Next Article in Journal
Stent Fractures: New Insights into an Old Issue
Next Article in Special Issue
Assessment of Gastrointestinal Symptoms and Dyspnea in Patients Hospitalized due to COVID-19: Contribution to Clinical Course and Mortality
Previous Article in Journal
How to Explore an Endocrine Cause of Hypertension
Previous Article in Special Issue
Acute Mesenteric Ischemia in COVID-19 Patients
 
 
jcm-logo
Article Menu

Article Menu

Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Nationwide COVID-19-EII Study: Incidence, Environmental Risk Factors and Long-Term Follow-Up of Patients with Inflammatory Bowel Disease and COVID-19 of the ENEIDA Registry

by
Yamile Zabana
1,2,*,
Ignacio Marín-Jiménez
3,
Iago Rodríguez-Lago
4,5,
Isabel Vera
6,
María Dolores Martín-Arranz
7,
Iván Guerra
8,9,
Javier P. Gisbert
2,10,11,
Francisco Mesonero
12,
Olga Benítez
1,
Carlos Taxonera
13,14,
Ángel Ponferrada-Díaz
15,
Marta Piqueras
16,
Alfredo J. Lucendo
2,11,17,
Berta Caballol
2,18,
Míriam Mañosa
2,19,
Pilar Martínez-Montiel
20,
Maia Bosca-Watts
21,
Jordi Gordillo
22,
Luis Bujanda
2,23,24,
Noemí Manceñido
25,
Teresa Martínez-Pérez
26,
Alicia López
27,
Cristina Rodríguez-Gutiérrez
28,
Santiago García-López
29,
Pablo Vega
30,
Montserrat Rivero
31,
Luigi Melcarne
32,
Maria Calvo
33,
Marisa Iborra
2,34,
Manuel Barreiro de-Acosta
35,
Beatriz Sicilia
36,
Jesús Barrio
37,
José Lázaro Pérez
38,
David Busquets
39,
Isabel Pérez-Martínez
40,
Mercè Navarro-Llavat
41,
Vicent Hernández
42,
Federico Argüelles-Arias
43,
Fernando Ramírez Esteso
44,
Susana Meijide
45,
Laura Ramos
46,
Fernando Gomollón
2,47,
Fernando Muñoz
48,
Gerard Suris
49,
Jone Ortiz de Zarate
50,
José María Huguet
51,
Jordina Llaó
52,
Mariana Fe García-Sepulcre
53,
Mónica Sierra
54,
Miguel Durà
55,
Sandra Estrecha
56,
Ana Fuentes Coronel
57,
Esther Hinojosa
58,
Lorenzo Olivan
59,
Eva Iglesias
60,
Ana Gutiérrez
2,61,
Pilar Varela
62,
Núria Rull
63,
Pau Gilabert
64,
Alejandro Hernández-Camba
65,
Alicia Brotons
66,
Daniel Ginard
67,
Eva Sesé
68,
Daniel Carpio
69,
Montserrat Aceituno
1,2,
José Luis Cabriada
4,5,
Yago González-Lama
6,
Laura Jiménez
8,9,
María Chaparro
2,10,11,
Antonio López-San Román
12,
Cristina Alba
13,14,
Rocío Plaza-Santos
15,
Raquel Mena
16,
Sonsoles Tamarit-Sebastián
17,
Elena Ricart
2,18,
Margalida Calafat
2,19,
Sonsoles Olivares
20,
Pablo Navarro
21,
Federico Bertoletti
22,
Horacio Alonso-Galán
23,24,
Ramón Pajares
25,
Pablo Olcina
26,
Pamela Manzano
1,
Eugeni Domènech
2,19,
Maria Esteve
1,2 and
on behalf of the ENEIDA registry of GETECCU
add Show full author list remove Hide full author list
1
Hospital Universitari Mútua Terrassa, 08221 Terrassa, Spain
2
Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
3
Hospital Gregorio Marañón, 218007 Madrid, Spain
4
Gastroenterology Department, Hospital Universitario de Galdakao, 48960 Galdakao, Spain
5
Biocruces Bizkaia Health Research Institute, 48960 Galdakao, Spain
6
Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain
7
Hospital Universitario La Paz, 28046 Madrid, Spain
8
Hospital Universitario de Fuenlabrada, 28942 Fuenlabrada, Spain
9
Instituto de Investigación Hospital Universitario La Paz (IdiPaz), 28046 Madrid, Spain
10
Department of Gastroenterology, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
11
Instituto de Investigación Sanitaria Princesa (IIS-IP), 28006 Madrid, Spain
12
Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
13
Hospital Clínico San Carlos, 28040 Madrid, Spain
14
Instituto de Investigación del Hospital Clínico San Carlos [IdISSC], 28040 Madrid, Spain
15
Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
16
Consorci Sanitari de Terrassa, 08227 Terrassa, Spain
17
Hospital General de Tomelloso, 13700 Tomelloso, Spain
18
Hospital Clínic de Barcelona-IDIBAPS, 08036 Barcelona, Spain
19
Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
20
Fundación Hospital Universitario Doce de Octubre, 28041 Madrid, Spain
21
Hospital Clinic Universitari de Valencia, 46010 Valencia, Spain
22
Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
23
Hospital Universitario Donostia, Instituto Biodonostia, 20014 San Sebastián, Spain
24
Universidad del País Vasco (UPV/EHU), 48940 Leioua, Spain
25
Hospital Universitario Infanta Sofía, 28703 San Sebastián de los Reyes, Spain
26
Hospital Virgen de la Luz, 16002 Cuenca, Spain
27
Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Hospital del Mar, 08003 Barcelona, Spain
28
Complejo Hospitalario de Navarra, 31008 Pamplona, Spain
29
Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
30
Complexo Hospitalario Universitario de Ourense, 32005 Ourense, Spain
31
Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
32
Hospital Universitari Parc Taulí, 08208 Sabadell, Spain
33
Hospital San Pedro-Logroño, 26006 Logroño, Spain
34
Hospital Universitario y Politécnico de la Fe de Valencia, 46026 Valencia, Spain
35
Hospital Clínico Universitario de Santiago, 15706 Santiago, Spain
36
Hospital Universitario de Burgos, 09006 Burgos, Spain
37
Hospital Universitario Río Hortega (HURH), 47012 Valladolid, Spain
38
Hospital Universitario Fundación de Alcorcón, 28922 Alcorcón, Spain
39
Hospital Universitari de Girona Doctor Josep Trueta, 17007 Girona, Spain
40
Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
41
Hospital de Sant Joan Despí Moisès Broggi, 08970 Sant Joan Despí, Spain
42
Hospital Álvaro Cunqueiro, 36213 Vigo, Spain
43
Hospital Universitario Virgen de la Macarena, Universidad de Sevilla, 41009 Sevilla, Spain
44
Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain
45
Hospital Universitario de Cruces, 48903 Barakaldo, Spain
46
Hospital Universitario de Canarias, 38320 San Cristobal de la Laguna, Spain
47
Hospital Clínico Universitario “Lozano Blesa” and IIS Aragón, 50009 Zaragoza, Spain
48
Hospital Universitario de Salamanca, 37007 Salamanca, Spain
49
Hospital Universitari de Bellvitge, 08907 L’Hospitalet de Llobregat, Spain
50
Hospital Universitario de Basurto, 48013 Bilbo, Spain
51
Consorcio Hospital General Universitario de Valencia, 46014 Valencia, Spain
52
Althaia Xarxa Assistencial Universitària de Manresa, 08243 Manresa, Spain
53
Hospital Universitario de Elche, 03203 Elche, Spain
54
Complejo Asistencial Universitario de León, 24071 León, Spain
55
Hospital Clínico de Valladolid, 47003 Valladolid, Spain
56
Hospital Universitario Álava, 01009 Gasteiz, Spain
57
Hospital Virgen de la Concha, 49022 Zamora, Spain
58
Hospital de Manises, 46940 Manises, Spain
59
Hospital Universitario San Jorge, 22004 Huesca, Spain
60
Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía de Córdoba, 14004 Cordoba, Spain
61
Hospital General Universitario de Alicante, 03010 Alicante, Spain
62
Hospital Universitario de Cabueñes, 33394 Gijón, Spain
63
Hospital Universitario Son Llàtzer, 07198 Palma, Spain
64
Hospital de Viladecans, 08840 Viladecans, Spain
65
Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
66
Hospital Vega Baja de Orihuela, 03314 Alicante, Spain
67
Hospital Universitario Son Espases, 07120 Palma, Spain
68
Hospital Universitari Arnau de Vilanova de Lleida, 25198 Lleida, Spain
69
Complexo Hospitalario de Pontevedra, 36071 Pontevedra, Spain
*
Author to whom correspondence should be addressed.
All Co-authors are part of ENEIDA.
J. Clin. Med. 2022, 11(2), 421; https://doi.org/10.3390/jcm11020421
Submission received: 9 December 2021 / Revised: 23 December 2021 / Accepted: 6 January 2022 / Published: 14 January 2022

