Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Published: 08 April 2021
  • Antoni Torres   ORCID: orcid.org/0000-0002-8643-2167 1 , 2 , 3 , 4 ,
  • Catia Cilloniz   ORCID: orcid.org/0000-0002-4646-9838 1 , 2 , 3 , 4 ,
  • Michael S. Niederman   ORCID: orcid.org/0000-0003-0293-386X 5 ,
  • Rosario Menéndez 6 ,
  • James D. Chalmers 7 ,
  • Richard G. Wunderink   ORCID: orcid.org/0000-0002-8527-4195 8 &
  • Tom van der Poll 9  

Nature Reviews Disease Primers volume  7 , Article number:  25 ( 2021 ) Cite this article

205k Accesses

222 Citations

696 Altmetric

Metrics details

  • Respiratory tract diseases

Pneumonia is a common acute respiratory infection that affects the alveoli and distal airways; it is a major health problem and associated with high morbidity and short-term and long-term mortality in all age groups worldwide. Pneumonia is broadly divided into community-acquired pneumonia or hospital-acquired pneumonia. A large variety of microorganisms can cause pneumonia, including bacteria, respiratory viruses and fungi, and there are great geographical variations in their prevalence. Pneumonia occurs more commonly in susceptible individuals, including children of <5 years of age and older adults with prior chronic conditions. Development of the disease largely depends on the host immune response, with pathogen characteristics having a less prominent role. Individuals with pneumonia often present with respiratory and systemic symptoms, and diagnosis is based on both clinical presentation and radiological findings. It is crucial to identify the causative pathogens, as delayed and inadequate antimicrobial therapy can lead to poor outcomes. New antibiotic and non-antibiotic therapies, in addition to rapid and accurate diagnostic tests that can detect pathogens and antibiotic resistance will improve the management of pneumonia.

Similar content being viewed by others

introduction pneumonia case study

Clinical features for diagnosis of pneumonia among adults in primary care setting: A systematic and meta-review

introduction pneumonia case study

Etiological and epidemiological features of acute respiratory infections in China

introduction pneumonia case study

Respiratory viral infections in pragmatically selected adults in intensive care units

Introduction.

Pneumonia is a common acute respiratory infection that affects the alveoli and distal bronchial tree of the lungs. The disease is broadly divided into community-acquired pneumonia (CAP) or hospital-acquired pneumonia (HAP, which includes ventilation-associated pneumonia (VAP)) (Box  1 ). Aspiration pneumonia represents 5–15% of all cases of CAP; however, its prevalence amongst patients with HAP is not known 1 . The lack of robust diagnostic criteria for aspiration pneumonia may explain why the true burden of this type of pneumonia remains unknown 1 .

The causative microorganisms for CAP and HAP differ substantially. The most common causal microorganisms in CAP are Streptococcus pneumoniae , respiratory viruses, Haemophilus influenzae and other bacteria such as Mycoplasma pneumoniae and Legionella pneumophila . Conversely, the most frequent microorganisms in HAP are Staphylococcus aureus (including both methicillin-susceptible S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA)), Enterobacterales, non-fermenting gram-negative bacilli (for example, Pseudomonas aeruginosa ), and Acinetobacter spp. 2 , 3 . In health-care-associated pneumonia (HCAP), owing to patient risk factors, the microbial aetiology is more similar to that in HAP than to that in CAP. However, difficulties in standardizing risk factors for this population, coupled with the heterogeneity of post-hospital health care worldwide, suggest that the concept of HCAP has little usefulness, and indeed, HCAP was not included in recent guidelines for CAP and HAP 3 , 4 , 5 .

Differences in microbiology between CAP and HAP depend on whether pneumonia was acquired in the community or health care environment and on host risk factors, including abnormal gastric and oropharyngeal colonization. In addition, the aetiopathogenesis of CAP is different from that of HAP. In general, mild CAP is treated on an outpatient basis, moderately severe CAP in hospital wards, and severe CAP in intensive care units (ICUs) with or without mechanical ventilation 6 . The need for mechanical ventilation is used as a sub-classification of interest for prognosis and stratification in randomized clinical trials.

Both CAP 7 and HAP 4 can occur in either immunosuppressed or immunocompetent patients. To date, most research data have been based on studies of immunocompetent patients and, therefore, we rely on such sources in this Primer. However, CAP, HAP and VAP in immunosuppressed patients have attracted the attention of researchers, and more investigation is to come.

In this Primer, we cover and summarize the most important and recent updates related to epidemiology, pathophysiology, diagnostic screening, prevention, management, quality of life, and research perspectives. Additionally, owing to the profound impact of the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome (SARS) coronavirus 2 (SARS-CoV-2), we summarize the main features of SARS-CoV-2 pneumonia (Box  2 ).

Box 1 Classifications of pneumonia

Community-acquired pneumonia (CAP)

Pneumonia acquired outside the hospital in individuals who have not been hospitalized during the month prior to symptom onset.

Hospital-acquired pneumonia (HAP)

Pneumonia acquired after at least 2 days of hospitalization and when no suspicion of disease incubation before hospital admission is present.

Ventilator-associated pneumonia (VAP)

HAP occurring >48 h after endotracheal intubation.

Aspiration pneumonia

Pneumonia occurring as a result of inhalation of contents from the stomach or mouth into the lungs. It is best considered as part of the continuum between CAP and HAP, and not as a distinct entity.

Health-care-associated pneumonia (HCAP)

Pneumonia acquired in non-hospital care institutions.

Box 2 COVID-19 features

Frequent symptoms

Shortness of breath

Less-common symptoms

Hyposmia (decreased sense of smell) and hypogeusia (decreased sense of taste)

Sore throat

Rhinorrhoea (runny nose)

Muscle pain

Diarrhoea and vomiting

Main complications

Acute respiratory distress syndrome (ARDS)

Sepsis and septic shock

Multiple organ failure

Secondary infection

Epidemiology

Global incidence.

Data from the 2019 Global Burden of Diseases (GBD) study 8 showed that lower respiratory tract infections (LRTIs) including pneumonia and bronchiolitis affected 489 million people globally. Children of <5 years of age and adults of >70 years of age are the populations most affected by pneumonia, according to the 2019 GBD study 8 . In 2019, there were 489 million incident cases of LRTI, and 11 million prevalent cases of LRTI. In the 2016 GBD study, the global incidence of LRTI was 155.4 episodes per 1,000 adults of >70 years of age and 107.7 episodes per 1,000 children of <5 years of age 9 . Finally, aspiration pneumonia contributes 5–15% of all cases of CAP and is associated with worse outcomes, especially in older patients with multiple comorbidities 10 , 11 . There is a lack of data about the incidence of aspiration pneumonia in patients with HAP 1 , 12 .

In the USA, the Etiology of Pneumonia in the Community (EPIC) study 13 found that the annual incidence of CAP was 2.4 cases per 1,000 adults, with the highest rates amongst adults of 65–79 years of age (6.3 cases per 1,000 individuals) and those of ≥80 years of age (16.4 cases per 1,000 people). In Europe, the annual incidence of CAP has been estimated at 1.07–1.2 cases per 1,000 people, increasing to 14 cases per 1,000 people amongst those of ≥65 years of age and with a preponderance in men 14 . Differences in epidemiology between the USA and Europe might be explained by the higher proportion of the adult population who received the pneumococcal vaccine in the USA (63.6% of adults of ≥65 years of age, compared with pneumococcal vaccination rates of 20% to 30% in most European countries 15 , 16 ); in addition, in 2015 in the USA, ~69% of adults of ≥65 years of age had received an influenza vaccine within the previous 12 months. Another possible contributing factor is the decreased rate of smoking in the USA: between 2005 and 2016, the percentage of smokers who quit increased from 51% to 59% 17 . Finally, marked differences between US and European health systems can influence epidemiological data.

The South American Andes region had the highest incidence of adults of >70 years of age with LRTIs (406.5 episodes per 1,000 people), while South Asia had the greatest number of LRTI episodes amongst adults of >70 years of age. Incidence per global region was 171.1 per 1,000 people in Central Europe, eastern Europe and central Asia; 234.4 per 1,000 people in Latin America and the Caribbean; 130.8 per 1,000 people in Southeast Asia, eastern Asia and Oceania; 246.6 per 1,000 people in North Africa and the Middle East; and 229.3 per 1,000 people in sub-Saharan Africa 9 .

According to the 2016 GBD study 9 , Oceania had the highest incidence of LRTI in children (171.5 per 1,000 children of <15 years of age), while South Asia had the greatest number of LRTI episodes amongst children of <5 years of age. Incidence per global region was: 107.1 per 1,000 children in Central Europe, eastern Europe, and central Asia; 94.9 per 1,000 children in Latin America and the Caribbean; 120.4 per 1,000 children in Southeast Asia, eastern Asia and Oceania; 133.2 per 1,000 children in North Africa and the Middle East; and 100.6 per 1,000 children in sub-Saharan Africa.

The epidemiology of pneumonia is constantly changing, owing to the development of molecular diagnostic tests, novel antimicrobial therapies and implementation of preventive measures. Since the beginning of the 21st century, pneumonia has been the most common cause of pandemic infections that have effects on its own epidemiology. In the 2009 influenza pandemic, the influenza virus A H1N1 infected ~200 million people and caused almost 250,000 deaths, with infectivity higher in children than in adults 18 . By contrast, in the current SARS-CoV-2 pandemic, 106 million people had been infected and >2 million had died worldwide by 9 February 2021. However, unlike the influenza virus A H1N1, SARS-CoV-2 affects adults more often than children 19 .

The annual incidence of HAP in adults ranges from 5 to 10 cases per 1,000 hospital admissions globally, whereas VAP affects 10–25% of all patients on mechanical ventilation 3 . HAP is the second most frequent hospital infection after urinary tract infection, and VAP is the most common cause of nosocomial infection and death in the ICU 3 , 4 . The incidence of HAP is highest amongst immunocompromised, post-surgical and older patients 20 . In the USA, the incidence of VAP is estimated to range from 2 to 6 cases per 1,000 ventilator-days 21 , and the incidence of non-ventilator-associated HAP is estimated to be 3.63 cases per 1,000 patient-days 22 . A 2018 systematic review and meta-analysis of studies of VAP in adults from 22 Asian countries found an overall incidence of 15.1 cases per 1,000 ventilator-days 23 . In 2015, data from the prospective French multicentre OUTCOMEREA database (1996–2012) indicated that the risk of VAP was ~1.5% per ventilator-day, decreasing to <0.5% per day after 14 days of mechanical ventilation 24 .

The 2019 GBD study 8 showed that LRTI was responsible for >2.49 million deaths, with mortality highest amongst patients of >70 years of age (1.23 million deaths). These data indicate that mortality due to LRTI is higher than mortality due to tuberculosis (1.18 million deaths) and HIV (864,000 deaths), making it the leading cause of infectious disease mortality worldwide. Indeed, data from a systematic review and meta-analysis on the global and regional burden of hospital admissions for pneumonia estimated that 1.1 million pneumonia-related hospital deaths occurred in 2015 amongst older adults 25 .

In 2016, the highest LRTI mortality rates amongst children of <5 years of age were in the Central African Republic (460 deaths per 100,000 children), Chad (425 deaths per 100,000) and Somalia (417 deaths per 100,000) 9 . Interestingly, data from the 2017 GBD study 26 showed that mortality due to LRTI decreased by 36.4% between 2007 and 2017 for children of <5 years of age, whereas it increased by an estimated 33.6% in adults of ≥70 years of age. LRTI-related deaths amongst children have substantially reduced as a result of the implementation of vaccines (against S. pneumoniae and H. influenzae ), antibiotic therapy, the continuous improvements in education, nutrition, water, sanitation and hygiene, and female empowerment. Nevertheless, in many areas the progress is slow; Nigeria, India, Pakistan, Ethiopia and the Democratic Republic of Congo are the five countries with the highest child mortality 27 .

Conversely, the increased mortality in adults of >70 years of age might be associated with the increasing longevity of the frail older population, chronic diseases, comorbidities 28 , multiple medication use and functional disability, especially in high-income countries. In low-income countries, the high mortality is associated with the effect of air pollution; smoke and alcohol consumption are the main risk factors for pneumonia in this age group.

Globally, amongst children and adults, mortality in those with CAP is related to the treatment setting: <1% in outpatient care, ~4–18% in hospital wards and up to 50% in the ICU 29 , 30 , 31 . However, in adults, age and comorbidities influence mortality. A study that investigated the effects of age and comorbidities on CAP mortality found a mortality of 5% in patients of <65 years of age, 8% amongst patients of 65–79 years and 14% amongst patients of ≥80 years of age 32 , and these rates increased to 20%, 42% and 43%, respectively, in patients with more than one comorbidity. On the basis of studies on long-term mortality across 1–10 years 33 , 34 , 35 , approximately one in three adults will die within one year of being hospitalized with CAP 36 . The estimated in-hospital mortality in patients with chronic obstructive pulmonary disorder (COPD) and CAP has been reported to be 6% during hospitalization and 12%, 24% and 33% within 30 days, 6 months and 1 year from discharge, respectively 37 . Interestingly, 30-day mortality amongst those with pneumococcal pneumonia remained fairly stable in a 20-year study 33 , and this was further confirmed in a review on the burden of pneumococcal CAP in Europe 38 .

Globally, HAP and VAP are considered the leading causes of death due to hospital-acquired infection 39 , 40 , 41 . The estimated global mortality due to HAP is 20–30%, whereas global mortality due to VAP is 20–50% 20 , 42 . Mortality due to VAP in the USA was ~13% 4 . By contrast, a prospective study in central Europe 43 indicated that 30-day mortality due to VAP was 30%. In a large French cohort of patients admitted to the ICU for >48 h, both non-ventilator-associated HAP and VAP were associated with an 82% and a 38% increase in the risk of 30-day mortality, respectively 44 . However, analysis of data from trials on antibiotic therapy for bacterial HAP and VAP to characterize all-cause mortality showed that mortality differed notably within and across studies; all-cause mortality at day 28 was 27.8% in bacterial HAP, 18% in bacterial VAP and 14.5% in non-ventilation-associated bacterial HAP 45 .

In a systematic review and meta-analysis 10 , aspiration pneumonia was significantly associated with increased in-hospital mortality (relative risk 3.62) and 30-day mortality (relative risk 3.57) in patients with CAP treated outside of the ICU. One of the largest studies in aspiration pneumonia demonstrated that mortality in patients with aspiration pneumonia (29%) was more than twice that in patients with CAP (12%) 11 .

Risk factors and differences in epidemiology

Children of <5 years of age 46 and older adults 13 , particularly those of of ≥65 years of age and with comorbidities 14 , 47 , have an increased risk of CAP (Table  1 ). In children, prematurity, malnutrition, household air pollution, ambient particulate matter or suboptimal breastfeeding are the main CAP-related risk factors 48 . In adults, respiratory disease (for example, COPD), diabetes mellitus, cardiovascular disease and chronic liver disease are the most frequent comorbidities that increase the risk of CAP 14 . Of note, men have a higher risk of CAP than women, which may be explained by differences in anatomy, and behavioural, socioeconomic and lifestyle factors 49 .

A US study on the incidence, outcomes and disease burden in >18,000 hospitalized patients with COPD 37 found that, during the 2-year study, 3,419 patients had pneumonia; the annual incidence for CAP was 93.6 cases per 1,000 in the COPD population. In patients without COPD, the incidence was 5.09 cases per 1,000. In the USA, 506,953 adults with COPD are estimated to be hospitalized every year due to pneumonia 37 .

Immunocompromised patients have a higher risk of CAP than the general population 7 , 14 . A secondary analysis of an international, multicentre study from 54 countries worldwide found that almost one in five patients hospitalized with CAP were not immunocompetent 7 . Amongst patients with CAP, 18% had one or more risk factors for immunodeficiency, with chronic steroid use (45%), haematological cancer (25%) and chemotherapy (22%) being the most frequent.

Several studies have also demonstrated an association between lifestyle factors and the risk of CAP, including smoking, high alcohol consumption, being underweight (owing to under-nutrition or underlying conditions that compromise the immune response), living conditions, such as a large household or regular contact with children, and others 14 . Smoking is associated with colonization by pathogenic bacteria and an increased risk of lung infection, especially by S. pneumoniae 50 . Consumption of 24 g, 60 g and 120 g of pure alcohol daily (one standard alcoholic beverage equals 10 ml or 8 g of pure alcohol, and it is the approximate amount of alcohol that the average adult can process in an hour) resulted in relative risks for CAP of 1.12, 1.33 and 1.76, respectively, compared with no consumption 51 . In addition, exposure to air pollution may increase the risk of pneumonia in the short and long term; a study in 345 hospitalized patients with CAP and 494 controls (patients who were admitted in the same period but for non-pneumonia reasons) demonstrated that long-term exposure (1–2 years) to high levels of air pollutants (particulate matter 2.5 μm and nitrogen dioxide) was associated with increased hospitalization in those of ≥65 years of age 52 .

Factors that increase the risk of HAP can be categorized into patient-related and treatment-related groups (Table  1 ). Oropharyngeal colonization is the main mechanism underlying HAP. However, much attention has been shifted to oropharyngeal colonization in critically ill patients (present at ICU admission or occurring during ICU stay) 53 . A study from Japan investigating oral colonization in residents in long-term care facilities found that 38% of these individuals were colonized with antibiotic-resistant pathogens, mainly Acinetobacter spp., Enterobacterales and Pseudomonas spp. The presence of these pathogens represents a potential risk for pneumonia 54 . Indeed, current international guidelines have suggested that previous colonization by antibiotic-resistant pathogens be considered when identifying patients with an increased risk of HAP due to such pathogens 3 , 4 .

Colonization and biofilm formation were present within 12 h of intubation and remained for >96 h in most patients 55 . Underpinning an important association between intubation and VAP pathogenesis, this study also showed that colonization in patients undergoing mechanical ventilation occurred in the oropharynx and stomach first, followed by the lower respiratory tract and, thereafter, the endotracheal tube 55 . Intubation and mechanical ventilation can increase the risk of developing VAP by 6–21-fold, with the highest risk within the first 5 days of intubation 53 . Endotracheal tubes enable the direct entry of bacteria into the lower respiratory tract, interfere with normal host defence mechanisms and serve as a reservoir for pathogenic microorganisms.

Multiple risk factors are related to aspiration pneumonia, each one increasing the chance of gastric contents reaching the lungs. The most frequent of these factors are impaired swallowing, decreased consciousness and an impaired cough reflex 1 (Table  1 ).

Microbial aetiology

Knowledge of pathogens associated with pneumonia is crucial to provide more targeted empiric antibiotic therapy, prevent the emergence of antimicrobial resistance through selection pressure and reduce health-care-associated costs.

The microbial aetiology of CAP differs by its severity at clinical presentation and by season 2 , 56 , 57 , 58 . However, the microbial aetiology of CAP is not detected in ~50% of patients; possible reasons include the failure to obtain a respiratory sample adequate for culture or before the initiation of antibiotic therapy and the inconsistent availability of newly improved molecular tests 59 . S. pneumoniae remains the most frequent pathogen in CAP, although a study in North America found that its incidence has decreased owing to the introduction of polysaccharide vaccines 60 and a reduced smoking rate 61 , 62 . No such decrease has been observed in Europe 2 , 63 , 64 , 65 (Fig.  1 ).

figure 1

a | Aetiology of community-acquired pneumonia (CAP) in the adult population in the USA from 2010 to 2012 (from 2,488 cases) 9 . b | Aetiology of CAP in the adult population in Europe from 2003 to 2014 (from 3,854 cases) 6 . Possible reasons that may explain the challenge in identifying the aetiology of pneumonia include difficulty in obtaining samples from the lower respiratory tract, the effect of antibiotic use prior to sample collection and low sensitivity of some diagnostic tests.

In a small proportion of patients, CAP is caused by MRSA and antibiotic-resistant gram-negative bacteria (for example, P. aeruginosa and Klebsiella pneumoniae ) 2 , 66 . As antibiotic resistance complicates clinical management, clinicians need to recognize risk factors for these pathogens and initiate adequate empirical therapy in response (Box  3 ). The main risk factors for multidrug-resistant (MDR) pathogens in CAP include immunosuppression, previous antibiotic use, prior hospitalization, use of gastric acid-suppressing agents, tube feeding and non-ambulatory status 67 . Various scoring systems can help to determine the risk of infection by antibiotic-resistant pathogens.

The P. aeruginosa , extended-spectrum β-lactamase (ESBL)-positive Enterobacterales and MRSA (PES) score 68 is based on several risk factors, including age 40–65 years and male sex (one point each), age >65 years, previous antibiotic use, chronic respiratory disorder and impaired consciousness (two points each), and chronic renal failure (three points). The PES score has been validated in general wards, ICUs and a very old population (age ≥80 years). One study 69 demostrated that there is an 80% probability of detecting a PES pathogen with the PES score, demonstrating good accuracy of the score. In another study 70 , the accuracy of the PES score in patients of ≥80 years of age with CAP was ~64%, highlighting differences in clinical characteristics of this population who are more susceptible to infections, recurrent pneumonia and sepsis.

The drug resistance in pneumonia (DRIP) score 71 is based on both major and minor risk factors. Major risk factors (two points each) include previous antibiotic use, residence in a long-term care facility, tube feeding and prior infection by a drug-resistant pathogen (within the past year). Minor risk factors (one point each) include hospitalization within the previous 60 days, chronic pulmonary disease, poor functional status, gastric acid suppression, wound care and MRSA colonization (within the past year).

The use of new diagnostic molecular techniques has led to an increased interest in the role of respiratory viruses as potential aetiological agents in CAP. Recent studies have reported that respiratory viruses account for 7–36% of CAP cases with a defined microbial aetiology 13 , 72 , 73 . A recent study from China reported that in patients with viral CAP, influenza virus, non-influenza virus and mixed viral infections were the cause of CAP in 63%, 27% and 10% of patients, respectively (Fig.  2 ). The outcomes were similar between patients with CAP due to influenza virus and those with CAP due to non-influenza viruses, although in patients with CAP due to non-influenza viruses the incidence of complications was higher 74 . In another study, 3% of all patients with a diagnosis of CAP admitted to the emergency department had pure viral sepsis 75 . Viral sepsis was present in 19% of those admitted to ICU, and sepsis was present in 61% of all patients with viral CAP.

figure 2

a | Aetiology of community-acquired pneumonia (CAP) in the adult population in India from 2013 to 2015 (from 225 cases) 54 . b | Aetiology of CAP in the adult population in China from 2004 to 2005 (from 593 cases) 55 . Possible reasons that may explain the challenge in identifying the aetiology of pneumonia include difficulty in obtaining samples from the lower respiratory tract, the effect of antibiotic use prior to sample collection and low sensitivity of some diagnostic tests. MRSA, methicillin-resistant Staphylococcus aureus ; MSSA, methicillin-susceptible Staphylococcus aureus .

Respiratory viruses are detected in more than half of children with CAP 76 . Respiratory viruses were the most frequent cause of pneumonia (66%) in children with an aetiological diagnosis in the USA, with respiratory syncytial virus, rhinovirus and metapneumovirus being the most common ones 76 . Bacterial pathogens were the cause of CAP in 8% of patients, with S. pneumoniae and S. aureus being the most common bacteria. Bacteria–virus co-infections were detected in 7% of patients.

Box 3 Pathogen-specific risk factors

Streptococcus pneumoniae : Dementia, seizure disorders, congestive heart failure, cerebrovascular disease, chronic obstructive pulmonary disease (COPD), HIV infection, overcrowded living conditions and smoking

Legionella pneumophila : Smoking, COPD, compromised immune system, travel to outbreak areas, residence in a health-care facility and proximity to cooling towers or whirlpool spas

MRSA : Previous MRSA infection or colonization, residence in a nursing home or long-term care facility and prior hospitalization within the previous 90 days

Pseudomonas aeruginosa : Pulmonary comorbidity

Enterobacterales: Residence in a nursing home

MRSA, methicillin-resistant Staphylococcus aureus

Data on microbial aetiology of HAP have mostly been obtained from patients with VAP. However, studies in patients with HAP or VAP with known microbial aetiology have shown that both HAP and VAP have similar microbial aetiology, with P. aeruginosa and S. aureus being the most frequent pathogens. Other pathogens such as Acinetobacter spp. and Stenotrophomonas spp. are more frequently reported in VAP 4 , 77 .

Antibiotic resistance is the main concern with HAP and VAP. Assessing risk factors for MDR organisms (resistant to at least one agent in three or more groups of antibiotics), extensively drug-resistant organisms (XDR; resistant to one or more agents in all but one or two antibiotic groups) and pandrug-resistant organisms (resistant to almost all groups of approved antibiotics) is central to managing patients with these pathogens 78 . In general, we can classify the risk into three categories: (1) local epidemiology (for example, ICU with high rates of MDR pathogens); (2) patient risk factors (including structural pulmonary diseases (for example, bronchiectasis), antibiotic use during the 90 days prior to HAP or VAP onset, hospitalization (2–5 days) during the 90 days prior to HAP or VAP onset, septic shock at VAP onset, acute respiratory distress syndrome (ARDS) preceding VAP, at least 5 days of hospitalization prior to VAP onset, and acute renal replacement therapy prior to VAP onset) 42 ; and (3) previous colonization or infection with MDR pathogens 42 . Anaerobes and gram-negative bacilli (for example, E. coli , K. pneumoniae and P. aeruginosa ) are the most frequent microorganisms found in aspiration pneumonia 1 .

Mechanisms/pathophysiology

From colonization to infection.

