: 22 rpm. Heart rate: 105 bpm. Systolic/diastolic blood pressure: 115/65 mmHg.

Good general condition. Marked skin and mucosal paleness. No rashes or petechiae. No respiratory distress. Good peripheral perfusion, capillary filling time <2 seconds, peripheral pulses are present and symmetrical. Cardiac auscultation: rhythmic sounds, systolic murmur II/VI. Lung auscultation: normal. Normal oropharynx. Normal bilateral otoscopy. No pathologic lymphadenopathies. Soft, non-tender abdomen, without masses or organ enlargement. Preserved peristalsis. No signs of peritoneal irritation. Glasgow coma scale 15/15, collaborating, negative meningeal signs, no neurological focus.

Work-up tests

Full blood count: Hb: 6.5 g/dl; MCV: 89 fl; leukocytes: 4.76×109/l; neutrophils: 0.98×109/l; lymphocytes: 3.61×109/l; monocytes: 0.16×109/l; platelets: 120×109/l; MPV: 10 fl. Reticulocytes: 21×109/l (0.7%).

Biochemistry: glucose: 80 mg/dl; urea: 25 mg/dl; creatinine: 0.4 mg/dl; sodium: 140 mEq/l; potassium: 4.1 mEq/l; chloride: 105 mEq/l; GOT: 29 IU/l; GPT: 21 IU/l; total bilirubin: 0.4 mg/dl; LDH: 206 IU/l; phosphorus: 5.5 mg/dl; total calcium: 10 mg/dl.

 

 

 

 

Acne

J.M. Azaña Defez, M.L. Martínez Martínez
Topics on
Continuous Training


J.M. Azaña Defez, M.L. Martínez Martínez

Doctors in Medicine and Surgery. Consultant Physicians at the Dermatology Service. Pediatric Dermatology Unit. University Hospital Complex of Albacete

Abstract

Acne is a chronic inflammatory skin disease of the pilosebaceous unit of multifactorial etiology characterized by increased sebaceous secretion, comedone formation, inflammatory lesions and risk of scarring sequelae. It is undoubtedly one of the most frequent dermatological processes in the daily clinical practice, especially in adolescence, although it can also appear in childhood and persist into adulthood. Adequate management of this pathology is relevant, as it can cause lower self-esteem and social dysfunction in patients, with the subsequent impact on quality of life.

 

Abstract

Acne is a chronic inflammatory skin disease of the pilosebaceous unit of multifactorial etiology characterized by increased sebaceous secretion, comedone formation, inflammatory lesions and risk of scarring sequelae. It is undoubtedly one of the most frequent dermatological processes in the daily clinical practice, especially in adolescence, although it can also appear in childhood and persist into adulthood. Adequate management of this pathology is relevant, as it can cause lower self-esteem and social dysfunction in patients, with the subsequent impact on quality of life.

 

Key words: Cutibacterium acnes; Grading and classification of acne; Acne management; Isotretinoin.

Palabras clave: Acné; Cutibacterium acnes; Graduación y clasificación del acné; Manejo del acné; Isotretinoina.

Pediatr Integral 2021; XXV (4): 166 – 175


Acne

Introduction

Acne is a frequent inflammatory skin disease of chronic course and polymorphous in its clinical expression.

Acne is a chronic skin disease of the pilosebaceous unit, of multifactorial etiology, characterized by its clinical polymorphism. It undoubtedly represents one of the dermatological processes of greatest interest in daily clinical practice.

Epidemiology

Acne can appear in all stages of life, although its prevalence is higher in adolescence and there seems to be a genetic predisposition.

Acne is one of the most frequent dermatological diseases, as it is estimated that around 85% of the population will present it throughout their lives(1). The prevalence of acne among the Spanish population aged 12 to 18 years is 74%, without significant differences regarding sex and with a peak between 14 and 16 years of age(2); therefore, it accounts for 25% of dermatological consultations. Acne is estimated to be of moderate / severe intensity in about 20% of patients.

Its highest prevalence and intensity occur around 14-15 years of age in females and somewhat later (16-18 years) in males. Despite its prevalence in adolescence, in 7-25% of patients it will persist into adulthood.

In females, it can manifest a longer course; whilst in males, more serious forms are identified(3).

There seems to be certain genetic predisposition to develop acne: history of acne is often found in parents, and in addition, there is a high concordance in monozygotic twins(3,4).

Seasonal variations in acne severity are observed, with a tendency to worsen in winter(5).

Etiopathogenesis

Acne is a multifactorial disease, produced by: increased sebaceous secretion, follicular epidermal hyperproliferation, comedogenesis, bacterial colonization and induction of inflammation.

Acne is a disease of the pilosebaceous follicle, induced by androgens of adrenal and gonadal origin: its onset correlates with the increase in sebaceous production triggered by this hormonal stimulus. The pilosebaceous unit is the target organ of acne, explained by the distribution of lesions in the areas with the highest concentration.

The factors involved in its development include: increased sebaceous secretion and follicular epidermal hyperproliferation, which leads to the formation of: comedones (comedogenesis), bacterial colonization by Cutibacterium acnes (formerly named Propionibacterium acnes) and induction of inflammation(6).

The onset and persistence of the activity of the sebaceous glands is mainly due to the action of androgens. After its activity in the postnatal period, due to the maternal hormonal influence, sebaceous glands remain minimized until puberty, when the size and number of lobes per gland increases, as a result of the androgenic stimulus (adrenarche). The presence of this glandular activity is a necessary requirement for the development of acne. There is a greater sebaceous secretion and, in addition, a qualitative alteration. These events have been related to the development of hypercornification of the sebaceous duct and changes in surface microorganisms. The consequence of hypercornification of the sebaceous duct is microcomedone: keratinocytes are grouped in dense clumps with monofilaments and lipid droplets, retaining the secreted sebum that distends the channel and the gland. This microcomedone is the primary acne lesion.

Acne is not an infectious process, but there are microorganisms that colonize and multiply in the follicular duct, and that can play a role in the pathogenesis. Cutibacterium acnes (C. acnes), which predominates in areas rich in sebaceous glands, can act as an opportunistic pathogen in acne. C. acnes is scantily present on the skin surface, whereas it is the dominant resident in the pilosebaceous unit; and the development of acne would be related, not with its proliferation, but with the selection of certain types, especially the IA1 phylotype, in a medium with increased sebaceous secretion in addition to balance alteration of the skin microbiome(7,8). Also, the formation of biofilm, an organized conglomerate of bacteria enhancing their survival, could increase its pathogenicity and resistance to antibiotics. These data open up new therapeutic possibilities in the management of acne (probiotics, anti-biofilm compounds…)(9).

The inflammation would not be caused by the presence of bacteria in the dermis, but by the action of biologically active mediators produced by C. acnes, which diffuse from the follicular channel and, later, by an inflammatory reaction to a foreign body triggered by the rupture of the ductal wall. The cell wall of C. acnes contains a carbohydrate antigen that stimulates the development of antibodies, which facilitate the inflammatory response.

There are multiple factors related to acne exacerbation episodes. Various studies postulate that diet could be considered a stimulating factor for acne, since foods that are high in sugar and other carbohydrates, dairy products or proteins, would affect serum insulin and insulin-like growth factor (IGF-1), which would induce an increase in the production of available androgens and the development of acne(3). However, currently, there is no scientific evidence with controlled studies to justify the restriction of specific foods(10).

The exacerbation of lesions in stressful situations is a known fact, in relation to the neuroendocrine regulation of sebocytes, the increase in adrenal secretion and the manipulation of lesions. Conversely, acne has an undoubted psychosocial impact, with consequent repercussions on quality of life.

About 70% of patients report a premenstrual acne exacerbation. This has been related to: increased hydration of the pilosebaceous duct, progressive decrease in estrogen levels with anti-inflammatory action and increased progesterone in this phase, with androgenic and pro-inflammatory effects.

70% of patients show acne improvement with sun exposure during the summer months. This amelioration could be potentially explained by the “camouflage” effect caused by tanning and an anti-inflammatory action of the immune suppression induced by ultraviolet light(5). This is the theoretical foundation of phototherapy for acne. However, ultraviolet radiation can also increase the comedogenic effect of sebaceous secretion and cause significant exacerbations of acne (“acne Mallorca”, acne aestivalis, tropical acne).

Activities in humid climates and occlusion determine acne worsening in up to 15% of patients. It could be due to a ductal hydration mechanism that would favor the obstruction. This is the mechanism that explains the exacerbation of acne lesions produced by the use of protective masks during the SARS-CoV-2 pandemic (“Mask acne”).

Various drugs, such as anabolic steroids or contraceptives containing progestogens with androgenic action, can exacerbate acne. In addition, there are numerous treatments such as: oral corticosteroids, isoniazid, lithium or certain anti-cancer drugs, capable of inducing acneiform eruptions.

The use of high lipid-based cosmetic formulations, aggressive cleaners or alkaline soaps can: alter the skin barrier, promote the formation of comedones and induce inflammation.

The term exposome defines the assorted environmental factors that influence the development and severity of a disease, in this case acne. Identifying the negative exposome factors can help reduce its impact and manage the disease(11).

Clinical manifestations

Acne is an inflammatory skin disease with two main characteristics: lesional polymorphism and chronic course.

In addition to seborrhea, open and/or closed comedones and inflammatory lesions are observed such as: papules, pustules and nodules, as well as residual lesions (scars and pigmentation alterations).

Regarding the distribution of the lesions, almost all the patients (99%) present facial localization, accompanied in more than half of the cases, by involvement of the back (60%) and, to a lesser extent, the pectoral area (15%). The majority of patients will refer to a gradual onset of the lesions around puberty, so that in cases in whom a sudden onset of these is described, an underlying cause must be ruled out. In women with severe, rapid-onset acne associated with hirsutism or menstrual irregularities, the existence of endocrinological pathology (hyperandrogenism) must be excluded.

Types of lesions

Non-inflammatory lesions

Closed comedones are small firm papules with a whitish surface in which, occasionally, the follicular opening is observed. Alternatively, open comedones present a blackish central plug, due to the deposit of melanin and the oxidation of sebum (Fig. 1).

Acné - J.M. Azaña Defez, M.L. Martínez Martínez

Figure 1. Comedonian acne: open and closed comedones (“blackheads”).

Inflammatory lesions

They derive from the previous ones and include:

Superficial lesions: papules and pustules (Fig. 2).

Acné - J.M. Azaña Defez, M.L. Martínez Martínez

Figure 2. Mild-moderate inflammatory acne: comedones, papules and pustules.

Deep lesions: deep pustules and nodules, when the inflammation affects the entire follicle. Nodules are firm, painful lesions that can be larger than 1 cm. They cons­titute the characteristic lesion of what is known as acne conglobata. Although epidermoid cysts can be seen in patients with acne, in most cases, the so-called “cysts” are not true cyst, but rather deep nodules (nodular acne).

Residual injuries

Residual lesions are observed in up to 90% of patients, although they significant in only 22% of cases are. Inflammatory lesions can leave sequelae such as erythematous macules that can persist for months, scars of various characteristics (depressed, varioliform, ice pick, hypertrophic, keloid) and dyschromias (post-inflammatory hyperpigmented and hypopigmented macules).

Types of acne

Based on the clinical presentation and the predominance of some of these lesions, classic clinical types of acne are distinguished: comedonian, papule-pustular and nodular. Acne conglobata is a severe and treatment-resistant form, more frequently found in men and with a more intense location on the trunk. It is characterized by deep papules and painful nodules, which can converge and form sinusoidal paths, and evolve to form depressed and keloid scars. Acne fulminans or malignant acne, initially described as a form of acne conglobata (“acute febrile ulcerative acne conglobata”), is a rare variant in which a systemic immune reaction against C. acnes takes place(12). It is observed especially in young men who, suddenly, develop inflammatory lesions predominantly in the trunk, with systemic manifestations (fever, anorexia, polyarthropathy, splenomegaly, aseptic osteolysis), together with marked leukocytosis. Blood cultures are sterile. Sometimes it can be induced by drugs (isotretinoin or androgens) and intercurrent infections (Epstein-Barr virus). In terms of treatment, in addition to that indicated for acne, the use of oral corticosteroids is required.

Special forms of acne of interest in Pediatrics

Infantile acne

It has been associated with transplacental stimulation of adrenal androgen production, at least in cases of onset before the first year of life. There are no other manifestations of hyperandrogenism, because sebaceous glands are the only ones capable of converting dehydroepiandrosterone to androstenedione and testosterone. Occasionally, drugs such as pheny­toin, corticosteroids and halogens have been implicated.

It prevails in males, usually as a localized form, with especial involvement of the cheeks. In the neonatal period, the main differential diagnosis must be established with benign cephalic pustulosis, which is much more frequent (20% of neonates), with onset within the first weeks of life, consisting of papulopustular lesions on the cheeks, but without comedones and associated with colonization by Malassezia (M. sympodialis and M. furfur).

The treatment of infantile acne is similar to that of acne vulgaris (see below), but excluding oral tetracyclines. Only exceptionally has oral isotretinoin been used in this form(13).

Excoriated acne

Although it is more frequent in young women, it can also be diagnosed in adolescence in patients who manipulate the lesions, causing erosions or ulcerations with the risk of superinfection and scarring. The most severe cases can hide relevant psychological disorders that will require specialized evaluation(14).

Drug-induced acne

It is not a true acne as it is a monomorphic process without comedones, which is why it is referred to as drug-induced acneiform eruptions. Its onset is chronologically related to the start of the treatment involved. Many drugs can be involved in its appearance, the most common being corticosteroids (Fig. 3), but also: antiepileptic drugs, antituberculosis drugs, lithium, vitamin B, halogenated compounds (iodides or bromides) and epidermal growth factor receptor inhibitors, among others.

Acné - J.M. Azaña Defez, M.L. Martínez Martínez

Figure 3. Monomorphic drug acne (oral corticosteroids).

Endocrine acne

The term is used in those women who besides acne associate other manifestations of hyperandrogenism, and where polycystic ovary syndrome is the most frequent condition. SAHA syndrome (Seborrhea, Acne, Hirsutism and Alopecia) is an acronym for the main manifestations of this dermatological androgenization entity(15). Hormonal investigations are necessary for its diagnosis and management.

Acne induced by topical substances

It includes different clinical forms, notably cosmetic acne, which predominates in women and is related to the use of comedogenic cosmetics. Also, excessive washing as an attempt to improve acne can aggravate it (detergent acne).

Mechanical acne

It refers to the appearance of acne lesions in areas subjected to prolonged friction. Continuous irritation of the superficial area of the pilosebaceous duct and excessive hydration of the area due to occlusion, would be the factors involved in its appearance. The distribution of lesions and the mechanical history facilitates the diagnosis. A current example would be acne exacerbated or triggered by the use of protective face masks(16), which was interestingly already referred to in Asian literature in a previous coronavirus epidemic(17).

Hidradenitis suppurativa

It is an inflammatory process with a chronic and recurrent course, which predominantly affects: armpits, groin and anogenital area, sometimes associated with severe inflammatory acne with which, as mentioned previously, it shares physiopathogenic mechanisms(18). It usually begins during puberty, although there are childhood cases and its etiopathogenesis involves familial, endocrine factors (obesity, hyperandrogenism) and local irritation(18). It is characterized by: comedonal lesions, papules, pustules, and painful nodules that converge into large abscesses with sinus discharge and a tendency to cicatrize in the areas previously described.

Diagnosis

The diagnosis is based on clinical examination. The use of scales to determine the predominant type of lesion and its severity will guide the treatment.

The diagnosis of acne is a clinical one, based on the presence of comedones and/or inflammatory lesions. Examination with Wood’s light will allow to observe the presence of C. acnes in the comedones, as it emits a red-orange fluorescence due to the production of porphyrins (Fig. 4). Only in case of suspected endocrine acne, will complementary examinations be carried out to rule out hyperandrogenism.

Acné - J.M. Azaña Defez, M.L. Martínez Martínez

Figure 4. Orange follicular fluorescence: relationship with porphyrins produced by C acnes. This indicates that the patient does not undergo treatment or that the microorganism is resistant.