Abstract

:
We aim to describe the incidence and source of contagion of COVID-19 in patients with IBD, as well as the risk factors for a severe course and long-term sequelae. This is a prospective observational study of IBD and COVID-19 included in the ENEIDA registry (53,682 from 73 centres) between March–July 2020 followed-up for 12 months. Results were compared with data of the general population (National Centre of Epidemiology and Catalonia). A total of 482 patients with COVID-19 were identified. Twenty-eight percent were infected in the work environment, and 48% were infected by intrafamilial transmission, despite having good adherence to lockdown. Thirty-five percent required hospitalization, 7.9% had severe COVID-19 and 3.7% died. Similar data were reported in the general population (hospitalisation 19.5%, ICU 2.1% and mortality 4.6%). Factors related to death and severe COVID-19 were being aged ≥ 60 years (OR 7.1, 95% CI: 1.8–27 and 4.5, 95% CI: 1.3–15.9), while having ≥2 comorbidities increased mortality (OR 3.9, 95% CI: 1.3–11.6). None of the drugs for IBD were related to severe COVID-19. Immunosuppression was definitively stopped in 1% of patients at 12 months. The prognosis of COVID-19 in IBD, even in immunosuppressed patients, is similar to that in the general population. Thus, there is no need for more strict protection measures in IBD.

Graphical Abstract

1. Introduction

The COVID-19 pandemic hit Spain at the end of February 2020 and is far from under complete control. Data on affected cases and mortality are continuously updated [1]. There is evidence that patients suffering from inflammatory bowel disease (IBD) have a greater risk for infections, some of them opportunistic, mainly favoured by immunosuppressive treatment [2,3,4]. For that reason, experts on IBD, worried by the potential severity of COVID-19 in these patients, recommended, during the initial phases of the pandemic, that whenever possible, starting immunosuppressants should be delayed and treatment deescalated [5,6,7]. Notwithstanding this information, more than one year after the start of the pandemic, factors related to deleterious prognosis of COVID-19 in patients with IBD are essentially the same as those of the general population (mainly older age and comorbidities), whereas those on immunosuppressants do not appear to have a greater risk for severe COVID-19, except for corticosteroids [8]. In this sense, the international self-reported registry SECURE-IBD (https://covidibd.org (accessed on 24 August 2021)) has provided valuable clinical and therapeutic information [9]. Nevertheless, retrospective studies and registries have important limitations, such as reporting bias, over- or underrepresentation of the more severe cases of COVID-19, and the possibility of including confounding factors that may influence the results.
In addition, some studies have reported a low incidence of COVID-19 in patients with IBD [10], suggesting that IBD and the type of immunosuppressants administered for disease control do not represent risk factors for COVID-19. However, few of these studies are population based and do not address important environmental epidemiological risk factors, such as variability in the incidence of the infection in different regions within the same country. Neither do they address factors that may facilitate the infection, such as occupational risk, or those that may reduce the risk, as they may be specific isolation measures that could be recommended to a particular diseased population [10,11,12,13,14,15,16]. Likewise, the impact of COVID-19 on patients with IBD in the long term has not yet been explored.
The present study (COVID-19-EII study) was conducted in the setting of the ENEIDA project, the Spanish registry of patients with IBD, promoted by the Spanish Working Group on Crohn’s disease and ulcerative colitis (GETECCU) [17]. The aims of the present study were (1) to describe the incidence of COVID-19 in the ENEIDA registry, the geographical distribution of the infection compared with the distribution in the general Spanish population and exposure factors that may favour or prevent the infection (occupational risk and lockdown measures) during the first wave of the pandemic and (2) to describe the clinical characteristics and the disease course, including a 12-month follow-up after COVID-19.