The mechanisms that drive LRTIs have become increasingly known. Most instances of bacterial pneumonia are caused by microorganisms that translocate from the nasopharynx to the lower respiratory tract 79 , 80 . Bacteria enter the nasopharynx after shedding from a colonized individual. Pathogens can spread between individuals via direct or indirect contact, droplets and aerosols 81 . Transmission success depends on many variables, including environmental conditions, gathering of people and host factors, such as the distribution of pattern recognition receptors in the epithelial cells of the airways 81 . Pathogen adherence to the upper airway epithelium is a crucial first step in colonization and subsequent infection. Once in the nasopharynx, bacteria escape from mucus and attach to the epithelium using multiple strategies to evade host clearance, including expression of host-mimicking or antigenically varying molecules 82 (that is, molecules that imitate the structure of host molecules or can vary their antigens to avoid recognition by host immune cells). Microorganisms gain entry to the lower airways through inhalation or, less frequently, by pleural seeding from blood. Selection of colonizing mutants that can evade immune clearance is considered to precede infection 79 . Infection occurs when host defences are impaired and/or there has been exposure to a highly virulent microorganism or a large inoculum. Several factors can facilitate the transition from colonization to infection, including preceding viral infection and chronic lung diseases. Other mechanisms involved in the increased susceptibility to infection include loss of barrier integrity and impaired host defences due to complex interactions amongst anatomical structures, microorganisms (and their virulence factors) and the host immune system 79 , 80 , 83 .

Of note, it has become clear that healthy lungs are not sterile; instead, they harbour a unique microbiota that includes ~100 different taxa 84 . The main genera in healthy lower airways are Prevotella , Streptococcus , Veillonella , Fusobacterium and Haemophilus 84 . The pathogenesis of pneumonia has been suggested to include a change in the lung microbiota, from a physiological, homeostatic state to dysbiosis, in association with a low microbial diversity and high microbial burden, and with corresponding immune responses 84 , 85 To further support this concept, longitudinal lung microbiota studies are required to document transitions from homeostatic to dysbiotic states during the development and resolution of pneumonia. An additional area of research lies in analysing the virome and mycobiome in airways and their influence on host defence against pneumonia. The mechanisms by which lung microbiota affect immunity in the airways have been partially elucidated. Bacteria present in the upper airways that potently stimulate nucleotide-binding oligomerization domain-containing (NOD)-like receptors ( Staphylococcus aureus and Staphylococcus epidermidis ) increase resistance to pneumonia through NOD2 and induction of release of granulocyte–macrophage colony-stimulating factor 86 .

Mechanisms of infection

A general mechanism of infection of the lower airways is difficult to define. The many different microorganisms that can cause pneumonia do not seem to express specific features. Even in specific populations (for example, young children, hospitalized patients, older individuals), a spectrum of pathogens, rather than a specific microorganism, can cause pneumonia. This finding has led to the assumptions that the development of pneumonia largely depends on the host response to the microbe in the airways, with pathogen characteristics playing a less prominent role 83 . Nonetheless, virulence factors expressed by microorganisms do contribute to the ability of specific pathogens to cause pneumonia 79 , 80 . For example, pneumolysin, a virulence factor expressed by S. pneumoniae , is a member of the cholesterol-dependent cytolysin family that can form large pores in (and thereby injure) eukaryotic cells with cholesterol-containing membranes 87 . S. aureus expresses several virulence factors, such as α-haemolysin (also known as α-toxin), a pore-forming toxin that causes cell death via activation of the inflammasome 88 . α-Haemolysin binds to the disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and results in disruption of the barrier function of the respiratory epithelium 88 . Finally, toxins secreted by the type III secretion system are a key element in P. aeruginosa virulence in the lung. Genes encoding type III-secreted toxins are induced in P. aeruginosa upon contact with host cells, eliciting a plethora of effects, including cytotoxicity 89 .

Once an LRTI has occurred, the maintenance of lung homeostasis whilst in the presence of microbes depends on an adequate balance between two seemingly opposing processes, immune resistance and tissue resilience, that are largely mediated by the same cell types. Whilst immune resistance seeks to eliminate invading microbes, tissue resilience strives to prevent or resolve tissue damage caused by the immune response, the pathogen or both 83 . The organized actions of immune resistance and tissue resilience determine whether and how an LRTI progresses or resolves. Inadequate or unfitting immune responses can result in adverse outcomes, such as ARDS, defined as the acute onset of non-cardiogenic pulmonary oedema, hypoxaemia and the need for mechanical ventilation 90 , 91 . Unbalanced immune responses during pneumonia can also result in extrapulmonary complications, some of which can occur up to years after the respiratory illness (see below).

Immune resistance

Anatomical barriers present the first line of defence against pneumonia. Mucociliary clearance, mediated by mucous and liquid layers and cilia on the surface of respiratory epithelial cells, is considered the primary innate defence mechanism 92 . The respiratory epithelium produces a robust barrier composed of secretory products, surface glycocalyces and membranes, and intercellular junctional proteins linked to the actin cytoskeleton 92 . Cell-associated and secreted mucins form a polymeric glycoconjugate layer that can bind and transport pathogens from the airways 92 . The branching bronchial tree provides an additional defence mechanism by preventing particles of >3 µm in diameter from entering the lower airways 92 . If microbes do reach the lower respiratory tract, the host defence becomes shaped by an interplay between resident and recruited immune cells and mechanisms (Fig.  3 ).

figure 3

Immune resistance aims to eradicate microorganisms that invade the airways. Respiratory epithelial cells are covered by cell-associated and secreted mucins that form a layer of polymeric glycoconjugates that remove pathogens from the airways. The epithelium can also remove pathogens through phagocytosis and intracellular killing. The quiescent alveolar space contains many alveolar macrophages that, upon activation, can phagocytose and kill pathogens, which is improved by apoptosis. Innate lymphoid cells (ILCs) are tissue-resident cells populating the pulmonary mucosa. Together with natural killer cells, ILCs boost host defence during airway infection. Neutrophils migrate to the airways attracted by chemotactic proteins released by respiratory epithelial cells and alveolar macrophages; these chemotactic proteins also promote the recruitment of other leukocyte subsets. The lung contains a marginated pool of neutrophils tethered to the vasculature, enabling rapid neutrophil recruitment into tissue upon infection. Adequate pulmonary immunity entails neutrophil-mediated killing of invading microbes by several effector mechanisms, including the release of neutrophil extracellular traps (NETs). Platelets can form complexes with leukocytes, facilitating NET formation and the release of microbicidal agents. Resident memory T (T RM ) cells are generated after exposure to pathogens and reside in the quiescent lung. ATI, alveolar type I cell; ATII, alveolar type II cell; BASC, bronchioalveolar stem cell; CXCs, CXC chemokines; DAMPs, damage-associated molecular patterns; NF-κB, nuclear factor-κB; RBC, red blood cell; SP, surfactant protein; T1R, G-protein-coupled sweet taste receptor; T2R, G-protein-coupled bitter taste receptor; TLR, Toll-like receptor.

Innate immunity

Various innate immune cells reside in quiescent airways to provide the next line of defence against pathogens. Lung epithelial cells can be triggered through a variety of receptors that recognize not only pathogens but host-derived molecules as well, including damage-associated molecular patterns (released upon cell injury) and cytokines. Many pattern recognition receptors (for example, toll-like receptors) then induce nuclear factor ĸB, which is a major driver of protective immunity in the epithelium 93 , 94 . In the alveoli, surfactant proteins SP-A and SP-D produced by type II epithelial cells can directly inhibit microbes 95 . Recently, G-protein-coupled bitter taste receptors (T2R) and sweet taste receptors (T1R) were identified in respiratory epithelial cells 96 ; bacterial quorum-sensing molecules can trigger bitter taste receptors, whilst sugars can activate sweet receptors, and these interactions may then modify host defence mechanisms 97 . IL-17 and IL-22 mediate protection during pneumonia largely through epithelial cell activation 98 . IL-17 stimulates the epithelium to secrete antimicrobial proteins and CXC chemokines that trigger neutrophil recruitment. The protective properties of IL-22 are linked to its function in stimulating epithelial cell proliferation, which is indispensable for repair following injury 99 .

Alveolar macrophages (AMs), which reside on lower airway surfaces, have essential roles in both immune resistance and tissue resilience 100 . During homeostasis, they limit the effect of potentially noxious environmental stimuli through anti-inflammatory effects. The crucial role of AMs in immune resistance during pneumonia is illustrated by studies showing impairment of the host defence when AM function is disrupted 94 . Microbes can activate AMs via several pattern recognition receptors and nuclear factor ĸB, leading to the production of pro-inflammatory cytokines that orchestrate subsequent, innate immune responses necessary for resistance. In addition, stimulated by AM apoptosis, activated AMs can phagocytose and kill pathogens 101 . By contrast, AM death via non-apoptotic pathways, such as necroptosis, impairs antibacterial defence during pneumonia 102 . The complex role of necroptosis in the host response to bacterial infection is illustrated by reports linking necroptosis to exaggerated inflammation and impaired bacterial clearance during S. aureus pneumonia 103 , whereas it has a protective, anti-inflammatory effect associated with improved bacterial clearance during systemic S. aureus infection 104 . Local conditions may instruct AMs in providing the most suitable response.

Innate lymphoid cells (ILCs) serve as counterparts to T cells by regulating immune responses via the production of effector cytokines and by influencing functions of other innate and adaptive immune cells 105 . These cells are especially abundant on the mucosal surfaces of the lung. There are three major groups of ILCs, namely, ILC1, ILC2 and ILC3. ILC classification reflects these cells’ capacity to secrete types 1, 2 and 17 cytokines, respectively. Beneficial roles for ILC1s and ILC2s have been reported in viral pneumonia models 106 , 107 ; lung ILC3s have a protective role in pneumonia by secreting IL-17 and IL-22 (refs 108 , 109 ). Mucosal-associated invariant T cells are other innate-like T lymphocytes that are abundant in the lung mucosa 110 . These cells probably have a role in protective immunity during airway infection through a variety of mechanisms, including production of pro-inflammatory cytokines, macrophage activation and recruitment of effector helper and cytotoxic T cells 111 .

When resident cells are unable to eradicate invading pathogens, mechanisms are activated to attract additional effector cells to the site of infection. Neutrophils are the first and most profusely recruited cells in response to infection 112 . Primed neutrophils have a strongly increased capacity to phagocytose microbes and initiate a respiratory burst response 112 . In addition, neutrophil products, such as elastase, proteinase 3 (also known as myeloblastin), cathepsin G, lactoferrin and LL-37, exert potent antimicrobial activities 113 . Neutrophil extracellular traps, comprising decondensed chromatin fibres that carry histones and antimicrobial peptides, are also released to kill pathogens 113 . The crucial role of neutrophils in pulmonary immune resistance is illustrated by the increased susceptibility found in patients with neutropenia or neutrophil deficiencies and mouse pneumonia models, in which neutrophil depletion has been shown to exacerbate infection with several pathogens 112 . In addition to AMs, newly recruited inflammatory monocytes–macrophages are involved in immune resistance during pneumonia 114 . In mice, induction of K. pneumoniae -associated pneumonia has been found to lead to the recruitment of inflammatory monocytes to the lungs where they mediate the influx of protective ILCs producing IL-17 through the release of tumour necrosis factor 109 . Innate-like B1 B cells mainly reside in the pleural space. In response to infection, B1a B cells migrate to the lung parenchyma to produce polyreactive immunoglobulin M and contribute to protective immunity 115 . Platelets also provide immune resistance during pneumonia through various mechanisms, including platelet–bacteria interactions and complex formation with leukocytes. Other mechanisms include facilitating neutrophil extracellular trap formation and stimulating the release of microbicidal agents that can directly lyse bacteria 116 . Thrombocytopenia is associated with impaired antibacterial defence during murine pneumonia 117 , 118 .

Finally, several distant organs can affect immune resistance in the respiratory tract. For example, depletion of gut microbiota by broad-spectrum antibiotics has been shown to impair host defence during viral and bacterial pneumonia in mice 119 , 120 . This protective gut–lung axis has been hypothesized to be mediated, at least in part, by gut-derived microbial products that can improve host defence mechanisms in other tissue 121 . The existence of a liver–lung axis has been suggested in many studies; pneumonia elicits a robust acute-phase protein response in the liver, probably mediated by cytokines released into circulation, and distinct acute-phase proteins can improve antibacterial defence through several mechanisms, for example, by enhancing opsonophagocytosis (phagocytosis mediated by opsonins) and respiratory burst activity by immune cells and by limiting iron availability to bacteria.

Adaptive immunity

Previous encounters with respiratory pathogens shape memory defence mechanisms against pneumonia. Evidence highlights roles of innate immune cells (for example, epithelial cells and AMs) that had been modified by a prior infection to trigger epigenetic alterations in a so-called process of ‘trained immunity’ 122 . Trained immunity has received attention within the context of pneumonia in humans. The Bacille Calmette–Guérin vaccination induces trained immunity. When administered to older patients after hospital discharge, the vaccination increased time to first infection, and most of the protection was observed against respiratory tract infections of probable viral origin 123 . Humoral response to microbes improves host defence by producing pathogen-specific antibodies, as illustrated by the efficacy of vaccines in diminishing the risk of pneumonia.

The airways contain pools of memory cells that are assembled in tertiary lymphoid organs in the upper airways and in bronchus-associated lymphoid tissue in the lower airways. Together, these cells protect against infection through local and systemic antibody production 124 . The majority of CD4 + T cells and CD8 + T cells in the quiescent lung have a memory phenotype (hence they are named resident memory T (T RM ) cells) and are generated in response to exposure to respiratory pathogens 125 . In experimental mouse models, the lung is enriched with T RM cells specific for multiple viral and bacterial pathogens following a respiratory infection, and these cells contribute to future protective immunity. For example, lobar pneumococcal pneumonia in mice leads to the accumulation of CD4 + T RM cells in the infected lobe, but not in other areas of the lung. This T RM cell-populated lobe expresses better defence against reinfection by S. pneumoniae than other lobes 126 .

Tissue resilience

Tissue resilience is essential in controlling excessive inflammation whilst sustaining effective protection against microbes (Fig.  4 ). AMs contribute to tissue resilience by producing anti-inflammatory cytokines, such as IL-10 and IL-1 receptor antagonist, and through the phagocytosis of apoptotic leukocytes. This process is named efferocytosis and protects tissue in two manners: by preventing the release of pro-inflammatory and toxic contents from dying cells and by concurrently prompting the release of anti-inflammatory and repair factors, including transforming growth factor β1, prostaglandin E 2 , and pro-resolving lipid mediators 100 . Pro-resolving lipid mediators (resolvins, protectins and maresins) can mediate a large variety of immune responses in pneumonia, both increasing bacterial killing and promoting tissue repair 127 . Such mediators have been shown to have important protective roles in mouse models of bacterial pneumonia 128 , 129 .

figure 4

Tissue resilience controls excessive inflammation whilst safeguarding protection against pathogens. The respiratory epithelium is an important player in tissue resilience. Maintenance of the structural integrity of the epithelial barrier is a crucial factor here. Alveolar macrophages also have an important role, via release of anti-inflammatory mediators and efferocytosis (phagocytosis of apoptotic leukocytes). Sessile macrophages adhere to the epithelium, where they probably contribute to tissue resilience. Cell types recruited to the site of infection during pneumonia that are involved in tissue resilience include myeloid-derived suppressor cells, regulatory T (T reg ) cells, type 2 ILC2s and natural killer (NK) cells. ATI, alveolar type I cell; ATII, alveolar type II cell; BASC, bronchioalveolar stem cell; FOXM1, forkhead box protein M1; ILC, innate lymphoid cell; LIF, leukaemia inhibitory factor; RBC, red blood cell; STAT3, signal transducer and activator of transcription 3.

The structural integrity of the epithelial barrier in the respiratory tract is crucial to tissue resilience. Contributors to epithelial resilience include β-catenin (also known as catenin β1) 130 , forkhead box protein M1 (FOXM1) 131 , tumour protein 63 (p63) 132 and signal transducer and activator of transcription 3 (STAT3) 133 , 134 . Interestingly, a deficiency of STAT3 in airway epithelial cells results in exaggerated lung injury during experimental pneumonia 133 , 134 . Epithelial cell-derived leukaemia inhibitory factor (LIF) has been implicated as an important inducer of STAT3 in the respiratory epithelium, and inhibition of LIF has been shown to increase lung injury in pneumonia 135 . Several immune cells recruited to the site of infection during pneumonia are known to contribute to tissue resilience, including myeloid-derived suppressor cells 136 , regulatory T cells 137 , ILC2s 138 and natural killer cells 139 , 140 .

Lung pathology

With respect to the histopathology of bacterial pneumonia, four stages have classically been described: congestion, red hepatization, grey hepatization and resolution (Fig.  5 ). The term hepatization refers to an increased firmness of inflamed lung tissue that renders the tissue consistency similar to that attributed to hepatic tissue. In the early stages of bacterial pneumonia, lung tissue shows mild intra-alveolar oedema and congestion of the capillaries within the alveolar septa 141 . This stage is followed by inflammatory exudation with an accumulation in the alveolar spaces of neutrophils, red blood cells and fibrin, and a subsequent, gradual disintegration of red blood cells and neutrophils. The exudates are eventually transformed into intra-alveolar fibromyxoid moulds, consisting of macrophages and fibroblasts, and gradual resolution follows thereafter.

figure 5

a | Early stage bacterial pneumonia, with congestion of septal capillaries (arrows) and intra-alveolar presence of oedema, neutrophils and a meshwork of fibrin strands (arrowheads). b | Early stage viral pneumonia, with interstitial lymphocytic infiltrates (arrowheads) and diffuse alveolar damage, as evidenced by the presence of hyaline membranes (arrows). c | Organizing pneumonia, with intra-alveolar fibroblast plugs (arrowheads) and few remnant fibrin deposits. Haematoxylin and eosin staining; original magnification ×20. Images in parts a – c courtesy of J.J.T.H. Roelofs, Amsterdam UMC, Netherlands.

Viral pneumonia is typically associated with interstitial inflammation and diffuse alveolar damage 142 . Interstitial inflammation involves the alveolar walls, which widen and usually contain an inflammatory infiltrate of lymphocytes, macrophages and plasma cells in some cases. Alveolar damage is characterized by pink hyaline membranes lining the alveolar septa that follow a pattern of organization and resolution similar to that of intra-alveolar inflammation in bacterial pneumonia.

In addition to these features, specific microorganisms may cause different histopathological changes such as granulomas, multinucleated giant cells or specific viral inclusions.

Extrapulmonary complications

Extrapulmonary complications are extremely common in patients with pneumonia, including those without sepsis. Such complications entail both acute and long-term adverse sequelae. Patients who have been hospitalized for pneumonia have higher rates of all-cause hospitalization and an increased mortality risk for 10 years after discharge 35 compared with matched patients hospitalized for other pneumonia-unrelated conditions.

Sepsis, defined as a life-threating organ dysfunction caused by a dysregulated host response to an infection 143 , is most often caused by pneumonia (up to half of all patients with sepsis) 144 . Conversely, of patients who are hospitalized with CAP 145 or HAP 146 , 36% and 48% have been reported to develop sepsis, respectively. Both pro-inflammatory and anti-inflammatory reactions characterize host response to sepsis, which varies strongly between individuals. Pro-inflammatory responses include the release of cytokines, activation of the complement and coagulation system (which could result in disseminated intravascular coagulation), and disruption of the normal barrier and anticoagulant function of the vascular endothelium. Anti-inflammatory responses can result in immune suppression, in part due to apoptotic loss of lymphoid cells 147 , 148 .

Cardiovascular disease

Pneumonia particularly affects the cardiovascular system, and its effects include depression of left ventricular function, myocarditis, arrhythmias, ischaemia and infarction 149 . Patients hospitalized for pneumonia have an increased short-term and long-term risk (up to ten years) of cardiovascular disease 150 . A meta-analysis of the incidence of cardiac events within 30 days of pneumonia diagnosis found new or worsening heart failure in 14% of all patients, new or worsening arrhythmias in 5% and acute coronary syndromes in 5% 151 . Approximately 90% of cardiac complications occur within 7 days of a pneumonia diagnosis, and more than half occur within the first 24 h 149 . In a multicentre study, one third of patients hospitalized for CAP experienced intrahospital cardiovascular events, mainly involving the heart, and such occurrence was associated with a fivefold increase in 30-day mortality. Independent risk factors for cardiovascular events were severity of pneumonia and pre-existing heart failure 152 . Additionally, hospitalization for pneumonia is associated with an increased risk of new-onset heart failure in the intermediate and long term, with a hazard ratio of 2 after 5 years 34 . In patients with pneumonia who require ICU treatment within 24 h of hospital admission, approximately half have diagnostic criteria for myocardial infarction 153 ; cardiac complications are the direct or main cause of death in 27% of patients hospitalized for pneumonia 154 . Notably, whilst the increased risk for myocardial infarction associated with pneumonia is proportional to disease severity, it is not restricted to patients with pneumonia-induced sepsis 155 . Even mild respiratory infection is associated with an increased risk of myocardial infarction for several months after the onset of infection 155 .

The mechanisms underlying an increased risk of cardiovascular disease after pneumonia are probably multifactorial. Hypoxaemia due to impaired gas exchange and ventilation–perfusion mismatching, as well as endothelial dysfunction causing vasoconstriction, may increase vulnerability to ischaemic events 149 . Systemic inflammation during pneumonia can increase inflammatory activity within coronary atherosclerotic plaques, rendering them prone to rupture 149 . The systemic host response during pneumonia also entails endothelial dysfunction and procoagulant changes, which can promote thrombus formation at the site of a ruptured coronary plaque 149 . Indeed, as reflected by elevated markers of coagulation activation in the circulation, the majority of patients admitted to hospital for pneumonia have a procoagulant phenotype 156 , 157 .

Patients with pneumonia and acute coronary syndromes show higher platelet-aggregating activity than patients with acute coronary syndromes without pneumonia 149 . Notably, the connection between pneumonia and cardiovascular disease is probably bidirectional. For example, pre-existing heart failure is a risk factor for pneumonia, perhaps partially related to impaired immune responses 149 . Preclinical investigations suggest that lung congestion can facilitate the growth of common respiratory pathogens in the airways 149 . With regard to long-term risk, investigations in mice predisposed to developing atherosclerosis 158 and post mortem examinations in humans 159 have suggested that infection can elicit pro-inflammatory responses in atherosclerotic lesions and result in increased vulnerability for coronary and cerebrovascular events. For example, acute lung inflammation induced by intratracheal administration of lipopolysaccharide in mice prone to atherosclerosis resulted in destabilization of atherosclerotic plaques; neutrophil depletion prevented this destabilization, suggesting a role for neutrophils in plaque weakness elicited by lung injury 160 . In addition, systemic inflammation and coagulation are sustained in many patients with pneumonia and have been associated with an increased risk of cardiovascular death 161 , 162 . Left ventricular dysfunction during pneumonia may be secondary to depressant activity of pro-inflammatory cytokines in circulation and/or altered vascular reactivity 149 .

Other complications

Additional extrapulmonary complications of pneumonia include a decline in cognition and functional status 163 , 164 . Pneumonia is associated with a 57% increase in the risk of dementia 164 . Encephalopathy associated with acute infectious disease has been studied in the context of sepsis 165 , 166 . Mechanisms involved include impaired circulation in the brain secondary to hypotension, a disturbed vasoreactivity, endothelial dysfunction and microvascular thrombosis, which can result in ischaemic and haemorrhagic lesions. The blood–brain barrier can be disturbed through increased activity of pro-inflammatory cytokines and reactive oxygen species produced at least in part by astrocytes. Activation of microglia can further contribute to neuronal damage in the brain 166 .

Approximately one fifth of patients hospitalized with pneumonia are readmitted to the hospital within 30 days; pneumonia, cardiovascular disease and (chronic obstructive) pulmonary disease are the most common diagnoses 167 . An increased susceptibility for infection after pneumonia may be related to a relatively immunocompromised state, as has been described in patients with sepsis 147 . Knowledge of immunological defects contributing to recurrent pneumonia (usually defined as a new episode of pneumonia within several months of the previous one, separated by at least a 1-month asymptomatic interval and/or radiographic clearing of the acute infiltrate) 168 is limited. A small study involving 39 patients suggested that immunoglobulin deficiency and an inability to react to polysaccharide antigens are associated with an increased incidence of recurrent pneumonia 169 . Further, a study in mice found a reduced capacity of AMs to phagocytose E. coli and S. aureus following recovery from primary pneumonia, a reduction mediated by signal-regulatory protein-α (also known as tyrosine–protein phosphatase non-receptor type substrate 1) and associated with an impaired host defence after secondary infection of the lower airways 170 .

Diagnosis, screening and prevention

The most common symptoms of pneumonia are cough, breathlessness, chest pain, sputum production and fatigue 171 , 172 . Symptoms are not a part of the initial severity assessment of patients, as the initial symptom burden does not influence outcome. Exceptions include delirium, which is associated with an increased risk of mortality 173 , and pleuritic chest pain, which is associated with an increased risk of para-pneumonic effusion and complicated (infected) para-pneumonic effusion 174 , 175 . Usually mild disease refers to patients with CAP who do not require hospitalization, moderate disease to those cared for in conventional hospital wards, and severe disease to those admitted to the ICU.

It is not possible to differentiate bacterial and viral pneumonia based on symptoms in adults or children, as patients report similar symptoms regardless of microbial aetiology 176 . A recent study found that artificial intelligence was also unable to differentiate microbial aetiology based on symptoms, clinical features and radiology 177 .