In addition to the predominant type of lesion and its location, there are multiple systems for assessing the severity of acne(19,20), although they are of limited use in daily practice. The simplest is a qualitative classification, taking into account the type of dominant lesion(1) (Table I), which can be completed according to the severity (mild, moderate, severe).

Treatment

The treatment of choice will be combined topical products containing antibiotics, retinoids, or benzoyl peroxide; while, in case of non-response or moderate-severe acne, oral treatment with antibiotics or isotretinoin will be prescribed.

Basic concepts

The treatment of acne should begin by knowing the clinical history and response to other drugs that have been administered to the patient, in order to design an individualized plan for maximum effectiveness. Low adherence to treatment sometimes conditions results not turning out as expected, hence it is essential to indicate and explain some of the most relevant aspects of this, specifically, on the progress of the process, which may require treatment during months or years.

Compliance improvement is increased by using topical products with fixed combinations, rather than multiple products separately. Combination products have two advantages: on the one hand, they act against several pathogenic factors of acne simultaneously and, on the other, they simplify the therapeutic regimen. Patients should be informed that clinical improvement will not be achieved immediately, but rather after 6-8 weeks of treatment.

In order to minimize irritation from topical treatments, it should be indicated that the initial application can be gradual to improve tolerance during the first few weeks. We must instruct the patient in the use of non-comedogenic moisturizers, along with the avoidance of products that contribute to the development of acne. The use of a mild daily wash soap is advisable, while excessive hygiene can alter the skin barrier and increase the irritation potential of the treatment.

In the follow-up clinics, the therapeutic plan should be reinforced and inquire the presence of possible triggers or aggravating factors. In patients refractory to treatment, folliculitis due to gram-negative organisms should be excluded, especially in patients receiving continuous antibiotic treatment. Regarding diet, as mentioned above, it is not necessary to make dietary restrictions, except in case the patient relates acne appearance to certain foods or in case of deterioration with the consumption of abundant skimmed milk, or foods with a high sugar load.

The main objective of acne treatment is to avoid the appearance of scars, so the treatment must be as early as possible, with treatment modality appropriate to the severity and extension. Table II shows a therapeutic algorithm(1) to be followed in patients with acne. In general, the use of an oral or topical antibiotic in monotherapy should be avoided, in order to reduce the risk of bacterial resistance.

Topical treatment

Topical treatment will be prescribed in all patients with acne, in monotherapy in mild acne and, combined with systemic treatment, in cases of moderate or severe acne. In addition, after systemic treatment, maintenance topical retinoids should be applied. The patient should be insisted on applying it not only to the lesions, but also to the areas susceptible of presenting them, so as to avoid their appearance.

Retinoids

Topical retinoids prevent the formation of comedones and inflammatory lesions, normalize the desquamation of keratinocytes, and have an anti-inflammatory effect. They include: retinoic acid, isotretinoin, adapalene, tazarotene (in Spain this is indicated for the treatment of psoriasis) and, recently, tripharo­tene. The latter has been approved as a 0.005% cream, being a fourth-generation retinoid for the treatment of acne vulgaris on the face and trunk, from 12 years of age onwards. Tripharotene selectively targets gamma retinoic acid (RAR-γ) receptors, the most common ones in the skin(21).

Topical retinoids also enhance the penetration of associated topical antimicrobials, thereby increasing their efficacy. They are used as monothera­­py for comedonal acne and as part of a combination therapy for mild-mode­rate papule-pustular acne(22). They achieve a 40-70% reduction in come­dones and inflammatory lesions(23). They are also recommended as a maintenance treatment to prevent recurrences. Different concentrations are used, depending on severity and clinical tolerance, in aqueous creams or gels. The recommendation is to start with low concentrations or short application times, and gradually increase according to tolerance.

The most frequent side effects are: irritant dermatitis and photosensitivity. In 20% of patients, a transient increase in inflammatory lesions may occur. It is contraindicated in pregnancy due to its proven teratogenic properties with oral administration.

Benzoyl peroxide

It is an antimicrobial agent with anti-inflammatory and comedolytic activity, its main action being the neutralization of C. acnes in the hair follicles, thus achieving a bacteriostatic and possibly bactericidal effect, similar to topical antibiotics and without being associated with antimicrobial resistance. It is marketed in different concentrations (from 2.5% to 10%) and galenic forms (creams, gels, cleansers), alone or in combination with other active ingredients.

Its main side effects are irritation, in addition to discoloring dark clothes or hair. Tolerance increases if applied not immediately after the skin wash, but minutes later. It can be used during pregnancy and lactation.

Topical antibiotics

They have antibacterial action, inhibiting the growth and activity of C. acnes and a direct and indirect anti-inflammatory effect. They are used as part of combination treatments for mild-moderate papulopustular acne.

The most frequently used are clindamycin and erythromycin, in concentrations of 1-4%. Nadifloxacin 1% is a topical quinolone that also appears to have certain antiandrogenic action in vitro, in addition to inhibiting the activation of T cells and keratinocytes.

Monotherapy use is not recommended, due to the possible development of resistance to antibiotics and the slower onset of action. In the case of macrolides, resistances greater than 50% of strains of C. acnes have been reported in some countries(24). For this reason, they should be discontinued once improvement is appreciated and, in case of ineffectiveness after 6 to 8 weeks, when another treatment should be considered. The combination of topical antibiotic with topical retinoid or benzoyl peroxide reduces the possibility of resistance(22).

Side effects are less frequent compared to those of oral antibiotics and generally mild and local (pruritus, xerosis). Pseudomembranous colitis is a rare complication associated with topical clindamycin. Nadifloxacin should not be used in children under 14 years of age, as it is a fluoroquinolone.

Azelaic acid

Azelaic acid is a dicarboxylic acid that exhibits antimicrobial and anti-comedogenic activity(25). It is used for comedonal and inflammatory acne. It is available as 20% cream and 15% gel, and its main side effect is mild irritation. It can be used in pregnancy and lactation.

Other topical treatments

Salicylic acid is used as a comedolytic and antibacterial. It is used in various galenic presentations at 0.5-2%, in non-inflammatory forms, being less effective than topical retinoids. Its main side effects are: erythema and scaling.

The α-hydroxy acids are used in the comedonian forms at various concentrations and, in general, with good tolerance. The most used one is glycolic acid, which can be found in presentations combined with tretinoin and topical clindamycin.

Niacinamide is the active form of vitamin B3, with anti-inflammatory properties. It has been used as 4% hydroalcoholic gel in the treatment of mild acne.

Combined treatment

Combinations with retinoids are recommended as a first line of treatment for mild-moderate and papulopustular acne. Combinations of various therapeutic agents allow to target multiple pathogenic factors of acne. On the other hand, the combination of antibiotics with benzoyl peroxide reduces the possibility of bacterial resistance. Marketed fixed-dose combinations include the following:

0.1% adapalene plus 5% benzoyl peroxide, applied daily as a gel.

Clindamycin 1% plus benzoyl peroxide 5% gel, which is more effective than both products used separately.

0.025% tretinoin plus 1% clindamycin gel.

0.02% tretinoin plus 4% glycolic acid, plus 0.8% clindamycin gel.

Systemic treatment

Indicated in patients with moderate-severe acne or in the absence of response to topical treatment in mild or mild-moderate acne. Also indicated in cases of extensive skin involvement.

Hormonal treatment

Most combined contraceptives (estrogen + progestogen) have the ability to improve acne and hirsutism to a greater or lesser extent, which is why they are useful in the patient with hyperandrogenism, but it is also an effective treatment in women with acne, regardless of androgen serum concentrations. Contraceptives improve: seborrhea, androgenic alopecia, SAHA syndrome and late-onset acne, in addition to regulating menstrual disturbances. Its main objective is to offset the effect of androgens on the sebaceous glands.

Its main indications in patients with acne are: failure of antibiotic treatment, when oral isotretinoin is contraindicated or inappropriate, and if, in addition to acne, it is necessary to control the menstrual cycle or as a contraceptive method. Better results seem to be obtained, even with normal serum androgen levels, in women with predominantly inflammatory lesions on the lower half of the face and neck who frequently present with premenstrual exacerbations of their acne.

Thrombosis is one of the most serious side effects of oral contraceptives which depends, over all, on the estrogens used and their dose. In order to reduce this side effect, the oral contraceptive of choice should be one that combines 30 µg or less of ethinyl estradiol (estrogen) with gestagen. If our aim is to treat signs of androgenization, the oral contraceptives of choice would be those with progestogens with an antiandrogenic effect: cyproterone acetate, chlormadinone acetate, drospirenone, dienogest, and nomegestrol acetate(26). According to a meta-analysis, cyproterone acetate and drospirenone were found to be the most effective progestogens in treating acne(27). The response is observed from the third month of treatment, and it should be maintained for a year, in the absence of side effects, with risk of recurrence upon discontinuation, thus its association to other treatments.

Oral antibiotics

They are used in moderate-severe inflammatory acne. They produce an anti-inflammatory action through the inhibition of the growth of C. acnes, in addition to reducing the amount of free fatty acids and, thus, their irritating effect. The most used ones are: oral tetracyclines (doxycycline, minocycline) and macrolides (erythromycin, azithromycin and josamycin). Doxycycline is the most widely used one, with doses ranging between 50-100 mg per day, the most frequent side effect being photosensitivity. They are generally well tolerated, and serious side effects are rare. Tetracyclines should not be used in children under 8 years of age, due to their effects on the developing skeleton and dentition.

Treatments are usually maintained for at least a month and a half, because as with other treatments, its clinical effect takes weeks to be appreciated, reducing the dose or suspending treatment once the appearance of inflammatory lesions diminishes, usually after 3 months.

They should be avoided as monotherapy, associating topical retinoids or benzoyl peroxide, if necessary: repeat treatment, use the same antibiotic if it was effective, and avoid the simultaneous use of topical and oral antibiotics, so as not to favor the appearance of resistance.

Oral isotretinoin

13-cis-retinoic acid of vitamin A or isotretinoin is indicated in severe nodule-cystic acne, refractory to other treatments or that can lead to scarring. It constitutes the therapeutic pillar of severe acne, but also for acne that has an important impact on the quality of life of the patient: inflammatory acne resistant to conventional treatment and chronic forms of recurrent tendency, in addition to gram-negative folliculitis, facial pyoderma and severe forms of rosacea.

It acts on all the factors involved in the pathophysiology of acne.

It is usually administered at doses of 0.5-0.6 mg/kg per day (from 0.1 to 2 mg/kg/day), maintaining the treatment until reaching a total dose of 120 to 150 mg/kg, so as to reduce the possibility of relapses. Doses can be modified based on clinical response and side effects. It should be taken with meals to enhance its absorption. Low-dose isotretinoin regimens (0.25-0.4 mg/kg/day) are effective, with fewer side effects, but higher recurrence rates(28).

The clinical response is excellent (Fig. 5), most of the cases responding to a single 6-month course, although, in general, the results are not evident until 1-2 months after starting the treatment, a period in which even a certain exacerbation can be observed.

Acné - J.M. Azaña Defez, M.L. Martínez Martínez

Figure 5. Deep inflammatory acne: before and after treatment with isotretinoin.

Side effects depend on the administered dose and, in general, are similar to those of hypervitaminosis A. Almost all patients present with cheilitis and, more than 50%, cutaneous and mucosal xerosis. Xerophthalmia, alteration of night vision, conjunctivitis, keratitis, headache and epistaxis can also be observed. Myalgias can affect 15% of patients, being the most frequent musculoskeletal manifestations. Rarely, long-term hyperostosis or osteoporosis has been described, hence radiological studies are not indicated in standard treatments. Exceptionally, an association with inflammatory bowel disease has been described, without finding, to date, an increased risk in patients treated with isotretinoin(29). The relationship with diabetes has been studied, identifying that treatment with isotretinoin significantly increases the level of serum adiponectin, but does not alter the state of insulin resistance in patients with acne(30). Its concomitant administration with tetracyclines is contraindicated, due to the increased risk of benign intracranial hypertension.

Digestive symptoms are rare. More frequent is the elevation of transaminases that can appear in 15% of the cases. In addition, triglycerides can increase in up to 25% of patients, especially in the first month, and they tend to decrease when the dose decreases. Triglyceride concentrations beyond 700-800 mg/dl is a criterion for stopping treatment. Full blood count can show: anemia, leukopenia, thrombocytosis or thrombopenia, and elevated ESR.

Psychiatric side effects such as: increased risk of depression, suicide, psychosis, and aggressive and violent behaviors have been described, although the causal relationship or mechanism of action remains unclear. A prevalence of depression of 1-11% has been described in patients receiving isotretinoin, a similar percentage to that observed in patients receiving oral antibiotic treatment, so it cannot be established as a cause(31). Despite this, the majority of patients experience improvement in the psychosocial repercussions related to acne, once treatment has begun. In any case, it is important that the patient and his family are aware of these data, in addition to identifying patients at risk and the possible appearance of any related symptoms. In case of severe headache, abnormal night vision or psychiatric manifestations, isotretinoin treatment must be stopped.

Isotretinoin is teratogenic, producing its maximum effect in the third week of gestation, so women of childbearing age should not start treatment until they have a negative pregnancy test, which should be repeated monthly. It is essential to recommend contraceptive methods from 1 month before to 1 month after the end of treatment. Patients must be adequately informed and a specific informed consent document should be completed.

The patient should be informed of the therapeutic effects of isotretinoin and taught to control or prevent side effects (adequate hydration, artificial tears to control dry eyes and nose, avoid irritants, elude alcoholic beverages, photoprotection, avert blood donation during the treatment…).

Recurrence of acne is not uncommon, in most patients with a good response to conventional therapy, but in some cases, a new cycle of treatment with isotretinoin must be indicated. It usually happens in the first year after treatment, and is rare after 3 years. Factors associated with risk of recurrence include: a low daily dose of isotretinoin (0.1-0.5 mg/kg) or not reaching a certain total dose (120-150 mg/kg), severe or prolonged acne, women of more than 25 years at the onset of treatment, endocrine abnormalities, patients under 16 years of age and acne located on the trunk. Maintenance treatment with topical retinoids can reduce recurrence risk by avoiding the formation of microcomedones.

The investigations to be carried out prior to prescribing the treatment include: full blood count, liver function tests, triglyceride concentrations and a urine pregnancy test in the case of females. The next check-up, including liver function and lipid profile, should take place after a month; if everything remains within normality, it is not necessary to repeat them, as long as we are dealing with a patient on usual doses and without other underlying pathology(32-33).

In conclusion, it is fair to say that isotretinoin is an effective medication, which can achieve the definitive “cure” in a high percentage of cases, with a comfortable administration and known dosage, and with controllable side effects which, in general, are well tolerated.

Other treatments

Phototherapy

Phototherapy targets C. acnes, a producer of porphyrins, especially coproporphyrin III. These porphyrins can be activated by light, at specific wavelengths (blue light at 415 nm and mixed blue-red light at 415 and 660 nm), producing a photodynamic effect capable of destroying these bacteria. It has been used in mild or moderate forms of inflammatory acne, with variable response.

Photodynamic therapy

The combination of topical aminolevulinic acid or methylaminolevulinate, metabolized to protoporphyrin IX in the pilosebaceous unit, a powerful photosensitizer, and the subsequent activation by a light source with a specific wavelength, would lead to: reduction of sebaceous secretion, destruction of C. acnes and also decreased ductal hyperkeratinization.

Intense pulsed light and laser

The pulsed dye laser at 585 nm, the neodymium: YAG at 1320 nm and the 1450 nm diode laser have been used in inflammatory acne, but with transient improvement, which together with the discomfort that it generates, makes it of limited performance.

Zinc

It does not improve comedones, but it does show some efficacy in treating inflammatory acne. Its action is due to: inhibition of neutrophil chemotaxis, inhibition of 5α-reductase and tumor necrosis factor. Oral route (200 mg/day, administered outside of meals) has been indicated. Its main side effects are gastrointestinal (nausea, vomiting, epigastric pain).