2. Materials and Methods

2.1. Design

This was an observational prospective cohort study (COVID-19-EII) within the Spanish ENEIDA IBD registry. It included all patients with IBD who had COVID-19 between March and July 2020 (in the first wave) from the participant centres.

2.2. Study Population

The potential population was all patients with IBD registered in ENEIDA. Patients with COVID-19 were identified by an active search from their IBD unit (systematically addressing all the patients with IBD from the unit by email or phone call) or by direct notification from the patient itself, the family physician, the emergency department or the hospitalisation unit.

2.3. Data Collection

A prospective module hosted on the ENEIDA platform was specially designed for this study. Data collected included clinical baseline characteristics such as type of IBD, date of IBD diagnosis and Montreal’s classification [18], extraintestinal manifestations, family history of IBD and smoking behaviour at time of infection. The following comorbidities were specifically registered: cirrhosis, chronic renal failure, chronic obstructive pulmonary disease, heart disease, stroke, diabetes mellitus, arterial hypertension, dyslipidaemia, neoplasia, congestive heart failure, dementia, HIV, rheumatological disease or immune-mediated disease. Charlson’s index [19] was also calculated. We decided to explore both individual comorbidities and the Charlson comorbidity score, as these two approaches examine different aspects of comorbidity and are complementary. Variables measuring the exposure risk to SARS-CoV-2 included occupational risk (such as health care workers, teachers, basic services as supermarket cashiers, market clerks or pharmacy workers, police and firepersons, workers of closed institutions, veterinaries, animal control workers or conservation and forest technicians), compliance with lockdown measures, social distancing and the route of contagion. At the time of COVID-19 diagnosis, IBD activity was evaluated using the Harvey-Bradshaw index [20] or partial Mayo score [21]. The IBD therapeutic regimen was registered at the time of infection and up to 3 and 12 months before it: systemic steroid treatment, aminosalicylates, immunosuppressants (thiopurines, cyclosporine, methotrexate, tacrolimus and tofacitinib) and biologics (anti-TNF, vedolizumab and ustekinumab). Regarding COVID-19, the data collected included symptoms associated with the infection at the time of diagnosis, diagnostic procedures and specific treatment. The variables registered 3 and 12 months after COVID-19 were IBD activity and COVID-19 sequelae, both physical and psychological. To assess the impact of COVID-19 on IBD treatment, any change in medical therapy, including withdrawal of immunosuppression, both definitive and temporary, was collected during follow-up.

2.4. Definitions

COVID-19 diagnosis was based on a typical clinical picture consisting of fever (>38 °C), respiratory symptoms (cough and/or dyspnoea), anosmia or dysgeusia within the epidemiological setting. COVID-19 was considered confirmed by a positive diagnostic test including polymerase chain reaction (PCR) taken by nasopharyngeal swab or serology (IgM or IgG) for SARS-CoV-2. COVID-19 was considered probable in patients with a typical clinical picture but negative or lacking diagnostic tests. Asymptomatic patients with positive PCR or serology were not included.
It was considered that any patient had a good compliance with the lockdown measures when maintaining social distance by staying at home almost exclusively since 14 March 2020, the date the Spanish government ordered a total lockdown to prevent the spread of SARS-CoV-2.
Sequelae due to COVID-19 were any sign or symptom that the patient and/or physician considered related directly to COVID-19 and that was present at 3 and 12 months after infection.

2.5. Outcomes

To assess the disease course and clinical evolution of COVID-19, the following outcomes were registered and analysed: hospitalisation due to COVID-19, intensive care unit (ICU) admission, sequelae, severe COVID-19 and death. Severe COVID-19 was considered a composite variable that included ICU admission and/or use of active amines and/or respiratory distress and/or invasive oxygen therapy and/or death [22]. Cases with systemic inflammatory response syndrome were also registered. Data on outcomes of our study were compared to those registered on the SECURE-IBD registry (accessed on 24 August 2021) [9], considered the worldwide IBD registry on COVID-19. These outcomes were also compared to those of the general population taking into account data from Catalonia [23].

2.6. Ethical Considerations

The Scientific Committee of ENEIDA approved the study on 16 March 2020. It was also approved by the Ethics Committee of Hospital Universitari Mútua Terrassa (coordinating centre). Informed consent was obtained from all subjects. The patients were not identified by name in the publication, and no one, except the investigators of this study, had access to their local data, in accordance with the local Law of Personal Data Protection.