CAP is usually clinically suspected in the presence of acute (≤7 days) symptoms of LRTI, such as cough, expectoration, fever and dyspnoea, as well as the presence of new infiltrates on chest radiographs (CXRs) 178 . In older patients, symptoms are typically less evident, and fever can be absent in as many as 30% of patients 179 . Symptoms may also be less evident in patients treated with steroids, NSAIDs and antibiotics 6 . Other pulmonary diseases — most frequently pulmonary embolism and lung cancer — may present with fever and pulmonary infiltrates that can mimic CAP. Interstitial and systemic diseases can also mimic CAP. When diagnosing CAP, it is extremely important to review prior chest CXRs if available, as an additional means to help rule out the disease.

Although HAP is also suspected clinically, symptoms may be hidden by either other medications or the cause of admission. No studies exist about symptom duration in HAP before diagnosis; however, it is usually suspected when patients present with pyrexia (fever) and/or tachypnoea (rapid breathing). HAP diagnosis is believed to be usually delayed, which could explain the higher mortality observed in this population than in patients with VAP.

VAP is suspected when there are at least two of the following symptoms: fever or hypothermia, leukocytosis or leukopenia, and evidence of purulent secretions in an endotracheal tube or tracheostomy 4 . For VAP diagnosis, clinicians often rely on clinical parameters; radiological and laboratory parameters help initiate antimicrobial treatment. Scores have been proposed to facilitate diagnosis. For example, the clinical pulmonary infection score (CPIS) 180 is the most common one, and it is based on points assigned to various signs and symptoms of pneumonia. A CPIS score of >6 suggests VAP, although score sensitivity and specificity are not perfect. In fact, the FDA does not accept this score to diagnose VAP in randomized controlled trials studying antibiotics. In patients with VAP, fever and pulmonary infiltrates can present as atelectasis (collapse of parts of the lung), alveolar haemorrhage and pulmonary thromboembolism, amongst other conditions. In a landmark study using immediate post mortem lung histopathology and microbiology as a gold standard, the presence of two clinical criteria plus the presence of infiltrates on CXRs had a 70% sensitivity and 75% specificity in the diagnosis of VAP 181 .

Radiographic confirmation is essential for the diagnosis of pneumonia. CXRs provide important information about the site, extent and associated features of pneumonia (for example, the lobes involved and the presence of pleural effusion and cavitation) 5 (Fig.  6 ). CXRs have a sensitivity and specificity of 43.5% and 93%, respectively, for detecting pulmonary opacities 182 . In CAP, sensitivity and specificity of 66% and 77%, respectively, have been reported 183 using CT scans as the gold standard. The presence of either pleural fluid or multilobar pneumonia serve as indicators of severity 5 . In CAP, the development of pulmonary infiltrates that were not previously present on a simple posterior–anterior (PA) CXR is essential for CAP diagnosis. The standard CXR for CAP consists of a PA and lateral images; the use of lateral projection images increases diagnostic performance of PA images. In HAP, radiographic evidence of infiltrates is usually determined by CXR examination alone. In VAP, new infiltrates are usually detected by anterior–posterior projection in the supine position; however, in this situation, CXRs are insufficiently sensitive and specific.

figure 6

Pneumonia in upper right lobe (arrow) (part a ); pleural effusion on the left side (arrow) (part b ); massive pleural effusion in the left lung (arrow) (part c ); bilateral pneumonia (arrows) (part d ); lateral image showing left parahilar cavitation with air–fluid level in the lower left lobe (arrow) (part e ); front-to-back image in the same individual as in part e .

In studies in patients hospitalized with CAP, CT identified up to 35% of patients with CAP who had not initially been caught by CXRs 184 . In many patients with COVID-19, CT scans detect pulmonary infiltrates not observed on simple CXRs 185 . In patients with CAP, CT scans serve as a practical complement to CXRs in several cases: when radiographic findings are non-specific, when pulmonary complications such as empyema (pus in the pleural space) or cavitation are present, when there is suspicion of an underlying lesion such as lung carcinoma, and when recurrent pneumonia or non-resolving pneumonia is present 186 . Although this supporting role of CT scans is assumed to apply to patients with HAP as well, supporting evidence is lacking.

Ultrasonography

Lung ultrasonography is a non-invasive imaging method that is now frequently used in many emergency departments and ICUs. Advantages over CT include the absence of radiation exposure, ready use at the bedside and reasonable diagnostic sensitivity and specificity 187 . However, the technique has a steep learning curve, especially in mechanically ventilated patients. In a systematic review, lung ultrasonography was shown to have a sensitivity of 88% and a specificity of 89%, with a ~90% probability of diagnosing pneumonia 188 . Echographic diagnosis is more complex in patients with VAP, and only a few observational studies have been conducted to date 188 . The best of these studies have shown that such diagnosis had better accuracy than the CPIS score alone; the addition of direct Gram stain examination in quantitative cultures of endotracheal aspirates further improved accuracy 189 , 190 . On the basis of on these results, the ventilator-associated pneumonia lung ultrasound score (VPLUS) was developed, and has a sensitivity of 71% and a specificity of 69% for VAP diagnosis 190 .

Microbiology and laboratory tests

Recommendations for microbiological diagnosis in CAP vary according to disease severity (Table  2 ). Of note, microbiological diagnosis in CAP cannot be obtained in up to 50% of patients 5 . In patients with CAP who do not need hospital admission, obtaining samples such as sputum and pharyngeal swabs is optional or not recommended in recent guidelines 5 . In patients requiring hospitalization, obtaining good-quality sputum and blood samples, as well as pharyngeal swabs (for PCR), is recommended. Sputum is the most common respiratory sample in patients with CAP, and samples should be collected before antibiotic treatment. The sensitivity of Gram staining for a sputum sample is ~80% in patients with pneumococcal pneumonia and 78% in patients with pneumonia caused by Staphylococcus spp., and the specificity is 93–96% 191 , 192 . Most health care institutions perform viral PCR on pharyngeal swabs during the influenza season. In the COVID-19 pandemic, it is recommended that all patients admitted with CAP receive a PCR test for the detection of SARS-COV-2.

In patients requiring ICU admission, in addition to all tests mentioned above, bronchoscopic samples, such as bronchoalveolar lavage (BAL) in intubated patients, are not difficult to obtain and provide information on the lower respiratory tract microbiota. Urinary antigen detection tests for S. pneumoniae and L. pneumophila have good sensitivity and specificity, are not extremely expensive and are recommended in all hospitalized patients.

In patients with HAP or VAP, international guidelines 4 recommend obtaining distal respiratory samples for semiquantitative or quantitative cultures (Table  3 ). In patients with HAP, bronchoscopy is not easy to perform, and sputum samples are not often collected. In patients with VAP, distal respiratory samples are preferred. BAL (performed with or without concomitant bronchoscopy) is the sample that provides most information, as, in addition to cultures, cellularity analysis and PCR can be performed on the fluid. A recent meta-analysis showed that Gram staining of BAL performs well in detecting S. aureus 193 . Respiratory samples from patients with HAP or VAP have to be collected before the initiation of a new antibiotic treatment to avoid false-negative cultures. International guidelines 4 do not recommend using procalcitonin (PCT) for the initial diagnosis of HAP or VAP, as several studies have shown that it lacks diagnostic value 194 .

Since the 2000s, owing to multiple outbreaks, epidemics and pandemics caused by respiratory viruses in particular, several molecular tests have been developed, which have contributed to widened availability of molecular testing for the aetiological diagnosis of CAP. Molecular tests have several advantages, including detecting low levels of microbial genetic material, remaining unaffected by prior antibiotic therapy, and providing results within a clinically relevant time frame 195 . Molecular tests based on multiplex PCR have been developed to simultaneously detect and quantify multiple respiratory pathogens, as well as some genes related to antimicrobial resistance. Several commercial multiplex platforms are currently available for comprehensive molecular testing for respiratory pathogens that cause pneumonia (respiratory viruses, bacteria and fungi) and for the main resistance genes of the most common bacteria causing pneumonia 195 , 196 , 197 , 198 .

The WHO currently recommends COVID-19 diagnosis by molecular tests that detect SARS-CoV-2 RNA. SARS-CoV-2 viral sequences can be detected by real-time reverse transcriptase (RT-PCR) in nasopharyngeal swab samples 199 . The disadvantage of this method is that it requires specialized equipment and trained personnel. Additionally, two types of rapid tests are available for COVID-19 diagnosis. The direct SARS-CoV-2 antigen test detects viral components present during infection in samples such as nasopharyngeal secretions, and, therefore, can indicate whether an individual is currently carrying the virus. The indirect antibody test detects antibodies that can be found in serum as part of the immune response against the SARS-CoV-2; thus, it can yield false-negative results if performed before the antibody response has developed and cannot distinguish between past and current infections. These two tests are relatively simple to perform and interpret, requiring limited test operator training 199 .

Some biomarkers may be helpful in identifying which patients are likely to have bacterial pneumonia, in deciding whether antibiotic therapy should be administered, in determining prognosis and in facilitating decisions related to the site of care. However, biomarkers should only be used as an adjunctive tool when managing CAP, as no biomarker has proven full utility in predicting clinical outcomes in patients.

The most widely used biomarkers are acute phase reactants such as C-reactive protein (CRP) and PCT 200 . However, their serum kinetics differ: CRP levels increase after the first 3 days of infection (peak time from infection is 36–50 h), whereas PCT levels rise rapidly (peak time from infection is 12–24 h) in response to microbial toxins or host responses. These properties are useful in differentiating CAP from other non-infectious causes. CRP levels increase in response to any inflammation, and can be modified by the presence of corticosteroids and previous antibiotic therapy, whereas PCT is more specific in bacterial pneumonia. Viral infection-related cytokines attenuate induction of CRP and PCT; however, some elevation in their levels can occur when pneumonia is caused by atypical pathogens (for example, Mycoplasma spp., Chlamydia spp. and Legionella spp.) 201 .

Both CRP and PCT can assist in the clinical diagnosis of pneumonia, but CRP and PCT cannot be used in isolation as a basis for treatment decisions. A second test after 24–48 h is mandatory to monitor for any increases. Clinicians should also consider the pattern in the days preceding symptom onset in patients with CAP and whether a patient is taking medication that could have modified these values. For patients with radiographic CAP, PCT levels can be used with clinical assessment to identify those individuals from whom antibiotic therapy can be safely withheld. This assessment can be combined with a PCR test to identify viral infection, especially as new data show that viruses can frequently be a cause of CAP 13 , 75 . However, caution should be used when a mixed viral–bacterial infection is considered. The new American Thoracic Society (ATS)/Infectious Diseases Society of America (IDSA) CAP 5 guidelines do not recommend using PCT to determine the need for initial antibacterial therapy. The current recommendation is that empirical antibiotic therapy should be initiated in adults with clinically suspected and radiographically confirmed CAP, regardless of initial PCT level.

In studies in patients with HAP or VAP, in whom biomarkers had been monitored serially since before infection, steady increases or persistent elevations in CRP levels were shown to be associated with a high risk of VAP 202 . However, no such pattern was shown for PCT values (crude values or kinetics), with poor diagnostic accuracy for VAP 203 . Thus, a recent international consensus concluded that a combination of clinical assessment including PCT levels in well-defined antibiotic stewardship algorithms could improve diagnosis of bacterial infections and support antibiotic effectiveness 204 .

Prevention of CAP

Many factors increase the risk of CAP and can generally be divided into host factors (for example, age, and the presence of COPD and other chronic pulmonary diseases, diabetes mellitus and chronic heart failure), unhealthy habits (for example, smoking and excessive alcohol consumption) and medications (for example, immunosuppressive drugs, sedating medications such as opioids, and proton pump inhibitors within the first 3 months of administration 205 ). Prevention of CAP is crucial, especially in individuals with these risk factors. Available preventive measures include smoking and alcohol use cessation, improvements in dental hygiene, physical exercise, avoiding contact with children with respiratory infections, and pneumococcal and influenza vaccinations 14 . Implementing these measures in primary and specialized care could help reduce the burden of CAP. Presently, pneumococcal and influenza vaccination are the cornerstones of CAP prevention.

The 23-valent pneumococcal polysaccharide vaccine (PPV23) and the 13-valent pneumococcal conjugate vaccine (PCV13) are currently used in adults. Owing to the demonstrated effectiveness of PPV23 in preventing invasive pneumococcal disease (IPD) in people of ≥65 years of age, the use of the vaccine in this population is recommended in many countries 206 . However, PPV23 effectiveness in preventing non-IPD or CAP due to any cause is much less clear. The effectiveness of PPV23 has been reported to range from 25% to 63% in pneumococcal pneumonia 207 , 208 ; the effectiveness of PCV13 in preventing the first episode of CAP, non-bacteraemic and non-invasive CAP, and IPD due to serotypes contained in the vaccine amongst adults of ≥ 65 years of age has been reported to be 45.6%, 45% and 75%, respectively 209 . Efficacy persisted through the mean follow-up period of 4 years 209 . A post-hoc analysis based on data from the CAPITA trial showed that the effectiveness of PCV13 ranged from 43% to 50.0% for pneumococcal CAP, 36% to 49% for non-bacteraemic and non-invasive pneumococcal CAP, and 67% to 75% for pneumococcal IPD 210 . Of note, the most important measure in reducing pneumococcal CAP burden (bacteraemic and non-bacteraemic) in adults is conjugate vaccine programmes in children. Vaccination with pneumococcal conjugate vaccine in children substantially reduces disease in adults owing to the interruption of transmission and herd protection 211 , 212 .

Influenza vaccination can reduce the risk of complications of influenza, such as pneumonia, and is associated with a decrease in severity, hospitalization, ICU admission and mortality associated with influenza 213 , 214 . All age groups can be affected by influenza virus infection; however, older individuals, young children, pregnant women and those with underlying medical conditions have the highest risk of severe complications. In 2019, a study 75 found that viral sepsis was present in 19% of patients with CAP admitted to ICU and in 61% of patients with viral CAP; influenza virus was the main aetiology. More recently, a study 215 found influenza virus in 23% of patients with LRTI; 57% of these patients had radiographically confirmed CAP. The authors reported 35% vaccine effectiveness against influenza virus LRTI and 51% against influenza-associated CAP. These data demonstrate the importance of an annual influenza vaccination, especially in at-risk groups.

Prevention of HAP

HAP is the leading cause of death from hospital-acquired infection; however, only limited effort has been made in developing prevention strategies. HAP occurs owing to pharyngeal colonization with pathogenic organisms and, in the case of VAP, subsequent aspiration. Thus, oral care and precautions against aspiration may attenuate some of the risk. Although oral and/or digestive decontamination with antibiotics may also be effective, this approach could increase the risk of selecting resistant organisms. Other preventive measures, including isolation practices, remain theoretical or experimental. Indeed, most potential prevention strategies for HAP remain unproven 216 .

The individual measures included in prevention bundles can be divided into non-pharmacological and pharmacological categories. To date, most of our knowledge in HAP prevention is extrapolated from prevention strategies for VAP. An important concept in these strategies is that no single measure is deemed adequate to ensure prevention, with prevention bundles advocated instead. A prospective, interventional, multicentre study in Spain, the Pneumonia Zero project 217 , which included 181 ICUs and built on the experience from a previous study 218 , suggested VAP prevention via a bundle of mandatory and highly recommended measures. The mandatory measures were education and training of medical staff in airway management, hand hygiene with alcohol solutions, oral hygiene with an antiseptic (chlorhexidine), semirecumbent positioning and promotion of procedures and protocols that safely avoid or reduce duration of mechanical ventilation. The highly recommended measures were aspiration of subglottic secretions (removal of secretions that accumulate above the endotracheal tube cuff, in patients who were expected to be mechanically ventilated for >72 h), selective digestive decontamination (SDD)), and selective oropharyngeal decontamination (SOD) (prophylactic strategies to prevent or minimize infections in critically ill patients, based on the application of non-absorbable antibiotics in the oropharynx and gastrointestinal tract (SDD) or oropharynx (SOD) of patients). When implemented, these measures enabled a decrease in adjusted frequency of VAP from 9.83 to 4.34 per 1,000 ventilator-days over 21 months; similarly, the percentage of patients with VAP significantly decreased from 2.4% to 1.9%. In the ICUs with prolonged participation in the study (19–21 months), the incidence of VAP significantly decreased further to just 1.2%. Finally, significant decreases were observed in VAP recurrence rates (from 10.9% to 7.7%).

Non-pharmacological measures

Good hand hygiene using alcohol solution before airway management is firmly established as a fundamental component of clinical practice. Its inclusion in the VAP care bundle represents an opportunity to audit compliance with, and optimize the quality of, hand hygiene practices 217 , 219 .

Remaining in the supine position 220 , the use of gastric tubes and the presence of contents in the stomach contribute to the reflux of gastric contents, aspiration and VAP. Semirecumbent positioning at 30–60° may help to avoid these problems, as found in a 2016 meta-analysis 221 . The lateral Trendelenburg body position (the patient is positioned inclined with head down and feet elevated) has shown no substantial benefit, with research even showing an increase in the number of adverse events 222 . However, based on the results of a post-hoc analysis of the Gravity VAP trial, patients without pulmonary infiltrates at intubation and with no contraindications for the approach may benefit from this position for a short period 222 . The prone position is used to improve hypoxaemia in patients with severe ARDS 223 . This measure is frequently used in COVID-19-associated ARDS 224 , 225 . This approach might decrease the incidence of VAP, as it facilitates the drainage of secretions compared with a semirecumbent position 226 . Further confirmation is needed to assess the beneficial effect in reducing VAP in patients with COVID-19.

Endotracheal tubes also have an important role in the pathogenesis of VAP, and removing contaminated oropharyngeal secretions can reduce the risk of VAP. In a meta-analysis from 2016, evidence supported the use of endotracheal tubes with subglottic secretion drainage to decrease the rate of VAP 227 . Maintaining cuff pressure at >25 cmH 2 O may further prevent the leakage of bacterial pathogens into the lower respiratory tract 217 , and continuous cuff pressure regulation could be superior to intermittent control for preventing VAP 228 . Finally, the tube cuff shape and material may have a role in the aspiration of secretions; a randomized, multicentre trial showed that cuffs made of polyurethane or of a conical shape were not superior to conventional cylindrical polyvinyl chloride cuffs in preventing tracheal colonization and VAP 229 .

Pharmacological measures

Oral washing with chlorhexidine seems to be effective in preventing VAP; however, a recent meta-analysis 230 showed a trend for increased mortality in patients who received chlorhexidine. Consequently, recent international guidelines 3 did not recommend its use. It is plausible that this increased mortality could be due to direct lung toxicity from aspirated chlorhexidine.

Furthermore, the use of either SOD or SDD remains controversial, with most studies to date being performed in settings with low prevalence rates of MDR or XDR microorganisms. These studies have shown a decrease in both the incidence of VAP and overall mortality 231 . However, in a recent cluster randomized clinical trial performed in units with high rates of MDR or XDR pathogens, SOD and SDD were not effective in decreasing bacteraemia caused by those microorganisms 232 . SDD and SOD are not applied in many centres in the USA and in Europe, primarily for fear of inducing microbial resistance. Owing to the unclear balance between a potential reduction in pneumonia rate and a potential increase in mortality, the 2017 international guidelines 3 decided not to issue a recommendation on the use of chlorhexidine for SOD in patients requiring mechanical ventilation until more safety data becomes available. However, the guidelines did suggest the use of SOD — but not SDD — in settings with low rates of antibiotic-resistant bacteria and low antibiotic consumption. Although establishing a cut-off value for low and high resistance settings is a dilemma, the committee felt that a 5% threshold was reasonable.

Prevention of recurrent pneumonia

Recurrent pneumonia affects ~9% of patients hospitalized with CAP 233 , 234 . The main factors related to recurrent pneumonia are age ≥65 years, lack of pneumococcal vaccination, previous episode of pneumonia, COPD and corticoid therapy. S. pneumoniae is the most frequently identified pathogen in patients with recurrent pneumonia 233 , 234 . The main preventive measures for recurrent pneumonia are vaccination and adequate control of prior comorbidities, especially in an older population who have an increased risk of infection.

Antibiotics are the mainstay of therapy for pneumonia; however, the agents used depend on a variety of host and pathogen factors. Ideally, therapy should be pathogen-directed, even though a pathogen is often not identified. Nevertheless, as therapy must be started promptly, empirical therapy directed at the most likely aetiological pathogens is required. Because empirical therapy may be more broad-spectrum than definitive therapy, it is often necessary to narrow and target antibiotics once diagnostic testing results become available, usually after 48–72 h. Such a strategy is referred to as a ‘de-escalation’ of therapy 235 . Rapid comprehensive multiplex molecular methods have been cleared by the FDA and provide results within 2–4.5 h, prior to obtaining final diagnostic testing data. These methods include antibiotic resistance markers and facilitate identification of specific viruses and bacteria, thereby aiding in therapeutic choices and the escalation, de-escalation or cessation of antibiotics.

Considerations for therapeutic choices

Relevant host factors for choosing the type of empirical therapy are severity of illness, the presence of specific medical comorbidities and certain historical data. In detail, these include: chronic lung, heart or liver disease; diabetes mellitus; asplenia; alcohol use disorder; malignancy; malnutrition; recent hospitalization, antibiotic use or colonization by drug-resistant bacteria; the presence of risk factors for aspiration of gastric contents into the lungs (such as impaired swallowing, vomiting, altered consciousness and impaired cough reflex); and recent contact with a health care environment (for example, patients requiring haemodialysis) 236 . It is also important to know epidemiological data regarding individual patients. Seasonal viruses such as influenza viruses are worth examining during the autumn and winter. Contact with someone known to have an illness transmitted by an airborne route (for example, tuberculosis) is also relevant. Similarly, residence in an area with endemic mycoses is a risk for certain fungal pneumonias. Finally, an ICU with a high rate of drug-resistant pathogens poses a risk factor for VAP caused by such organisms 3 .

The site of pneumonia acquisition is also an important consideration, namely, in the community, hospital or ICU, or whilst on mechanical ventilation. Since the late 1990s, guidelines have been developed for patients with pneumonia in each of these settings; however, recent data suggest that patient risk factors, and not the site of infection, should be the main determinant for empirical antibiotic choice. Recently, a unified algorithm based on these risk factors has been proposed for all patients with pneumonia 236 .

In addition to choosing an antibiotic that is likely to target the aetiological pathogens, it is equally important to determine the right dose and route of administration, to ensure that the drug penetrates into the site of infection. In general, oral therapy is used in patients with less severe illness, whilst intravenous therapy is administered in patients with more serious illness. Aerosolized therapy can be used to boost drug delivery to infected lung tissue, especially if the chosen drugs penetrate into the lung poorly. When treating a critically ill patient with pneumonia and a MDR pathogen, it may be necessary to use high doses to ensure reaching bactericidal drug concentrations at the site of infection. Continuous or prolonged infusion may be needed in the case of β-lactam antibiotics to maximize the time during which the drug concentration exceeds the minimum inhibitory concentration (MIC) of the target organism. Other drugs, such as aminoglycosides, kill bacteria in a concentration-dependent fashion and are best administered at high dosages given once daily 237 . In young patients with pneumonia and sepsis, drug clearance by the kidney may be accelerated (augmented renal clearance), and dosing will need to be increased appropriately 238 . In those with renal impairment, dosing or the frequency of administration may need to be reduced and can be optimized by therapeutic drug monitoring, if available.

CAP therapy

Guidelines for CAP recommend empirical therapy based on the severity of illness and presence of risk factors for specific complex pathogens 5 , 53 , 239 (Table  4 ). In the past, patients with risk factors that included contact with a health care environment (haemodialysis, recent hospitalization, residence in a nursing home) were considered to have HCAP and were treated differently from patients with CAP. The new guidelines have eliminated HCAP as a category and recommended that these patients be treated as having CAP. Without forgoing consideration of the local frequency of penicillin and macrolide resistance, every patient with CAP should be treated for pneumococcus in most parts of the world. In addition, atypical pathogens may have a role, often as co-infecting agents; studies showed improved patient outcomes when macrolides or quinolones were added to β-lactam therapy in patients with CAP, particularly those with more severe illness 240 , suggesting a need to treat atypical pathogens in many patients with CAP. Patients with more severe illness may need empirical therapy for MRSA and/or P. aeruginosa , especially if colonization had occurred previously following influenza (in the case of MRSA) or after prior use of broad-spectrum antibiotics (for both pathogens) 241 .

Although in many patients CAP may have a viral aetiology, either as a single pathogen or as part of a mixed infection, antiviral therapy is not routinely recommended. However, for documented influenza-associated pneumonia, current guidelines recommend the use of an anti-influenza agent such as oseltamivir, regardless of illness duration 5 . Nonetheless, the benefit of these agents is greatest within the first 48 h of infection onset. Thus, in patients with a high suspicion of influenza, therapy should be started, whilst results from diagnostic testing are pending. Additionally, even with documented influenza, antibiotics should be used empirically to account for possible bacterial superinfection 5 .

Outpatients

For outpatients without comorbidities or risk factors for MDR pathogen infection, current guidelines recommend monotherapy with respiratory fluroquinolone or combination with amoxicillin–clavulanate or a cephalosporin and macrolide or doxycycline 5 . Regardless of the prevalence of resistance, good experience with macrolide monotherapy has been reported, suggesting that in vitro resistance is not always clinically relevant unless it is high-level resistance (resulting from a ribosomal mechanism) and not lower-level resistance (caused by efflux pumps) 242 . For example, in a Canadian study, patients with CAP who received macrolide therapy (usually as monotherapy) had lower mortality and hospitalization rates than those receiving alternative therapies 243 . For outpatients with comorbid illnesses, current guidelines recommend therapy with a β-lactam and macrolide combination or monotherapy with a respiratory fluoroquinolone, even though recent concerns about fluoroquinolone toxicity have limited their use 5 .