Corticosteroids

Its use is associated with the appearance of acne lesions (corticoid drug acne); however, they may be indicated in certain cases. Thus, in severe inflammatory forms, liable to exacerbation at the beginning of treatment with isotretinoin, a short course of oral corticosteroids rapidly reduces the number of lesions and the risk of exacerbating them with retinoid. Similarly, intralesional injection of corticosteroids may be useful in large, recent-onset inflammatory lesions.

Adjuvant treatment

Comedone removal

This technique achieves immediate improvement, which must be maintained with topical retinoids that prevent the formation of new microcomedones. Macrocomedones are a cause of therapeutic failure and do not usually respond to treatment with topical or oral retinoids; hence the need to eliminate them.

Chemical exfoliation (“chemical peels”)

It is indicated once acne is controlled, for the treatment of superficial scars or residual hyperpigmentation. The following are used: α-hydroxy acids (especially glycolic acid), trichloroacetic acid or salicylic acid.

Cryotherapy

Cryotherapy of liquid nitrogen and spray applicators has been used, especially in scar lesions.

Surgery

Surgery can be performed in case of scar lesions. It would include: removal of depressed scars (“lift technique”), classic dermabrasion, the use of filling material in deep scars with a non-fibrotic base or laser therapy.

Role of the Primary Care pediatrician

Acne in the majority of patients can be managed in the Primary Care setting. A correct treatment includes: early onset and appropriate therapy to the type of acne, use of combined treatments, never use antibiotics in monotherapy nor associate topical and oral antibiotics and, in the follow-up, reinforce the therapeutic plan and assess the response and tolerance to the treatment. Referral to the dermatologist would be considered in the following cases:

Severe forms of acne.

Moderate forms without response to prescribed topical and/or oral treatments.

Patients with important psychosocial repercussions of the disease.

Suspicion of an associated underlying endocrine disorder.

Although rare, differential diagnosis with other processes.

Finally, whenever treatment with isotretinoin is considered, as it is a drug that requires prescription by a specialist and a medical inspector visa. In these cases, the follow-up will be coordinated with the specialist.

Bibliography

The asterisks show the interest of the article in the opinion of the authors.

1.*** Zaenglein AL, Pathy AL, Schlosser BJ, Alikhan A, Baldwin HE, Berson DS, et al. Guidelines of care for the management of acne vulgaris. J Am Acad Dermatol. 2016; 74: 945-73.

2. Guerra A. Estudio epidemiológico descriptivo transversal sobre la prevalencia del acné en la población adolescente española. Act Dermatol. 2001; 11: 1-6.

3. Heng AHS, Chew FT. Systematic review of the epidemiology of acne vulgaris. Sci Rep. 2020; 10: 5754.

4. Lichtenberger R, Simpson MA, Smith C, Barker J, Navarini AA. Genetic architecture of acne vulgaris. J Eur Acad Dermatol Venereol. 2017; 31: 1978-90.

5. González-Cantero A, Arias-Santiago S, Buendía-Eisman A, Molina-Leyva A, Gilaberte Y, Fernández-Crehuet P, et al. ¿Existe variación en los diagnósticos dermatológicos entre la temporada de frío vs calor? Un subanálisis del estudio DIADERM (España 2016). Actas Dermosifiliogr. 2019; 110: 734-43.

6. Zaenglein AL. Acne vulgaris. N Engl J Med. 2018; 379: 1343-52.

7. Scholz CF, Kilian M. The natural history of cutaneous propionibacteria, and reclassification of selected species within the genus Propionibacterium to the proposed novel genera Acidipropionibacterium gen.nov., Cutibacterium gen.nov. and Propionibacterium gen.nov. Int J Syst Evol Microbiol. 2016; 66: 4422-32.

8.*** Dréno B, Pécastaings S, Corvec S, Veraldi S, Kharmari A, Roques C. Cutibacterium acnes (Propionibacterium acnes) and acne vulgaris: a brief look at the latest updates. J Eur Acad Dermato Venereol. 2018; 32: 5-14.

9. Pécastaings S, Roques C, Nocera Th, Peraud C, Mengeaud V, Khamari A, et al. Characterisation of Cutibacterium acnes phylotypes in acne and in vivo exploratory evaluation of Myrtacine. J Eur Acad Dermatol Venereol. 2018; 32: 15-23.

10. Claudel JP, Auffret N, Leccia MT, Poli F, Dréno B. Acne and nutrition: hypothesis, myths and facts. J Eur Acad Dermatol. 2018: 32: 1631-7.

11. Dréno B, Bettoli V, Araviiskaia E, Sánchez Viera M, Bouloc A. The influence of exposome on acne. J Eur Acad Dermatol. 2018; 32: 812-9.

12. Bocquet-Trémoureux S, Corvec S, Khammari A, Daqniele MA, Boisrobert A, Drenó B. Acne Fulminans and Cutibacterium acnes phylotypes. J Eur Acad Dermatol. 2020; 34: 827-33.

13. Miller IM Echeverria B, Torrelo A, Jemec GB. Infantile acne treated with oral isotretinoin. Pediatr Dermatol. 2013; 30: 513-18.

14. Anzengruber F, Ruhwinkel K, Ghosh A, Klaghofer R, Lang UE, Navarini AA. Wide range of age of onset and low referral rates to psychiatry in a large cohort of acne excoriée at a Swiss tertiary hospital. J Dermatol Treat. 2018; 29: 277-80.

15. Orfanos CE, Adler YD, Zouboulis CC. The SAHA syndrome. Horm Res. 2000; 54: 251-8.

16. Han C, Shi J, Chen Y, Zhang Z. Increased flare of acne caused by long-time mask wearing during COVID-19 pandemic among general population. Dermatol Ther. 2020; 29: e13704.

17. Tan KT, Greaves MW. N95 acne. Int J Dermatol. 2004; 43: 522-3.

18. Bandera A, de Lucas R. Aspectos epidemiológicos, clínicos y terapéuticos en situaciones especiales: hidradenitis supurativa infantil. Actas Dermosifiliogr. 2016; 107: 51-60.

19. Puig L, Guerra-Tapia A, Conejo-Mir J, Toribio J, Berasategui C, Zsolt I. Validation of the Spanish Acne Severity Scale (Escala de Gravedad del Acné Española – EGAE). Eur J Dermatol. 2013; 23: 233-40.

20. 0’Brien SC, Lewis JB, Cunlife WJ. The Leeds revised acne grading system. J Dermatol Treat. 1998; 9: 215-20.

21. Scott LJ. Trifarotene: First Approval. Drugs. 2019; 79: 1905-9

22. Thiboutot D, Gollnick H, Bettoli V, Dréno B, Kang S, Leyden JJ, et al. Global Alliance to Improve Outcomes in Acne. New insights into the management of acne: an update from the Global Alliance to Improve Outcomes in Acne group. J Am Acad Dermatol. 2009; 60: S1-50.

23. Haider A, Shaw JC. Treatment of acne vulgaris. JAMA. 2004; 292: 726-35.

24. Walsh TR, Efthimiou J, Dréno B. Systematic review of antibiotic resistance in acne: an increasing topical and oral threat. Lancet Infect Dis. 2016; 16: e23-33.

25. Williams HC, Dellavalle RP, Garner S. Acne vulgaris. Lancet. 2012; 379: 361-72.

26. Requena C, Llombart B. Oral Contraceptives in Dermatology. Actas Dermosifiliogr. 2020; 111: 351-6.

27. Arowojolu AO, Gallo MF, López LM, Grimes DA. Combined oral contraceptive pills for treatment of acne. Cochrane Database Syst Rev. 2012; 7: CD004425.

28. Lee JW, Yoo KH, Park KY, Han TY, Li K, Seo SJ, et al. Effectiveness of conventional, low-dose and intermittent oral isotretinoin in the treatment of acne: a randomized, controlled comparative study. Br J Dermatol. 2011; 164: 1369-75.

29. Lee SY, Jamal MM, Nguyen ET, Bechtold ML, Nguyen DL. Does exposure to isotretinoin increase the risk for the development of inflammatory bowel disease? A meta-analysis. Eur J Gastroenterol Hepatol. 2016; 28: 210-6.

30. Sai TY, Liu HW, Chao YC, Huang YC. Effects of isotretinoin on glucose metabolism in patients with acne: A systematic review and meta-analysis. J Dtsch Dermatol Ges. 2020; 18: 539-45.

31. Huang YC, Cheng YC. Isotretinoin treatment for acne and risk of depression: A systematic review and meta-analysis. J Am Acad Dermatol. 2017; 76: 1068-76.e9.

32. Lee YH, Scharnitz TP, Muscat J, Chen A, Gupta-Elera G, Kirby JS. Laboratory Monitoring during Isotretinoin Therapy for Acne: A Systematic Review and Meta-analysis. JAMA Dermatol. 2016; 152: 3544.

33. Barbieri JS, Shin DB, Wang S, Margo­lis DJ, Takeshita J. The clinical utility of laboratory monitoring during isotretinoin therapy for acne and changes to monitoring practices over time. J Am Acad Dermatol. 2020; 82: 72-9.

34. de Lucas Laguna R. Acné. Pediatr Integral. 2016; XX(4): 227-33.

Recommended bibliography

– Zaenglein AL, Pathy AL, Schlosser BJ, Alikhan A, Baldwin HE, Berson DS, et al. Guidelines of care for the management of acne vulgaris. J Am Acad Dermatol. 2016; 74: 945-73.

Interesting article describing the current recommendations in the management of acne. They highlight the importance of topical retinoid in all patients with mild-moderate acne, combined either with benzoyl peroxide or topical antibiotic, always avoiding the latter in monotherapy, in order to reduce the risk of bacterial resistance. For moderate-severe acne, oral antibiotic treatment should be added, or in case of lack of response to it, oral isotretinoin in monotherapy should be started.

– Dréno B, Pécastaings S, Corvec S, Veraldi S, Kharmari A, Roques C. Cutibacterium acnes (Propionibacterium acnes) and acne vulgaris: a brief look at the latest updates. J Eur Acad Dermato Venereol. 2018; 32: 5-14.

The bacteria Cutibacterium acnes (formerly known as Propionibacterium acnes) is part of a healthy skin, however, it can also act as an opportunistic pathogen in the appearance of acne vulgaris. The novelties identified in the etiopathogenesis of acne place C. acnes in a different position than previously considered. The proliferation of C. acnes would not act as a trigger, since acne patients do not harbor more C. acnes in their follicles than normal individuals. Instead, the loss of microbial diversity in the skin, coupled with the activation of innate immunity, could lead to this chronic inflammatory condition.

Clinical case

17-year-old female patient, with no relevant personal history. She presents facial injuries since a few years, which have experienced aggravation in recent months. She had been diagnosed of acne, and received treatment with doxycycline in monotherapy and various topical treatments later, with poor response. She had her menarche at age 11; and associates menstrual irregularities. On examination she presents: open and closed comedones, papules, pustules and nodules on the cheeks and, to a lesser extent on the forehead; seborrhea and hirsutism in the examined areas (scored 10 points on the Ferriman / Gallwey scale).

Acné - J.M. Azaña Defez, M.L. Martínez Martínez

 

 

Atopic dermatitis and seborrheic dermatitis

T. Pozo Román*, B. Mínguez Rodríguez**
Topics on
Continuous Training


T. Pozo Román*, B. Mínguez Rodríguez**

*Lead of the Dermatology Unit at Río Hortega University Hospital. Valladolid. **Consultant in Pediatrics at San Joan de Deu Hospital. Barcelona

Abstract

Atopic dermatitis is a common and frequently familial inflammatory dermatosis, which usually appears during infancy or early childhood and is often associated with other atopic diseases such as asthma, allergic rhinoconjunctivitis, food allergies or eosinophilic esophagitis. It is a complex genetic disease with environmental influences, characterized by intense pruritus and a chronic course in flares.
In infants and young children, cheeks, scalp and extensor surfaces of the extremities are affected and in older children and adults the lesions are usually located in the flexion folds, or in specific locations such as the perioral area, eyelids, hands or feet. In most patients the disease disappears when puberty arrives, but it can also appear in adulthood.
Seborrheic dermatitis is a common inflammatory dermatosis, with an infantile and an adult clinical form. Lesions are located on the scalp, ears, face, central part of the chest and intertriginous areas. There is an etiological relationship with active sebaceous glands, alterations in sebum composition and the genus Malassezia (Pityrosporum).
Malassezia (Pityrosporum).

 

Key words: Atopic dermatitis; Eczema; Atopy; Seborrheic dermatitis; Malassezia (Pityrosporum).

Palabras clave: Dermatitis atópica; Eczema; Atopia; Dermatitis seborreica; Malassezia (Pityrosporum).

 

 

Pediatr Integral 2021; XXV (3): 119 EN – 127 EN

 


Atopic dermatitis and seborrheic dermatitis

Atopic dermatitis


Introducción/definición

Atopic dermatitis is characterized by flares of inflammatory, itchy lesions, with a characteristic distribution and personal and / or family history of atopy (allergic rhinoconjunctivitis, asthma, food allergies, etc.).

Atopic dermatitis (AD) and food allergy have a predilection for infants and young children; whereas, asthma prevails in older children and rhinoconjunctivitis predominates in adolescents. This characteristic age-dependent sequence is called “atopic march”, however, it does not always manifest, as these diseases may or may not appear, and do so simultaneously or at different ages.

It is more prevalent in children (10-20%) than in adults (1-3%) and, in 90% of cases, it appears in childhood (45% during the first 6 months of life and 60% before the first year of life). At least 50% of atopic children will continue to express certain manifestation of the disease during adolescence and 20% also in adult life(1,2).

Most of the cases can be considered mild, but 10% of the patients suffer a severe form which is more prevalent in the adult population. The prevalence of severe atopic dermatitis in adults in Spain is estimated to be 0.08%(3).

In the most severe forms, the sleep cycle is disturbed leading to irritability, which affects school and sport performance, self-esteem, social relationships, as well as routine and leisure activities.

On the other hand, AD, and especially severe AD, implies a significant economic expense, both direct (medical visits, pharmacological cost) and indirect costs (loss of school hours and productivity), and both at a personal level as well as for health systems. This emphasizes the need to evaluate its impact on the family environment and on the patients’ caregivers. Hence, it should be recognized as a “family” disease, rather than as an individual one and, as such, it must be evaluated(4).

Etiopathogenesis

A deficient skin barrier action, an abnormal immune response, alteration of the cutaneous microbiome and an important psychosomatic influence are the main etiopathogenic factors.

It is a complex genetic disease, with interactions between different genes and of these with the environment.

In patients with AD, the lesional skin and, to a lesser extent, the non-lesional skin, present a defective cutaneous barrier, with: increased transepidermal water loss, alteration of skin lipids, increased epidermal proliferation, reduction of Filaggrin expression, inflammation, and increased number of IgE receptors on Langerhans cells.

The uninjured skin also shows different immunological profiles to normal skin (17% more T cells than normal skin and increased expression of Th1, Th2 and Th22 lymphocytes). Antigens that cross the skin reach the ganglia (via dendritic cells) and stimulate Th2 response with the consequent increase in the production of IgE and several other mediators from various inflammatory and epidermal cells. Immunity mediated by the Th1 pathway (which is attenuated) and the innate immune system contribute to skin inflammation with the release of cytokines and a deficiency of antimicrobial peptides.

Patients with AD show important changes in skin microbiome, mainly involving a decrease in Staphylococcus Epidermidis and a dominant colonization of Staphylococcus Aureus (in up to 90% of the lesions), which correlates with the severity of the disease. Conversely, recovery of microbiome diversity precedes resolution of flare-ups.

All these abnormalities interact with each other; so that: barrier dysfunction alters the skin’s microbiome and immune response; the skin microbiome alters the immune response and the skin barrier; and, lastly, immune dysregulation also alters the cutaneous microbiome and the skin’s barrier function(5).

Manifestations and diagnosis

The diagnosis of AD is mainly clinical and remains based on the classical criteria described by Hanifin and Rajka(6): 3 or more major criteria, and 3 or more minor criteria (Table I).

The diagnosis of AD is primarily clinical (Figs. 1-5).

Figura 1. Atopic dermatitis in the acute phase.

Figura 2. Impetiginized eczema.

Figura 3. Retroauricular eczema with fissuring.