2.7. Statistical Analysis

Quantitative variables were compared with Student’s t test and the Mann–Whitney test, and the results are expressed as the means (±standard deviation) or median (±interquartile range (IQR) 25–75 percentiles). Quantitative variables were compared using Student’s t test for parametric data and the Mann–Whitney test for nonparametric data, while qualitative variables were compared using the Chi2 test or Fisher’s exact test, as appropriate. Univariate and multivariate logistic binary regression analyses were performed to explore the variables related to the need for hospitalisation, ICU admission, severe COVID-19, death and sequelae found at the 3- and 12-month follow-ups. The intensity of the significant associations was measured by calculating the OR and its 95% confidence interval. The multivariate models included significant variables in univariate analysis at the p < 0.1 level. In addition, for the outcomes with a small number of events (death and ICU admission), only the 2 most significant covariates in the univariate analysis were included. As the use of aminosalicylates was more frequent in patients with ulcerative colitis (UC) than in Crohn’s disease (CD), the model was adjusted to UC diagnosis when this drug was independently associated with a specific outcome.
Due to the great variability in the incidence of COVID-19 between the Spanish territories, the number of cases of both IBD and the general population are shown per province. Data on COVID-19 incidence in the general population as well as the age at COVID-19 diagnosis, age of hospitalized patients (including ICU admission) and age of patients with fatal outcomes were obtained from the National Centre of Epidemiology (CNE) [24,25]. The age- and sex-standardised incidence of every outcome in the IBD cohort was obtained based on all the patients actively followed in each participant centre of ENEIDA.

3. Results

A total of 73 out of the 86 centres adhered to the ENEIDA registry at the time of the study and decided to participate. This registry had, at that moment, 60,512 patients actively followed-up (data as for 15 July 2020), with 53,682 coming from the 73 participating centres (89% of the whole registry). Finally, 482 cases of COVID-19 were reported (251 males (52%); median 52 years (IQR: 42–61); cumulative incidence of 8.97 per 1000 patients with IBD, taking into account the population at risk from the participating centres). Ten centres that agreed to participate did not register any COVID-19 cases during the study period, despite being aware.

3.1. Clinical Baseline Characteristics

Table 1 and Table 2 show the most important clinical characteristics of the patients regarding IBD at the time of COVID-19 diagnosis. The activity of the disease and treatment are provided separately for UC and CD (Table 2). Notably, 80% of patients were in remission, and 9% had moderate–severe disease activity. Regarding IBD treatment at the time of the infection, 42% of the patients were on aminosalicylates, 5.4% were receiving systemic steroids, 36% were receiving immunosuppressants, 36% were receiving biologics and 12% were receiving combination therapy. Eleven percent of the patients required steroids within the 3 months before COVID-19 diagnosis.
Forty-four percent had at least one comorbidity, and 64% had a Charlson score of one or more. The most frequent comorbidity was arterial hypertension (22% (106/482)), followed by dyslipidaemia (15% (74/482)) and immune-mediated diseases (11% (53/482)) other than IBD (Supplementary Table S1).

3.2. Geografical Distribution of COVID-19 and Epidemiological Risk Factors of Exposure

The geographical distribution of cases is shown in Figure 1A, allowing a comparative approach with the incidence of COVID-19 in the Spanish general population (Figure 1B).
The majority of cases were reported in ENEIDA centres of the communities of Madrid and in those of the metropolitan area of Barcelona (red colour), where there is also the highest proportion of certified IBD units [26]. These two regions have the highest population density in Spain (844 and 743 inhabitants per km2, respectively, as of January 2021 [27]) and register the highest incidence of COVID-19 in the general population.
Compared to the Spanish population in March 2020 [24], the median age of IBD cases was similar: 52 years old (IQR, 42–61) in patients with IBD vs. 54 years old (IQR, 39–70) in the general population. When considering specific outcomes, a similar trend was observed: the age of hospitalised patients with IBD was 59 years old (IQR, 50–72) vs. 66 years old (IQR, 51–79) in the general population, and the age of ICU/death was 72 years old (IQR, 57–80) in patients with IBD vs. 70 years old (IQR, 59–80 years) in the general population
Regarding risk factors for COVID-19, almost half of the patients declared good adherence to lockdown measures (48% (229/482)). The circumstances that ensured an appropriate domiciliary lockdown were having preventive sick leave (31% (71/229)), being retired (26% (60/229)), doing telecommuting (18% (42/229)) and being unemployed (6.6% (15/229)). Table 3 summarizes the risk for SARS-CoV-2 infection related to occupational risk.
Almost one-third of the patients had a job position considered as posing a high risk of infection, which was the main cause for not having proper adherence to lockdown measures. Health care professions were the most frequent occupational hazard (18% (85/482). Table 4 shows the relationship between infection and occupational risk in patients with and without good adherence to a total lockdown.
Despite declaring good adherence, patients became infected mainly by intrafamilial transmission, particularly during the first 2 weeks (March 2020) after the Spanish government established lockdown.

3.3. COVID-19 Diagnosis and Treatment

Symptoms of COVID-19 diagnosis can be found in Supplementary Table S2, with fever (69% (336/482)) and cough (63% (305/482)) as the most frequently observed symptoms. Diarrhoea was reported in 26% (126/482) of patients, with no significant differences between patients with active or inactive IBD.
Supplementary Table S3 includes the tests performed for COVID-19 diagnosis. Notably, 90% of IBD patients had a diagnostic test performed, whether PCR (80% (388/482)) or SARS-CoV-2 serology (35% (167/482)). Only 49 patients did not have any diagnostic test performed. It is also important to emphasize that 28% of patients (85/301) had a positive PCR after one or more negative PCRs, with no difference between immunosuppressed and non-immunosuppressed patients (19% vs. 21%, p = 0.67).
COVID-19 treatment followed the trend used and recommended by health authorities at that time. It included chloroquine/hydroxychloroquine in 41% (198/482), antibiotics in 38% (182/482), antivirals in 18% (88/482), systemic corticosteroids in 12% (58/482) and biologic therapy in 2.9% (14/482) (tocilizumab in 1.7% (8/482), interferon in 1.2% (6/482) and anakinra in 0.2% (1/482)). Antifungal therapy was used in 1.9% (9/482), vasoactive amines in 1.2% (6/482) and invasive oxygen therapy in 2.9% (14/482) of the patients (one patient received invasive oxygen therapy in a conventional floor due to occupation limitations of the ICU).