Hospitalized patients

In patients with CAP in hospital wards, therapy should be a β-lactam–macrolide combination or a quinolone (levofloxacin or moxifloxacin) alone (Table  4 ). In areas with a high prevalence of endemic tuberculosis, caution should be exercised with the use of a quinolone, as it can mask the presence of tuberculosis and select for drug-resistant tuberculosis. β-Lactams include ceftriaxone, ceftaroline and ampicillin–sulbactam, whilst macrolides should comprise azithromycin or clarithromycin; some recent data have shown more frequent cardiac complications with the use of erythromycin 244 . Many studies have shown that the addition of a macrolide to the β-lactam, particularly in those with moderately severe illness or with Legionella spp. infection, is associated with a lower mortality rate than β-lactam monotherapy 245 .

All ICU-admitted patients should receive a combination therapy of a β-lactam and either a macrolide or a quinolone. Admission to ICU should be guided by the presence of one of two major criteria (need for mechanical ventilation or septic shock requiring vasopressors) or three of nine minor criteria, as per the 2007 ATS/IDSA guidelines 239 . In this population, a macrolide is generally preferred, although some studies have shown that a quinolone may prove more effective if Legionella spp. infection is highly suspected or documented 246 . If the patient has risk factors for P. aeruginosa or MRSA infection, then treatment for such pathogens should be added.

HAP therapy

Patients can develop HAP in or outside the ICU and can be managed with or without mechanical ventilation, although as many as 30% of patients with HAP who are not initially ventilated will require mechanical ventilation 247 . In patients with a predicted mortality risk of <15% based on the presence or absence of septic shock, monotherapy is associated with lower mortality than combination therapy. In patients with a predicted mortality risk of >25%, combination therapy is associated with reduced mortality; the type of therapy has no effect on mortality in those with a predicted mortality risk of 15–25% 248 . MDR pathogen infection should be considered in patients with a history of prior antibiotic therapy or prolonged hospitalization in the previous 3 months, as well as patients hospitalized in an ICU with a >25% rate of MDR pathogen infections. Although empirical therapy can be guided by patient features, each ICU has its own unique bacteriology; thus, therapy should be guided by knowledge of the local antibiogram 3 , 249 .

Patients with a low mortality risk (estimated from published data in relation to the presence of sepsis and shock) and no MDR pathogen risk factors should receive monotherapy (Table  5 ). In patients with a mortality risk of >15% and/or risk factors for MDR pathogens but who are not in septic shock, monotherapy can be adequate (provided that the chosen antibiotic can target >90% of the gram-negative pathogens in the ICU). Although there is controversy in many hospitals about the need for combination therapy, two agents are often necessary to provide a >90% likelihood of appropriate therapy, especially in the high-risk population and in those with septic shock. The combination regimen should target P. aeruginosa and ESBL-producing Enterobacterales. In all patients with HAP, anti-MRSA therapy should be considered and, if necessary, administered with either vancomycin or linezolid. Depending on local epidemiology, some patients will be at risk of infection with Acinetobacter baumanii , carbapenem-producing Enterobacterales or Stenotrophomonas maltophilia , each one requiring a unique therapy approach. For VAP due to MDR pathogens, such as Acinetobacter baumanii , adjunctive inhaled antibiotics (amikacin or colistin) have been added to systemic therapy, with no proven mortality benefit; efficacy may vary with the type of aerosol delivery system used 250 .

The duration of HAP therapy is between 7 and 14 days, although most patients are successfully treated within only 7 days 251 . Although not all experts agree, the European guidelines list the following groups as exceptions to short duration therapy: patients with MDR pathogen infection, such as P. aeruginosa and Acinetobacte r spp.; those who received inappropriate therapy initially; those who are severely immunocompromised; and those receiving second-line antibiotic agents 217 , 252 , 253 . Current guidelines do not strongly endorse biomarkers such as PCT to guide therapy duration for HAP and VAP, although some randomized trial data do show efficacy for this approach 254 .

Therapy in immunocompromised patients

Immunocompromised patients can develop pneumonia due to the common community and nosocomial pathogens present in the setting as well as other pathogens related to a specific type of immune dysfunction and/or resistant bacteria, viruses, fungi and parasites. Common conditions that impair the immune system include malignancy, HIV infection with a CD4 + T cell count of <200 cells per mm 3 , and solid organ or stem cell transplantation. Therapies that cause immune suppression include prednisone, biological disease modifiers, and chemotherapeutic agents such as azathioprine, methotrexate and cyclophosphamide.

Although empirical therapy is often used, the range of possible pathogens in this population is so broad that aggressive diagnostic testing is necessary, including sampling of deep lower respiratory tract secretions with bronchoscopy in most patients 255 . In patients with HIV infection and a low CD4 + T cell count or with recent corticosteroid tapering, therapy should target common pathogens and Pneumocystis jirovecii 256 . Patients with severe neutropenia, steroid-induced immune suppression and those receiving biologic response modifiers (such as tumour necrosis factor inhibitors) can be infected with fungi such as Aspergillus spp. or Mucorales. Diagnostic testing in those with malignancy or drug-induced immune suppression should also consider other opportunistic pathogens, including cytomegalovirus, Varicella zoster virus, Nocardia spp., parasites such as Strongyloides stercoralis and Toxoplasma gondii , and Mycobacterium tuberculosis (for example, owing to a re-emergence of latent infection).

Aspiration pneumonia therapy

Patients with witnessed macro-aspiration of gastric or oral contents into the lung can develop chemical or bacterial pneumonitis, or simply have bland aspiration. If bacterial pneumonia occurs, patients should receive antibiotics aimed at common community or nosocomial pathogens that were likely to be colonizing the oral and gastric tract at the time of aspiration. In community aspiration, therapy is the same as in CAP unless the patient has poor dentition, which can make infection by anaerobic pathogens possible owing to favourable growth conditions for such microbes in the patient’s mouth. When patients with poor dentition have a lung infiltrate after a witnessed or clinically suspected aspiration event, therapy should be a β-lactam such as ampicillin–sulbactam or amoxicillin–clavulanate, or a quinolone, such as levofloxacin or moxifloxacin. Any of these drugs could also be used if dentition is normal; alternatively, ceftriaxone would be effective 1 . For those with nosocomial aspiration, therapy should be based on the presence of risk factors for MDR pathogens and aimed at common, local and drug-resistant organisms, similar to therapy in other forms of nosocomial pneumonia. There is no need to add specific anti-anaerobic coverage, as these organisms are uncommon in patients who aspirate whilst in hospital or chronic care facilities 257 .

Adjunctive therapy

In addition to antibiotics, patients with severe illness might benefit from adjunctive corticosteroid therapy. In general, this therapy should be restricted to those with severe CAP and a high inflammatory response 258 . In one trial, methylprednisolone was more effective than placebo, leading to less treatment failure (especially late failure) in a population with both severe CAP and elevated CRP levels in the serum 259 . However, before using corticosteroids, it is necessary to rule out influenza, as it may worsen with this line of therapy 260 . By contrast, studies in patients with COVID-19 and hypoxaemic respiratory failure have shown a benefit of corticosteroid therapy with dexamethasone 261 . Similarly, IgM-enriched immunoglobulin may be useful in patients with severe CAP, and high CRP levels and low IgM levels in the serum. In a randomized, double-blind, placebo-controlled trial, IgM-enriched immunoglobulin led to a reduction in mortality and an increase in ventilator-free days in this population, when compared with placebo 262 .

Another adjunctive and supportive therapy includes management of hypoxaemia with respiratory failure, which may necessitate mechanical ventilator support. However, some studies show that patients with CAP can be managed with either non-invasive ventilation or high-flow oxygen. Either modality can reduce the need for mechanical ventilation and, therefore, avoid some of the complications associated with endotracheal intubation and ventilation 263 .

Follow-up of patients after pneumonia

In some patients with CAP, pneumonia can be the start of an inexorable downhill course. In one study, the long-term mortality of patients of >65 years of age hospitalized with CAP far exceeded the in-hospital mortality (33.6% and 11%, respectively) 264 . In some studies, this long-term effect has been attributed to cardiac events that were initiated by acute lung infection 155 .

Pneumonia recurrence can occur in all forms of pneumonia. Recurrence should be classified on the basis of the site of infection. If re-infection occurs at the same site as the original infection, consideration should be given to local factors such as endobronchial obstruction (due to a tumour or foreign body), focal bronchiectasis, insufficient duration of therapy, or infection with a drug-resistant or inadequately treated pathogen. Recurrence elsewhere could be due to immune impairment (due to comorbid illness or certain medications), a non-infectious pulmonary process or recurrent aspiration.

Routine follow-up chest radiography after CAP is not generally recommended. However, if it is prescribed (to monitor resolution of a pleural effusion or infiltrate suggestive of a possible lung mass), it should be delayed for 4–6 weeks if the patient is responding well to therapy 5 . During follow-up, patients should be monitored for undiagnosed or ineffectively managed comorbid illness and encouraged to avoid cigarette smoking. Patients should also have up-to-date pneumococcal and influenza vaccinations. The 30-day readmission rate for patients with CAP has been found to vary from 16.8% to 20.1% 167 . Pneumonia itself was the cause of readmission in only 17.9–29.4% of patients; however, other common causes were exacerbations of congestive heart failure or COPD 167 . Patients with health-care-associated risk factors have a higher probability of readmission than patients with uncomplicated CAP 265 .

Quality of life

The effect of pneumonia is heavily influenced by both the origin of the disease (within the community or in health care environments) and its severity 266 . Most data regarding the effect on quality of life have been obtained in patients with CAP 171 . Antibiotic treatment starts to improve pneumonia symptoms rapidly; acute symptoms typically improve within 3–5 days in patients with mild CAP (outpatients) and 5–10 days in hospitalized patients with more severe CAP not requiring ICU admission; however, return to baseline levels of symptoms and function seems to take substantially longer 172 , 267 , 268 , 269 . In mild-to-moderate CAP, in most patients symptoms such as cough and breathlessness resolve within 14 days, although up to 6 months are required for full recovery 267 . Thus, the greatest burden seems to be a loss of function in the long term. Delayed recovery is associated with the number of comorbid conditions. In most cases, the presence of ongoing health impairment is largely related to a decompensation of underlying diseases rather than the ongoing acute symptoms of CAP 267 . A modelling study showed that in hospitalized patients with CAP, these acute symptoms reduced in intensity by ~50% within the first 3–5 days, and resolved in nearly all patients by day 28 (ref. 268 ). There does not seem to be a meaningful difference in symptom intensity or time to symptom resolution between viral and bacterial pneumonia 270 .

A French study in patients with pneumococcal pneumonia followed for 12 months after hospital discharge used the EQ-5D-3L questionnaire to evaluate health status 271 . Patients experienced a progressive improvement in quality of life after discharge, plateauing at six months. Importantly, quality of life either did not improve or deteriorated after discharge in 34% of patients; recovery was worse in old patients than in young patients. In a US study in patients with CAP, on average, patients were able to return to normal productivity in 3 weeks and missed 2 weeks of work 272 . Recovery was slowest in patients with comorbidities such as COPD, leading to recovery times of 2 months on average. Even after recovery, symptom scores in patients with CAP are worse than those in the general population, partially because CAP has a long-term effect on health. Another partial reason for these lower scores is the development of CAP in patients with high-risk comorbidities, which make these patients more symptomatic than the general population 273 . Lastly, long-term mortality is increased in patients with CAP compared with the general population 35 . LRTIs without radiographic infiltrates (non-CAP LRTIs) are associated with a similar impairment in quality of life to CAP 274 .

Studies comparing quality of life between patients with CAP and the general population have shown consistently worse quality of life up to 12 months after CAP. With a few exceptions, most of these studies used generic quality of life and productivity tools. A systematic review identified five CAP-specific, patient-reported outcome measures, of which the CAP symptom questionnaire (CAP-sym) was the most widely used 275 . This review concluded that most CAP-specific tools have thus far been evaluated in highly specific populations and may not be fully representative, and it recommends continuing to use generic tools until better tools are available.

Improved diagnostics

The key to a switch to pathogen-specific therapy is an accurate aetiological diagnosis, and the availability of rapid molecular diagnostic tests makes clinical trials and subsequent clinical use of these targeted therapies feasible. Most progress in diagnostics can be observed in two areas: rapid identification of pathogens in positive blood cultures and detection of respiratory viral pathogens. However, bacteraemia is uncommon in pneumonia and, therefore, the effect of these molecular assays on management is limited. By contrast, PCR diagnosis of respiratory viral infections has now become the standard of care. The greatest issue with these assays obtained from nasopharyngeal specimens is whether results reflect upper respiratory tract infections only or accurately detect the cause of pneumonia. In addition, negative nasopharyngeal samples have occurred in patients with positive concurrent bronchoalveolar samples for influenza and SARS-CoV-2 (ref. 276 ).

Several multiplex PCR platforms are available for clinical use for bacterial pneumonia, with approval based on comparison with standard diagnostic tools, specifically culture 277 , 278 . However, as culture itself is not a gold standard, the true operating characteristics of the tests remain unknown. One alternative is metagenomics sequencing to determine all microbiota present; clinically relevant platforms are available 279 , 280 . Generally, these molecular assays are more sensitive than culture, especially for fastidious microorganisms; nevertheless, none of the current multiplex assays detect all of the relevant pathogens and, therefore, cannot replace cultures. In addition, a limited ability to provide information on antibiotic susceptibility is a major weakness. Despite such limitations, substantial impact on antibiotic prescription is possible. Most evaluations to date comprise observational studies and analyses of the theoretical benefit if antibiotic decisions based on molecular assays were applied prospectively. Perhaps the best demonstration of such potential is to limit the use of vancomycin or linezolid for suspected MRSA pneumonia 197 . Multiple sensitive and specific gene targets for S. aureus identification are available, whilst the absence of the mecA gene detection essentially excludes methicillin resistance in that isolate; thus, a negative assay eliminates the need for MRSA coverage. However, the greatest hurdle for molecular assays is clinicians’ willingness to base antimicrobial treatment on results obtained from these novel diagnostic platforms; even a BAL assay with a 98% negative predictive value did not result in a decrease in empirical treatment of VAP 281 . Implementation trials are required to demonstrate the true benefit of more accurate diagnostics.

Improved diagnostic testing may enable a host of unanswered epidemiological matters surrounding pneumonia to be addressed. A leading question in the field of pneumonia is its cause in immunocompromised patients; only expert opinion guides treatment recommendations 256 . The COVID-19 pandemic also illustrates the probable high frequency of additional viral agents that may cause CAP of seemingly unknown aetiology 13 . The role of fungal superinfection of viral pneumonia also remains controversial owing to diagnostic uncertainty 282 .

Antibiotic therapy

For most of the ~75-year history of antibiotic treatment of pneumonia, the backbone of therapy has been a β-lactam 283 . The emergence of bacterial resistance to β-lactams has been tackled with two strategies: newer generations or types of β-lactams (penicillins, cephalosporins and carbapenems) 284 , 285 , 286 , 287 , 288 and combinations with β-lactamase inhibitors (BLIs). Ceftolozane is the newest β-lactam on the market; it has improved activity against P. aeruginosa compared with other cephalosporins 284 . Each BLI has slightly different activity against the variety of resistance mechanisms in Enterobacterales, including carbapenem-resistant and ESBL-producing Enterobacterales, which may affect local efficacy owing to geographical differences in resistance patterns 289 .

Each new drug had been intended to replace the prior generation, gain a large proportion of market share and, therefore, justify the large development costs for the pharmaceutical industry. However, the majority of infections, especially community-acquired 13 , remain susceptible to cheap generic antibiotics even today, and the probability of a new blockbuster drug that would garner a large market share is progressively in decline 290 . This and multiple other factors, including increased costs for registration trials, a regulatory environment and challenges in clinical trial design, have led many pharmaceutical firms to abandon antibiotic development, as it offers a poor return on investment 291 .

Nevertheless, the paradigm for antibiotic development has shifted and, since the 2000s, niche antibiotics, particularly for gram-negative pathogens, have progressively emerged, developed by small biotech companies. These niche antibiotics specifically address gaps in standard antibiotic treatment coverage, yet leverage high prices to compensate for a small market share. The future success of these niche antibiotics could be increased by the emergence of rapid diagnostic tests that can detect specific pathogens or specific resistance markers immediately.

New antibiotics

The first generation of niche antibiotics were new β-lactams or BLIs developed for individual MDR or XDR pathogens 292 . The greatest unmet need for pneumonia due to gram-negative pathogens is for treatment of carbapenem-resistant Acinetobacter spp.; the only agent in development specifically for Acinetobacter spp. is a combination of two BLIs 293 . Both BLIs also have intrinsic β-lactam activity but are being studied in combination with a carbapenem for serious Acinetobacter spp. infections, including pneumonia.

Agents specific for Pseudomonas spp. are also in development. Murepavadin is the first of a new class of antibiotics that inhibit the outer membrane assembly of P. aeruginosa ; other drugs targeting outer membrane assembly are in development, including phage-derived endolysins 294 . Small molecule inhibitors of the type-III secretion apparatus in P. aeruginosa , a crucial component of its pathogenesis, are also in development.

One exception to the niche drug approach is cefiderocol, an extremely broad-spectrum agent with activity against almost all MDR pathogens. Cefiderocol links ceftazidime and cefepime together, maintaining the β-lactam bactericidal mechanism whilst enhancing bacterial uptake 295 . Bacteria take up cefiderocol through iron channels, and this mechanism is extremely appealing, as many MDR gram-negative pathogens, including P. aeruginosa , Acinetobacter spp. and Stenotrophomonas spp., avidly take up iron, and a major component of the acute-phase host response is to sequester iron from pathogens. Cefiderocol was non-inferior to high-dose extended-infusion meropenem for HAP due to gram-negative pathogens 296 , but it was associated with a higher mortality than the best available therapy for pneumonia and bacteraemia, specifically due to carbapenem-resistant Acinetobacter spp. 292 .

Lefamulin is the first truly new antibiotic class since the oxazolidinone linezolid. The mechanism of action of lefamulin is via protein synthesis inhibition, and lefamulin is approved for the treatment of CAP based on equivalence to moxifloxacin 297 , 298 . This drug can be used as a single agent to target MRSA and other CAP pathogens resistant to macrolide, β-lactam and fluoroquinolone antibiotics, and possibly in cases of treatment failure and/or in patients with multiple drug allergies. Unfortunately, lefamulin does not have substantial activity against ESBL-producing gram-negative pathogens, which is an unmet need in CAP.

Non-antibiotic therapy

Monoclonal or polyclonal antibodies to specific MDR pathogens, including S. aureus and P. aeruginosa , are the ultimate narrow-spectrum agents, being both extremely safe and having the great advantage of not disturbing the commensal microbiota 299 , 300 . Antibodies against the P. aeruginosa type-III secretion apparatus, alginate and other unique targets have entered clinical trials. Several anti- S. aureus antibodies have also been developed 301 . The challenge for specific antibodies is whether they should be used for prevention or as adjuncts to antibiotic therapy. The lack of sensitive risk factors or predictive markers for pneumonia caused by a specific pathogen make prophylactic trials difficult and potential clinical use expensive; thus, development for preventive indications has been abandoned for several agents, and attention has shifted to adjunctive use, despite this being associated with loss of the microbiota-sparing effect with this strategy.

Case reports have been published on bacteriophage therapy as an alternative to antibiotics in patients with extremely difficult-to-treat pneumonia 302 . However, major logistic issues must be overcome before phage therapy becomes a legitimate option 303 : the individual patient’s isolate must be tested for susceptibility against a battery of bacteria-specific phages; a cocktail of at least three phages is usually needed, owing to the emergence of resistance to any single phage; and the availability of phages and susceptibility testing facilities remain extremely limited. Furthermore, the optimal delivery method, namely aerosolization, instillation or venous infusion, remains unclear. No large-scale clinical trials have been completed.

Lastly, the COVID-19 pandemic has generated a large number of studies of adjuvant treatments focusing on host response to SARS-CoV-2. It remains unclear whether any adjuvant treatments other than corticosteroids that may provide benefit in SARS-CoV-2 infection can be used for influenza or other serious viral pneumonias. However, the COVID-19 pandemic has clearly increased interest in both host-directed therapy and newer antivirals.

Mandell, L. A. & Niederman, M. S. Aspiration pneumonia. N. Engl. J. Med. 380 , 651–663 (2019). A review article about aspiration pneumonia, including new insights about microbial aetiology and antibiotic treatment .

CAS   PubMed   Google Scholar  

Cillóniz, C. et al. Microbial aetiology of community-acquired pneumonia and its relation to severity. Thorax 66 , 340–346 (2011).

PubMed   Google Scholar  

Torres, A. et al. International ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia. Eur. Respir. J. 50 , 1700582 (2017). In these international European and Latin American guidelines, a panel of experts present recommendations about diagnosis, risk factor for antibiotic resistance and type and duration of treatment for HAP and VAP. PICO questions and GRADE methodology were used .

Kalil, A. C. et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin. Infect. Dis. 63 , e61–e111 (2016). These guidelines provide risk factors for suspected MDR or XDR microorganisms and recommendations for empirical treatments, use of biomarkers and duration of antibiotic administration .

PubMed   PubMed Central   Google Scholar  

Metlay, J. P. et al. Diagnosis and treatment of adults with community-acquired pneumonia. An Official Clinical Practice Guideline of the American Thoracic Society and Infectious Diseases Society of America. Am. J. Respir. Crit. Care Med. 200 , e45–e67 (2019). These guidelines include new recommendations for microbiological diagnostic tests, in particular for empirical treatments in outside and in-hospital patients .

Prina, E., Ranzani, O. T. & Torres, A. Community-acquired pneumonia. Lancet 386 , 1097–1108 (2015).

Di Pasquale, M. F. et al. Prevalence and etiology of community-acquired pneumonia in immunocompromised patients. Clin. Infect. Dis. 68 , 1482–1493 (2019).

GBD 2019 Diseases and Injuries Collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet 396 , 1204–1222 (2020).

Google Scholar  

GBD 2016 Lower Respiratory Infections Collaborators. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of lower respiratory infections in 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect. Dis. 18 , 1191–1210 (2018).

Komiya, K. et al. Prognostic implications of aspiration pneumonia in patients with community acquired pneumonia: a systematic review with meta-analysis. Sci. Rep. 6 , 38097 (2016).

CAS   PubMed   PubMed Central   Google Scholar  

Lindenauer, P. K. et al. Variation in the diagnosis of aspiration pneumonia and association with hospital pneumonia outcomes. Ann. Am. Thorac. Soc. 15 , 562–569 (2018).

Neill, S. & Dean, N. Aspiration pneumonia and pneumonitis: a spectrum of infectious/noninfectious diseases affecting the lung. Curr. Opin. Infect. Dis. 32 , 152–157 (2019).

Jain, S. et al. Community-acquired pneumonia requiring hospitalization among U.S. adults. N. Engl. J. Med. 373 , 415–427 (2015).

Torres, A., Peetermans, W. E., Viegi, G. & Blasi, F. Risk factors for community-acquired pneumonia in adults in Europe: a literature review. Thorax 68 , 1057–1065 (2013).

Norris, T., Vahratian, A. & Cohen, R. A. Vaccination coverage among adults aged 65 and over: United States, 2015. NCHS Data Brief No. 281 (CDC, 2017).

Fedson, D. S. et al. Pneumococcal polysaccharide vaccination for adults: new perspectives for Europe. Expert. Rev. Vaccines 10 , 1143–1167 (2011).

Jamal, A. et al. Current cigarette smoking among adults – United States, 2016. MMWR 67 , 53–59 (2018).

Louie, J. K. et al. Factors associated with death or hospitalization due to pandemic 2009 influenza A(H1N1) infection in California. JAMA 302 , 1896–1902 (2009).

Wu, C. et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern. Med. 180 , 934–943 (2020).

Barbier, F., Andremont, A., Wolff, M. & Bouadma, L. Hospital-acquired pneumonia and ventilator-associated pneumonia: recent advances in epidemiology and management. Curr. Opin. Pulm. Med. 19 , 216–228 (2013).

Rosenthal, V. D. et al. International Nosocomial Infection Control Consortium (INICC) report, data summary of 36 countries, for 2004-2009. Am. J. Infect. Control. 40 , 396–407 (2012).

Giuliano, K. K., Baker, D. & Quinn, B. The epidemiology of nonventilator hospital-acquired pneumonia in the United States. Am. J. Infect. Control. 46 , 322–327 (2018).

Bonell, A. et al. A systematic review and meta-analysis of ventilator-associated pneumonia in adults in Asia: an analysis of national income level on incidence and etiology. Clin. Infect. Dis. 68 , 511–518 (2019).

Bouadma, L. et al. Ventilator-associated events: prevalence, outcome, and relationship with ventilator-associated pneumonia. Crit. Care Med. 43 , 1798–1806 (2015).

Shi, T. et al. Global and regional burden of hospital admissions for pneumonia in older adults: a systematic review and meta-analysis. J. Infect. Dis. 222 , S570–S576 (2020).

GBD 2017 Causes of Death Collaborators. Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 392 , 1736–1788 (2018).

JustActions. The Missing Piece. Why Continued Neglect of Pneumonia Threatens the Achivement of Health Goals (JustActions, 2018).

Nunes, B. P., Flores, T. R., Mielke, G. I., Thumé, E. & Facchini, L. A. Multimorbidity and mortality in older adults: a systematic review and meta-analysis. Arch. Gerontol. Geriatr. 67 , 130–138 (2016).

Arnold, F. W. et al. Mortality differences among hospitalized patients with community-acquired pneumonia in three world regions: results from the Community-Acquired Pneumonia Organization (CAPO) International Cohort Study. Respir. Med. 107 , 1101–1111 (2013).