Figura 4. Labial and perioral eczema.

Figura 5. Chronic eczema from scratching (neurodermatitis).

Characteristically, the typical lesions of atopic eczema are erythematous areas of skin, often poorly defined, with intense itching, although their distribution and characteristics vary with age. There are also some characteristic clinical manifestations that must be known (Table II).

It is also important to distinguish between acute, subacute and chronic forms, as the topical treatment differs (Table II) and to identify other clinical manifestations of AD, which are very common (Table III).

Differential diagnosis

The symptoms of AD are highly characteristic, thus most of the possible differential diagnoses can be excluded by the medical history (scabies, seborrheic dermatitis, irritant contact dermatitis and ichthyosis).

In some cases, it may be necessary to perform biopsies (cutaneous T-cell lymphomas and psoriasis) or to perform contact tests (allergic contact or airborne dermatitis), or special studies (photosensitive diseases, diseases due to deficiencies of the immune system, erythroderma due to other causes, etc.). It is worth mentioning that the usefulness of atopy patch tests used in the last 20 years for the diagnosis of food and aeroallergens allergies is very limited, because although they are specific, they are not sensitive. The latter holds true especially in children with gastrointestinal symptoms related to food allergy and, in the case of aeroallergens that could trigger AD, there is insufficient data(7,8).

Assessment of AD severity

In recent years, the objective assessment of the severity of AD has become especially important in the daily clinic, with the appearance of new treatments for severe AD and their high cost, as the fundamental criterion for deciding: first, its administration (limited to severe forms and with lack of response to classical treatments); and, subsequently, the continuity of its administration.

There are multiple assessment scales for the disease and quality of life, which were used primarily in research and which have now been transferred to routine clinical practice, although as we have already mentioned, only for patients with severe AD clinical pictures(9 ) (Table IV).

Approach / Treatment

In the management of AD, the treatment of the disease is as important or more as the prevention of the factors that trigger the flare-ups.

Recommendations include: avoiding triggers, making a regular use of emollient creams and treat flare-ups, so as to contain subclinical inflammation and symptomatic exacerbations(9,10).

Prevention

In preventing flare-ups, the education of children and parents, although time consuming, is particularly important. A study of an educational program for children, showed that 97% of those who received education on atopic dermatitis, obtained a significant decrease in SCORAD (Scoring Atopic Dermatitis) (Table IV) after 6 months(11).

In addition to educating on the characteristics and likely progress of the disease along with the basic measures to prevent flares, it must be taken into account that, in our society where “everyone” has an opinion, there are many things that are unproven and therefore must be avoided. For example:

Only in patients with a proven food allergy, have exclusion diets proven certain degree of usefulness, although unable to fully control AD flare-ups. Only double-blind, placebo-controlled provocation tests are reliable. Allergy tests, RAST and prick tests, are positive in more than 40% of patients, without this involving clinical relevance. Epicutaneous tests to foods, which can be useful in patients with negative RAST and prick tests, although specific, they are not very sensitive.

It has been found that the exclusion of the most common allergens (cow’s milk, eggs, nuts, soy and fish) in high-risk children or in their mothers during pregnancy and lactation, decreases the prevalence of atopy during the first 2 years of life, although these differences are not sustained in the long term. These restrictive diets are very difficult to maintain overtime in children and can even turn out to be harmful (for instance, there is an increase in the frequency of allergy to peanut by delaying its introduction)(12). Some epidemiological studies have shown a significant association between food diversity given during the 1st year of life and protection against AD(13).

Exclusive breastfeeding is recommended up to 4 months of age, as a method to prevent against food allergy associated with AD, but with a C level of evidence; that is, only one notch above experts´ opinion (D). Also, in children at high risk (first-degree relative with allergic symptoms), if breastfeeding is not possible, the use of hypoallergenic milk formulas is recommended; in this case, with a somewhat higher level of evidence (B)(9).

Studies looking into the potential benefit of prebiotics and probiotics, remain currently inconclusive(14-16).

Treatments not recommended by the doctor should be avoided (as they have not been proven to be effective or safe), such as: montelukast (leukotriene antagonist), topical capsaicin, essential fatty acids (topical or oral), phytotherapy (Chinese herbs), acupuncture, autologous blood, bioresonance, homeopathy, massage therapy or aromatherapy, salt baths and balneotherapy, vitamins and minerals, vitamin B12 in avocado oil, etc.(10).

There is insufficient evidence to support that the use of non-sedating antihistamines reduces itching in patients with AD; however, they can be useful prior to exercise, because they reduce the itchiness caused by sweating. Similarly, sedative antihistamines, such as hydroxyzine, do not seem to control AD symptoms, however, by inducing sleep, they can improve rest and reduce scratching(17).

 Healthy skin care is essential in these patients given the alterations in their skin barrier and increased transepidermal water loss, therefore: showers or baths should be brief (5 minutes) and with lukewarm water; bathing only 1-2 times a week is not a good idea, because it increases the skin proliferation of Staphylococcus Aureus; mildly irritating soaps should be used (oatmeal, oil soaps, etc.) and limited in extension (to surfaces that could be dirtier); the use of daily moisturizers or oil baths is essential; and the causes of itching and / or flare-ups must be avoided, such as the use of wool clothing directly on the skin as well as clothing or footwear that promote sweating, as well as exposure to clinically relevant allergens (diagnosed in allergy tests ).

When relapses are frequent, it is useful to apply calcineurin inhibitors or topical corticosteroids (two / three times a week) between flares, in the areas usually affected by atopic dermatitis. The usefulness of twice weekly bleach baths is more doubtful (various studies have shown no differences with water-only baths) and there are no references to the usefulness of nasal mupirocin in AD(18).

Treatment

Anti-inflammatory treatment is necessary, even in subclinical inflammation, in order to restore the balance in the inflamed skin as soon as possible.

Although eczema flare-ups tend to heal spontaneously within 1-2 weeks, all flare-ups need to be treated; because, if disregarded, they tend to become subintrant and spread out. Treatment must be adapted to the characteristics of the patient and the lesions they present and, hence, we will distinguish various situations with their corresponding treatment(19).

In mild forms (limited to 4 – 6 sites), anti-inflammatory treatment should be topical (Tables V and VI) and corticosteroids are the first choice, although calcineurin inhibitors are an option in case of subacute lesions.

In severe forms (with a significant extension or resistant to topical treatment, or with intense involvement of particularly important areas, such as the hands or face), it is necessary to maintain topical treatment and to add systemic treatment: oral corticosteroids, cyclosporine, methotrexate, azathioprine, mycophenolate mofetil or phototherapy. Since January 31st, 2020, a biological drug, dupilumab, is also approved, initially only for children over 12 years of age, but from December 2nd, 2020 also for those over 6 years of age, which has shown results in children similar to those found in adults(20).

Microbial colonization of lesions and superinfection (Staphylococcus Aureus throughout the body and Malassezia furfur in head and neck lesions) seem to play a role in exacerbating flares and thus, justify the association of antimicrobials when these situations take place.

Treatments used

Topical corticosteroids are the first-line anti-inflammatory treatment in all phases of AD, but especially in the acute exacerbations. Except in chronic forms, where high-potency corticosteroids, such as clobetasol propionate are necessary, in the rest, low and medium-potency corticosteroids, such as clobetasone (low potency) or methylprednisolone aceponate (medium-high potency), usually suffice.

Calcineurin inhibitors (tacrolimus and pimecrolimus) are the alternative to topical corticosteroids and, more commonly nowadays, they are used in association to them so as to reduce their adverse effects. Tacrolimus 0.1% has an anti-inflammatory potency similar to that of a medium-potency corticosteroid and is clearly superior to pimecrolimus 1%.

Crisaborole 2% is an inhibitor of phosphodiesterase 4, mainly 4B, which acts by reducing the release of pro-inflammatory cytokines, such as: TNF, IL12 and IL23. It has been approved for children over 2 years of age by the EMA (European Medicines Agency, March 27th, 2020), although it does not yet have the mandatory “therapeutic positioning report”. Its use is restricted to a maximum of 40% of the body surface and, until now, its superiority against topical corticosteroids or calcineurin inhibitors has not been demonstrated.

Other topical treatments that have demonstrated their usefulness, as adjuvants in the treatment of pruritus, are: doxepin (antihistamine), topical agonists of cannabinoid receptors, topical antagonists of l-opioid receptors or topical anesthetics, but they are NOT to be routinely recommended given their adverse effects, but also because its use is not approved for AD.

Systemic corticosteroids (at initial doses no greater than 0.5 mg/kg/day) produce rapid, albeit temporary, improvements with prompt relapses, which is why they should be used in acute exacerbations or when a quick response is required. “Corticophobia” is relatively frequent, both among doctors and patients, which requires pausing to explain the advantages and disadvantages of these drugs, as well as careful managing, clearly specifying the dose and duration.

Phototherapy (narrow band ultraviolet B and ultraviolet A1) is only authorized from 12 years old onwards, but it is very useful, although the need to travel and time schedule difficulties are an obstacle, sometimes insurmountable.

Cyclosporine is the only drug with authorized indication in Spain and the fastest acting one. Its efficacy is 53-95%, but this efficacy is limited by its medium and long-term side effects (fatigue, gingival hyperplasia, increased blood pressure, hypertrichosis, kidney failure, etc.).

Azathioprine or methotrexate, both of them used off-label in severe AD, are drugs with theoretically similar efficacy (26-39% and 42%, respectively), which are indicated in severe cases of AD. They are slow acting (it takes more than a month for improvement to be noticeable), but are generally well tolerated for months, although they require periodic analytical tests.

Other alternatives without approved indication for AD, are: mycophenolate mofetil, alitretinoin (for hand eczema), rituximab, omalizumab, ustekinumab, tofacitinib or apremilast, with scarce or variable response.

Targeted immunotherapy against specific allergens (dust mites or pollens) can be considered in sensitized patients with severe AD and a history of clinical exacerbations following exposure to these allergens.

Psychotherapy may be indicated in patients in whom exacerbations are related to episodes of stress (exams, relationship problems, etc.).

Dupilumab is a human monoclonal antibody that is administered subcutaneously. It blocks the alpha chain common to IL4 and IL13 and decreases not only the production of IgE, but also the inflammatory response mediated by Th2 cells. It is indicated in severe forms that do not respond to cyclosporine or that present some contraindication to its use. Dupilumab is the first in a series of new drugs, which are being released now for the treatment of moderate-severe AD. Some have already been approved by the EMA, such as baricitinib (which has the therapeutic positioning report of the Spanish Agency for Medicines and Health Products pending), and others that have not yet been approved or are still in advanced stages of clinical trials, which will radically modify the prognosis of these patients.

Seborrheic dermatitis



Introduction

Seborrheic dermatitis is a common eczema, self-limited in childhood and with a chronic and relapsing course in adults.

Seborrheic dermatitis (SD) is a common eczema, with two clinical forms, the one found in the infant and that of the adult. The first is self-limited to the first 3 months of life; while, the second is chronic and, although it can present in puberty, its frequency is highest during the fourth to sixth decades of life. The latter affects men more than women.

Etiopathogenesis

There is a relationship between seborrheic dermatitis and: overproduction of sebum (seborrhea), alterations in its composition and commensal yeasts of the genus Malassezia (Pityrosporum).

The etiopathogenesis is not fully known, but there is a relationship with: overproduction of sebum (seborrhea), alterations in its composition and commensal yeasts of the genus Malassezia (Pityrosporum). In babies, sebum is produced for a few weeks after birth, and the adult form of SD does not develop before puberty, supporting a role of androgens in the activation of the sebaceous glands. However, patients with SD may have normal sebum production, and those with excessive sebum production usually do not have SD. Malassezia Furfur and other species can be isolated from SD lesions, including infant SD, but there is no relationship between the number of yeasts and the severity of SD (unaffected skin may have a microorganism load similar to that of lesions). Anyhow, with antifungal treatment, skin lesions improve and the number of yeasts decreases and, these increase again when SD relapses. It has also been discovered that a major component of the resident skin microflora, Propionibacterium acnes, is greatly diminished in SD; thus, it could be associated with an imbalance of the microbial flora.

Clinical presentation and diagnosis

The diagnosis of seborrheic dermatitis is a clinical one. The lesions, generally not very bothersome, are located in areas with an abundance of sebaceous glands and, less frequently, in skinfolds.

The lesions consist of erythematous areas, generally well defined, with an unctuous sensation to the touch, and loose and moderate scaling. Vesiculation and crusting are rare to find. It is located in areas with an abundance of sebaceous glands (scalp, face, ears, presternal region) and, less frequently, in folds, but generalized and even erythrodermic forms can occur.

Seborrheic dermatitis of the infant

It usually begins a week after birth and can persist for several months. Initially, they are greasy scales, attached to the vertex and anterior fontanel, which can spread to the entire scalp (cradle cap) and face (Fig. 6).

Figura 6. Seborrheic dermatitis. Cradle cap.

Lesions in armpits, inguinal folds, neck, and retroauricular folds are usually more inflamed, although well defined. Lesions may become superinfected by Candida and, more rarely, by group A Streptococcus. A widespread rash of psoriasiform-appearing erythematous-scaly lesions may develop after an intensely inflammatory or superinfected SD, especially in the diaper area.

Seborrheic dermatitis in adults

In the scalp, the lesions are predominantly located in the parietal and vertex regions, but also in the occipital region, with a quite diffuse pattern. On the face, the lesions are located symmetrically in: nasogenial folds, eyebrows and retroauricular folds, but also in: the forehead, on the implantation edge of the scalp, in the glabella region and, occasionally, on the back of the neck, and also on the hairline. The lesions are not usually infiltrated and the scaling is fine and loose. Lesions on the trunk are located in the presternal area (in men) and in the center of the back where they tend to adopt a petaloid morphology. They are rarer in the armpits. Flare-ups lasting 1-2 weeks are more or less recurrent and they are frequently related to stress. Pruritus is usually moderate, but may be severe, and folliculitis (Pityrosporum) and inflammation of the meibomian glands in the palpebral tarsus (meibomitis) may be seen.

Differential diagnosis

The main differential diagnosis of seborrheic dermatitis is with atopic dermatitis.

Infantile SD differs from atopic dermatitis by: its earlier onset, the different distribution pattern and the absence of itching, irritability and insomnia. Irritant diaper rash is limited to the diaper area and usually does not affect the folds. Yeast (Candida) diaper rash preferentially affects the folds, with occasional fissuring, and satellite lesions are usually present around the main lesions. In inverted psoriasis of the diaper area, the lesions are more infiltrated and demarcated, and the presence of typical lesions in other locations aids in the diagnosis.

Pityriasis amiantacea (thick asbestos-like flakes which adhere to strands of hair on the scalp, in an irregular shape) which could be confused with cradle cap, occurs in atopic dermatitis and psoriasis.

Other rare disorders, which must also be considered in the differential diagnosis, especially in treatment-resistant forms, are: Langerhans cell histiocytosis, acrodermatitis enteropathica, and Leiner’s disease. The latter, previously considered the maximal expression variant of infantile SD, is now considered erythroderma in the context of underlying immunosuppression.

Another differential diagnosis, to be considered especially in prepubertal children, and primarily in black ethnicity, with scaling of the scalp, is tinea capitis (due to Trichophyton tonsurans).

When SD is extensive and severe in adults, HIV infection should be ruled out. The main differential diagnosis is with psoriasis of the scalp. In this case, the lesions are erythematous plaques covered by whiter and adherent scales, especially in the occipital and temporal regions.

Pityriasis simplex (dandruff) is defined as a diffuse, more or less intense, flaking of the scalp and beard, but without significant erythema or irritation. This common disorder can be considered the mildest form of seborrheic dermatitis of the scalp.

Treatment

Washing the lesions with a 2% ketoconazole shampoo and the application of an emollient cream suffice in most cases.

Both, infantile and adult forms of SD satisfactorily respond to washing with mild shampoos and application of emollients. Ketoconazole (2%) or ciclopirox olamine cream or shampoo is indicated in more extensive or persistent cases. In acute phases, short courses of low-potency topical corticosteroids or calcineurin inhibitors (although not approved for this indication) can be used to suppress inflammation.