3.4. Outcomes

Patients attended their primary care facility in 55% of the cases (266/482), received emergency room assistance in 52% (251/482) and required hospitalisation due to COVID in 35% (167/482). Eleven patients (0.2%) required IBD-related hospitalisation during the study period. Twenty-four patients had respiratory distress (4.9%), 56 (12%) presented with systemic inflammatory response syndrome upon admission and 6.4% (31/482) presented with systemic inflammatory response syndrome during hospitalisation. Thirteen (2.5%) required ICU admission, 38 (7.9%) fulfilled the criteria of severe COVID-19 and 21 patients died during the study. Of those who died, 18 (3.7%) died due to COVID-19 and 3 due to causes other than COVID-19, 2 of them during the first wave of the pandemic (one case with signet-ring cell adenocarcinoma of digestive origin and one case of pulmonary neoplasia) and the third 9 months after COVID-19 infection due to urinary sepsis. Only one death occurred outside the hospital (80-year-old female with inactive ileal CD treated with oral aminosalicylates). Compared to the general population in Spain, the severe outcomes were similar, with a mortality proportion of 4.6% and ICU requirement proportion of 2.1% with a slightly lower proportion of patients requiring hospitalisation (19.5%) [23].
Univariate and multivariate analyses of factors associated with hospitalisation, ICU admission, severe COVID-19 and death are detailed in Supplementary Tables S4–S7. In that case, predictive factors for hospitalisation due to COVID-19 were being 50 years of age or more (OR 2.09, 95% CI 1.3–3.4), having at least one comorbidity (OR 2.28, 95% CI 1.4–3.6) and being treated with steroids for IBD within the 3 months before COVID-19 diagnosis (OR 1.3, 95% CI 1.1–1.6). Predictors for ICU admission were having a Charlson score of at least 2 (OR 5.4, 95% CI 1.5–20.1) and the use of aminosalicylates (OR 4.6, 95% CI 1.2–17). However, when adjusted for diagnosis, the effect of aminosalicylates disappeared (OR 3.6, 95% CI 0.85–15.2, p = 0.08). Independent risk factors related to death due to COVID-19 were being 60 years of age or more (OR 7.1, 95% CI 1.8–27.4) and having at least 2 comorbidities (OR 3.9, 95% CI 1.3–11.6). The only predictor for severe COVID-19 was being 60 years of age or more (OR 4.59, 95% CI 1.3–15.9), while having CD with an inflammatory behaviour was protective for this outcome (OR 0.29, 95% CI 0.09–0.89). There were no differences in the proportion of hospitalisation, ICU admission, severe COVID-19 or death between patients found under active search (18% of the centres) vs. those that were found by direct notification from the patient itself, the family physician, the emergency department or the hospitalisation unit (data not shown). The proportion of patients under specific IBD treatment, taking into account each predefined outcome, is shown in Table 5.
There were no differences in any outcomes between patients with probable or confirmed COVID-19 (data not shown). To compare the results of the present study with those of the SECURE-IBD (accessed on 24 August 2021) [9], the results of the two cohorts are provided in the same table. In Supplementary Table S8, we show the incidence of adverse outcomes of COVID-19 in patients with IBD, taking into account sex and age. Of note, being ≥50 years old increases the incidence of the reported adverse outcomes from 3 to 7, with a greater frequency in males than in females.

3.5. Follow-Up

IBD treatment and the presence of sequelae related to COVID-19 at the 3- and 12-month follow-up are described in Table 6.
Only 13% of patients had short-term immunosuppression withdrawal due to COVID-19 (12% partial and 1.4% definitive), and only 1% kept their withdrawal of immunosuppression in the long term.
At the 3-month follow-up, 65 patients (13%) presented COVID-19 sequelae, of which 4% (20/482) were psychological and 11% (55/482) were physical (Table 6). At the 12-month follow-up, 72 patients (15%) were considered to have sequelae, of which 3.1% (15/482) were psychological and 14% (67/482) were physical. The most frequent physical sequelae were asthenia, myalgia/arthralgia and anosmia. The only predictive factor for having physical sequelae at the 3-month follow-up was the use of steroid treatment for IBD within the 3 months before COVID-19 (OR 1.4, 95% CI 1.07–1.7) (Supplementary Table S9). No predictive factor for physical sequelae was found at the long-term follow-up (12 months) (Supplementary Table S10). Only one patient reported SARS-CoV-2 reinfection 6 months after the index diagnosis.
IBD activity both at COVID-19 diagnosis and at 3- and 12-month follow-ups is shown in Figure 2. Two-thirds of patients (68%, 330/482) remained in remission throughout the study period.