Heo, J. Y. & Song, J. Y. Disease burden and etiologic distribution of community-acquired pneumonia in adults: evolving epidemiology in the era of pneumococcal conjugate vaccines. Infect. Chemother. 50 , 287–300 (2018).

Cillóniz, C. et al. Community-acquired pneumonia in outpatients: aetiology and outcomes. Eur. Respir. J. 40 , 931–938 (2012).

Luna, C. M. et al. The impact of age and comorbidities on the mortality of patients of different age groups admitted with community-acquired pneumonia. Ann. Am. Thorac. Soc. 13 , 1519–1526 (2016).

Cillóniz, C. et al. Twenty-year trend in mortality among hospitalized patients with pneumococcal community-acquired pneumonia. PLoS ONE 13 , e0200504 (2018).

Corrales-Medina, V. F. et al. Intermediate and long-term risk of new-onset heart failure after hospitalization for pneumonia in elderly adults. Am. Heart J. 170 , 306–312 (2015).

Eurich, D. T., Marrie, T. J., Minhas-Sandhu, J. K. & Majumdar, S. R. Ten-year mortality after community-acquired pneumonia. A prospective cohort. Am. J. Respir. Crit. Care Med. 192 , 597–604 (2015).

Ramirez, J. A. et al. Adults hospitalized with pneumonia in the United States: incidence, epidemiology and mortality. Clin. Infect. Dis. 65 , 1806–1812 (2017).

Bordon, J. et al. Hospitalization due to community-acquired pneumonia in patients with chronic obstructive pulmonary disease: incidence, epidemiology and outcomes. Clin. Microbiol. Infect. 26 , 220–226 (2020).

Torres, A. et al. Burden of pneumococcal community-acquired pneumonia in adults across Europe: a literature review. Respir. Med. 137 , 6–13 (2018).

Magill, S. S. et al. Multistate point-prevalence survey of health care-associated infections. N. Engl. J. Med. 370 , 1198–1208 (2014).

Micek, S. T., Chew, B., Hampton, N. & Kollef, M. H. A case-control study assessing the impact of nonventilated hospital-acquired pneumonia on patient outcomes. Chest 150 , 1008–1014 (2016).

Melsen, W. G. et al. Attributable mortality of ventilator-associated pneumonia: a meta-analysis of individual patient data from randomised prevention studies. Lancet Infect. Dis. 13 , 665–671 (2013).

Bassetti, M. et al. Risk stratification and treatment of ICU-acquired pneumonia caused by multidrug-resistant/extensively drug-resistant/pandrug-resistant bacteria. Curr. Opin. Crit. Care 24 , 385–393 (2018).

Herkel, T. et al. Epidemiology of hospital-acquired pneumonia: results of a Central European multicenter, prospective, observational study compared with data from the European region. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc Czech Repub. 160 , 448–455 (2016).

Ibn Saied, W. et al. A comparison of the mortality risk associated with ventilator-acquired bacterial pneumonia and nonventilator ICU-acquired bacterial pneumonia. Crit. Care Med. 47 , 345–352 (2019).

Talbot, G. H. et al. Evidence-based study design for hospital-acquired bacterial pneumonia and ventilator-associated bacterial pneumonia. J. Infect. Dis. 219 , 1536–1544 (2019).

McAllister, D. A. et al. Global, regional, and national estimates of pneumonia morbidity and mortality in children younger than 5 years between 2000 and 2015: a systematic analysis. Lancet Glob. Health 7 , e47–e57 (2019).

Weir, D. L., Majumdar, S. R., McAlister, F. A., Marrie, T. J. & Eurich, D. T. The impact of multimorbidity on short-term events in patients with community-acquired pneumonia: prospective cohort study. Clin. Microbiol. Infect. 21 , 264.e7–264.e13 (2015).

CAS   Google Scholar  

Bradley, J. S. et al. The management of community-acquired pneumonia in infants and children older than 3 months of age: clinical practice guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin. Infect. Dis. 53 , e25–e76 (2011).

Barbagelata, E. et al. Gender differences in community-acquired pneumonia. Minerva Med. 111 , 153–165 (2020).

Mutepe, N. D. et al. Effects of cigarette smoke condensate on pneumococcal biofilm formation and pneumolysin. Eur. Respir. J. 41 , 392–395 (2013).

Samokhvalov, A. V., Irving, H. M. & Rehm, J. Alcohol consumption as a risk factor for pneumonia: a systematic review and meta-analysis. Epidemiol. Infect. 138 , 1789–1795 (2010).

Neupane, B. et al. Long-term exposure to ambient air pollution and risk of hospitalization with community-acquired pneumonia in older adults. Am. J. Respir. Crit. Care Med. 181 , 47–53 (2010).

American Thoracic Society & Infectious Diseases Society of America. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am. J. Respir. Crit. Care Med. 171 , 388–416 (2005).

Le, M. N.-T. et al. Oral colonisation by antimicrobial-resistant Gram-negative bacteria among long-term care facility residents: prevalence, risk factors, and molecular epidemiology. Antimicrob. Resist. Infect. Control. 9 , 45 (2020).

Feldman, C. et al. The presence and sequence of endotracheal tube colonization in patients undergoing mechanical ventilation. Eur. Respir. J. 13 , 546–551 (1999).

Cilloniz, C. et al. Seasonality of pathogens causing community-acquired pneumonia. Respirology 22 , 778–785 (2017).

Para, R. A., Fomda, B. A., Jan, R. A., Shah, S. & Koul, P. A. Microbial etiology in hospitalized North Indian adults with community-acquired pneumonia. Lung India 35 , 108–115 (2018).

Tao, L.-L. et al. Etiology and antimicrobial resistance of community-acquired pneumonia in adult patients in China. Chin. Med. J. 125 , 2967–2972 (2012).

Shoar, S. & Musher, D. M. Etiology of community-acquired pneumonia in adults: a systematic review. Pneumonia 12 , 11 (2020).

Moberley, S., Holden, J., Tatham, D. P. & Andrews, R. M. Vaccines for preventing pneumococcal infection in adults. Cochrane Database Syst. Rev. 2013 , CD000422 (2013).

PubMed Central   Google Scholar  

Centers for Disease Control and Prevention (CDC). Current cigarette smoking among adults - United States, 2011. MMWR 61 , 889–894 (2012).

Luca, D. L. et al. Impact of pneumococcal vaccination on pneumonia hospitalizations and related costs in Ontario: a population-based ecological study. Clin. Infect. Dis. 66 , 541–547 (2017).

Johansson, N., Kalin, M., Tiveljung-Lindell, A., Giske, C. G. & Hedlund, J. Etiology of community-acquired pneumonia: increased microbiological yield with new diagnostic methods. Clin. Infect. Dis. 50 , 202–209 (2010).

Rozenbaum, M. H. et al. The role of Streptococcus pneumoniae in community-acquired pneumonia among adults in Europe: a meta-analysis. Eur. J. Clin. Microbiol. Infect. Dis. 32 , 305–316 (2013).

Huijts, S. M. et al. Diagnostic accuracy of a serotype-specific antigen test in community-acquired pneumonia. Eur. Respir. J. 42 , 1283–1290 (2013).

Aliberti, S. et al. Multidrug-resistant pathogens in hospitalised patients coming from the community with pneumonia: a European perspective. Thorax 68 , 997–999 (2013).

Shindo, Y. et al. Risk factors for drug-resistant pathogens in community-acquired and healthcare-associated pneumonia. Am. J. Respir. Crit. Care Med. 188 , 985–995 (2013).

Prina, E. et al. Risk factors associated with potentially antibiotic-resistant pathogens in community-acquired pneumonia. Ann. Am. Thorac. Soc. 12 , 153–160 (2015).

Ceccato, A. et al. Validation of a prediction score for drug-resistant microorganisms in community-acquired pneumonia. Ann. Am. Thorac. Soc. 18 , 257–265 (2021).

Cilloniz, C. et al. Difficult to treat microorganisms in patients over 80 years with community-acquired pneumonia: the prevalence of PES pathogens. Eur. Respir. J. 56 , 2000773 (2020).

Webb, B. J. et al. Derivation and multicenter validation of the drug resistance in pneumonia clinical prediction score. Antimicrob. Agents Chemother. 60 , 2652–2663 (2016).

Karhu, J., Ala-Kokko, T. I., Vuorinen, T., Ohtonen, P. & Syrjälä, H. Lower respiratory tract virus findings in mechanically ventilated patients with severe community-acquired pneumonia. Clin. Infect. Dis. 59 , 62–70 (2014).

Wu, X. et al. Incidence of respiratory viral infections detected by PCR and real-time PCR in adult patients with community-acquired pneumonia: a meta-analysis. Respiration 89 , 343–352 (2015).

Zhou, F. et al. Disease severity and clinical outcomes of community acquired pneumonia caused by non-influenza respiratory viruses in adults: a multicenter prospective registry study from CAP-China Network. Eur. Respir. J. 54 , 1802406 (2019).

Cillóniz, C. et al. Pure viral sepsis secondary to community-acquired pneumonia in adults: risk and prognostic factors. J. Infect. Dis. 220 , 1166–1171 (2019).

Jain, S. et al. Community-acquired pneumonia requiring hospitalization among U.S. children. N. Engl. J. Med. 372 , 835–845 (2015). This study is a prospective multicentre investigation of the CAP microbial aetiology in hospitalized patients. Very importantly, PCR tests for the detection of viral pathogens , Legionella spp. and Mycoplasma pneumoniae were systematically used in the diagnostic work-up. With this approach, viruses represented the first cause of CAP .

Weber, D. J. et al. Microbiology of ventilator-associated pneumonia compared with that of hospital-acquired pneumonia. Infect. Control. Hosp. Epidemiol. 28 , 825–831 (2007).

Magiorakos, A.-P. et al. Multidrug-resistant, extensively drug-resistant and pandrug-resistant bacteria: an international expert proposal for interim standard definitions for acquired resistance. Clin. Microbiol. Infect. 18 , 268–281 (2012).

Parker, D., Ahn, D., Cohen, T. & Prince, A. Innate immune signaling activated by MDR bacteria in the airway. Physiol. Rev. 96 , 19–53 (2016).

Grousd, J. A., Rich, H. E. & Alcorn, J. F. Host-pathogen interactions in gram-positive bacterial pneumonia. Clin. Microbiol. Rev. 32 , e00107-18 (2019).

Kutter, J. S., Spronken, M. I., Fraaij, P. L., Fouchier, R. A. & Herfst, S. Transmission routes of respiratory viruses among humans. Curr. Opin. Virol. 28 , 142–151 (2018).

Siegel, S. J. & Weiser, J. N. Mechanisms of bacterial colonization of the respiratory tract. Annu. Rev. Microbiol. 69 , 425–444 (2015).

Quinton, L. J., Walkey, A. J. & Mizgerd, J. P. Integrative physiology of pneumonia. Physiol. Rev. 98 , 1417–1464 (2018).

Dickson, R. P., Erb-Downward, J. R. & Huffnagle, G. B. Towards an ecology of the lung: new conceptual models of pulmonary microbiology and pneumonia pathogenesis. Lancet Respir. Med. 2 , 238–246 (2014). A review–opinion article about new insights into the aetiopathogenesis of pneumonia based on changes in the microbiota .

Pettigrew, M. M., Tanner, W. & Harris, A. D. The lung microbiome and pneumonia. J. Infect. Dis. https://doi.org/10.1093/infdis/jiaa702 (2020).

Article   Google Scholar  

Brown, R. L., Sequeira, R. P. & Clarke, T. B. The microbiota protects against respiratory infection via GM-CSF signaling. Nat. Commun. 8 , 1512 (2017).

Nishimoto, A. T., Rosch, J. W. & Tuomanen, E. I. Pneumolysin: pathogenesis and therapeutic target. Front. Microbiol. 11 , 1543 (2020).

von Hoven, G., Qin, Q., Neukirch, C., Husmann, M. & Hellmann, N. Staphylococcus aureus α-toxin: small pore, large consequences. Biol. Chem. 400 , 1261–1276 (2019).

Hauser, A. R. The type III secretion system of Pseudomonas aeruginosa: infection by injection. Nat. Rev. Microbiol. 7 , 654–665 (2009).

Ferguson, N. D. et al. The Berlin definition of ARDS: an expanded rationale, justification, and supplementary material. Intensive Care Med. 38 , 1573–1582 (2012).

Matthay, M. A. et al. Acute respiratory distress syndrome. Nat. Rev. Dis. Prim. 5 , 18 (2019).

Whitsett, J. A. & Alenghat, T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat. Immunol. 16 , 27–35 (2015).

Cheng, D. et al. Airway epithelium controls lung inflammation and injury through the NF-κB pathway. J. Immunol. 178 , 6504–6513 (2007).

Quinton, L. J. et al. Functions and regulation of NF-κB RelA during pneumococcal pneumonia. J. Immunol. 178 , 1896–1903 (2007).

Han, S. & Mallampalli, R. K. The role of surfactant in lung disease and host defense against pulmonary infections. Ann. Am. Thorac. Soc. 12 , 765–774 (2015).

Carey, R. M. & Lee, R. J. Taste receptors in upper airway innate immunity. Nutrients 11 , 2017 (2019).

CAS   PubMed Central   Google Scholar  

Lee, R. J. & Cohen, N. A. The emerging role of the bitter taste receptor T2R38 in upper respiratory infection and chronic rhinosinusitis. Am. J. Rhinol. Allergy 27 , 283–286 (2013).

McAleer, J. P. & Kolls, J. K. Directing traffic: IL-17 and IL-22 coordinate pulmonary immune defense. Immunol. Rev. 260 , 129–144 (2014).

Aujla, S. J. et al. IL-22 mediates mucosal host defense against Gram-negative bacterial pneumonia. Nat. Med. 14 , 275–281 (2008).

Allard, B., Panariti, A. & Martin, J. G. Alveolar macrophages in the resolution of inflammation, tissue repair, and tolerance to infection. Front. Immunol. 9 , 1777 (2018).

Preston, J. A. et al. Alveolar macrophage apoptosis-associated bacterial killing helps prevent murine pneumonia. Am. J. Respir. Crit. Care Med. 200 , 84–97 (2019).

González-Juarbe, N. et al. Pore-forming toxins induce macrophage necroptosis during acute bacterial pneumonia. PLoS Pathog. 11 , e1005337 (2015).

Kitur, K. et al. Toxin-induced necroptosis is a major mechanism of Staphylococcus aureus lung damage. PLoS Pathog. 11 , e1004820 (2015).

Kitur, K. et al. Necroptosis promotes Staphylococcus aureus clearance by inhibiting excessive inflammatory signaling. Cell Rep. 16 , 2219–2230 (2016).

Panda, S. K. & Colonna, M. Innate lymphoid cells in mucosal immunity. Front. Immunol. 10 , 861 (2019).

Kaiko, G. E., Phipps, S., Angkasekwinai, P., Dong, C. & Foster, P. S. NK cell deficiency predisposes to viral-induced Th2-type allergic inflammation via epithelial-derived IL-25. J. Immunol. 185 , 4681–4690 (2010).

Jayaraman, A. et al. IL-15 complexes induce NK- and T-cell responses independent of type I IFN signaling during rhinovirus infection. Mucosal Immunol. 7 , 1151–1164 (2014).

Van Maele, L. et al. Activation of type 3 innate lymphoid cells and interleukin 22 secretion in the lungs during Streptococcus pneumoniae infection. J. Infect. Dis. 210 , 493–503 (2014).

Xiong, H. et al. Innate lymphocyte/ly6c(hi) monocyte crosstalk promotes Klebsiella pneumoniae clearance. Cell 165 , 679–689 (2016).

Hinks, T. S. C. et al. Steroid-induced deficiency of mucosal-associated invariant T cells in the chronic obstructive pulmonary disease lung. Implications for nontypeable Haemophilus influenzae infection. Am. J. Respir. Crit. Care Med. 194 , 1208–1218 (2016).

Meierovics, A. I. & Cowley, S. C. MAIT cells promote inflammatory monocyte differentiation into dendritic cells during pulmonary intracellular infection. J. Exp. Med. 213 , 2793–2809 (2016).

Liu, J. et al. Advanced role of neutrophils in common respiratory diseases. J. Immunol. Res. 2017 , 6710278 (2017).

Castanheira, F. V. S. & Kubes, P. Neutrophils and NETs in modulating acute and chronic inflammation. Blood 133 , 2178–2185 (2019).

Xiong, H. et al. Distinct contributions of neutrophils and CCR2+ monocytes to pulmonary clearance of different Klebsiella pneumoniae strains. Infect. Immun. 83 , 3418–3427 (2015).

Winter, C. et al. Important role for CC chemokine ligand 2-dependent lung mononuclear phagocyte recruitment to inhibit sepsis in mice infected with Streptococcus pneumoniae. J. Immunol. 182 , 4931–4937 (2009).

Weber, G. F. et al. Pleural innate response activator B cells protect against pneumonia via a GM-CSF-IgM axis. J. Exp. Med. 211 , 1243–1256 (2014).

de Stoppelaar, S. F. et al. Thrombocytopenia impairs host defense in gram-negative pneumonia-derived sepsis in mice. Blood 124 , 3781–3790 (2014).

van den Boogaard, F. E. et al. Thrombocytopenia impairs host defense during murine Streptococcus pneumoniae pneumonia. Crit. Care Med. 43 , e75–e83 (2015).

Ichinohe, T. et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl Acad. Sci. USA 108 , 5354–5359 (2011).

Schuijt, T. J. et al. The gut microbiota plays a protective role in the host defence against pneumococcal pneumonia. Gut 65 , 575–583 (2016).

Haak, B. W. & Wiersinga, W. J. The role of the gut microbiota in sepsis. Lancet Gastroenterol. Hepatol. 2 , 135–143 (2017).

Netea, M. G., Schlitzer, A., Placek, K., Joosten, L. A. B. & Schultze, J. L. Innate and adaptive immune memory: an evolutionary continuum in the host’s response to pathogens. Cell Host Microbe 25 , 13–26 (2019).

Giamarellos-Bourboulis, E. J. et al. Activate: randomized clinical trial of BCG vaccination against infection in the elderly. Cell 183 , 315–323.e9 (2020).

Hwang, J. Y., Randall, T. D. & Silva-Sanchez, A. Inducible bronchus-associated lymphoid tissue: taming inflammation in the lung. Front. Immunol. 7 , 258 (2016).

Snyder, M. E. & Farber, D. L. Human lung tissue resident memory T cells in health and disease. Curr. Opin. Immunol. 59 , 101–108 (2019).

Smith, N. M. et al. Regionally compartmentalized resident memory T cells mediate naturally acquired protection against pneumococcal pneumonia. Mucosal Immunol. 11 , 220–235 (2018).

Serhan, C. N. & Levy, B. D. Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J. Clin. Invest. 128 , 2657–2669 (2018).

Flitter, B. A. et al. Pseudomonas aeruginosa sabotages the generation of host proresolving lipid mediators. Proc. Natl Acad. Sci. USA 114 , 136–141 (2017).

Sham, H. P. et al. 15-epi-lipoxin A4, resolvin D2, and resolvin D3 induce NF-κB regulators in bacterial pneumonia. J. Immunol. 200 , 2757–2766 (2018).

Zemans, R. L. et al. Neutrophil transmigration triggers repair of the lung epithelium via β-catenin signaling. Proc. Natl Acad. Sci. USA 108 , 15990–15995 (2011).

Liu, Y. et al. FoxM1 mediates the progenitor function of type II epithelial cells in repairing alveolar injury induced by Pseudomonas aeruginosa. J. Exp. Med. 208 , 1473–1484 (2011).

Kumar, P. A. et al. Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell 147 , 525–538 (2011).

Matsuzaki, Y. et al. Stat3 is required for cytoprotection of the respiratory epithelium during adenoviral infection. J. Immunol. 177 , 527–537 (2006).

Quinton, L. J. et al. Alveolar epithelial STAT3, IL-6 family cytokines, and host defense during Escherichia coli pneumonia. Am. J. Respir. Cell Mol. Biol. 38 , 699–706 (2008).

Quinton, L. J. et al. Leukemia inhibitory factor signaling is required for lung protection during pneumonia. J. Immunol. 188 , 6300–6308 (2012).

Poe, S. L. et al. STAT1-regulated lung MDSC-like cells produce IL-10 and efferocytose apoptotic neutrophils with relevance in resolution of bacterial pneumonia. Mucosal Immunol. 6 , 189–199 (2013).

D’Alessio, F. R. et al. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in mice and are present in humans with acute lung injury. J. Clin. Invest. 119 , 2898–2913 (2009).

Monticelli, L. A. et al. Innate lymphoid cells promote lung-tissue homeostasis after infection with influenza virus. Nat. Immunol. 12 , 1045–1054 (2011).

Laidlaw, B. J. et al. CD4+ T cell help guides formation of CD103+ lung-resident memory CD8+ T cells during influenza viral infection. Immunity 41 , 633–645 (2014).

Xu, X. et al. Conventional NK cells can produce IL-22 and promote host defense in Klebsiella pneumoniae pneumonia. J. Immunol. 192 , 1778–1786 (2014).

Kradin, R. L. & Digumarthy, S. The pathology of pulmonary bacterial infection. Semin. Diagn. Pathol. 34 , 498–509 (2017).

Pritt, B. S. & Aubry, M. C. Histopathology of viral infections of the lung. Semin. Diagn. Pathol. 34 , 510–517 (2017).

Singer, M. et al. The third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA 315 , 801–810 (2016).

Angus, D. C. & van der Poll, T. Severe sepsis and septic shock. N. Engl. J. Med. 369 , 840–851 (2013).

Dremsizov, T. et al. Severe sepsis in community-acquired pneumonia: when does it happen, and do systemic inflammatory response syndrome criteria help predict course? Chest 129 , 968–978 (2006).

Giuliano, K. K. & Baker, D. Sepsis in the context of nonventilator hospital-acquired pneumonia. Am. J. Crit. Care 29 , 9–14 (2020).

Hotchkiss, R. S. et al. Sepsis and septic shock. Nat. Rev. Dis. Prim. 2 , 16045 (2016).

van der Poll, T., van de Veerdonk, F. L., Scicluna, B. P. & Netea, M. G. The immunopathology of sepsis and potential therapeutic targets. Nat. Rev. Immunol. 17 , 407–420 (2017).

Corrales-Medina, V. F., Musher, D. M., Shachkina, S. & Chirinos, J. A. Acute pneumonia and the cardiovascular system. Lancet 381 , 496–505 (2013).

Corrales-Medina, V. F. et al. Association between hospitalization for pneumonia and subsequent risk of cardiovascular disease. JAMA 313 , 264–274 (2015). The short-term and long-term risk of cardiovascular diseases after CAP hospitalization is shown in this capital study .

Corrales-Medina, V. F. et al. Cardiac complications in patients with community-acquired pneumonia: a systematic review and meta-analysis of observational studies. PLoS Med. 8 , e1001048 (2011).

Violi, F. et al. Cardiovascular complications and short-term mortality risk in community-acquired pneumonia. Clin. Infect. Dis. 64 , 1486–1493 (2017).

Ramirez, J. et al. Acute myocardial infarction in hospitalized patients with community-acquired pneumonia. Clin. Infect. Dis. 47 , 182–187 (2008).

Mortensen, E. M. et al. Causes of death for patients with community-acquired pneumonia: results from the Pneumonia Patient Outcomes Research Team cohort study. Arch. Intern. Med. 162 , 1059–1064 (2002).

Musher, D. M., Abers, M. S. & Corrales-Medina, V. F. Acute infection and myocardial infarction. N. Engl. J. Med. 380 , 171–176 (2019). A review article showing the evidence of acute respiratory viral infection and the increased risk of myocardial infarction .

Milbrandt, E. B. et al. Prevalence and significance of coagulation abnormalities in community-acquired pneumonia. Mol. Med. 15 , 438–445 (2009).

van Vught, L. A. et al. Comparative analysis of the host response to community-acquired and hospital-acquired pneumonia in critically Ill patients. Am. J. Respir. Crit. Care Med. 194 , 1366–1374 (2016).

Naghavi, M. et al. Influenza infection exerts prominent inflammatory and thrombotic effects on the atherosclerotic plaques of apolipoprotein E-deficient mice. Circulation 107 , 762–768 (2003).

Madjid, M., Vela, D., Khalili-Tabrizi, H., Casscells, S. W. & Litovsky, S. Systemic infections cause exaggerated local inflammation in atherosclerotic coronary arteries: clues to the triggering effect of acute infections on acute coronary syndromes. Tex. Heart Inst. J. 34 , 11–18 (2007).

Jaw, J. E. et al. Lung exposure to lipopolysaccharide causes atherosclerotic plaque destabilisation. Eur. Respir. J. 48 , 205–215 (2016).

Yende, S. et al. Inflammatory markers at hospital discharge predict subsequent mortality after pneumonia and sepsis. Am. J. Respir. Crit. Care Med. 177 , 1242–1247 (2008).

Yende, S. et al. Elevated hemostasis markers after pneumonia increases one-year risk of all-cause and cardiovascular deaths. PLoS ONE 6 , e22847 (2011).

Iwashyna, T. J., Ely, E. W., Smith, D. M. & Langa, K. M. Long-term cognitive impairment and functional disability among survivors of severe sepsis. JAMA 304 , 1787–1794 (2010).

Shah, F. A. et al. Bidirectional relationship between cognitive function and pneumonia. Am. J. Respir. Crit. Care Med. 188 , 586–592 (2013).

Girard, T. D., Dittus, R. S. & Ely, E. W. Critical illness brain injury. Annu. Rev. Med. 67 , 497–513 (2016).