The role of the Primary Care pediatrician

The role of the Primary Care pediatrician is to recognize the typical and less typical forms of both diseases, in order to be able to treat mild or moderate forms, and refer the most serious forms or those that do not respond to treatment to the specialist. In the case of AD, he plays a fundamental role in the prevention of flare-ups, in the education of patients and their families (it should not be forgotten that it is often a familial disease) as well as in screening for other diseases associated with AD (asthma, environmental allergy, etc.).

Bibliography

The asterisks show the interest of the article in the opinion of the authors.

1.** Eichenfield LF, Tom WL, Chamlin SL, Feldman SR, Hanifin JM, Simpson EL, et al. Guidelines of care for the management of atopic dermatitis: Section 1.Diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014; 70: 338-51.

2.* Spergel JM. Epidemiology of atopic dermatitis and atopic march in children. Immunol Allergy Clin N Am. 2010; 30: 269-80.

3.* Sicras-Mainar A, Navarro-Artieda R, Sánchez L, Sastre J. Prevalence of Severe Atopic Dermatitis in Adults in 3 Areas of Spain. J Investig Allergol Clin Immunol. 2018; 28: 195-97.

4.* Sicras-Mainar A, Navarro-Artieda R, Carrascosa Carrillo JM. Impacto económico de la dermatitis atópica en adultos: estudio de base poblacional (estudio IDEA). Actas Dermo-sifiliográficas. 2018; 109: 35-46.

5.** Patrick GJ, Archer NK, Miller LS. Which Way Do We Go? Complex Interactions in Atopic Dermatitis Pathogenesis. J Investig Dermatol. 2021; 14: 274-84.

6. ** Hanifin JM, Rajka G. Diagnostic features of atopic dermatitis. Acta Dermatol Venereol. 1980; 92: 44-7.

7. * Luo Y, Zhang GQ, Li ZY. The diagnostic value of APT (atopy patch test) for food allergy in children: a systematic review and meta-analysis. Pediatric Allergy and Immunology. 2019; 30: 451-61.

8. * Dickel H, Kuhlmann L, Bauer A, Bircher AJ, Breuer K, Fuchs T. Atopy patch testing with aeroallergens in a large clinical population of dermatitis patients in Germany and Switzerland, 2000-2015: a retrospective multicentre study. J Eur Acad Dermatol Venereol. 2020; 34: 2086-95.

9. *** Wollenberg A, Barbarot S, Bieber T, Christen-Zaech S, Deleuran M, Fink-Wagner A, et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part I. J Eur Acad Dermatol Venereol.2018; 32: 657-82.

10. *** Wollenberg A, Barbarot S, Bieber T, Christen-Zaech S, Deleuran M, Fink-Wagner A, et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part II. J Eur Acad Dermatol Venereol. 2018; 32: 850-78.

11.** Heratizadeh A, Werfel T, Wollenberg A, Abraham S, Sybylle PH, Schnopp C, et al. Effects of structured patient education in adult atopic dermatitis: Multi-center randomized controlled trial. J Allergy Clin Immunol. 2017; 140: 845-53.

12. * Du Toit G, Roberts G, Sayre PH, Bahnson HT, Radulovic S, Santos AF, et al. Randomized trial of peanut consumption in infants at risk for peanut allergy.N Eng J Med. 2015; 372: 803-13.

13. ** Roduit C, Frei R, Loss G, Büchele G, Weber J, Depner M, et al. Development of atopic dermatitis according to age of onset and association with early-life exposures. J Allergy Clin Immunol. 2012; 130: 1306e5.

14. * Fölster-Holst R, Müller F, Schnopp N, Abeck D, Kreiselmaier I, Lentz T, et al. Prospective, randomized controlled trial on Lactobacillus rhamnosus in infants with moderate to severe atopic dermatitis.Br J Dermatol. 2006; 155: 1256-61.

15.* Rosenfeldt V, Benfeldt E, Nielsen SD, Michaelsen KF, Jepessen DL, Valerius NH, et al. Effect of probiotic Lactobacillus strains in children with atopic dermatitis. J Allergy Clin Immunol. 2003; 111: 389-95.

16. * Grüber C. Probiotics and prebiotics in allergy prevention and treatment: future prospects. Exp Rev Clin Immunol.2012; 8: 17-9.

17. ** He A, Feldman SR, Fleischer AB. An assessment of the use of antihistamines in the management of atopic dermatitis. J Am Acad Dermatol. 2018: 79: 92-6.

18. ** Hon KL, Tsang YCK, Lee VWY, Pong NH, Ha G, Lee ST, et al. Efficacy of sodium hypochlorite (bleach) baths to reduce Staphylococcus aureus colonization in childhood onset moderate-to-severe eczema: A randomized, placebo-controlled cross-over trial. J Dermatolog Treat. 2016; 27: 156-62.

19. *** Paller AS, Siegfried EC, Vekeman F, Gadkari A, Kaur M, Mallya UG, et al. Treatment patterns of pediatric patients with atopic dermatitis: A claims data analysis. J Am Acad Dermatol.2020; 82: 651-60.

20.** Igelman S, Kurta AO, Sheikh U, McWilliams A, Armbrecht E, Jackson Cullison SR, et al. Off-label use of dupilumab for pediatric patients with atopic dermatitis: A multicenter retrospective review. J Am Acad Dermatol. 2020; 82: 407-11.

21. Prieto-Torres L, Torrelo A. Dermatitis atópica y otras erupciones eccematosas. Pediatr Integral. 2016; XX(4): 216-26.

Recommended bibliography

- Patrick GJ, Archer NK, Miller LS. Which Way Do We Go? Complex Interactions in Atopic Dermatitis Pathogenesis. J Investig Dermatol. 2021; 14: 274-84.

It summarizes the current knowledge on the etiopathogenesis of atopic dermatitis.

- Wollenberg A, Barbarot S, Bieber T, Christen-Zaech S, Deleuran M, Fink-Wagner A, et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part I. J Eur Acad Dermatol Venereol.2018; 32: 657-82.

- Wollenberg A, Barbarot S, Bieber T, Christen-Zaech S, Deleuran M, Fink-Wagner A, et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part II. J Eur Acad Dermatol Venereol. 2018; 32: 850-78.

They both reflect the current European consensus in the treatment of atopic dermatitis.

- Paller AS, Siegfried EC, Vekeman F, Gadkari A, Kaur M, Mallya UG, et al. Treatment patterns of pediatric patients with atopic dermatitis: A claims data analysis. J Am Acad Dermatol. 2020; 82: 651-60.

It highlights how things are currently being done and what has to be improved.

 

Clinical case

 

11-year-old patient who has noticed for months, the appearance of cracks in the 1st and 2nd toes of both feet. The examination is shown in figure 7.

 

Figure 7.

 

New approach in the treatment of children with asthma

J. Pellegrini Belinchón*, C. Ortega Casanueva**, S. de Arriba Méndez***
Topics on
Continuous Training


J. Pellegrini Belinchón*, C. Ortega Casanueva**, S. de Arriba Méndez***

*Primary Care Pediatrician. Pizarrales Health Center. Salamanca. **Pediatrician and Allergist. Child Allergy and Pneumology Unit. Quironsalud San José Hospital. Madrid. ***Pediatrician specialized in Childhood Allergology and Pulmonology. Salamanca University Clinical Hospital

Resumen

Asthma is the most common chronic disease among children.
The national and international consensus, the manuals and clinical practice guidelines agree that the ultimate goal of treatment is to achieve and maintain total control of asthma.
In order to avoid limitations on the children’s normal activity and guarantee lack of symptoms and exacerbations, and a normal lung function, it is necessary to optimize the use of the resources available to fight this disease, with the education of the patients and their relatives playing a key role.
This chapter reviews the latest news in the Spanish Guideline on the Management of Asthma, including the current pharmacological treatment of the crisis, the maintenance treatment of the disease and the education of the patient.

 

 

Key words: Childhood asthma; Crisis; Treatment; Anti-asthma drugs.

Palabras clave: Asma infantil; Crisis; Tratamiento; Fármacos antiasmáticos.

 

Pediatr Integral 2021; XXV (2): 67 – 75 – EN

 


New approach in the treatment of children with asthma

Introduction

Asthma is one of the most prevalent chronic diseases in childhood(1).

According to the ISAAC study (International Study of Asthma and Allergies in Childhood), its prevalence in Spain is 10%, a similar number to that of the European Union, being more frequent in males aged 6-7 years(2).

Asthma constitutes a public health problem(3-5), entailing high social and health costs, as it is a frequent reason for consultation, both in pediatric emergency services and in the Primary Care setting.

The cost of asthma in Pediatrics in Spain depends on the severity of the disease. Blasco Bravo et al.(6) identified, a decade ago, that the total cost of asthma in Pediatrics in Spain was about 532 million euros, but it could range between 392 and 693 million euros. Direct costs (healthcare costs) represented 60% of the total cost whereas indirect costs (care provider’s time) 40%. A problem of this magnitude requires the optimization of resources against this disease and enhances the relevance of educating patients and their families.

In May 2020, the new update of the Spanish Guide for Asthma Management(7) (GEMA5.0) was published. Like the previous versions, it is a very practical, independent and consensual guide; but on this occasion it has been agreed on by twenty-one societies and scientific groups, among which is the Spanish Society of Primary Care Pediatrics (SEPEAP). This multidisciplinary guide allows a consensual approach and treatment of asthma, differentiating the management in those older or younger than 3-4 years of age, and it has become the main reference for the management of this disease among pediatricians in different areas of care.

The Global Initiative for asthma (GINA)(8), updated in 2020, is another essential guide for the management of asthma that provides treatment guidelines, according to the age of the child: those five years old or younger, and conversely, those aged six-years-old and above who will follow recommendations equal to those of adolescents and adults.

This manuscript will address: the evaluation and pharmacological treatment of exacerbations, the indications for maintenance treatment, the drugs used for this purpose, the treatment of exercise-induced bronchospasm and, finally, the education of the patient with asthma.

Assessment and treatment of asthma exacerbation

In a child with symptoms of asthma attack, once other diagnoses have been excluded, a rapid assessment of the severity of the exacerbation must be carried out to determine if immediate action is necessary and to apply treatment.

The severity assessment is mainly based on clinical criteria (respiratory rate, presence of wheezing and existence of retractions of the sternocleidomastoid muscle). Although no clinical scales are well validated(9,10), the Pulmonary Score (Table I) is simple and applicable to all ages(11).

The combination of symptoms together with oxygen saturation (SaO2) will allow to estimate the severity of the episode(7) (Table II).

Figure 1 represents the treatment proposed by the Spanish pediatric consensus and the GEMA5.0 Guide to treat asthma attacks according to their severity(7,12).

Figure 1. Treatment of asthma exacerbation in children (Spanish Guide for Asthma Management:
GEMA5.0).
kg: kilogram; min: minute; mg: milligram; µg: microgram; SaO2: oxyhemoglobin saturation; max.: maximum. SABA: Short-acting inhaled β2 adrenergic agonists.

It is advisable to individualize the dose of drugs according to the severity of the exacerbation and the response to treatment. Generally, for mild and moderate exacerbations, MDI (Metered Dose Inhaler) with chamber is preferred over nebulizer. Therefore, to avoid logistical problems, health care providers at primary care and at hospitals should encourage children and their families to bring their spacer and inhaler when attending the emergency room.

During the SARS-CoV-2 pandemic, avoiding nebulizers and using MDI with spacer, becomes even more relevant.

Medications

The main characteristics of the drugs used in the treatment of acute asthma attacks are described below.

Short-acting inhaled β2 adrenergic agonists (SABA)

They are the bronchodilator drugs with the greatest effectiveness, onset of action and fewer side effects, which is why they constitute the first line of treatment(13).

In children, salbutamol is mainly used in primary care centers and hospitals, however the patient can use terbutaline if prescribed, unless the asthma attack is so severe that it prevents him/her from properly inhaling through the turbuhaler system, which is how the latter drug is marketed in Spain.

Ipratropium bromide

Together with rescue β2 agonists, it is indicated during the first 48 hours of a severe asthma attack. Its early use has been associated with a decrease in the number of hospitalizations. Its onset of action is slower, between 30 and 60 minutes. The nebulized dose is 250 µg / 4-6 hours in patients under 30 kg and 500 µg / 4-6 hours in patients over 30 kg(15). The inhaler (with chamber) dose is 40-80 µg / 4-6 hours (2-4 puffs).

Systemic glucocorticoids

They are used for the treatment of moderate or severe exacerbations in short schemes (3-5 days or until resolution). In this case, they are administered at a dose of 1-2 mg / kg / day of prednisone or equivalent, with a maximum of 50 mg / day, as a single morning dose. Oral route is preferred, whenever possible(8). Steroids used during short periods can be withdrawn abruptly, as they seem not to affect the hypothalamus-pituitary-adrenal axis.

Alternatively, dexamethasone can be used. The effect of administering a single dose of dexamethasone orally (0.3 mg / kg) is not inferior to that of prednisolone at a dose of 1-2 mg / kg / day (maximum 40 mg) for 3 to 5 days, or up to resolution(16).

With regards to the additional use of inhaled glucocorticoids, there is currently insufficient evidence to recommend it(17).

Evaluation and maintenance treatment

One of the novelties of GEMA5.0 when initiating maintenance treatment relates to the asthmatic terms: occasional episodic, frequent episodic, moderate persistent or severe persistent, when classifying asthma the first time in a child who does not receive preventive treatment. These terms are replaced by mild, moderate or severe asthma, depending on the clinical assessment of the patient(7).

In case the child has mild and infrequent daytime symptoms, does not have inter-exacerbation symptoms, tolerates exercise well and does not present nocturnal symptoms, he or she will be allowed to receive only short-acting β2 adrenergic bronchodilators on demand. In all other cases, an anti-inflammatory maintenance treatment will be commenced.

Figure 2 shows escalation of treatment in the pediatric age depending on its degree of control (GEMA5.0).

Figure 2. Escalation treatment of asthma in pediatric age depending on the degree of control (Spanish Guide for Asthma Management: GEMA5.0 ). IGC: inhaled glucocorticoids; LTRAs: Leukotriene receptor antagonists; LABA: long-acting β2 adrenergic agents; GC: glucocorticoid; *: from 6 years-old; **: off indication.

To facilitate the evaluation of symptom control, 2-3 months after commencing treatment, specific questionnaires can be offered to the parents and patient, the purpose of which is to try to verify the response to this initial treatment.

The Child Asthma Control (CAN) questionnaire (Fig. 3a), considers a patient to be poorly controlled when he or she has a score equal to or greater than 8 points(7).

Figure 3a. Asthma Control Questionnaire in Children (CAN).

Another available questionnaire is the c-ACT (Childhood Asthma Control Test) validated in Spanish (Fig. 3b).

Figura 3b. Pediatric Asthma Control Test (ACT) [also available validated in Spanish (Spanish Guide for Asthma Management: GEMA5.0)].

This questionnaire is aimed at children between 4-11 years-old and consists of 7 questions, 4 aimed at children (answers expressed with smiley icons) and 3 at parents / caregivers. A patient is considered to be poorly controlled when the score is below 20(7). The ACT for over 12-year-olds consists of 5 questions, which are completed only by the adolescent and it considers poorly controlled asthma when the score is 20 or less, and well but insufficient control from 21 to 24 points.

GINA recommends a shorter questionnaire of only four questions for asthma control(8) (Table III).

If after frequent clinical reviews to ensure adherence to treatment, correct use of inhalers, and if after two to three months asthma is not controlled, escalation of treatment must be considered. Of course, the correct diagnosis must have been confirmed and modifiable risk factors addressed.

After three months of total asthma control, reduction of treatment can be contemplated. The decrease will be gradual, for instance, in the opposite direction to how it was ascended, although it shall be individualized based on the response obtained to different drugs. For example, for those patients who were not responders to leukotriene receptor antagonists in monotherapy, it would not make sense now to leave them on it again.

Inhaled glucocorticoids (IGC)

They are the recommended first line treatment from stage 2 of asthma in the pediatric age(7).