4. Discussion

We report the largest cohort of patients with IBD and symptomatic COVID-19 prospectively recruited with a one-year follow-up after infection. This is a national, multicentre study that was conducted within the ENEIDA project and included 482 patients with COVID-19 among 53,682 patients with IBD, giving a cumulative incidence of 8.97 per 1000. These data are in the upper limit of the wide range of incidence previously described [10,14,28], ranging from 0.95 [29] to 100 per 1000 [15]. The universal access to health care within the National Health System in Spain (less than 3% of IBD patients with private insurance never use public services [30]) and the adherence of most centres to the nationwide certification programme in IBD [26] provide homogeneity to the cohort, minimizing potential bias in the clinical characteristics. We did not calculate the comparative incidence of COVID-19 between the IBD cohort and data obtained from the general population because the identification of cases did not use the same methodology. However, it is clearly observed in Figure 1 that the number of cases with COVID-19 and IBD is the highest in areas with the highest incidence of infection. The high variability in incidence between relatively close geographical areas was also observed, with a maximum in those areas with the highest population density. We decided to exclude asymptomatic patients, as there was no policy for a universal testing in our patients nor in the general population during the first wave. Therefore data on asymptomatic patients with IBD and positive SARS-CoV-2 test are scarce and can imply a selection bias.
It has been suggested that patients with IBD are at lower risk of having COVID-19, and most studies support this assertion [10,31,32,33,34,35]. However, only a few of these studies performed in Denmark and Sweden were population based [36,37], which is how differences in the incidence of infection between population groups should be addressed. In our cohort, we recorded the influence of occupational risk, lockdown strategies and other risk factors, in addition to IBD treatment, that may have influenced, either increasing or decreasing, the risk of contagion. We observed, for example, that one-third of infected patients received special protection measures such as sick leave simply because they were considered a risk group. However, many patients became infected, perhaps because they were infected early before mandatory lockdown or through close family-infected contact. Lockdown has been demonstrated to be the most effective strategy precluding SARS-CoV-2 expansion [38,39,40], also in patients with IBD [41]. Thus, the low incidence of COVID-19 in IBD cohorts does not necessarily reflect a lower susceptibility to infection but a higher protection attitude towards IBD patients based on recommendations [6,42] or because they spontaneously adopt more rigorous self-protecting measures [43]. It has been shown that patients under biologic drugs perceive themselves to be at greater risk of SARS-CoV-2 infection, but, despite that, they do not perform more strict social distancing practices than patients with IBD without biologics or in remission [43]. However, in our cohort, only 49% exhibited good adherence to lockdown measures. Currently, data on the exposure environment of noninfected patients are very limited [44]. The study also showed that one-third of IBD patients with COVID-19 had occupational risks, mainly working in health care facilities. It has been reported that health care providers bore a great burden during the pandemic, as shown in data coming from Italy and China [22,45]. Nonetheless, we did not find that occupational risk or failure to meet lockdown measures were predictors for a worse evolution of COVID-19 in patients with IBD.
The reported outcomes of COVID-19 are highly variable [10,28,33,36,46] and may depend on many factors, such as age, comorbidity, ethnicity, socioeconomic status and the quality of health care. In our cohort, as in others, the most important factors influencing outcomes were age and comorbidity. Thirty-five percent of the patients required hospitalisation, 7.9% had severe COVID-19, 2.5% required ICU admission and 3.7% died. These figures are similar to those extracted from the general population in Spain [23] (hospitalisation 19.5%, ICU 2.1% and mortality 4.6%) and are also identical to those reported in the first publication of SECURE-IBD, including the first 525 registered cases [8].
Age is known to be one of the most consistent risk factors for severe COVID-19 and death worldwide [47,48], both in the general population and in patients with IBD [10,49]. We found that patients 50 years old or older were at greater risk for hospitalization, while those 60 years old or older were more prone to severe COVID-19 or death due to COVID-19. Comorbidities have also been considered the other important risk factor for deleterious COVID-19 evolution, both in patients with IBD [16,29,33,36,46] and in the general population [22,50], and we also found consistent results in our cohort. Thus, differences in the percentages of parameters of the severity of COVID-19 between cohorts can be largely explained by these two factors, not only by themselves. The median age of patients in the initial SECURE-IBD cohort was 41 years versus 52 years in our cohort, and the percentage of comorbidities was 36% versus 44%, respectively. Data from SECURE-IBD accessed on 24 August 2021 [9], showed significantly improved outcomes compared to previous outcomes, with a fifty percent reduction in indicators of severity, including hospitalisation, severe COVID-19 and death (more than 70% of the patients were younger than 49 years). Although it can be speculated that improved knowledge of COVID-19 management may account for a better outcome over time, important selection bias accounting for differences in this type of registry cannot be ruled out.
The only protective factor for severe COVID-19 was CD with inflammatory behaviour. As this was found to be independent of IBD activity and treatment, we speculate that this is because this pattern has less disease burden, as it was also related to a shorter duration of the disease (11 years of disease in inflammatory IBD vs. 18 years in stricturing or penetrating disease, p < 0.0001).
As stated previously, IBD-related immunosuppression has not been found to be a risk factor for severe COVID-19 or death [28,29,51]. Our cohort confirms that there is no relationship between anti-TNF or any other form of therapeutic immunosuppression and COVID-19 severity. Some authors suggested that the risk of hospitalisation is higher in patients under biologics, but this may reflect a precaution more than COVID-19 severity itself, as overall mortality related to COVID has not been demonstrated to be increased in ours and other previous studies [28,36,46,51,52]. This consistent evidence reinforces the message that biologics can be safely continued in most cases. The use of steroids in this pandemic has been controversial [53,54]. In contrast to SECURE-IBD [8], we did not find that current treatment with systemic steroids was related to a worse COVID-19 evolution, an outcome that we have previously found related to other relevant infections in patients with IBD [3]. This might be due to the small proportion of patients under this treatment (5.4%) or the type of schedule administered. However, the use of steroids three months before COVID-19 diagnosis was an independent risk factor for hospitalisation and physical sequelae (at the 3-month follow-up). This could be indirectly related to a probable active IBD that was challenging to treat and required the use of systemic steroids.
Finally, we describe the evolution of patients at 3 and 12 months after COVID-19. Thirteen percent withdrew IBD medication during COVID-19. This is less than previously reported, ranging between 27% [15] and 34% [55]. This is certainly encouraging, as the first guidelines on the treatment of patients with IBD during the pandemic were very clear in recommending the withdrawal of immunosuppressants and biologics in infected individuals [6]. However, as our experience increased, it was assured that the inappropriate cessation of effective agents for IBD treatment due to unjustified fear of adverse events could lead to IBD relapse and then to the use of steroids or hospitalisation, thus increasing the risk of COVID-19 exposure and infection. The last outcome that we explored was COVID-19 sequelae, present in 13% at 3 months and 15% at 12 months in our cohort. The only predictive factor for having physical sequelae at the 3-month follow-up was the use of steroid treatment for IBD 3 months before COVID-19 (OR 1.4, 95% CI 1.07–1.7). It has been shown that severe sequelae were lower in patients with IBD when matched to non-IBD controls [49], so IBD does not seem to be a disease linked to more sequelae due to COVID-19. In contrast, a recent population-based Danish study has suggested that sequelae are a common phenomenon, affecting almost 44% of patients [56]. It has to be noted that this study accessed sequelae only in 222 of the 516 patients included and that they were self-reported sequalae, limiting the confirmation of a real effect that might be caused by COVID-19. In addition, another study found that patients with IBD, hospitalized due to COVID-19, have a greater risk of severe infections requiring further hospitalisation [57], a situation that has not been found in our cohort.
Our study has several limitations to be aware of. First, only 18% of centres performed an active search of cases (82% of cases were collected by direct notification from the patient itself, the family physician, the emergency department or the hospitalisation unit, as stated in the Methods section). Thus, it is possible that very mild cases did not consult their IBD unit; thus, mild COVID-19 cases might be underrepresented. However, data on hospitalisation, severe COVID-19, ICU admission and death from the general population are similar to our cohort and we did not find differences in specific outcomes between these two searching strategies; therefore, this bias is probably small. On the other hand, the majority of participant centres were certified IBD units that had open access to their outpatient clinics, nurse-led advice lines and/or emails for emergencies as mandatory quality criteria [26]. Second, this study, including only the first wave of an ongoing pandemic, is observational and cannot establish causation or account for unmeasured confounders. Finally, discovering the real number of infected patients remains a global challenge because PCR or serology were not performed universally at the beginning and there is evidence of false negative results. This occurred in 10% of this cohort. Notwithstanding these issues, there are some important strengths. First, it has national coverage with active participation of almost 90% of the IBD units from the ENEIDA registry. However, the main strength relies on prospective data, collecting IBD activity, and other important variables. In addition, although nationwide series on COVID-19 and IBD have been published before [29,33], our study is the largest cohort of patients with IBD and COVID-19 with the longest follow-up after infection.