Chung, H.-Y., Wickel, J., Brunkhorst, F. M. & Geis, C. Sepsis-associated encephalopathy: from delirium to dementia? J. Clin. Med. 9 , 703 (2020).

Prescott, H. C., Sjoding, M. W. & Iwashyna, T. J. Diagnoses of early and late readmissions after hospitalization for pneumonia. A systematic review. Ann. Am. Thorac. Soc. 11 , 1091–1100 (2014).

Dang, T. T., Majumdar, S. R., Marrie, T. J. & Eurich, D. T. Recurrent pneumonia: a review with focus on clinical epidemiology and modifiable risk factors in elderly patients. Drugs Aging 32 , 13–19 (2015).

Ekdahl, K., Braconier, J. H. & Svanborg, C. Immunoglobulin deficiencies and impaired immune response to polysaccharide antigens in adult patients with recurrent community-acquired pneumonia. Scand. J. Infect. Dis. 29 , 401–407 (1997).

Roquilly, A. et al. Alveolar macrophages are epigenetically altered after inflammation, leading to long-term lung immunoparalysis. Nat. Immunol. 21 , 636–648 (2020).

Lamping, D. L. et al. The community-acquired pneumonia symptom questionnaire: a new, patient-based outcome measure to evaluate symptoms in patients with community-acquired pneumonia. Chest 122 , 920–929 (2002).

Metlay, J. P. et al. Measuring symptomatic and functional recovery in patients with community-acquired pneumonia. J. Gen. Intern. Med. 12 , 423–430 (1997).

Chalmers, J. D. et al. Severity assessment tools for predicting mortality in hospitalised patients with community-acquired pneumonia. Systematic review and meta-analysis. Thorax 65 , 878–883 (2010).

Chalmers, J. D. et al. Risk factors for complicated parapneumonic effusion and empyema on presentation to hospital with community-acquired pneumonia. Thorax 64 , 556–558 (2009).

Falguera, M. et al. Predictive factors, microbiology and outcome of patients with parapneumonic effusion. Eur. Respir. J. 38 , 1173–1179 (2011).

Bhuiyan, M. U. et al. Combination of clinical symptoms and blood biomarkers can improve discrimination between bacterial or viral community-acquired pneumonia in children. BMC Pulmonary Med. 19 , 71 (2019).

Lhommet, C. et al. Predicting the microbial cause of community-acquired pneumonia: can physicians or a data-driven method differentiate viral from bacterial pneumonia at patient presentation? BMC Pulmonary Med. 20 , 62 (2020).

Torres, A., & Cillóniz, C. Clinical Management of Bacterial Pneumonia (Springer, 2015).

Cilloniz, C., Ceccato, A., San Jose, A. & Torres, A. Clinical management of community acquired pneumonia in the elderly patient. Expert Rev. Respir. Med. 10 , 1211–1220 (2016).

Schurink, C. A. M. et al. Clinical pulmonary infection score for ventilator-associated pneumonia: accuracy and inter-observer variability. Intensive Care Med. 30 , 217–224 (2004).

Fàbregas, N. et al. Clinical diagnosis of ventilator associated pneumonia revisited: comparative validation using immediate post-mortem lung biopsies. Thorax 54 , 867–873 (1999). The most complete immediate post-mortem study of VAP to validate clinical diagnosis .

Self, W. H., Courtney, D. M., McNaughton, C. D., Wunderink, R. G. & Kline, J. A. High discordance of chest x-ray and computed tomography for detection of pulmonary opacities in ED patients: implications for diagnosing pneumonia. Am. J. Emerg. Med. 31 , 401–405 (2013).

Laursen, C. B. et al. Diagnostic performance of chest X-ray for the diagnosis of community acquired pneumonia in acute admitted patients with respiratory symptoms. Scand. J. Trauma. Resusc. Emerg. Med. 21 , A21 (2013).

Claessens, Y.-E. et al. Early chest computed tomography scan to assist diagnosis and guide treatment decision for suspected community-acquired pneumonia. Am. J. Respir. Crit. Care Med. 192 , 974–982 (2015).

Ding, X., Xu, J., Zhou, J., Long, Q. & Chest, C. T. findings of COVID-19 pneumonia by duration of symptoms. Eur. J. Radiol. 127 , 109009 (2020).

Franquet, T. Imaging of community-acquired pneumonia. J. Thorac. Imaging 33 , 282–294 (2018).

D’Amato, M. et al. Assessment of thoracic ultrasound in complementary diagnosis and in follow up of community-acquired pneumonia (CAP). BMC Med. Imaging 17 , 52 (2017).

Long, L., Zhao, H.-T., Zhang, Z.-Y., Wang, G.-Y. & Zhao, H.-L. Lung ultrasound for the diagnosis of pneumonia in adults: a meta-analysis. Medicine 96 , e5713 (2017).

Mongodi, S. et al. Lung ultrasound for early diagnosis of ventilator-associated pneumonia. Chest 149 , 969–980 (2016).

Bouhemad, B., Dransart-Rayé, O., Mojoli, F. & Mongodi, S. Lung ultrasound for diagnosis and monitoring of ventilator-associated pneumonia. Ann. Transl. Med. 6 , 418 (2018).

Musher, D. M., Montoya, R. & Wanahita, A. Diagnostic value of microscopic examination of Gram-stained sputum and sputum cultures in patients with bacteremic pneumococcal pneumonia. Clin. Infect. Dis. 39 , 165–169 (2004).

Fukuyama, H., Yamashiro, S., Kinjo, K., Tamaki, H. & Kishaba, T. Validation of sputum Gram stain for treatment of community-acquired pneumonia and healthcare-associated pneumonia: a prospective observational study. BMC Infect. Dis. 14 , 534 (2014).

Ranzani, O. T. et al. Diagnostic accuracy of Gram staining when predicting staphylococcal hospital-acquired pneumonia and ventilator-associated pneumonia: a systematic review and meta-analysis. Clin. Microbiol. Infect. 26 , 1456–1463 (2020).

Torres, A., Artigas, A. & Ferrer, R. Biomarkers in the ICU: less is more? No. Intensive Care Med. 47 , 97–100 (2021).

Torres, A., Lee, N., Cilloniz, C., Vila, J. & Van der Eerden, M. Laboratory diagnosis of pneumonia in the molecular age. Eur. Respir. J. 48 , 1764–1778 (2016). In-depth revision of available molecular diagnostic techniques for bacterial and viral pneumonia .

Schulte, B. et al. Detection of pneumonia associated pathogens using a prototype multiplexed pneumonia test in hospitalized patients with severe pneumonia. PLoS ONE 9 , e110566 (2014).

Paonessa, J. R. et al. Rapid detection of methicillin-resistant Staphylococcus aureus in BAL: a pilot randomized controlled trial. Chest 155 , 999–1007 (2019).

Gastli, N. et al. Multicentric evaluation of BioFire FilmArray Pneumonia Panel for rapid bacteriological documentation of pneumonia. Clin. Microbiol. Infect. https://doi.org/10.1016/j.cmi.2020.11.014 (2020).

Article   PubMed   Google Scholar  

Centers for Disease Control and Prevention. Overview of Testing for SARS-CoV-2 (COVID-19). https://www.cdc.gov/coronavirus/2019-ncov/hcp/testing-overview.html (CDC, 2020).

Karakioulaki, M. & Stolz, D. The case of procalcitonin for lower respiratory tract infections. BRN Rev. 5 , 277–293 (2019).

Krüger, S. et al. Inflammatory parameters predict etiologic patterns but do not allow for individual prediction of etiology in patients with CAP: results from the German Competence Network CAPNETZ. Respir. Res. 10 , 65 (2009).

Ramirez, P. et al. Sequential measurements of procalcitonin levels in diagnosing ventilator-associated pneumonia. Eur. Respir. J. 31 , 356–362 (2008).

Luyt, C.-E. et al. Usefulness of procalcitonin for the diagnosis of ventilator-associated pneumonia. Intensive Care Med. 34 , 1434–1440 (2008).

Schuetz, P. et al. Procalcitonin (PCT)-guided antibiotic stewardship: an international experts consensus on optimized clinical use. Clin. Chem. Lab. Med. 57 , 1308–1318 (2019).

Liapikou, A., Cilloniz, C. & Torres, A. Drugs that increase the risk of community-acquired pneumonia: a narrative review. Expert Opin. Drug Saf. 17 , 991–1003 (2018).

Niederman, M. S. et al. Efficacy and effectiveness of a 23-valent polysaccharide vaccine against invasive and non-invasive pneumococcal disease and related outcomes: a review of available evidence. Expert Rev Vaccines https://doi.org/10.1080/14760584.2021.1880328 (2021).

Maruyama, T. et al. Efficacy of 23-valent pneumococcal vaccine in preventing pneumonia and improving survival in nursing home residents: double blind, randomised and placebo controlled trial. BMJ 340 , c1004 (2010).

Falkenhorst, G. et al. Effectiveness of the 23-valent pneumococcal polysaccharide vaccine (PPV23) against pneumococcal disease in the elderly: systematic review and meta-analysis. PLoS ONE 12 , e0169368 (2017).

Bonten, M. J. M. et al. Polysaccharide conjugate vaccine against pneumococcal pneumonia in adults. N. Engl. J. Med. 372 , 1114–1125 (2015).

Patterson, S. et al. A post hoc assessment of duration of protection in CAPiTA (Community Acquired Pneumonia immunization Trial in Adults). Trials Vaccinol. 5 , 92–96 (2016).

Millar, E. V. et al. Indirect effect of 7-valent pneumococcal conjugate vaccine on pneumococcal colonization among unvaccinated household members. Clin. Infect. Dis. 47 , 989–996 (2008).

Hammitt, L. L. et al. Indirect effect of conjugate vaccine on adult carriage of Streptococcus pneumoniae: an explanation of trends in invasive pneumococcal disease. J. Infect. Dis. 193 , 1487–1494 (2006).

Chung, J. R. et al. Effects of influenza vaccination in the United States during the 2018-2019 influenza season. Clin. Infect. Dis. 71 , e368–e376 (2020).

Restivo, V. et al. Influenza vaccine effectiveness among high-risk groups: a systematic literature review and meta-analysis of case-control and cohort studies. Hum. Vaccin. Immunother. 14 , 724–735 (2018).

Chow, E. J. et al. Vaccine effectiveness against influenza-associated lower respiratory tract infections in hospitalized adults, Louisville, Kentucky, 2010-2013. Open. Forum Infect. Dis. 7 , ofaa262 (2020).

Lyons, P. G. & Kollef, M. H. Prevention of hospital-acquired pneumonia. Curr. Opin. Crit. Care 24 , 370–378 (2018).

Álvarez-Lerma, F. et al. Prevention of ventilator-associated pneumonia: the multimodal approach of the Spanish ICU “Pneumonia Zero” Program. Crit. Care Med. 46 , 181–188 (2018).

Palomar, M. et al. Impact of a national multimodal intervention to prevent catheter-related bloodstream infection in the ICU: the Spanish experience. Crit. Care Med. 41 , 2364–2372 (2013).

Ma, S. et al. A meta analysis of the effect of enhanced hand hygiene on the morbidity of ventilator-associated pneumonia. Zhonghua Wei Zhong Bing. Ji Jiu Yi Xue 26 , 304–308 (2014).

Drakulovic, M. B. et al. Supine body position as a risk factor for nosocomial pneumonia in mechanically ventilated patients: a randomised trial. Lancet 354 , 1851–1858 (1999).

Wang, L. et al. Semi-recumbent position versus supine position for the prevention of ventilator-associated pneumonia in adults requiring mechanical ventilation. Cochrane Database Syst. Rev. 2016 , CD009946 (2016).

Li Bassi, G. et al. Randomized, multicenter trial of lateral Trendelenburg versus semirecumbent body position for the prevention of ventilator-associated pneumonia. Intensive Care Med. 43 , 1572–1584 (2017).

Guérin, C. et al. Prone positioning in severe acute respiratory distress syndrome. N. Engl. J. Med. 368 , 2159–2168 (2013).

Douglas, I. S. et al. Safety and outcomes of prolonged usual care prone position mechanical ventilation to treat acute coronavirus disease 2019 hypoxemic respiratory failure. Crit. Care Med. 49 , 490–502 (2021).

Shelhamer, M. C. et al. Prone positioning in moderate to severe acute respiratory distress syndrome due to COVID-19: a cohort study and analysis of physiology. J. Intensive Care Med. 36 , 241–252 (2021).

Sud, S., Sud, M., Friedrich, J. O. & Adhikari, N. K. J. Effect of mechanical ventilation in the prone position on clinical outcomes in patients with acute hypoxemic respiratory failure: a systematic review and meta-analysis. CMAJ 178 , 1153–1161 (2008).

Mao, Z. et al. Subglottic secretion suction for preventing ventilator-associated pneumonia: an updated meta-analysis and trial sequential analysis. Crit. Care 20 , 353 (2016).

Marjanovic, N. et al. Multicentre randomised controlled trial to investigate the usefulness of continuous pneumatic regulation of tracheal cuff pressure for reducing ventilator-associated pneumonia in mechanically ventilated severe trauma patients: the AGATE study protocol. BMJ Open 7 , e017003 (2017).

Philippart, F. et al. Randomized intubation with polyurethane or conical cuffs to prevent pneumonia in ventilated patients. Am. J. Respir. Crit. Care Med. 191 , 637–645 (2015).

Klompas, M., Speck, K., Howell, M. D., Greene, L. R. & Berenholtz, S. M. Reappraisal of routine oral care with chlorhexidine gluconate for patients receiving mechanical ventilation: systematic review and meta-analysis. JAMA Intern. Med. 174 , 751–761 (2014).

de Smet, A. M. G. A. et al. Decontamination of the digestive tract and oropharynx in ICU patients. N. Engl. J. Med. 360 , 20–31 (2009).

Wittekamp, B. H. et al. Decontamination strategies and bloodstream infections with antibiotic-resistant microorganisms in ventilated patients: a randomized clinical trial. JAMA 320 , 2087–2098 (2018).

Dang, T. T., Eurich, D. T., Weir, D. L., Marrie, T. J. & Majumdar, S. R. Rates and risk factors for recurrent pneumonia in patients hospitalized with community-acquired pneumonia: population-based prospective cohort study with 5 years of follow-up. Clin. Infect. Dis. 59 , 74–80 (2014).

Garcia-Vidal, C. et al. Aetiology of, and risk factors for, recurrent community-acquired pneumonia. Clin. Microbiol. Infect. 15 , 1033–1038 (2009).

Liu, P. et al. Frequency of empiric antibiotic de-escalation in an acute care hospital with an established Antimicrobial Stewardship Program. BMC Infect. Dis. 16 , 751 (2016).

Maruyama, T. et al. A therapeutic strategy for all pneumonia patients: a 3-year prospective multicenter cohort study using risk factors for multidrug-resistant pathogens to select initial empiric therapy. Clin. Infect. Dis. 68 , 1080–1088 (2018).

Abdul-Aziz, M. H., Lipman, J. & Roberts, J. A. Antibiotic dosing for multidrug-resistant pathogen pneumonia. Curr. Opin. Infect. Dis. 30 , 231–239 (2017).

Tsai, D., Lipman, J. & Roberts, J. A. Pharmacokinetic/pharmacodynamic considerations for the optimization of antimicrobial delivery in the critically ill. Curr. Opin. Crit. Care 21 , 412–420 (2015).

Mandell, L. A. et al. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the management of community-acquired pneumonia in adults. Clin. Infect. Dis. 44 (Suppl 2), S27–S72 (2007).

Sligl, W. I. et al. Macrolides and mortality in critically ill patients with community-acquired pneumonia: a systematic review and meta-analysis. Crit. Care Med. 42 , 420–432 (2014).

Torres, A. et al. Challenges in severe community-acquired pneumonia: a point-of-view review. Intensive Care Med. 45 , 159–171 (2019).

Niederman, M. S. Macrolide-resistant pneumococcus in community-acquired pneumonia. Is there still a role for macrolide therapy? Am. J. Respir. Crit. Care Med. 191 , 1216–1217 (2015).

Asadi, L. et al. Guideline adherence and macrolides reduced mortality in outpatients with pneumonia. Respir. Med. 106 , 451–458 (2012).

Postma, D. F. et al. Cardiac events after macrolides or fluoroquinolones in patients hospitalized for community-acquired pneumonia: post-hoc analysis of a cluster-randomized trial. BMC Infect. Dis. 19 , 17 (2019).

Garin, N. et al. β-Lactam monotherapy vs β-lactam-macrolide combination treatment in moderately severe community-acquired pneumonia: a randomized noninferiority trial. JAMA Intern. Med. 174 , 1894–1901 (2014).

Gershengorn, H. B., Keene, A., Dzierba, A. L. & Wunsch, H. The association of antibiotic treatment regimen and hospital mortality in patients hospitalized with Legionella pneumonia. Clin. Infect. Dis. 60 , e66–e79 (2015).

Niederman, M. S. Antibiotic treatment of hospital-acquired pneumonia: is it different from ventilator-associated pneumonia? Curr. Opin. Crit. Care 24 , 353–360 (2018).

Kumar, A., Safdar, N., Kethireddy, S. & Chateau, D. A survival benefit of combination antibiotic therapy for serious infections associated with sepsis and septic shock is contingent only on the risk of death: a meta-analytic/meta-regression study. Crit. Care Med. 38 , 1651–1664 (2010).

Martin-Loeches, I. et al. Potentially resistant microorganisms in intubated patients with hospital-acquired pneumonia: the interaction of ecology, shock and risk factors. Intensive Care Med. 39 , 672–681 (2013).

Niederman, M. S. et al. Inhaled amikacin adjunctive to intravenous standard-of-care antibiotics in mechanically ventilated patients with Gram-negative pneumonia (INHALE): a double-blind, randomised, placebo-controlled, phase 3, superiority trial. Lancet Infect. Dis. 20 , 330–340 (2020).

Chastre, J. et al. Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA 290 , 2588–2598 (2003). A seminal article comparing 8 or 15 days of antibiotic treatment in VAP .

Garnacho-Montero, J. et al. Task force on management and prevention of Acinetobacter baumannii infections in the ICU. Intensive Care Med. 41 , 2057–2075 (2015).

Timsit, J.-F., Pilmis, B. & Zahar, J.-R. How should we treat hospital-acquired and ventilator-associated pneumonia caused by extended-spectrum β-lactamase-producing enterobacteriaceae? Semin. Respir. Crit. Care Med. 38 , 287–300 (2017).

de Jong, E. et al. Efficacy and safety of procalcitonin guidance in reducing the duration of antibiotic treatment in critically ill patients: a randomised, controlled, open-label trial. Lancet Infect. Dis. 16 , 819–827 (2016).

Sousa, D. et al. Community-acquired pneumonia in immunocompromised older patients: incidence, causative organisms and outcome. Clin. Microbiol. Infect. 19 , 187–192 (2013).

Ramirez, J. A. et al. Treatment of community-acquired pneumonia in immunocompromised adults: a consensus statement regarding initial strategies. Chest 158 , 1896–1911 (2020).

El-Solh, A. A. et al. Microbiology of severe aspiration pneumonia in institutionalized elderly. Am. J. Respir. Crit. Care Med. 167 , 1650–1654 (2003).

Siemieniuk, R. A. C. et al. Corticosteroid therapy for patients hospitalized with community-acquired pneumonia: a systematic review and meta-analysis. Ann. Intern. Med. 163 , 519–528 (2015).

Torres, A. et al. Effect of corticosteroids on treatment failure among hospitalized patients with severe community-acquired pneumonia and high inflammatory response: a randomized clinical trial. JAMA 313 , 677–686 (2015).

Rodrigo, C., Leonardi-Bee, J., Nguyen-Van-Tam, J. & Lim, W. S. Corticosteroids as adjunctive therapy in the treatment of influenza. Cochrane Database Syst. Rev. 3 , CD010406 (2016).

Recovery Collaborative Group. et al. Dexamethasone in hospitalized patients with Covid-19. N. Engl. J. Med. 384 , 693–704 (2021).

Welte, T. et al. Efficacy and safety of trimodulin, a novel polyclonal antibody preparation, in patients with severe community-acquired pneumonia: a randomized, placebo-controlled, double-blind, multicenter, phase II trial (CIGMA study). Intensive Care Med. 44 , 438–448 (2018).

Frat, J.-P. et al. High-flow oxygen through nasal cannula in acute hypoxemic respiratory failure. N. Engl. J. Med. 372 , 2185–2196 (2015).

Kaplan, V. et al. Pneumonia: still the old man’s friend? Arch. Intern. Med. 163 , 317–323 (2003).

Shorr, A. F. et al. Readmission following hospitalization for pneumonia: the impact of pneumonia type and its implication for hospitals. Clin. Infect. Dis. 57 , 362–367 (2013).

Chalmers, J. D. et al. Epidemiology, antibiotic therapy, and clinical outcomes in health care-associated pneumonia: a UK cohort study. Clin. Infect. Dis. 53 , 107–113 (2011).

El Moussaoui, R. et al. Long-term symptom recovery and health-related quality of life in patients with mild-to-moderate-severe community-acquired pneumonia. Chest 130 , 1165–1172 (2006).

Wootton, D. G. et al. A longitudinal modelling study estimates acute symptoms of community acquired pneumonia recover to baseline by 10 days. Eur. Respir. J. 49 , 1602170 (2017).

Marrie, T. J., Lau, C. Y., Wheeler, S. L., Wong, C. J. & Feagan, B. G. Predictors of symptom resolution in patients with community-acquired pneumonia. Clin. Infect. Dis. 31 , 1362–1367 (2000).

Almirall, J. et al. Epidemiology of community-acquired pneumonia in adults: a population-based study. Eur. Respir. J. 15 , 757–763 (2000).

Andrade, L. F. et al. Health related quality of life in patients with community-acquired pneumococcal pneumonia in France. Health Qual. Life Outcomes 16 , 28 (2018).

Wyrwich, K. W., Yu, H., Sato, R. & Powers, J. H. Observational longitudinal study of symptom burden and time for recovery from community-acquired pneumonia reported by older adults surveyed nationwide using the CAP Burden of Illness Questionnaire. Patient Relat. Outcome Meas. 6 , 215–223 (2015).

Carratala, J. et al. Outpatient care compared with hospitalization for community-acquired pneumonia: a randomized trial in low-risk patients. Ann. Intern. Med. 142 , 165–172 (2005).

Mangen, M.-J. J., Huijts, S. M., Bonten, M. J. M. & de Wit, G. A. The impact of community-acquired pneumonia on the health-related quality-of-life in elderly. BMC Infect. Dis. 17 , 208 (2017).

Lloyd, M., Callander, E., Karahalios, A., Desmond, L. & Karunajeewa, H. Patient-reported outcome measures in community-acquired pneumonia: a systematic review of application and content validity. BMJ Open. Respir. Res. 6 , e000398 (2019).

Gao, C. A. et al. Comparing nasopharyngeal and BAL SARS-CoV-2 assays in respiratory failure. Am. J. Respir. Crit. Care Med. 203 , 127–129 (2021).

Peiffer-Smadja, N. et al. Performance and impact of a multiplex PCR in ICU patients with ventilator-associated pneumonia or ventilated hospital-acquired pneumonia. Crit. Care 24 , 366 (2020).

Murphy, C. N. et al. Multicenter evaluation of the biofire filmarray pneumonia/pneumonia plus panel for detection and quantification of agents of lower respiratory tract infection. J. Clin. Microbiol. 58 , e00128-20 (2020).

Pendleton, K. M. et al. Rapid pathogen identification in bacterial pneumonia using real-time metagenomics. Am. J. Respir. Crit. Care Med. 196 , 1610–1612 (2017).

Chiu, C. Y. & Miller, S. A. Clinical metagenomics. Nat. Rev. Genet. 20 , 341–355 (2019).

Hellyer, T. P. et al. Biomarker-guided antibiotic stewardship in suspected ventilator-associated pneumonia (VAPrapid2): a randomised controlled trial and process evaluation. Lancet Respir. Med. 8 , 182–191 (2020).

Blot, S. I. et al. A clinical algorithm to diagnose invasive pulmonary aspergillosis in critically ill patients. Am. J. Respir. Crit. Care Med. 186 , 56–64 (2012).

Bassetti, M., Welte, T. & Wunderink, R. G. Treatment of Gram-negative pneumonia in the critical care setting: is the beta-lactam antibiotic backbone broken beyond repair? Crit. Care 20 , 19 (2016).

Kollef, M. H. et al. Ceftolozane-tazobactam versus meropenem for treatment of nosocomial pneumonia (ASPECT-NP): a randomised, controlled, double-blind, phase 3, non-inferiority trial. Lancet Infect. Dis. 19 , 1299–1311 (2019). A randomized clinical trial comparing ceftolozane–tazobactam with meropenem in ventilated HAP and VAP. A post-hoc analysis in ventilated HAP demonstrated superiority of ceftolozane–tazobactam .

Kollef, M. H. et al. A randomized trial of 7-day doripenem versus 10-day imipenem-cilastatin for ventilator-associated pneumonia. Crit. Care 16 , R218 (2012).

File, T. M. et al. FOCUS 1: a randomized, double-blinded, multicentre, phase III trial of the efficacy and safety of ceftaroline fosamil versus ceftriaxone in community-acquired pneumonia. J. Antimicrob. Chemother. 66 , iii19–iii32 (2011).

Biedenbach, D. J., Kazmierczak, K., Bouchillon, S. K., Sahm, D. F. & Bradford, P. A. In vitro activity of aztreonam-avibactam against a global collection of Gram-negative pathogens from 2012 and 2013. Antimicrob. Agents Chemother. 59 , 4239–4248 (2015).