Given their high affinity and selectivity for the receptor, they allow a powerful local anti-inflammatory effect, sustained therapeutic actions, prolonged presence in the lung and low oral bioavailability. They reduce asthma symptoms and the number of exacerbations.

The available IGC in Spain are: beclomethasone dipropionate, budesonide, fluticasone propionate, fluticasone furoate, mometasone furoate and ciclesonide (the latter 3 are only authorized in Spain for age 12 years and above). Budesonide and fluticasone are the most widely used in current consensuses. The lowest effective dose of IGC should always be used(18,19).

Table IV shows the comparable doses of fluticasone propionate and budesonide(7).

Leukotriene receptor antagonists (LTRAs)

The only drug of this group authorized in Spain for children (from six months of age) is montelukast. It is used orally and in a single nightly dose. Its metabolism does not seem to be altered by large or fatty meals. LTRAs are considered to improve exercise-induced and allergen-induced asthma.

In preschoolers with asthma or viral-triggered episodes, they modestly reduce symptoms and the need for oral glucocorticoids.

In addition to inhaled corticosteroids, they appear to improve lung function and reduce the number of exacerbations. When evaluating the usefulness of antileukotrienes combined with inhaled corticosteroids, a complementary anti-inflammatory effect has been observed, which allows the reduction of the corticosteroid dose. This effect seems inferior to that observed in the association of long-acting β2 adrenergic bronchodilators with corticosteroids. If symptoms are not controlled with low-dose IGC, increasing the dosage of IGC to medium dose is more effective than its combination with montelukast. In monotherapy, it also seems to have a beneficial effect, but inferior to inhaled corticosteroids(20).

Association of long-acting β2 adrenergic agonists (LABA) and inhaled glucocorticoids (GCI)

They must always be used in combination with an inhaled glucocorticoid and must never be administered as monotherapy.

Its use in Spain is authorized for children over the age of 4 years. The following combinations are available(19): salmeterol / fluticasone propionate (from 4 years-old onward), formoterol / budesonide (from 6 years-old onward), formoterol / fluticasone propionate (from age 12 years), vilanterol / furoate fluticasone (from age 12 years) and formoterol / beclomethasone (from 18 years of age).

The recommended formoterol dose is 4.5 – 9 μg, twice a day; whereas it is 50 μg, twice daily, for salmeterol. Currently, they are not recommended as rescue medication for childhood asthma.

Long-acting muscarinic receptor antagonists (LAMA)

In this group, tiotropium bromide stands out. It has beneficial effects in the maintenance treatment of asthma, through selective and prolonged blockade of M3 receptors.

It can be used in children from 6 years of age with severe asthma that is poorly controlled with high dose IGC plus a LABA(19). The dose is 5 µg per day.

Theophylline

In monotherapy, it is less effective than IGC, although its anti-inflammatory effect allows the possibility of individually using it in cases of severe asthma(7).

Monoclonal antibodies

Anti-IgE (Omalizumab)

It has shown therapeutic efficacy (reduction of IGC dose, improvement of quality of life, reduction of exacerbations and hospitalizations) in IgE-mediated asthma in children from 6 years of age with severe asthma (step 6), insufficiently controlled with high doses of GCI and LABA(21,22). It is subcutaneously administered every 2-4 weeks with dose-titration according to total IgE and body weight.

Anti-IL5 (mepolizumab)

Pediatric population studies are currently very scarce. Despite this, Mepolizumab (anti-IL5) is recommended in children from 6 years of age with severe eosinophilic asthma, insufficiently controlled with high doses of IGC and LABA(22).

Immunotherapy

Immunotherapy reduces symptoms, rescue and maintenance medications, and bronchial hyperresponsiveness (both specific and nonspecific), provided that biologically standardized extracts are used and in appropriately selected sensitized patients(7).

In our country, approximately 10% of children are asthmatic(2) and 85% of them have an allergic etiology(7,19). Asthma triggered by an allergic mechanism will be treated in the same way as asthma triggered by other causes, but in addition, avoidance of specific allergens and the therapeutic option of immunotherapy should be taken into account,.

Subcutaneous or sublingual immunotherapy, with allergen vaccines, is an effective treatment for well-controlled allergic asthma (steps 2-4), provided that clinically relevant IgE-mediated sensitization to common aeroallergens has been demonstrated. Immunotherapy should not be prescribed to patients with uncontrolled asthma, due to the high risk of serious adverse reactions. Subcutaneous immunotherapy should only be administered by trained personnel and in centers where the necessary means are available to treat potential anaphylaxis. The patient should be observed for 30 minutes after the subcutaneous injection; since it is then when the rare, although possible, serious reactions have been described.

Immunotherapy prevents the development of new sensitizations and asthma in children with rhinitis(23).

Treatment of exercise-induced bronchospasm

It is essential to convey to the child and his parents the fact that, with general measures and treatment, he will be able and should continue to exercise.

When a child presents with exercise-induced bronchospasm, the most frequent possibility is that it is a not completely controlled asthma, and once other diagnoses have been ruled out, the baseline treatment should be increased.

When the child or adolescent exclusively presents symptoms with physical exercise, a preventive treatment must be indicated.

Sedentary lifestyle should be avoided and the adolescent should be explained how a proper treatment will prevent the appearance of symptoms(24). Fitness and aerobic capacity should necessarily improve. Practicing sports will be beneficial for the progression of asthma, if performed properly. Certain sports, such as swimming, are usually better tolerated and, in addition, improve lung function in adolescents and children. Other recommendations include: an adequate and progressive warm-up beforehand, progression of exercise, the use of scarf if the environment is cold and dry, and avoidance of mouth breathing as much as possible.

Regarding medications, short-acting β2 are recommended, 10-15 minutes prior to exercise. When used very frequently or continuously, a progressive loss of effectiveness may develop (tachyphylaxis), thus in these cases, the recommendation is to associate maintenance inhaled corticosteroids. Likewise, in children in whom the timing of physical exercise is unpredictable, a baseline treatment is also recommended. Antileukotrienes are beneficial in a non-negligible percentage of these patients, hence a therapeutic trial can be performed(20).

Inhalation delivery devices

Regardless of the child’s age, the inhaled route is the most suitable for administering medication in the treatment of asthma.

No pharmacological treatment with inhaled medication can be delivered correctly, if the child (whether young or adolescent) and family do not use the different inhalation systems correctly(25).

Table V serves as a guide to the inhalation devices recommended according to age. Between nebulizers or inhalers with chamber/spacer, the latter system is preferred, relegating nebulizers for very specific cases of young, uncooperative children.

Periodic review of the inhalation technique is necessary, as well as the consideration to transition from one system to another, depending on age, preferences of the older child, or whenever asthma does not evolve correctly.

Asthma education

Educating the child with asthma and his family increases the quality of life, and reduces the risk of exacerbations and healthcare costs, which is why it is one of the fundamental pillars of treatment.

The Primary Care pediatrician is key in the education of the asthmatic child

The educational approach is responsibility of all health professionals: pediatricians, pediatric allergists and pulmonologists, nurses, physiotherapists and pharmacists. However, the Primary Care pediatrician and the health center nurse, due to their proximity, accessibility and trust, play a fundamental role(12,18).

Having an accessible pediatrician, who provides continuous care to the child from birth and knows the patient’s socio-family environment(26) and his illness, will make the health center the ideal setting to respond to educational and health needs, including control of his asthma.

At present, the role of new technologies, teachers and trainers trained in asthma and the so-called “expert patient” that, through groups or associations of patients, can be useful in the education of children and their families, cannot be ignored, albeit these require supervision by health professionals(26,27).

The key points to educate on are specified in table VI.

Objectives and educational sequence

For education to be effective, it is essential to identify the educational needs and the factors that impact the behavior of the patient and / or his family.

After the educational diagnosis and the identification of the needs and, depending on them and the available resources, the objectives must be established based on agreement between the child, his family and the educator(26-28). The general objective of education is to increase the quality of life of the child or adolescent and their families. Among the specific objectives are, in addition to the adequate training of the healthcare personnel involved in the program: to improve communication between patients and healthcare personnel, reduce anxiety, clarify doubts and overcome false beliefs and expectations. By increasing the knowledge of the child and his family, the aim is to induce behavioral changes and skills they need to: reduce the number of visits to the emergency department, avoid hospital admissions, improve clinical symptoms, develop prevention behaviors by identifying triggering factors, handle his condition according to his needs and future projects, and ultimately, improve quality of life in the short and long term(29).

Therefore, by means of a structured methodology termed “educational sequence”, the contents will be applied and developed in the following stages:

• Educational diagnosis.

• Awareness of illness and possible risks.

• Adherence to information. The way in which the information is presented and the empathy are essential in this stage.

• Search for solutions.

Given that asthma is a chronic disease, at this stage it is essential that the pediatrician abandons the role of expert, and switches to a more horizontal model, where solutions are sought by mutual consensus, agreeing to changes in habits and behavior modifications that promote autonomy of the child or adolescent.

In each visit, the maintenance treatment, the inhalation technique, recognition of the symptoms and approach in the event of a possible exacerbation will be reminded. It is essential that the educational program is developed in the first 6 months after diagnosis and, a minimum of three educational sessions to train and empower the child in a personalized self-management program are considered necessary(29).

Other relevant aspects include: making an escalated information plan, with a clear and understandable language adapted to each family, using personalized written information and based on graphic materials or instruments, such as chambers, placebos, explanatory rings on inflammation or bronchoconstriction, which may be helpful.

Adherence to treatment

Asthma, as other chronic diseases with long asymptomatic periods, has a high rate of therapeutic non-compliance.

The degree of adherence to treatment in pediatric asthma can be defined as the extent to which the child and / or his family truly follow the advice and use the medication that is indicated and agreed with the health care personnel, and does so correctly, using adequate techniques. At the present time, there are no effective measures for the assessment of pharmacological compliance of asthma in Pediatrics.

There are no known biochemical parameters to indicate with certainty the degree of treatment compliance and optimal disease control. An indirect attempt can be made by measuring the medication consumed and prescriptions made, using the symptom diary and interviewing the patient and his family.

Bibliography

The asterisks indicate the interest of the article considered by the authors.

1. World Health Organization. Accessed July 30, 2020. Available at: https://www.who.int/news-room/fact-sheets/detail/asthma

2. Carvajal-Urueña I, García-Marcos L, Busquets-Monge R, Morales M, García de Andoine N, Batlles-Garrido J, et al. Geographic variations in the prevalence of asthma symptoms in Spanish children and adolescents. International Study of Asthma and Allergies in Childhood (ISAAC) phase III Spain. Arch Bronchoneumol. 2005; 41: 659-66.

3. Mallol J, García.-Marcos L. Solé D, Brand P; EISL Study Group. International prevalence of recurrent wheezing during the first year of life: variability, treatment patterns and use of health resources. Thorax. 2010; 65: 1004-9.

4. Pellegrini J, Miguel G, Dios de B, Vicente E, Lorente F, García-Marcos L. Study of wheezing and its risk factors in the first year of life in the Province of Salamanca, Spain. The EISL Study. Allergol Immunopathol (Madr). 2012; 40: 164-71.

5. Hansen TE, Evjenth B, Holt J. Increasing prevalence of asthma, allergic rhinoconjunctivitis and eczema among schoolchildren: three surveys during the period 1985-2008. Acta Paediatr. 2013; 102: 47-52.

6. Blasco Bravo AJ, Pérez-Yarza EG, Lázaro y de Mercado P, Bonillo Perales A, Díaz Vázquez CA, Moreno Galdó A. Cost of asthma in Pediatrics in Spain: a cost assessment model based on prevalence. An Pediatr (Barc). 2011; 74: 145-53.

7.*** GEMA5.0. Spanish guide for the management of asthma. Ed. Luzán 5, SA Madrid 2020. Accessed July 30, 2020. Available at: www.gemasma.com.

8.*** Global Initiative for asthma (GINA). Accessed July 30, 2020.Available at: http://www.ginasthma.org/.

9. Bekhof J, Reimink R, Brand PL. Systematic review: insufficient validation of clinical scores for the assessment of acute dyspnoea in wheezing children. Paediatr Respir Rev. 2014; 15: 98-112.

10. Eggink H, Brand P, Reimink R, Bekhof J. Clinical Scores for Dyspnoea Severity in Children: A Prospective Validation Study. PLoS One. 2016; 11: e0157724.

11. Smith SR, Baty JD, Hodge D. Validation of the pulmonary score. An asthma severity score for children. Acad EmergMed. 2002; 9: 99-104.

12.*** Castillo Laita JA, De Benito Fernández J, Escribano Montaner A, Fernández Benítez M, García de la Rubia S, Garde Garde J, et al. Consensus on the treatment of asthma in Pediatrics. An Pediatr (Barc). 2007; 67: 253-73.

13. Robertson CF, Smith F, Beck R, Levison H. Response to frequent low doses of nebulized salbutamol in acute asthma. J Pediatr. 1985; 106: 672-4.

14. Griffits B, Ducharme FM. Combined inhaled anticholinergics and short-acting beta2-agonists for initial treatment of acute asthma in children. Cochrane Database Syst Rev. 2013; 8: CD000060.

15. Vézina K, Chauhan BF, Ducharme FM. Inhaled anticholinergics and short-acting beta2-agonists versus short-acting beta2-agonists alone for children with acute asthma in hospital. Cochrane Database Syst Rev. 2014; 7: CD010283.

16. Mathew JL. Oral Dexamethasone versus Oral Prednisolone in Acute Asthma: A New Randomized Controlled Trial and Updated Meta-analysis: Evidence-based Medicine Viewpoint. Indian Pediatr. 2018; 55: 155-9.

17. Kearns N, Maijersl, Harper J, Beasley R, Weatherall M. Inhaled corticosteroids in acute asthma: systemic review and meta-analysis. J. Allergy Clin Immunol Pract. 2020; 8: 605-17.

18.** De Arriba Méndez S, Pellegrini Belinchón J, Ortega Casanueva. Treatment of the asthmatic child. Comprehensive Pediatric. 2016; XX (2): 94-102.

19.** Torres Borrego J, Ortega Casanueva C, Tortajada-Girbés M. Treatment of pediatric asthma. treatment of asthma attack. Diagnostic protocol pediatr. 2019; 2: 117-32.

20. Hussein HR, Gupta A, Broughton S, Ruiz G, Brathwaite N, Bossley CJ. A meta-analysis of montelukast for recurrent wheeze in preschool children. Eur J Pediatr. 2017; 176: 963-9.

21. Corren J, Kavati A, Ortiz B, Colby JA, Ruiz K, Maiese BA, et al. Efficacy and safety of Omalizumab in children and adolescents with moderate-to-severe asthma: A systematic literature review. Allergy Asthma Proc. 2017; 38: 250-63.

22. Ahmed H, Turner S. Severe Asthma in children-a review of definitions, epidemiology and treatment options in 2019. Pediatr Pulmonol. 2019; 54: 778-87.

23. Kristiansen M, Dhami S, Netuveli G. Allergen immunotherapy for the prevention of allergy: A systematic review and meta-analysis. Pediatr Allergy Immunol. 2017; 28: 18-29.

24.*** Ortega Casanueva C, Pellegrini Belinchón J, De Arriba Méndez S. Asthma and adolescence. Adolescere. 2018; VI (3): 14-26.

25.*** Ortega Casanueva C, Pellegrini Belinchón J, de Arriba Méndez S. Inhalation devices in inhaled medication. Diagnostic protocol pediatr. 2019; 2: 51-64.

26. Pinnock H, Parke HL, Panagioti M, Daines L, Pearce G, Epiphaniou E, et al. Systematic meta-review of supported self-management for asthma: a health care perspective. BMC Med. 2017; 15:64.

27.*** GEMA educators. Asthma Educator Manual. Madrid: Luzán 5. 2010.

28. Ortega Casanueva C, Pellegrini Belinchón J. Asthma: health education, self-control and preventive measures. Comprehensive Pediatrics. 2012; XVI (2): 141-8.

29. Gillete C, Rockich-Winston N, Shepherd M, Flesher S. Children with asthma and their caregivers help improve written asthma action plans: A pilot mixed-method study. J. Asthma. 2008; 55: 609-14.