5. Conclusions

In conclusion, we have shown that occupational risk and intrafamilial transmission are relevant epidemiological risk factors and that a high proportion of patients receive preventive sick leave. We have also demonstrated that IBD does not worsen COVID-19 prognosis, even when immunosuppressants and biological drugs are used. Age and comorbidity are the most important prognostic factors for more severe COVID-19 in patients with IBD and are even more relevant than epidemiological risk factors such as occupational risk. Finally, COVID-19 is not a condition that affects the prognosis of IBD or its treatment, either in the short or the long term and is not a cause of significant sequelae in patients already suffering from IBD. Therefore, there is no need for more strict protection measures than those adopted for the general population.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/jcm11020421/s1, Table S1. Comorbidities; Table S2. Symptoms at COVID-19 diagnosis; Table S3. Tests performed for COVID-19 diagnosis; Table S4. Clinical characteristics and treatment for IBD in patients who required hospitalization; Table S5. Clinical characteristics and treatment for IBD in patients who required ICU admission; Table S6. Clinical characteristics and treatment for IBD in patients who died due to COVID-19; Table S7. Clinical characteristics and treatment for IBD in patients with severe COVID-19; Table S8. Incidence of adverse outcomes of COVID-19 patients in the ENEIDA registry; Table S9. Clinical characteristics and treatment for IBD in patients with 3-month physical sequelae; Table S10. Clinical characteristics and treatment for IBD in patients with 12-month physical sequelae.

Author Contributions

Study design: Y.Z., I.M.-J., I.R.-L., M.E.; study conduction: Y.Z., P.M.; data collection: Y.Z., I.M.-J., I.R.-L., I.V., M.D.M.-A., I.G., J.P.G., F.M. (Francisco Mesonero)., O.B., C.T., Á.P.-D., M.P., A.J.L., B.C., M.M., P.M.-M., M.B.-W., J.G., L.B., N.M., T.M.-P., A.L., C.R.-G., S.G.-L., P.V. (Pablo Vega), M.R., L.M., M.C. (Maria Calvo), M.I., M.B.d.-A., B.S., J.B., J.L.P., D.B., I.P.-M., M.N.-L., V.H., F.A.-A., F.R.E., S.M., L.R., F.G., F.M. (Fernando Muñoz), G.S., J.O.d.Z., J.M.H., J.L., M.F.G.-S., M.S., M.D., S.E., A.F.C., E.H., L.O., E.I., A.G., P.V. (Pilar Varela), N.R., P.G., A.H.-C., A.B., D.G., E.S., D.C., M.A., J.L.C., Y.G.-L., L.J., M.C. (María Chaparro), A.L.-S.R., C.A., R.P.-S., R.M., S.T.-S., E.R., M.C. (Margalida Calafat), S.O., P.N., F.B., H.A.-G., R.P., P.O., P.M., E.D., M.E.; data analysis and interpretation: Y.Z., I.M.-J., I.R.-L., M.E.; manuscript drafting: Y.Z., I.M.-J., I.R.-L., M.E. All authors have read and agreed to the published version of the manuscript, including the authorship list.

Funding

This study is funded by the Carlos III Health Institute (COV20/00227: Co-IP Dra. Maria Esteve and Dra. Yamile Zabana), FEDER (Fondo Europeo de Desarrollo Regional) and supported by GETECCU. The ENEIDA Registry of GETECCU is supported by Takeda, Pfizer, Galápagos, AbbVie and Biogen.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Scientific Committee of ENEIDA on 16 March 2020. It was also approved by the Ethics Committee of Hospital Universitari Mútua Terrassa (coordinating centre).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data underlying this article are available in the article and Supplementary Materials.

Acknowledgments

The authors would like to thank the centres that registered no cases during the study period but performed an active search of IBD patients with COVID-19: Hospital Sant Joan de Deu, Hospital Sant Jaume de Calella, Hospital de Castellón, Hospital Materno-Infantil de Málaga, Hospital de Granollers, Hospital Joan XXIII, Hospital Niño Jesús, Hospital de Cáceres, Hospital de Vic, and Complejo Hospitalario La Mancha Centro.