Awad, S. S. et al. A phase 3 randomized double-blind comparison of ceftobiprole medocaril versus ceftazidime plus linezolid for the treatment of hospital-acquired pneumonia. Clin. Infect. Dis. 59 , 51–61 (2014).

David, S. et al. Epidemic of carbapenem-resistant Klebsiella pneumoniae in Europe is driven by nosocomial spread. Nat. Microbiol. 4 , 1919–1929 (2019).

Watkins, R. R. & File, T. M. Lefamulin: a novel semisynthetic pleuromutilin antibiotic for community-acquired bacterial pneumonia. Clin. Infect. Dis. 71 , 2757–2762 (2020).

Spellberg, B., Bartlett, J., Wunderink, R. & Gilbert, D. N. Novel approaches are needed to develop tomorrow’s antibacterial therapies. Am. J. Respir. Crit. Care Med. 191 , 135–140 (2015).

Matteo Bassetti, R. E. et al. Efficacy and safety of cefiderocol for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): results of a phase 3 randomised, open-label, parallel-assigned, pathogen-focused study. Lancet 21 , 226–240 (2021).

Barnes, M. D. et al. Targeting multidrug-resistant Acinetobacter spp.: sulbactam and the diazabicyclooctenone β-lactamase inhibitor ETX2514 as a novel therapeutic agent. mBio 10 , e00159-19 (2019).

Lehman, K. M. & Grabowicz, M. Countering gram-negative antibiotic resistance: recent progress in disrupting the outer membrane with novel therapeutics. Antibiotics (Basel) 8 , 163 (2019).

Wu, J. Y., Srinivas, P. & Pogue, J. M. Cefiderocol: a novel agent for the management of multidrug-resistant gram-negative organisms. Infect. Dis. Ther. 9 , 17–40 (2020).

Wunderink, R. G. et al. Cefiderocol versus high-dose, extended-infusion meropenem for the treatment of Gram-negative nosocomial pneumonia (APEKS-NP): a phase 3, randomised, double-blind, non-inferiority study. Lancet Infect. Dis. 21 , 213–225 (2020).

File, T. M. et al. Efficacy and safety of IV-to-oral lefamulin, a pleuromutilin antibiotic, for treatment of community-acquired bacterial pneumonia: the phase 3 LEAP 1 trial. Clin. Infect. Dis. 69 , 1856–1867 (2019).

Alexander, E. et al. Oral lefamulin vs moxifloxacin for early clinical response among adults with community-acquired bacterial pneumonia: the LEAP 2 randomized clinical trial. JAMA 322 , 1661–1671 (2019).

Que, Y.-A. et al. Assessment of panobacumab as adjunctive immunotherapy for the treatment of nosocomial Pseudomonas aeruginosa pneumonia. Eur. J. Clin. Microbiol. Infect. Dis. 33 , 1861–1867 (2014).

François, B. et al. Safety and pharmacokinetics of an anti-PcrV PEGylated monoclonal antibody fragment in mechanically ventilated patients colonized with Pseudomonas aeruginosa: a randomized, double-blind, placebo-controlled trial. Crit. Care Med. 40 , 2320–2326 (2012).

François, B. et al. Safety and tolerability of a single administration of AR-301, a human monoclonal antibody, in ICU patients with severe pneumonia caused by Staphylococcus aureus: first-in-human trial. Intensive Care Med. 44 , 1787–1796 (2018).

Maddocks, S. et al. Bacteriophage therapy of ventilator-associated pneumonia and empyema caused by Pseudomonas aeruginosa. Am. J. Respir. Crit. Care Med. 200 , 1179–1181 (2019).

Wunderink, R. G. Turning the phage on treatment of antimicrobial-resistant pneumonia. Am. J. Respir. Crit. Care Med. 200 , 1081–1082 (2019).

Sicot, N. et al. Methicillin resistance is not a predictor of severity in community-acquired Staphylococcus aureus necrotizing pneumonia – results of a prospective observational study. Clin. Microbiol. Infect. 19 , E142–E148 (2013).

Download references

Acknowledgements

A.T. is the recipient of ICREA award from Generalitat de Catalunya. C.C. is the recipient of the SEPAR fellowship 2018, a grant 2019 from the Fondo de Investigación Sanitaria (PI19/00207), and the SEPAR fellowship “Programa Mentor”. We thank J.J.T.H. Roelofs (Department of Pathology, Amsterdam UMC, Amsterdam, Netherlands) for his invaluable assistance with the section on lung pathology and in providing representative histopathology slides.

Author information

Authors and affiliations.

Department of Pneumology, Hospital Clinic of Barcelona, Barcelona, Spain

Antoni Torres & Catia Cilloniz

August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain

University of Barcelona, Barcelona, Spain

Biomedical Research Networking Centers in Respiratory Diseases (CIBERES and CIBERESUCICOVID study), Barcelona, Spain

Division of Pulmonary and Critical Care, New York Presbyterian/Weill Cornell Medical Center, New York City, NY, USA

Michael S. Niederman

Department of Pneumology, Hospital Universitario y Politécnico La Fe, Valencia, Spain

Rosario Menéndez

Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK

James D. Chalmers

Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA

Richard G. Wunderink

Center of Experimental and Molecular Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands

Tom van der Poll

You can also search for this author in PubMed   Google Scholar

Contributions

Introduction (C.C. and A.T.); Epidemiology (C.C. and R.M.); Mechanisms/pathophysiology (T.v.d.P.); Diagnosis, screening and prevention (C.C. and A.T.); Management (M.S.N.); Quality of life (J.D.C.); Outlook (R.G.W); Overview of Primer (A.T. and C.C.).

Corresponding authors

Correspondence to Antoni Torres or Catia Cilloniz .

Ethics declarations

Competing interests.

A.T. has been a paid consultant to Pfizer, Jansen, and MSD, and a speaker for Pfizer and MSD. M.S.N. has received research grants from Shionogi, Bayer and Merck. He has been a paid consultant to Bayer, Merck, Paratek, Abbvie, Nabriva, and Thermo-Fisher. J.D.C. has received research funding from Astrazeneca, Boehringer-Ingelheim, Gilead Sciences, Glaxosmithkline, Insmed and Novartis; he has received consultancy fees from Chiesi, Grifols and Zambon. R.G.W. is a consultant to Merck, Shionogi, Polyphor, Microbiotix, bioMerieux, Curetis, KBP Biosciences, Idorsia and Accelerate. All other authors declare no competing interests.

Additional information

Peer review information.

Nature Reviews Disease Primers thanks Y. Arabi, C. Ginocchio, K. Klugman, M. Metersky, N. Suttorp and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Torres, A., Cilloniz, C., Niederman, M.S. et al. Pneumonia. Nat Rev Dis Primers 7 , 25 (2021). https://doi.org/10.1038/s41572-021-00259-0

Download citation

Accepted : 26 February 2021

Published : 08 April 2021

DOI : https://doi.org/10.1038/s41572-021-00259-0

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

This article is cited by

Risk factors and predicting nomogram for the clinical deterioration of non-severe community-acquired pneumonia.

  • Cheng-bin Xu
  • Shan-shan Su

BMC Pulmonary Medicine (2024)

Validating the accuracy of deep learning for the diagnosis of pneumonia on chest x-ray against a robust multimodal reference diagnosis: a post hoc analysis of two prospective studies

  • Jeremy Hofmeister
  • Nicolas Garin
  • Virginie Prendki

European Radiology Experimental (2024)

Hospitalization, case fatality, comorbidities, and isolated pathogens of adult inpatients with pneumonia from 2013 to 2022: a real-world study in Guangzhou, China

  • Zhufeng Wang
  • Jinping Zheng

BMC Infectious Diseases (2024)

A methodical exploration of imaging modalities from dataset to detection through machine learning paradigms in prominent lung disease diagnosis: a review

  • Sunil Kumar
  • Harish Kumar
  • Manoj Diwakar

BMC Medical Imaging (2024)

Diagnostic value of plasma heparin-binding protein and the heparin-binding protein-to-albumin ratio in patients with community-acquired Pneumonia: a retrospective study

  • Xueqin Xiao
  • Zhaozhong Xu

BMC Infectious Diseases (2023)

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

introduction pneumonia case study

Nursing Case Study for Pneumonia

Watch More! Unlock the full videos with a FREE trial

Included In This Lesson

Study tools.

Access More! View the full outline and transcript with a FREE trial

Charles is a 72-year-old male patient admitted via the emergency department to the medical surgical floor at 2220 with a diagnosis of community acquired pneumonia (CAP).  He arrives in the room via stretcher with oxygen (O2) via nasal cannula (NC) and is able to transfer to the bed with minimal assistance. He does get short of breath (SOB) with exertion.

What assessment findings does the nurse expect for this patient? Should there be a particular focus to the assessment?

  • The great majority of patients with CAP present with fever. Other systemic symptoms such as chills, fatigue, malaise, chest pain (which may be pleuritic, nurse should ask questions about onset/radiation/provoking factors), and anorexia are also common. Tachycardia, leukocytosis with a leftward shift, or leukopenia are also findings that are mediated by the systemic inflammatory response. Focus should be respiratory in nature (auscultation, adventitious lung sounds like crackles & rhonchi) along with checking labs, CXR. Level of consciousness (LOC) on admission is important to monitor for changes.

Are there any ER results the nurse should ask about during the bedside report?

  • Since the patient has a cough and fever, the nurse should check to see if a COVID test was done. It would also be beneficial to ask about chest x-rays (CXR), if blood and/or sputum cultures were taken, AND if antibiotics were already started. Most healthcare systems treat pneumonia as a core measure and must meet very strict metric guidelines (following a protocol, doing certain things within a narrow time frame window, etc)

What orders does the nurse expect the admitting provider to give?

  • After admission, it is common for providers to give continuous pulse oximetry, O2 titration orders (i.e. titrate O2 to keep O2 saturation above 92%), and IV fluids. If cultures are not done already, those should be ordered. There may be a daily CXR to monitor changes. The nurse should check the med administration record for antibiotics and there may be medications for the cough/sputum (tesslon perles, guaifenesin, etc.) and fever control (acetaminophen 1000 mg q 6 hrs PRN fever greater than 101 F for example)

After screening and assessing the patient, the nurse has the following data:

Patient AA&Ox4. SOB noted with speaking and after he moves around. IV 20g noted in left arm. Productive cough with moderate sputum production occasionally. Lung sounds in all fields indicate crackles; no barrel chest noted. Skin is warm and dry. He voids per urinal no assistance needed. Verbalizes understanding of call light use.

BP 120/60 SpO2 93% on NC 2L HR 100 bpm and regular Ht 172 cm RR 18 bpm Wt 60 kg Temp 38.3°C

CXR – posteroanterior and lateral chest radiographs obtained (two view). Radiographic findings consistent with the diagnosis of CAP including minor lobar consolidations, moderate interstitial infiltrates

CBC (abnormal/significant only listed, if NOT listed then the value falls within expected limits or is not significant for this patient), for reference see nursing.com Lab Value cheat sheet: WBC 15,000 cells/mcL Bands 10% Neutrophils 60% Eosinophils 1% Basophils 1% Lymphocytes 20%

Prioritize the top nursing interventions.

  • Frequent respiratory reassessments are vital for this patient along with continued LOC monitoring. The patient may need assistance with managing secretions so suction should be readily available. Positioning in the bed may alleviate symptoms and provide comfort – patient should not lie flat if there are airway concerns. The nurse should also ensure core measures are being followed. Blood cultures are needed as well as sputum so CORRECT collection and time of collection are necessary for proper monitoring. Timely administration of antibiotics helps improve outcomes.

What should the nurse be on the lookout for as the shift progresses? What warrants a call to the provider?

  • Watch for airway compromise or O2 sat decline. Ensure LOC does not decline inappropriately as this may indicate worsening of condition (respiratory failure). If the provider has given parameters to call, the nurse must either hesitate nor apologize for reporting these to the provider. If there is a sepsis algorithm, this should be followed since the patient is at risk for this condition. Fever uncontrolled with antipyretics needs to be reported. Worsening chest pain or sense of impending doom may indicate a pulmonary embolism or other complication and should be reported.

Are there other orders the nurse might anticipate and/or suggest?

  • A baseline lactic acid level (blood test) helps screen for sepsis. Strict I&O collection should begin since patients may have decreased appetite and/or thirst. Decreased urine output could be a sign of inadequate perfusion, so monitoring output is important. Respiratory therapy should be consulted for O2 titration, breathing treatments, O2 equipment/humidification and incentive spirometry (IS). Placing another IV may be helpful in case of patient deterioration (one in each upper extremity). Consider asking for Urinary antigen testing for S. pneumoniae

Are there past medical history concerns specific to this patient and his background that may aid in the plan of care?

  • Checking pneumococcal vaccine history is important – the patient may need education if he did not receive this vaccine. If he is homeless there is a greater risk he has tuberculosis or possibly non-compliant at discharge (due to lack of resources) so should be screened. His travel history may be relevant as treatment progresses and for Infectious diseases to consider. If he has a tobacco use history, this must be documented (i.e. 20 pack history) because that puts him at higher risk of chronic lung disease. If he is unable to care adequately for himself then case management may need to be involved for short-term acute care post-hospitalization.

ABG values:

pH 7.30 PaCO2 50 mmHg HCO3- 23 mEq/L PaO2 88 mmHg

What does this ABG indicate? How do you know? What may have caused this value? What can you do?

  • This is respiratory acidosis (pH < 7.35; PaCO2 > 45). It can be from hypoventilation -CNS depression -Pulmonary edema -Respiratory arrest -Airway obstruction. He may have an airway obstruction (thick sputum in throat) or his respiratory status could be worsening overall. Obtain VS, increase RR by arousing/waking him, reposition patient, maintain patent airway

The nurse gets the CNA to assist and repositions Charles in his hospital bed and he now sits straight but comfortably up in bed with pillows to bolster him. A yankauer at the bedside that he can use for a productive cough allows him to clear his airway after a brief teaching session. He had been frequently removing his NC due to ear discomfort, so RT brought padding for the tubing and Charles reports improved comfort. RT and the nurse teach him about “turn, cough, deep breath” aka TCDB.

After sleeping on and off through the shift, Charles is able to consume approximately 75% of his breakfast. He can properly demonstrate use of his IS and TCDB techniques. His NC remains in place with the padding on ears and his O2 sat is 95% on 2 L. He has two functioning IVs, one in each arm. Physician rounds and advises to continue plan of care including medication regimen based on test results after he and nurse discuss patient during rounds with the clinical pharmacist.

Describe examples of interdisciplinary team collaboration that may be useful in this patient’s care.

  • The nurse should maintain open communication and good rapport with respiratory therapy and work as a team for the best outcomes. Pharmacy may be consulted for an appropriate antibiotic regimen and/or if core measures are being used. Nursing assistants can assist with repositioning and I&O adherence. A team of physicians may be on this patient’s case (i.e. Pulmonology, Infectious Diseases, etc), so being able to give appropriate updates to a variety of professionals is vital (i.e. SBAR).

View the FULL Outline

When you start a FREE trial you gain access to the full outline as well as:

  • SIMCLEX (NCLEX Simulator)
  • 6,500+ Practice NCLEX Questions
  • 2,000+ HD Videos
  • 300+ Nursing Cheatsheets

“Would suggest to all nursing students . . . Guaranteed to ease the stress!”

References:

View the full transcript, nursing case studies.

Jon Haws

This nursing case study course is designed to help nursing students build critical thinking.  Each case study was written by experienced nurses with first hand knowledge of the “real-world” disease process.  To help you increase your nursing clinical judgement (critical thinking), each unfolding nursing case study includes answers laid out by Blooms Taxonomy  to help you see that you are progressing to clinical analysis.We encourage you to read the case study and really through the “critical thinking checks” as this is where the real learning occurs.  If you get tripped up by a specific question, no worries, just dig into an associated lesson on the topic and reinforce your understanding.  In the end, that is what nursing case studies are all about – growing in your clinical judgement.

Nursing Case Studies Introduction

Cardiac nursing case studies.

  • 6 Questions
  • 7 Questions
  • 5 Questions
  • 4 Questions

GI/GU Nursing Case Studies

  • 2 Questions
  • 8 Questions

Obstetrics Nursing Case Studies

Respiratory nursing case studies.

  • 10 Questions

Pediatrics Nursing Case Studies

  • 3 Questions
  • 12 Questions

Neuro Nursing Case Studies

Mental health nursing case studies.

  • 9 Questions

Metabolic/Endocrine Nursing Case Studies

Other nursing case studies.

Open Access is an initiative that aims to make scientific research freely available to all. To date our community has made over 100 million downloads. It’s based on principles of collaboration, unobstructed discovery, and, most importantly, scientific progression. As PhD students, we found it difficult to access the research we needed, so we decided to create a new Open Access publisher that levels the playing field for scientists across the world. How? By making research easy to access, and puts the academic needs of the researchers before the business interests of publishers.

We are a community of more than 103,000 authors and editors from 3,291 institutions spanning 160 countries, including Nobel Prize winners and some of the world’s most-cited researchers. Publishing on IntechOpen allows authors to earn citations and find new collaborators, meaning more people see your work not only from your own field of study, but from other related fields too.

Brief introduction to this section that descibes Open Access especially from an IntechOpen perspective

Want to get in touch? Contact our London head office or media team here

Our team is growing all the time, so we’re always on the lookout for smart people who want to help us reshape the world of scientific publishing.

Home > Books > Pneumonia

Introductory Chapter: Pneumonia

Published: 11 May 2022

DOI: 10.5772/intechopen.103675

Cite this chapter

There are two ways to cite this chapter:

From the Edited Volume

Edited by Nima Rezaei

To purchase hard copies of this book, please contact the representative in India: CBS Publishers & Distributors Pvt. Ltd. www.cbspd.com | [email protected]

Chapter metrics overview

254 Chapter Downloads

Impact of this chapter

Total Chapter Downloads on intechopen.com

IntechOpen

Total Chapter Views on intechopen.com

Author Information

Aysan moeinafshar.

  • Cancer Immunology Project Interest Group (CIP), Universal Scientific Education and Research Network (USERN), Iran
  • School of Medicine, Tehran University of Medical Sciences, Iran
  • Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Iran

Nima Rezaei *

  • Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Iran
  • Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Iran

*Address all correspondence to: [email protected]

1. Introduction

Pneumonia is an umbrella term regarding a variety of syndromes with different etiologies and refers to infection of the lung parenchyma. Pneumonia can be caused by a variety of microorganisms including bacterial, viral, and fungal pathogens [ 1 ]. Bacterial pneumonia can be divided into typical and atypical infections. Typical pneumonia is caused by microorganisms with the possibility of culturing on standard media or observation using gram staining techniques, such as Streptococcus pneumonia , Staphylococcus aureus, Haemophylus influenza, Moraxella catarrhalis, Group A streptococci, and gram-negative bacteria (both anaerobic and aerobic species). Atypical pneumonia is caused by pathogens that do not fit the aforementioned criteria; such as Mycoplasma pneumoniae, Chlamydia pneumoniae, and Legionella [ 2 ]. A variety of viruses, of both RNA and DNA virus families, can lead to pneumonia characteristics in patients. Some examples of these viruses include respiratory syncytial virus (RSV), rhinovirus, influenza viruses, parainfluenza viruses, adenovirus, varicella-zoster virus (VZV), cytomegalovirus (CMV), especially in HIV-infected patients, measles, and coronavirus family [ 3 ]. Fungal infections, though mostly overlooked, are important sources of pneumonia in immunocompromised patients. Some of the important organisms in this group include Histoplasma, Blastomyces, and Coccidioides [ 4 ].

2. Types of pneumonia

Pneumonia is classified into three groups based on etiology, disease characteristics, and clinical setting of the pathogen transmission. These subtypes include community-acquired pneumonia (CAP), hospital-acquired pneumonia (HAP), and ventilator-associated pneumonia (VAP) [ 1 ].

CAP is a type of pneumonia that is acquired in a community setting, caused by both atypical and typical bacterial organisms, viruses, and fungi [ 1 , 5 ]. HAP is considered a type of pneumonia acquired 48 h after hospital admission, with no incubation at time of admission [ 6 ]. On the other hand, VAP is a pneumonia acquired in patients under endotracheal incubation 48 h after the procedure [ 7 ]. The underlying pathogens responsible for HAP and VAP include gram-negative bacilli ( Escherichia coli , Pseudomonas Aeruginosa, Acinetobacter, Enterobacter, Klebsiella, etc.) and gram-positive cocci such as S. aureus [ 8 ]. Also, aspiration of both small (micro-aspiration) and large (macro-aspiration) amounts of oropharyngeal and upper gastrointestinal secretions is responsible for aspiration pneumonia, which accounts for approximately 5–15% of CAP cases [ 9 ].

Pneumonia’s pattern of pulmonary involvement in these infectious diseases varies widely and can be categorized into lobar pneumonia, lobular pneumonia, and focal/diffused interstitial pneumonia [ 10 ].

Risk factors predisposing patients to each of these pneumonia subtypes are summarized in Table 1 .

Rrisk factors of pneumonia. (DM = diabetes mellitus, CVD = cardiovascular disease, ARDS = acute respiratory distress syndrome, p superscript: pediatric cases, A superscript: adult cases).

3. Disease burden

The results of the Etiology of Pneumonia in the Community (EPIC) study in the united states indicated the annual incidence of CAP to be 2.4 cases per 1000 adults, mostly in age groups of >65 years old [ 12 ]. Similar studies in Europe estimated the annual incidence to be 1.07–1.2 cases per 1000 people [ 13 ]. The annual incidence of HAP is about 5–10 patients per 1000 hospital admissions worldwide and VAP cases include 10–25% of patients under ventilation [ 14 ].

Mortality of CAP in outpatient care, hospital wards, and ICU is <1%, 4–18%, and up to 50% respectively [ 15 , 16 , 17 ]. HAP and VAP are the most common causes of death in hospital-acquired infections with global mortality rates of 20–10% and 20–50% respectively [ 18 , 19 , 20 , 21 , 22 ].

4. Pathophysiology

Inability of the immune system in clearance of pathogens from the lower respiratory system is the basis for the incidence of pneumonia [ 23 ]. In addition to the pathogens, both local and systemic immune responses lead to parenchymal damage, constitutional symptoms, fluid congestion, pus formation in lungs, and reduction in alveolar compliance [ 24 ].

Pathological findings throughout this process consist of four stages including congestion, due to intra-alveolar edema, red hepatization, gray hepatization, both with characteristics of increased firmness of the parenchyma, and resolution [ 25 ].

5. Diagnosis

Most important symptoms stated by patients include fever, chills, diaphoresis, fatigue, myalgia, malaise, productive or non-productive coughs, dyspnea, and pleuritic chest pain [ 23 ]. Clinical data used in the definition of pneumonia include symptoms of acute lower respiratory tract disease for periods of less than 7 days, minimum of one systemic symptom, and newly appeared signs in chest examination unexplained by other conditions. Along with clinical criteria, laboratory tests, such as complete blood count (CBC), serum electrolytes, procalcitonin levels, renal and liver function tests, and radiologic findings, such as consolidations, abnormal silhouettes, opacities, and infiltrates, are also of importance in defining pneumonia [ 26 ].

6. Management

Management of CAP is carried out after a risk stratification process mostly using CURB-65 criteria. This scale consists of five criteria; Confusion, Uremia (BUN>20 mg/dl), respiratory rate > 30/minute, blood pressure < 90/60 mmHg, and ages>65 years. Treatments of HAP and VAP take more time and are more complicated. First-line treatment of pneumonia includes empirical antibiotic therapy using broad-spectrum antibiotics [ 1 ].

Figure 1 summarizes the approaches in the management of pneumonia.

introduction pneumonia case study

Approach to pneumonia [ 1 ].