Recommended bibliography

- GEMA5.0. Spanish guide for the management of asthma. Ed. Luzán 5, SA Madrid 2020. Accessed October 15, 2020. Available at: www.gemasma.com.

Essential for the management of this pathology, both in adults and children. Consensus guide developed by 17 Spanish scientific societies and groups, and with an international scope. The pediatric part was agreed by: SEPEAP, SEN, SEICAP and AEPap. Updated in May 2020, it provides the latest available evidence and expert consensus.

- Castillo Laita JA, De Benito Fernández J, Escribano Montaner A, Fernández Benítez M, García de la Rubia S, Garde Garde J, et al. Consensus on the treatment of asthma in Pediatrics. An Pediatr (Barc). 2007; 67: 253-73.

Childhood asthma consensus developed by 5 Spanish Scientific Societies related to asthma: SEN, SEICAP. SEPEAP, AEPap and SEUP. It describes in detail with the education of children and their families, as a fundamental basis for the treatment of asthma.

- GEMA educators. Asthma Educator Manual. Madrid: Luzán 5. 2010.

It is an essential guide to approach asthma education.

- Global Initiative for asthma (GINA). Accessed October 25, 2020. Available at: http://www.ginasthma.org/.

International consensus on the diagnosis and treatment of asthma, prepared by the National Heart, Lung and Blood Institute of the USA, with the collaboration of specialists representing different parts of the world.

- Ortega Casanueva C, Pellegrini Belinchón J, De Arriba Méndez S. Asthma and adolescence. Adolescere 2018; VI (3): 14-26.

The treatment of asthma in adolescents is addressed in a detailed and updated manner.

- Ortega Casanueva C, Pellegrini Belinchón J, de Arriba Méndez S. Inhalation devices in inhaled medication. Diagnostic protocol pediatr. 2019; 2: 51-64.

Updated review of the inhalation devices approved and marketed in Spain, along with the systematics for their handling.

 

Clinical case

 

21-month-old infant with episodes of coughing and wheezing coinciding with catarrhal processes, predominantly in autumn and winter.

Family history: healthy mother with no relevant history. Father has atopic dermatitis, rhinoconjunctivitis and allergic asthma due to Alternaria fungus, currently receiving immunotherapy treatment. Three-year-old brother has cow’s milk allergy and is under desensitizing therapy.

Personal history: adequately vaccinated, negative neonatal metabolic screening results (including cystic fibrosis). RSV positive bronchiolitis at 3 months of age, requiring PICU admission and non-invasive mechanical ventilation for 72 hours. Afterwards, during the first year of life he presented episodes of wheezing during four catarrhal episodes, manifesting good response to salbutamol. Ever since, he has received fluticasone (50 micrograms every 12 hours in a spacer chamber). In the following fall, she attended her pediatrician again because she was wheezing three times a month approximately. Likewise, his mother stated that at the beginning of September, the boy had an asthma attack, whose symptoms lasted 11 days, and that required ER care. He was then treated with salbutamol and methylprednisolone. After confirming correct adherence and inhaler technique, we increased the dose of fluticasone to 100 micrograms every 12 hours. In the following reviews up to now, three months later, we verify that he continues to be fully controlled.

 

 

Persistent cough

M.A. Zafra Anta*, C. Merchán Morales**
Topics on
Continuous Training


M.A. Zafra Anta*, C. Merchán Morales**

*Paediatrician. Paediatric Pulmonology Department. Assistant Doctor at University Hospital Fuenlabrada. Madrid. Professor-Honorary Collaborator at the Rey Juan Carlos University. Madrid.
** Resident of Paediatrics. University Hospital Fuenlabrada. Madrid

Abstract

Cough is a common medical consultation in paediatrics. A distinction must be made between recurrent acute cough episodes due to repeated respiratory infections and prolonged or chronic cough. An assessment should be made if coughing persists beyond 4 weeks. History taking must include particularities depending on age, mode of presentation, the epidemiological situation as well as if the cough is dry or productive and association of symptoms.

A decision algorithm is suggested for its management. When dealing with chronic cough, different aetiologies must be taken into consideration depending on age. For patients aged 14 years and younger, paediatric protocols will be followed. For adolescents aged 15 years or more, many authors suggest adhering to adult protocols. Training is advisable in order to recognise the sometimes termed “specific cough points”. Early diagnosis of underlying lung disorders (asthma, bronchiectasis, foreign body aspiration, persistent bacterial bronchitis and others) improves clinical outcomes.

 

Key words: Chronic cough; Pediatrics; Antitussive agents.

Palabras clave: Tos crónica; Pediatría; Antitusígenos.

 

Pediatr Integral 2021; XXV (1): 5 EN – 12 EN

 


 

Persistent cough

Prolonged and persistent or chronic cough (lasting more than 4 weeks) is a common reason for consultation; although this has varied during the SARS-COV-2 pandemic. Sometimes it is not sufficiently well studied by clinicians. Parents’ expectations should be taken into account, along with possible contact with smokers, and the most likely causes by age and area of care (rural, urban, region).

Cough is a very common reason for consultation in the Paediatric Department, especially in uncomplicated and self-limited acute viral infections. Prolonged and persistent or chronic cough is also a common reason for consultation. The estimated prevalence(1,2) is 5-7% in preschool age, and 12-15% in older children. Sometimes, however, it is not sufficiently well studied by clinicians. In this regard, acute slowly resolving cough and acute recurrent cough should be differentiated.

In recent years, a great deal of research has been carried out into the causes of chronic cough in adults and children(3). In children, for example, the most common in terms of incidence are post-infectious cough, persistent bacterial bronchitis, bronchiectasis, airway malacia and asthma. In contrast, causes typically encountered in adults, such as gastro-oesophageal reflux, asthma and upper airway syndrome, are predominant in adolescents. This difference is related to the inherent characteristics of children compared to adults regarding the respiratory tract, immune system, and nervous system(4).

In older children, particularly during adolescence, there appears to be a greater susceptibility for the development of psychosomatic symptoms, and the personal impact of chronic symptoms should not be ignored if they affect the social sphere(5,6).

A duration of more than 3-4 weeks is the threshold for distinguishing between acute and prolonged or chronic cough in most paediatric guidelines, and that is the period of time over which it should be evaluated and followed-up. In adults, this threshold is from 8 weeks onwards(7,8).

Only the British Thoracic Society considers cough in paediatric cases that lasts for more than 8 weeks to be chronic, thus bringing it into line with chronic cough in adults.

At consultation, there is usually a disagreement between the symptoms and the subjective assessment of parents, i.e. their expectations. Parental smoking is a factor found in persistent cough in up to 50% of children under 11 years of age, with this value decreasing somewhat in adolescents.

The different aetiologies for each age group must be taken into account when managing chronic cough. Thus, for patients aged 14 years and younger, paediatric protocols will be used, whereas for adolescents aged 15 years or older, many authors recommend that adult protocols be used(7,9,10).

The use of algorithms must take into account the place of assessment, in other words primary care or specialised paediatric pneumology, and when implementing protocols, it is essential to have training which is especially aimed at recognising the specific aspects of cough and their clinical significance.

Aetiology

After assessing whether it is an expected cough or within normal limits and frequency, in prolonged cough we can consider two main groups:

• Specific cough: secondary to pulmonary and extra-pulmonary pathophysiological processes.

Non-specific cough: with no apparent cause after an initial assessment(2,11,12).

From highest to lowest prevalence, in paediatric cases non-specific chronic cough usually has the following origin: post-infectious, cough as an asthmatic equivalent, psychogenic cough and chronic cough due to upper airway syndrome.

Isolated chronic cough without wheezing

This appears in apparently healthy children. Its causes include:

• Pulmonary abnormality or gastro-oesophageal reflux in infants.

• Cough as the only manifestation of asthma.

• Post-infectious cough.

• Respiratory infections: simple recurrent viral bronchitis or prolonged or pertusoid cough: Bordetella pertussis, Parapertussis, Mycoplasma, Chlamydia, Cytomegalovirus, Adenovirus, and Ureaplasma.

• In the current SARS Cov-2 or Covid-19 coronavirus pandemic, prolonged cough for more than one month is occasionally seen in paediatric cases(13). A study (Covid-19 PCR) is recommended in the event of an underlying disease or comorbidity and known contact with coronavirus.

• Rhinosinusitis. Allergic or not, which may or may not be associated with asthma.

• Psychogenic cough (somatic cough syndrome). Coughing habit (cough as a tic).

• Chronic upper airway cough syndrome (previously known as post-nasal drip syndrome).

Chronic cough in children with an underlying lung disease

• Cough as a manifestation of asthma.

• Gastro-oesophageal reflux, with or without aspiration.

• Persistent bacterial bronchitis (PBB).

• Chronic bronchitis: eosinophilic or not.

• Other diseases: tuberculosis, cystic fibrosis, foreign body aspiration, immunodeficiency, bronchopulmonary malformations, ciliary dysfunction, alpha-1-antitrypsin deficiency, tracheomalacia, obliterative bronchiolitis, chronic bronchitis, etc.

See Table I for the most frequent aetiology by age group.

Clinical Symptoms

The case history should be directed towards warning signs (neonatal age, association with general symptoms, aspiration and signs of suppurative bronchial inflammation), as well as by age, appearance, if the cough is dry or productive, and according to the epidemiological situation.

At consultation, questions about prolonged or chronic cough will be targeted(1,2,6) towards:

• Onset of the cough, age and whether or not it was associated with a respiratory infection.

• Characteristics of the cough: usual coughing style, to classify it as a dry or productive cough.

• If it is discontinuous, metallic, with stridor, paroxysmal, occurs regularly, is nocturnal or disappears during sleep, related to ingestion, has an abrupt onset, or is related to exercise or exertion (crying, laughing).

• If there is a possibility of distracting the cough or if there is a disproportionate concern about the cough. If it is related to: humidity, pets, smoke, environmental pollutants.

• Consideration should be given to: smoking, vaping (electronic cigarettes), use of inhaled drugs, use of irritants or chemical pollutants, such as hobbies (glues, paints) or in the workplace.

• Warning signs: onset in or near the neonatal period, or associated with general symptoms (weight loss, feeding difficulties, chronic diarrhoea, clubbing) or if there is haemoptysis.

• Other relevant information: fever, rhinorrhoea, catarrh, choking or aspiration of a foreign body, habitual vomiting, sputum production, risk of contact with tuberculosis or HIV, parental smoking, possible allergy and vaccination status.

• Personal history: prior diseases, especially pulmonary (pneumonia, congenital cardiopulmonary and autoimmune disorders, neoplasms, immunodeficiencies and medications). Where appropriate, geographic origin and possibility of endemic parasitic diseases: tuberculosis, histoplasmosis (USA), toxocara (China), echinococcosis (rural areas without adequate sanitary control).

• Previously performed additional examinations must be evaluated: chest X-rays or laboratory tests. Associated heart or neurological diseases or others, comorbidities (obesity, sleep apnoea, atopy). Also assess, where appropriate, what relieved the cough (inhaled corticosteroid, antibiotics, antihistamines)(1,2,6).

• When deciding treatment, it is important to take into account the impact on the child or adolescent’s quality of life in their various life environments, and it is necessary to enquire about this, as well as the parents’ expectations(5). In paediatric cases, there are multidimensional questionnaires (physical sphere, emotional and school and social functioning), such as the PedsQL or KIDSCREEN (kidscreen.org), which are recommended for use outside of research studies.

Complications

Complications due to chest-abdominal pressure caused by coughing, and due to the impact on social life, include: vomiting, petechiae, subconjunctival haemorrhage, cough syncope, urinary incontinence, musculo-skeletal pain, hoarseness, exhaustion, shyness, altered social relationships, insomnia, headache, dizziness, excessive perspiration, and concern that “something is wrong”, altered quality of life, lifestyle changes, abdominal hernias; and, less frequently: rib fractures (especially from 5th to 7th) induced by cough, pulmonary interstitial emphysema, pneumothorax, arrhythmias, etc.(1,2).

Diagnosis

This is based on a systematic clinical evaluation, with medical history and general physical examination. A first visit should be conducted to ask about: the existence of previous acute respiratory infections, the type of cough, the presence of signs of specific diseases and whether there are indicators of a specific cough or risk factors (infection, asthma, foreign body aspiration, etc.)(1,2,14).

This first visit, as well as the second assessment at 7-14 days, are key to initiating a diagnostic orientation and to consider additional targeted examinations.

Periodic reassessments and clinical control to determine the natural evolution or response to the initially targeted treatment are essential.

It should be remembered that prolonged coughing can improve spontaneously, thus overdiagnoses and unnecessary treatments should be avoided. In addition, at times, there may be two or more causes for the cough (e.g., asthma, rhinitis, persistent bacterial bronchitis, congenital airway abnormalities, and smoking).

Physical examination should be comprehensive, highlighting: weight/stature changes, skin colour, signs of atopy, thoracic deformity, clubbing, cardiopulmonary auscultation and a detailed examination of the pharynx, nose and ears, including the neck (explore the existence of goitre). Persistent auscultatory abnormality may be more associated with wet cough and other symptoms in addition to the cough(14,15)

Additional tests

In the diagnostic study for chronic cough, the different predominant aetiology according to age must be taken into account, and the diagnosis and targeted treatment for risk conditions must not be delayed. Paediatric protocols should be used for patients aged 14 years or younger.

A sequential approach can be considered guided by: case history, physical examination, and by performing an x-ray and a forced spirometry. See table II for study phases or levels of prolonged cough and the management algorithm (7,9,10). In older children and adolescents, if there is a suspected diagnosis of rhinitis or cough as an expression of “postnasal drip”, an initial therapeutic trial in primary care could be initiated.

Chest X-ray: a frontal projection is usually requested (AP up to 6 years, PA from that age onwards). In the event that the location of a lesion or imaging of the lung bases (e.g., with COVID-19) is required, two projections (PA and lateral) should be requested. This enables: asymmetries to be detected and congenital anomalies, radiopaque foreign bodies, infiltrates and atelectasis, increased cardiac silhouette to be seen. Other imaging studies depending on clinical suspicion: a sinus X-ray if sinupathy is suspected, ultrasound if the mediastinum is to be assessed, computed tomography can be useful if a more comprehensive study of the paranasal sinuses or alterations of the upper airway is desired, in the event of an altered chest X-ray or suspected lung disease. Magnetic resonance imaging will be useful in vascular disorders.

Spirometry at baseline and after bronchodilation in collaborators and those older than 5-6 years (if within normal parameters, consider spirometry after treatment with inhaled or systemic corticosteroids). In the event of suspected asthma, the record of variability in peak expiratory flow or peak-flow and bronchoprovocation tests (this is only available in specialised centres) may reveal reversible obstruction and hyper-responsiveness, or suggest anatomical abnormalities (stenosis of the airways or malacia).

• Measurement of exhaled nitric oxide (FeNO) levels, available in specialised centres, which helps the diagnosis of eosinophilic asthma. The cut-off point generally proposed as pathological is 50 ppm. It has a high negative predictive value of up to 93%.

Mantoux reaction. Tuberculosis screening could also be done with an IGRA (Interferon-Gamma Release Assay, such as Quantiferon)(16). Depending on the geographic area or risk of exposure, many physicians believe that tuberculin should be performed during the initial or first level assessment

Study of allergies to aeroallergens in those aged 3-4 years and older, with a suspected diagnosis, perhaps more specifically for fungi or mites. Studies according to availability: in vivo test such as skin tests (“prick”), or in vitro (blood test) Phadiatop, Cap system (specific IgE in allergen groups) or similar, which are feasible in many primary care centres. The pneumoallergen profile, or specific IgE for individualised allergens, would be used. A negative Phadiatop indicates a very low probability of allergic sensitisation.

Full blood count with ESR, basic biochemistry and TSH. In the event of a possible active infection. Goitre or thyroiditis may exceptionally be the cause of a cough as the only apparent initial symptom. During the SARS-Cov2 coronavirus pandemic, a serological study may be considered.

Gastroesophageal reflux study. Reflux and chronic cough only coincide in 3-8% of paediatric cases; this is more common in infants, who do not usually manifest with cough alone, and in older children or adolescents, especially if there are other symptoms such as heartburn or regurgitation. A therapeutic trial with a proton pump inhibitor (e.g., omeprazole) for 8 weeks can be considered, and if there is no response, referral to a paediatric gastroenterologist, who may suggest: a digestive endoscopy, an oesophageal pH measurement if appropriate with double channel intra-luminal impedance or others (upper digestive tract or oesophageal scintigraphy). Achalasia has been described as a cause of chronic cough.

• Assessment by an ENT specialist. If chronic sinupathy or upper airway alteration is suspected. With nasofibroscopy, a diagnosis of turbinate hypertrophy with rhinitis and polyposis can be made.

• If sputum can be obtained, a gram and cultures are performed. And if available: the study of eosinophils and metachromatic cells in the induced sputum. This contributes to the diagnosis of asthma and eosinophilic inflammation of the airway.

Serological tests: study of past viral infection, Mycoplasma pneumoniae, B pertussis, others.

• Determination of immunoglobulins and their classes, and other immunocompetence tests are suggested in cases of chronic infection. HIV infection study.

Bronchoscopy. Rigid bronchoscope or fibre-optic bronchoscope. The indications are: extraction of a foreign body, in the event of congenital anomalies, to obtain samples from the lower airways (bronchoalveolar lavage [BAL]), for culture and microscopy. An increase in lipids in bronchoalveolar lavage may indicate microaspiration. Fibre-optic bronchoscopy with BAL for cytological study, culture and PCR, constitutes the definitive test for the diagnosis of persistent bacterial bronchitis, although its limited availability means that it is reserved for those patients with an insufficient response to antibiotic treatment or who present a recurring PBB (more than 3 episodes per year).

Tracheobronchomalacia and PBB are often related (48-74% of cases).

The sweat test to rule out cystic fibrosis (CF) should be performed in the event of prolonged cough if there is malnutrition or a chronic productive cough. Over the past 11 years (since 2009), neonatal screening with immunoreactive trypsin in blood has become widespread in all of the Spanish territory. However, it must be considered that adolescents or migrants may not have undergone neonatal screening for CF.

• Primary ciliary dyskinesia is excluded only in specialised centres and requires in vivo and in vitro tests, electron microscopy and epithelial cultures. In the event of neonatal onset of cough, this should be considered in addition to situs inversus or congenital anomalies of the airway.

• In people with sickle cell disease, asthma can be a comorbidity that increases the severity of haemoglobinopathy; however, cough is not usually a sole symptom of complications.

Cough assessment in paediatric cases in primary care

It is important to assess whether cough is apparently normal or not, to consider the parents’ expectations, and to reassure most families. In the paediatric primary care consultation, in the presence of a child who is known, history taking of the characteristics of the cough, the presence of other signs of disease and the physical examination will likely guide us to suspect an underlying cause. Do not forget to consider persistent bacterial bronchitis as a non-uncommon cause of wet cough in children under 6 years of age. A chest X-ray should be performed, as well as additional examinations aimed at ruling out tuberculosis, coronavirus SARS-Cov 2, and spirometry in children aged more than 5 years.

The Phase I study and the left and central side of the algorithm should be used.

For appropriate treatment of prolonged cough in paediatric cases, an attempt should be made to clarify the cause, excluding serious underlying diseases. The presence of a long-standing cough should be investigated until it has fully disappeared or until a definitive diagnosis has been reached. Partial or temporary improvements in the cough should not be considered as a definitive therapeutic success. With an adequate approach, up to 80% of the causes of chronic cough in paediatric cases can be determined, and an improvement is seen in 90% of cases when treated(7,9-11,17).

Treatment

General measures

Oral hydration and nasal washes. Avoid exposure to tobacco and irritants.

Treatment with environmental measures

There is no evidence that home environmental management, including deionisers, vaporisers, humidifiers, air filters or others, are effective in the treatment of prolonged non-specific cough in paediatric cases.

Aetiological treatment

Once the cause is identified.

• Antibiotics for an established or suspected infection, for symptoms of PBB (2 weeks with amoxicillin-clavulanic acid at 80 mg/kg/day –adult dose, 875 mg amoxicillin/125 mg clavulanate–, as first choice), or cotrimoxazole, cephalosporins or macrolides (azithromycin 10 mg/kg/day, and up to 500 mg every 24 h, 3 days, or clarithromycin). Good response to antibiotic therapy also characterises and contributes to the diagnosis of PBB(12).

• Macrolides in the case of whooping cough (would not shorten the full-blown disease after 2 weeks of onset of the clinical coughing symptoms).

• For rhinitis or chronic cough syndrome of the upper airway, first-generation antihistamines due to their anticholinergic effect, or nasal corticosteroids.

• Treatment of allergic disease with environmental measures and inhaled steroids. For dry cough, the inhaled corticosteroid test for 4 weeks or leukotriene inhibitor (montelukast in those aged more than 6 months, as per the summary of product characteristics) as second option. If the anti-asthma treatment trial has failed, it must be discontinued.

• Surgical treatment for anatomical anomalies.

• Reflux can be treated medically (ranitidine in infants or omeprazole and lifestyles avoiding acidic drinks, colas, consumption immediately before bedtime, raised pillow), or surgically (Nissen fundoplication). In cases where there is no response to conservative treatment, chronic isolated cough will exceptionally be an indication for reflux surgery.

• Others: some authors have found a response to nebulised lidocaine for dry refractory cough in adults and children. In adults with chronic cough that does not respond to treatment, guanfacine or gabapentin have been used off-label (not indicated in the summary of product characteristics). Guanfacine is a selective alpha2A adrenergic receptor agonist, a centrally acting antihypertensive that is also approved for ADHD. Gabapentin is an anti-seizure drug that is also commonly used for the treatment of neuropathic pain.

Psychological support. Adequate information

For prolonged non-specific cough that is not indicative of any disease, it is important to inquire about the need for individualised information for the patient and family. Fears or prejudices may sometimes not be reported at first visits, and should be studied and properly addressed by counselling

The treatment for psychogenic cough (somatic cough syndrome) will be aimed at identifying the underlying conflict, reassuring parents and relatives, clearly explaining the benign nature of the process and not interrupting schooling. Relaxation techniques to decrease musculo-skeletal tension in the neck and chest can be successful. In extreme cases, refer to the psychiatrist and consider family hypnosis. Suggestion techniques may improve cough with a psychogenic origin: cough as a vicious circle triggered by irritants, which no longer works, concentrating on progressively spacing the cough, manoeuvres to relieve irritation, etc.(18).

Pharmacological cough treatment

Cough suppressants should be used sparingly, in specific circumstances, never as first-line treatment and respecting the indications and contraindications according to age.

Cough suppressants are sometimes helpful for intractable cough, especially due to viral infections, but they should not be used as first-line treatment for prolonged cough as they can delay a specific diagnosis and its treatment. The most common cough-suppressant drugs in primary care are collected in Table III.

Adolescents and their parents should receive information about the drugs and their partial effectiveness, as well as the likelihood of adverse effects associated with their use. Codeine is the most important centrally, but since 2015 the Spanish Medicines Agency has recommended not to use codeine in children under 12 years of age, or in adolescents aged between 12 and 18 years who present compromised respiratory function of a pulmonary, neuromuscular, traumatic or surgical aetiology. There are also a variety of home remedies with a predominant placebo effect.

Mucolytics, including bromhexine and acetylcysteine, should not be used for chronic cough as there is no evidence of efficacy and, in addition, they can cause irritation of the airway. Inhaled recombinant human DNase (alpha-dornase) may be effective in children with purulent viscous secretions and is indicated in cystic fibrosis.

• There are numerous current lines of research on new cough suppressants to act on the multiplicity of receptors or potential targets of the cough mechanism.

• For cough associated with treatment with antihypertensive angiotensin II inhibitors (ACEI), the latter must be suspended and, where appropriate, ARBs such as losartan or valsartan, which do not inhibit quinine metabolism, should be used. This is not common at a paediatric age.

Bibliography

The asterisks indicate the interest of the article in the authors’ opinion.

1.*** Pascual Sánchez MT, Urgelles Fajardo E. Tos persistente. Protoc diagn ter pediatr. 2017; 1: 1-14. Acceso el 26 septiembre de 2020. Available at: http://www.neumoped.org/docs/PROTOCOL_DIAGN_TERAP.pdf.

2.*** Chang AB, Marchant JM, Mallory GB, Hoppin AG. Approach to chronic cough in children. UpToDate 2020. Version October 2018. Accessed 26 October 2020.

3.* Kantar A, Seminara M, Why chronic cough in Children is different. Pulmonary Pharmacol Therapeut. 2019; 56; 51-6.

4. Mazzone SB, Undem BJ. Vagal afferent innervation of the airways in Health and Disease. Physiol Rev. 2016; 96: 975-1024.

5.* Newcombe PA, Sheffield JK, Petsky HL, Marchant JM, Willis C, Chang AB. A child chronic cough-specific quality of life measure: development and validation. Thorax. 2016; 71: 695-700.

6.** Zafra Anta MA. Tos crónica. En: Hidalgo Vicario MI, Rodríguez Molinero L, Muñoz Calvo MT. Eds. Medicina de la adolescencia. Atención integral. Majadahonda (Madrid): Ergón. 3ª ed; 2021. p. 585-90.

7.* Chang AB, Oppenheimer JJ, Weinberger M, Weir K, Rubin BK, Irwin RS. Use of management pathways or algorithms in children with chronic cough: Systematic reviews. Chest. 2016; 149: 106-19.

8.* Silvestri RC, Weinberger SE, Barnes PJ, King TE, Hollingsworth H. Evaluation of subacute and chronic cough in adults. UpToDate 2018. Version October 2018. Accessed 26 October 2020.

9.** Moneo Hernández I, Asensi Monzó M, Rodríguez Fernández-Oliva CR. Guía de Algoritmos en Pediatría de Atención Primaria. Tos crónica en AP. AEPap. 2017 (en línea). Consultado el 14 de noviembre de 2020. Available at: algoritmos.aepap.org.

10. O’Grady KAF, Grimwood K, Torzillo PJ, Rablin S, Lovie-Toon Y, Kaus M, et al. Effectiveness of a chronic cough management algorithm at the transitional stage from acute to chronic cough in children: a multicenter, nested, single-blind, randomised controlled trial. Lancet Chil Adolesc Health. 2019; 3: 889-98.

11.** Ridao Redondo M. Tos crónica en Pediatría. Pediatr Integral. 2016; XX(1): 7-15.

12.*** Busquets Monge RM, Castillo Corullón S, Aguilar Fernández AJ. Tos húmeda: bronquitis bacteriana persistente, enfermedad supurativa bronquial y bronquiectasias. Protoc diagn ter pediatr. 2017; 1: 15-29. Accessed 20 September 2020. Available at: http://www.neumoped.org/docs/PROTOCOL_DIAGN_TERAP.pdf.

13.** Devilla JG, Song E, Ouellette CP, Edwards MS, Torchia MM. Coronavirus Disease 2019 (COVID-19): clinical manifestations and diagnosis in Children. UptoDate 2020. Version november 2020. Accessed 12 november 2020.

14.* Zafra Anta MA. Semiología Respiratoria. Regreso a las Bases. Pediatr Integral. 2016; XX(1): 62.e1-62.e12.

15. Fernández Manso B, Albañil Ballesteros MR, Zafra Anta MA. Auscultación patológica persistente: no todo son broncoespasmos. Rev Pediatr Aten Primaria. 2019; 21: 173-9.

16. Zafra Anta MA, Rivero Calle I, García Begoña S. Tuberculosis (v.3/2020). En Guía-ABE. Infecciones en Pediatría. Guía rápida para la selección del tratamiento antimicrobiano empírico (en línea). Consultado el 3 de abril de 2020. Available at: https://www.guia-abe.es; https://guia-abe.es/files/pdf/Guia-ABE_Tuberculosis_v.3_2020.pdf.

17.** Seoane M, Sanz V, Villa JR. Tos crónica en la adolescencia. Adolescere. 2018; VI(3): 6-13.

18. Weinberger M, Lockshin B. When is cough functional, and how should it be treated? Breathe. 2017; 13: 22-30.

Recommended bibliography

– Pascual Sánchez MT, Urgelles Fajardo E. Tos persistente. Protoc diagn ter pediatr. 2017; 1: 1-14.

Article that summarises the fundamental characteristics and recommendations for the management of persistent cough in our environment, both in primary and specialised care.

– Chang AB, Marchant JM, Mallory GB, Hoppin AG. Approach to chronic cough in children. UpToDate 2020. Version October 2018.

Review with evidence-based medicine methodology of the clinical and epidemiological characteristics of prolonged cough, carried out by one of the leading international experts on the subject, which is constantly updated.

– Zafra Anta MA. Tos crónica. En: Hidalgo Vicario MI, Rodríguez Molinero L, Muñoz Calvo MT. Eds. Medicina de la adolescencia. Atención integral. Majadahonda (Madrid): Ergón. 3ª ed. 2021; p. 585-90.

Updated text specifically aimed at the 10-20 years age group.

– Moneo Hernández I, Asensi Monzó M, Rodríguez Fernández-Oliva CR. Guía de Algoritmos en Pediatría de Atención Primaria. Tos crónica en AP. AEPap. 2017 (on line). Available at: algoritmos.aepap.org.

Guide that serves as support and endorsement in the decision to carry out additional studies and referral where appropriate.

– Busquets Monge RM, Castillo Corullón S, Aguilar Fernández AJ. Tos húmeda: bronquitis bacteriana persistente, enfermedad supurativa bronquial y bronquiectasias. Protoc diagn ter pediatr. 2017; 1: 15-29.

Comprehensive analysis of chronic wet cough which is not as exceptional as one might think, and whose early treatment reduces the risk of chronic pulmonary complications.

– Chang AB, Oppenheimer JJ, Irwin RS; CHEST Expert Cough Panel. Managing Chronic Cough as a Symptom in Children and Management Algorithms: CHEST Guideline and Expert Panel Report. Chest. 2020; 158: 303-329.

Excellent review of the recent guidelines compared to the 2006 one with levels of evidence. It recommends following diagnostic guidelines by age. Diagnostic and treatment recommendations are presented.

– Morice AH, Millqvist E, Bieksiene K, et al. ERS guidelines on the diagnosis and treatment of chronic cough in adults and children. Eur Respir J. 2020; 55: 1901136 Available at: [https://doi.org/10.1183/13993003.01136-2019].

Chronic cough is recognised as a distinct entity. This European guide incorporates recent advances in pharmacological and non-pharmacological pathophysiology, diagnosis and the treatment of chronic cough.

Links of interest:

- GEMA Guide. Asthma management: https://www.gemasma.com/.

- Spanish Society of Paediatric Pneumology website: http://www.neumoped.org/.

- AEPap Respiratory Tract group: http://www.respirar.org/.

Clinical case

 

Tomás is 5 years old and visits primary care because, in the context of catarrhal symptoms, he started with a persistent wet cough more than 4 weeks ago. His parents report that he occasionally suffers from fatigue for a short time at the end of frequent coughing spells. He has no history of choking during a meal, or foreign body aspiration. There are no symptoms of Covid-19 in the family. There are no smokers in the home. He has not lost any weight. He has been in treatment for moderate asthma for two years with budesonide medium-dose chamber spray and montelukast. Upon examination he is afebrile, with disperse expiratory wheezing but no respiratory distress; the rest of the examination is normal. Treatment with salbutamol and an oral corticosteroid was initiated for 3 days, resulting in an improved auscultation but not the cough, which persisted. A chest X-ray was performed, with no findings reported. Poorly controlled asthma is considered and budesonide is replaced with a salmeterol-fluticasone spray with a chamber. There are no changes in the cough. Saturation with a finger pulse oximetry on several occasions is 96-99%. Oral amoxicillin-clavulanic acid was added to the treatment for 2 weeks, with improvement in the first 2-3 days, the cough disappearing and auscultation normalised.

 

 

 

Copyright © 2024 Sociedad Española de Pediatría Extrahospitalaria y Atención Primaria