  • 1. Jain V, Vashisht R, Yilmaz G, Bhardwaj A. Pneumonia Pathology. FL: Treasure Island; 2021
  • 2. Sattar SBA, Sharma S. Bacterial Pneumonia. FL: Treasure Island; 2021
  • 3. Freeman AM, Leigh Townes RJ. Viral Pneumonia. FL: Treasure Island; 2021
  • 4. Hage CA, Knox KS, Wheat LJ. Endemic mycoses: Overlooked causes of community acquired pneumonia. Respiratory Medicine. 2012; 106 (6):769-776
  • 5. Mandell LA, Wunderink RG, Anzueto A, Bartlett JG, Campbell GD, Dean NC, et al. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the management of community-acquired pneumonia in adults. Clinical Infectious Diseases: An Offical Publication Infectious Diseases Socical American. 2007; 44 (Suppl 2):S27-S72
  • 6. Kalil AC, Metersky ML, Klompas M, Muscedere J, Sweeney DA, Palmer LB, et al. Executive summary: Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clinical Infectious Diseases: An Offical Publication Infectious Diseases Socical American. 2016; 63 (5):575-582
  • 7. Canadian Critical Care Trials Group. A randomized trial of diagnostic techniques for ventilator-associated pneumonia. The New England Journal of Medicine. Dec 2006; 355 (25):2619-2630
  • 8. Jones RN. Microbial etiologies of hospital-acquired bacterial pneumonia and ventilator-associated bacterial pneumonia. Clinical Infectious Diseases: An Offical Publication Infectious Diseases Socical American. 2010; 51 (Suppl 1):S81-S87
  • 9. Mandell LA, Niederman MS. Aspiration pneumonia. The New England Journal of Medicine. 2019; 380 (7):651-663
  • 10. Gharib AM, Stern EJ. Radiology of pneumonia. The Medical Clinics of North America. 2001; 85 (6):1461-1491
  • 11. Torres A, Cilloniz C, Niederman MS, Menéndez R, Chalmers JD, Wunderink RG, et al. Pneumonia. Nature Reviews Disease Primers [Internet]. 2021; 7 (1):25. DOI: 10.1038/s41572-021-00259-0
  • 12. Jain S, Self WH, Wunderink RG, Fakhran S, Balk R, Bramley AM, et al. Community-acquired pneumonia requiring hospitalization among US adults. The New England Journal of Medicine. 2015; 373 (5):415-427
  • 13. Torres A, Peetermans WE, Viegi G, Blasi F. Risk factors for community-acquired pneumonia in adults in Europe: A literature review. Thorax. 2013; 68 (11):1057-1065
  • 14. Torres A, Niederman MS, Chastre J, Ewig S, Fernandez-Vandellos P, Hanberger H, et al. International ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia: Guidelines for the management of hospital-acquired pneumonia (HAP)/ventilator-associated pneumonia (VAP) of the European Respiratory Society (ERS), European Society of Intensive Care Medicine (ESICM), European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and Asociación Latinoamericana del Tórax (ALAT). The European Respiratory Journal. 2017; 50 (3):1700582
  • 15. Arnold FW, Wiemken TL, Peyrani P, Ramirez JA, Brock GN. Mortality differences among hospitalized patients with community-acquired pneumonia in three world regions: Results from the Community-Acquired Pneumonia Organization (CAPO) International Cohort Study. Respiratory Medicine. 2013; 107 (7):1101-1111
  • 16. Heo JY, Song JY. Disease burden and etiologic distribution of community-acquired pneumonia in adults: Evolving epidemiology in the era of pneumococcal conjugate vaccines. Infection & Chemotherapy. 2018; 50 (4):287-300
  • 17. Cillóniz C, Ewig S, Polverino E, Marcos MA, Prina E, Sellares J, et al. Community-acquired pneumonia in outpatients: Aetiology and outcomes. The European Respiratory Journal. 2012; 40 (4):931-938
  • 18. Magill SS, Edwards JR, Bamberg W, Beldavs ZG, Dumyati G, Kainer MA, et al. Multistate point-prevalence survey of health care–associated infections. The New England Journal of Medicine. 2014; 370 (13):1198-1208
  • 19. Micek ST, Chew B, Hampton N, Kollef MH. A case-control study assessing the impact of nonventilated hospital-acquired pneumonia on patient outcomes. Chest. 2016; 150 (5):1008-1014
  • 20. Melsen WG, Rovers MM, Groenwold RHH, Bergmans DCJJ, Camus C, Bauer TT, et al. Attributable mortality of ventilator-associated pneumonia: A meta-analysis of individual patient data from randomised prevention studies. The Lancet Infectious Diseases. 2013; 13 (8):665-671
  • 21. Barbier F, Andremont A, Wolff M, Bouadma L. Hospital-acquired pneumonia and ventilator-associated pneumonia: Recent advances in epidemiology and management. Current Opinion in Pulmonary Medicine. 2013; 19 (3):216-228
  • 22. Bassetti M, Righi E, Vena A, Graziano E, Russo A, Peghin M. Risk stratification and treatment of ICU-acquired pneumonia caused by multidrug-resistant/extensively drug-resistant/pandrug-resistant bacteria. Current Opinion in Critical Care. 2018; 24 (5):385-393
  • 23. Kaysin A, Viera AJ. Community-acquired pneumonia in adults: Diagnosis and management. American Family Physician. 2016; 94 (9):698-706
  • 24. Cilloniz C, Martin-Loeches I, Garcia-Vidal C, San Jose A, Torres A. Microbial etiology of pneumonia: Epidemiology, diagnosis and resistance patterns. International Journal of Molecular Sciences. Dec 2016; 17 (12):2120
  • 25. Kradin RL, Digumarthy S. The pathology of pulmonary bacterial infection. Seminars in Diagnostic Pathology. 2017; 34 (6):498-509
  • 26. Ticona JH, Zaccone VM, McFarlane IM. Community-acquired pneumonia: A focused review. American Journal of Medical Case Reports. 2021; 9 (1):45-52. Available from: https://pubmed.ncbi.nlm.nih.gov/33313398 [Accessed: April 11, 2020]

© 2022 The Author(s). Licensee IntechOpen. This chapter is distributed under the terms of the Creative Commons Attribution 3.0 License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Continue reading from the same book

By Sachin M. Patil

810 downloads

By Kien T. Nguyen, Suol T. Pham, Thu P.M. Vo, Chu X. ...

521 downloads

By Timothy R. Borgogna and Jovanka M. Voyich

284 downloads

  • Search Menu
  • Volume 2024, Issue 4, April 2024
  • Bariatric Surgery
  • Breast Surgery
  • Cardiothoracic Surgery
  • Colorectal Surgery
  • Colorectal Surgery, Upper GI Surgery
  • Gynaecology
  • Hepatobiliary Surgery
  • Interventional Radiology
  • Neurosurgery
  • Ophthalmology
  • Oral and Maxillofacial Surgery
  • Otorhinolaryngology - Head & Neck Surgery
  • Paediatric Surgery
  • Plastic Surgery
  • Transplant Surgery
  • Trauma & Orthopaedic Surgery
  • Upper GI Surgery
  • Vascular Surgery
  • Author Guidelines
  • Submission Site
  • Open Access
  • Reasons to Submit
  • About Journal of Surgical Case Reports
  • Editorial Board
  • Advertising and Corporate Services
  • Journals Career Network
  • Self-Archiving Policy
  • Journals on Oxford Academic
  • Books on Oxford Academic

Issue Cover

Article Contents

Introduction, case report, conflict of interest statement, data availability, consent for publication.

  • < Previous

Exploring a rare case of juvenile psammomatoid ossifying fibroma in the ethmoid: a case study and review

ORCID logo

  • Article contents
  • Figures & tables
  • Supplementary Data

Taha Yassine Aaboudech, Hafsa El Ouazzani, Habiba Kadiri, Leila Essakalli, Ayoub Bouteyine, Hanae Benadbdenbi, Naji Rguieg, Nadia Cherradi, Exploring a rare case of juvenile psammomatoid ossifying fibroma in the ethmoid: a case study and review, Journal of Surgical Case Reports , Volume 2024, Issue 4, April 2024, rjae242, https://doi.org/10.1093/jscr/rjae242

  • Permissions Icon Permissions

Juvenile ossifying fibroma (JOF) and its variants, including juvenile psammomatoid ossifying fibroma (JPOF), represent rare yet clinically significant benign fibro-osseous lesions that primarily occur in children and young adolescents. They can be found in diverse anatomical sites such as the jaw, nasal cavity, paranasal sinuses, and orbit. JOF exhibits an aggressive nature, necessitating early radiological detection and surgical intervention. Similarly, JPOF, with a locally malignant potential, requires surgical removal, typically conducted through endoscopic approaches. We report a case of a 5-year-old girl with JPOF arising in the ethmoid, revealed by recurrent epistaxis and proptosis. The text emphasizes the importance of early diagnosis through histopathology as a diagnostic tool and underscores the need for appropriate management.

Juvenile ossifying fibroma (JOF) is a rare condition primarily observed in children and adolescents. This benign fibro-osseous tumor commonly occurs within the sinonasal region. Its distinctions from the conventional ossifying fibroma lie in the age of onset, anatomical site, locally aggressive behavior, and a notable propensity for recurrence. Clinical manifestations vary based on the tumor’s location, potentially presenting as nasal obstruction, facial swelling, or proptosis. The World Health Organization (WHO) classifies JOF into two main subtypes: juvenile trabecular ossifying fibroma and juvenile psammomatoid ossifying fibroma (JPOF) [ 1 ]. The psammomatoid subtype is a rare, benign tumor found in the head and neck region, often affecting children and young adults. It tends to develop in areas such as the nasal cavity, paranasal sinuses, or the orbit. Although typically non-cancerous, it may display locally aggressive behavior, potentially damaging nearby structures. Surgical removal is the preferred treatment, with either endoscopic or external approaches being considered, with a preference for the latter. Due to the risk of significant bleeding during surgery, precautions to secure blood products beforehand are essential [ 2 ].

This article outlines a case of JPOF diagnosed in a child.

A 5-year-old girl presented with progressive right eye proptosis and recurrent epistaxis over the past 6 months, along with a history of nasal obstruction persisting for several months. During the examination, non-axial proptosis was observed, and both extraocular movements and visual acuity were found to be intact.

Flexible nasofibroscopy revealed a protrusion filling the right nasal fossa, closely approaching the nasal septum. Computed tomography (CT) and magnetic resonance imaging scans indicated a well-defined osteolytic lesion with a peripheral sclerotic rind, measuring 4.1 × 3.8 × 3.7 cm and originating from the right ethmoid, expanding into the ipsilateral nasal cavity, orbit, and maxillary sinus ( Fig. 1 ).

CT showing an expansible, well-demarcated, osteolytic lesion with a peripheral sclerotic rind on the right side of the ethmoid bone, expanding into the ipsilateral nasal cavity, orbit, and maxillary sinus; (A) coronal CT scan; (B) axial CT scan.

CT showing an expansible, well-demarcated, osteolytic lesion with a peripheral sclerotic rind on the right side of the ethmoid bone, expanding into the ipsilateral nasal cavity, orbit, and maxillary sinus; (A) coronal CT scan; (B) axial CT scan.

Upon admission to the Ear, Nose, and Throat department, the patient underwent an initial Frozen Section procedure. The sample was promptly sent to the pathology laboratory for histological analysis. The findings indicated the presence of a benign mesenchymal tumor accompanied by roughly round-shaped calcifications. Given the potential for further complications, total tumor removal was deemed necessary to both prevent complications and establish a definitive histological diagnosis.

The pathology laboratory received a white to reddish fragmented tumor with an elastic to hard consistency. Histological examination disclosed a proliferation of spindle-shaped or ovoid cells with vesicular nuclei and rare mitotic figures. Cells were arranged in sheets, with numerous rounded purplish formations corresponding to psammomatous body-type calcifications. A few giant cells were also observed. The stroma of the tumor was fibrous and collagenous. No necrosis was found ( Fig. 2 ).

H&E staining of the pathological specimen at different magnifications shows a benign fibro-osseous proliferation characterized by a large number of spherical basophilic calcified entities with no osteoblastic rimming resembling psammoma bodies (arrowheads), within a fibroblastic stroma of uniform stellate or spindle-shaped cells; (A and B) low power magnifications; (C) x20 power magnification; (D) high power (x40) magnification.

H&E staining of the pathological specimen at different magnifications shows a benign fibro-osseous proliferation characterized by a large number of spherical basophilic calcified entities with no osteoblastic rimming resembling psammoma bodies (arrowheads), within a fibroblastic stroma of uniform stellate or spindle-shaped cells; (A and B) low power magnifications; (C) x20 power magnification; (D) high power (x40) magnification.

Immunohistochemical studies were conducted, revealing that the tumor cells did not express Cytokeratin, CD34, S100, smooth and striatal muscle markers, MDM2, progesterone receptor (PR), or epithelial membrane antigen (EMA). Vimentin was diffusely expressed, with a very low ki67 proliferation index estimated at 4% ( Fig. 3 ).

Immunostaining reveals that the tumor tissue is (A) diffusely positive for Vimentin in the stromal component; (B) low Ki-67 proliferation index; (C and D) negative staining for EMA and CD34.

Immunostaining reveals that the tumor tissue is (A) diffusely positive for Vimentin in the stromal component; (B) low Ki-67 proliferation index; (C and D) negative staining for EMA and CD34.

This case illustrates a typical clinical history, radiographic appearance, and classic histological findings of JPOF presented in the ethmoid.

JPOF predominantly affects younger individuals, with reported average ages ranging from 16 to 33 years. However, Malek’s review of 86 JPOF cases revealed an age range from 3 to 49 years, with a mean age of 17.7 years and a slight male predominance (1.2:1) [ 3–5 ]. This tumor is typically found in the nasal cavity, paranasal sinuses, orbits, or the fronto-ethmoid complex, with an exceedingly rare occurrence in the base or vault of the skull [ 6 ]. The primary clinical presentation often involves bony expansion, which is frequently observed. This expansion can manifest through a range of symptoms including proptosis, nasal obstruction, headaches, facial swelling, pain, and recurring sinus infections [ 7 ]. Additionally, it may exhibit an aggressive clinical course [ 8 ].

Radiologically, ossifying fibroma typically appears as a mass with a distinct border, unlike fibrous dysplasia. Moreover, ossifying fibroma lacks ground-glass opacity and instead exhibits a mixed density resembling both compact bone and fibrous tissue [ 9 ].While the appearance of the lesion on imaging might offer diagnostic insights, distinguishing it can be challenging due to its resemblances to other osteo-fibroid lesions within the facial skeleton [ 10 ].

Histologically, JPOF is characterized by a large number of spherical calcified entities with no osteoblastic rimming. Some of these calcifications have psammoma-like concentric lamellae with basophilic cores and an eosinophilic osteoid rim. The surrounding fibrous stroma is composed of loosely to densely collagenized tissue that contains a dense proliferation of fibroblast-like, spindle-shaped cells with hyperchromatic nuclei [ 11 ].

JPOF must be distinguished from other fibro-osseous lesions such as fibrous dysplasia, osteoblastoma, low-grade central osteosarcoma, and primary aneurysmal bone cyst. Additionally, it is important to consider intraosseous cavernous hemangioma and eosinophilic granuloma due to their similarities in radiological and histological features [ 6 , 12 ]. Misdiagnosis may occur with extracranial meningioma featuring psammoma bodies, which typically tests positive for EMA, unlike JPOF [ 7 ]. JPOF typically does not express S100 and CD34 [ 7 ].

Despite its benign and slow growth, this subtype exhibits local malignancy with a tendency to infiltrate surrounding structures. Therefore, accurate diagnosis is crucial, and treatment should involve complete tumor removal. Incomplete or partial resection is associated with a heightened recurrence risk [ 2 ].

Our report underscores the rarity of JPOF, focusing on its clinical, radiological, and primarily histological aspects. Despite its uncommon occurrence, JPOF poses diagnostic and management challenges due to its potential aggressiveness and recurrence. Histopathological examination remains pivotal for confirming the diagnosis, highlighting the importance of thorough evaluation and appropriate management guided by histological findings.

None declared.

No external funding sources were obtained for this submission.

No new data were generated or analyzed in support of this research.

Written informed consent for publication of their clinical details and/or clinical images was obtained from the patient.

El-Naggar AK , Chan JKC , Grandis JR , et al.  World Health Organization Classification of Tumours , 4th edn. Lyon, France : International Agency for Research on Cancer, 2017 , 251 – 2 .

Google Scholar

Google Preview

Kuźmińska M , Gotlib T , Niemczyk K . Juvenile psammomatoid ossifying fibroma–case report . Polski Przegląd Otorynolaryngologiczny 2021 ; 10 : 57 – 61 .

Chrcanovic BR , Gomez RS . Juvenile ossifying fibroma of the jaws and paranasal sinuses: a systematic review of the cases reported in the literature . Int J Oral Maxillofac Surg 2020 ; 49 : 28 – 37 .

Sarode SC , Sarode GS , Waknis P , et al.  Juvenile psammomatoid ossifying fibroma: a review . Oral Oncol 2011 ; 47 : 1110 – 6 .

Makek M . In: Karger S (ed). Clinical pathology of fibro-osteo-cemental lesions of the cranio-facial skeleton and jaw bones . Basel : Switzerland , 1983 , 128 – 227 .

López CB , Zabala AB , Bareño EU . Cranial juvenile psammomatoid ossifying fibroma: case report . J Neurosurg Pediatr 2016 ; 17 : 318 – 23 .

Wenig BM , Vinh TN , Smirniotopoulos JG , et al.  Aggressive psammomatoid ossifying fibromas of the sinonasal region: a clinicopathologic study of a distinct group of fibro-osseous lesions . Cancer 1995 ; 76 : 1155 – 65 .

Brademann G , Werner JA , Jänig U , et al.  Cemento-ossifying fibroma of the petromastoid region: case report and review of the literature . J Laryngol Otol 1997 ; 111 : 152 – 5 .

Sontakke SA , Karjodkar FR , Umarji HR . Computed tomographic features of fibrous dysplasia of maxillofacial region . Imaging Sci Dent 2011 ; 41 : 23 – 8 .

Bertrand B , Eloy P , Cornelis JP , et al.  Juvenile aggressive cemento-ossifying fibroma: case report and review of the literature . Laryngoscope 1993 ; 103 : 1385 – 90 .

Owosho AA , Hughes MA , Prasad JL , et al.  Psammomatoid and trabecular juvenile ossifying fibroma: two distinct radiologic entities . Oral Surg Oral Med Oral Pathol Oral Radiol 2014 ; 118 : 732 – 8 .

Nakajima R , Saito N , Uchino A , et al.  Juvenile psammomatoid ossifying fibroma with visual disturbance: a case report with imaging features . J Neuroimaging 2013 ; 23 : 137 – 40 .

Email alerts

Citing articles via, affiliations.

  • Online ISSN 2042-8812
  • Copyright © 2024 Oxford University Press and JSCR Publishing Ltd
  • About Oxford Academic
  • Publish journals with us
  • University press partners
  • What we publish
  • New features  
  • Open access
  • Institutional account management
  • Rights and permissions
  • Get help with access
  • Accessibility
  • Advertising
  • Media enquiries
  • Oxford University Press
  • Oxford Languages
  • University of Oxford

Oxford University Press is a department of the University of Oxford. It furthers the University's objective of excellence in research, scholarship, and education by publishing worldwide

  • Copyright © 2024 Oxford University Press
  • Cookie settings
  • Cookie policy
  • Privacy policy
  • Legal notice

This Feature Is Available To Subscribers Only

Sign In or Create an Account

This PDF is available to Subscribers Only

For full access to this pdf, sign in to an existing account, or purchase an annual subscription.

COMMENTS

  1. (PDF) A Case Study: Pneumonia

    A Case Study: Pneumonia. Muhammad Zeeshan Zafar * ... Introduction. Community-acquired pneumonia (CAP) is a common and. potentially serious illness t hat is associated with morbidity and.

  2. Treatment of Community-Acquired Pneumonia: A Case Report and Current

    1. Introduction. Each year, over 4 million ambulatory patients are treated for community-acquired pneumonia (CAP) in the United States (US) [], with approximately 80% treated on an outpatient basis [].Community-acquired bacterial pneumonia (CABP) is a common presenting illness in the urgent care setting, yet many providers underappreciate the mortality associated with pneumonia and underrate ...

  3. Case Study: 33-Year-Old Female Presents with Chronic SOB and Cough

    Case Presentation. History of Present Illness: A 33-year-old white female presents after admission to the general medical/surgical hospital ward with a chief complaint of shortness of breath on exertion. She reports that she was seen for similar symptoms previously at her primary care physician's office six months ago.

  4. Case 24-2020: A 44-Year-Old Woman with Chest Pain, Dyspnea, and Shock

    On examination, the temperature was 36.4°C, the heart rate 103 beats per minute, the blood pressure 79/51 mm Hg, the respiratory rate 30 breaths per minute, and the oxygen saturation 99% while ...

  5. Scenario 1: A Patient with Mild Community-Acquired Pneumonia

    To focus on clinical trial design issues pertinent to the population of patients with mild pneumonia, a typical clinical-trial candidate patient is described below. Case Presentation. Present illness. A 35-year-old male resident of Boston, Massachusetts, presents with fever and cough.

  6. PDF Pneumonia

    Introduction Community-acquired pneumonia (CAP) is a prevalent and po-tentially life-threatening infection with high morbidity and fatality rates. Only half of the cases have an aetiology microorganism iden-Citation: Gamya Shree V and K Suresh. "Pneumonia - Case Study". Acta Scientific Clinical Case Reports 3.4 (2022): 37-41.

  7. A case report on management of severe childhood pneumonia in low

    1. Introduction. Globally, Pneumonia sustains as the leading cause of death among under five years old children [].The recent World Health Organization (WHO) global report (2013) deemed that pneumonia accounts for approximately 120 million cases every year [], among which 14 million (12%) progress to severe pneumonia []; and developing countries belong into the most vulnerable vicinity (95%) [].

  8. Case 18-2021: An 81-Year-Old Man with Cough, Fever, and Shortness of

    An 81-year-old man presented with fever, cough, and shortness of breath. Within a few hours after presentation, chest pain and respiratory distress developed. A chest radiograph showed bilateral pa...

  9. Pneumonia

    A US study on the incidence, outcomes and disease burden in >18,000 hospitalized patients with COPD 37 found that, during the 2-year study, 3,419 patients had pneumonia; the annual incidence for ...

  10. PDF Community-Acquired Pneumonia: A Focused Review

    American Journal of Medical Case Reports, vol. 9, no. (120 21): 45-52. doi: 10.12691/ajmcr-9-1-12. 1. Introduction . CAP is one of the most common reasons for hospitalization in the US. It can affect individuals of any age and cause ... of Louisville Pneumonia Study was the main study we came across that was used to argue the burden

  11. Nursing Case Study for Pneumonia

    We're going to go through a case study for pneumonia together. Let's get started in this scenario. We have a 72-year-old patient who is male. He was admitted via the emergency department to the med-surg floor with a diagnosis of community-acquired pneumonia. He arrives in the room by a stretcher with oxygen flowing through a nasal cannula.

  12. PDF Impact of the change in WHO's severe pneumonia case definition on

    To improve the case management of pneumonia in low- and middle-income countries, the World Health Or-ganization (WHO) developed the Integrated Management of Childhood Illness guidelines in 1995. In 2005, these guidelines4 defined severe pneumonia as the presence of cough or dificulty in breathing and tachypnoea (> 50 breaths per minute for ...

  13. Case 32-2015

    For example, one study showed that such cross-reactivity occurred in 90% of patients with blastomycosis. 19 Thus, on the basis of the results of microbiologic tests, the diagnosis was B ...

  14. Introductory Chapter: Pneumonia

    Introduction. Pneumonia is an umbrella term regarding a variety of syndromes with different etiologies and refers to infection of the lung parenchyma. ... Micek ST, Chew B, Hampton N, Kollef MH. A case-control study assessing the impact of nonventilated hospital-acquired pneumonia on patient outcomes.

  15. Overview: Pneumonia

    Introduction. Pneumonia is an inflammation of the air sacs in the lungs and the surrounding tissue.It often leads to a sudden high fever, the feeling that you are very unwell, a cough and shortness of breath. Because pneumonia is usually caused by bacteria, it can generally be treated effectively with antibiotics.Vaccinations that can prevent infection by certain germs are also available.

  16. Scenario 1: A Patient with Hospital-Acquired Bacterial Pneumonia

    A 65-year-old man presents to the emergency department a few hours after the gradual onset of left-sided weakness and expressive aphasia. Vital signs in the emergency department are a blood pressure of 140/85 mm Hg in both arms, temperature of 37°C, respiratory rate of 16 breaths per minute, and a regular pulse of 80 beats per min.

  17. Educational Case: Aspiration Pneumonia

    The following fictional case is intended as a learning tool within the Pathology Competencies for Medical Education (PCME), a set of national standards for teaching pathology. ... Educational Case: Aspiration Pneumonia. Candice C. Black ... Pulmonary disease due to aspiration of food and other particulate matter: a clinicopathologic study of 59 ...

  18. Introduction

    Introduction. Pneumonia is an infection of the lower respiratory tract that can be caused by bacteria, fungi, viruses, protozoa, or parasites. Community-acquired pneumonia (CAP) occurs in patients who have gotten the infection in the community compared to nosocomial pneumonia which is acquired in a healthcare setting such as a hospital or a nursing facility.

  19. Smart'n

    Pneumonia case study 2. Here is the second pneumonia case study nursing: Pneumonia case study 2 initial presentation. An 85-year-old resident of a long-term care facility is brought to the emergency department with confusion, rapid breathing, and a fever. The patient's baseline health is compromised due to advanced age and underlying conditions.

  20. Educational Case: Aspiration Pneumonia

    Abstract. The following fictional case is intended as a learning tool within the Pathology Competencies for Medical Education (PCME), a set of national standards for teaching pathology. These are divided into three basic competencies: Disease Mechanisms and Processes, Organ System Pathology, and Diagnostic Medicine and Therapeutic Pathology.

  21. Nursing 704C

    Click Next to start the Case Study. Introduction. Pneumonia is an infection of the lower respiratory tract that can be caused by bacteria, fungi, viruses, protozoa, or parasites. Community-acquired pneumonia (CAP) occurs in patients who have gotten the infection in the community compared to nosocomial pneumonia which is acquired in a healthcare ...

  22. A Case Study of Pediatric Pneumonia With Empyema

    In addition, the introduction of the H. influenzae type b vaccine has decreased the percentage of pneumonia caused by this organism. Recent studies also indicate an increased incidence of empyema causedbyS.aureus,particularlymethicillin-resistantS.aureus (MRSA) (Schultz et al., 2004). The introduction of the H. influenzae type b vaccine and

  23. Exploring a rare case of juvenile psammomatoid ossifying fibroma in the

    Exploring a rare case of juvenile psammomatoid ossifying fibroma in the ethmoid: a case study and review Taha Yassine Aaboudech, Taha Yassine Aaboudech Pathology Department, Rabat Specialty Hospital, Morocco Mohammed V University in Rabat ... Introduction. Juvenile ossifying fibroma (JOF) is a rare condition primarily observed in children and ...

  24. Impact of the change in WHO's severe pneumonia case definition on

    Findings. At six of the seven sites, the percentages of all-cause pneumonia hospitalizations due to severe pneumonia were significantly less (P < 0.001) according to the 2013 WHO definition compared with the 2005 definition.However, the percentage of severe pneumonia hospitalizations, according to the two definitions of severe pneumonia, with primary end-point consolidation varied little ...