Conflicts of Interest

Y. Zabana has received support for conference attendance, speaker fees, research support and consulting fees from Abbvie, Adacyte, Almirall, Amgen, Dr Falk, FAES Pharma, Ferring, Jannsen, MSD, Otsuka, Pfizer, Shire, Takeda and Tillotts. I. Marín-Jiménez has received financial support for travelling and educational activities from or has served as an advisory board member to MSD, Pfizer, Abbvie, Amgen, Takeda, Janssen, Sandoz, Shire Pharmaceuticals, Ferring, Dr. Falk Pharma, and Otsuka Pharmaceutical. I. Rodríguez-Lago has received financial support for travelling and educational activities from or has served as an advisory board member to MSD, Pfizer, Abbvie, Takeda, Janssen, Tillotts Pharma, Roche, Shire Pharmaceuticals, Ferring, Dr. Falk Pharma, Adacyte and Otsuka Pharmaceutical. MD Martín-Arranz has received fees as a speaker, consultant and advisory member for or has received funding from MSD, AbbVie, Hospira, Pfizer, Takeda, Janssen, Shire Pharmaceuticals, Tillotts Pharma, Faes farma. J. P. Gisbert has served as speaker, consultant, and advisory member for or has received research funding from MSD, Abbvie, Pfizer, Kern Pharma, Biogen, Mylan, Takeda, Janssen, Roche, Sandoz, Celgene, Gilead/Galapagos, Ferring, Faes Farma, Shire Pharmaceuticals, Dr. Falk Pharma, Tillotts Pharma, Chiesi, Casen Fleet, Gebro Pharma, Otsuka Pharmaceutical and Vifor Pharma. F. Mesonero has served as a speaker or has received education funding or advisory fees from MSD, Abbvie, Takeda, Janssen, Pfizer, Kern Pharma, Ferring and Dr. Falk Pharma. O. Benítez has received financial support for travelling and educational activities from Janssen, Biogen and MSD. C. Taxonera has served as a speaker, consultant and advisory board member for or has received research funding from MSD, AbbVie, Pfizer, Taked, Janssen, Ferring, Faes Pharma, Shire Pharmaceuticals, Dr Falk Pharma, Galapagos and Tillotts. M. Piqueras has received financial support or has served as speaker for Takeda, Abbvie and Janssen. N. Manceñido has been a consultant or speaker for Abbvie, Janssen, Takeda, Ferring, Dr. Falk Pharma and Tillotts Pharma. M. Rivero has served as a speaker, consultant and advisory member for Merck Sharp and Dohme, Abbvie, Pfizer, Takeda and Janssen. M. Barreiro has served as a speaker, consultant and advisory member for or has received research funding from MSD, AbbVie, Janssen, Kern Pharma, Celltrion, Takeda, Gillead, Celgene, Pfizer, Sandoz, Biogen, Fresenius, Ferring, Faes Farma, Dr. Falk Pharma, Chiesi, Gebro Pharma, Adacyte and Vifor Pharma. F. Argüelles has received financial support for travelling and educational activities from or has served as an advisory board member to MSD, Pfizer, Abbvie, Amgen, Takeda, Janssen, Sandoz, Shire Pharmaceuticals, Ferring and Dr Falk Pharma. JM. Huguet has participated in educational activities, research projects, scientific meetings or advisory boards sponsored by Merck Sharp Dohme (MSD), Ferring, Abbvie, Janssen, Biogen, Sandoz, Kern Pharma, Faes Farma, Vifor Pharma and Takeda. A. Hernández-Camba has served as speaker, or has received education funding from AbbVie, Takeda, Kern Pharma, Pfizer, Janssen, Adacyte Therapeutics and Ferring. D. Ginard has received support for conference attendance, speaker fees, research support and consulting fees from Abbvie, Adacyte, Janssen, Takeda, Sandoz, Ferring, Pfizer and Biogen. M. Aceituno has received educational grants from Ferring, Pfizer, Janssen and AbbVie. M. Chaparro has served as a speaker and consultant and has received research or education funding from MSD, Abbvie, Hospira, Pfizer, Takeda, Janssen, Ferring, Shire Pharmaceuticals, Dr. Falk Pharma, Tillotts Pharma, Faes Pharma, Biogen and Gilead. H. Alonso-Galán has received financial support for travelling and educational activities from or has served as an advisory board member to Janssen, MSD, Abbvie, Takeda, Ferring, Pfizer and Dr. Falk Pharma. E. Domènech has served as a speaker, or has received research or education funding or advisory fees from AbbVie, Adacyte Therapeutics, Biogen, Celltrion, Gilead, Janssen, Kern Pharma, MSD, Pfizer, Roche, Samsung, Takeda, and Tillotts. M. Esteve: has received support for conference attendance, speaker fees, research support and consulting fees from Abbvie, Ferring, Janssen, MSD, Otsuka, Pfizer, Takeda and Tillotts. I.V. era, I. Guerra, A. Ponferrada-Díaz, A. Lucendo, B. Caballol, M.Mañosa, P. Martínez-Montiel, M. Bosca-Watts, J. Gordillo, L. Bujanda, T. Martínez-Pérez, A. López, C. Rodríguez, S. García-López, P. Vega, L. Melcarne, M. Calvo, M. Iborra, B. Sicilia, J. Barrio, JL. Pérez, D. Busquets, I. Martínez-Pérez, M. Navarro-Llavat, V. Hernández, F. Ramírez Esteso, S. Mejilde, L. Ramos, F.Gomollón, F. Muñoz, G. Suris, J. Ortiz de Zarate, J. Llaó, M. García-Sepulcre, M. Sierra, M. Durà. S. Estrecha, A. Fuentes Coronel, E. Hinojosa, L. Olivan, E. Iglesias, A. Gutiérrez, P. Varela, N. Rull, P. Gilabert, A. Brotons, E. Sesé, D. Carpio, JL Cabriada, Y. González-Lama, L. Giménez, A. López-San Román, C. Alba, R. Plaza-Santos, R. Mena, S. Tamarit-Sebastián, E. Ricart, M. Calafat, S. Olivares, P. Navarro, F. Bertoletti, R. Pajares, P. Olcina and P. Manzano have no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. John Hopkins University Coronavirus Resource Center. Coronavirus COVID-19 Global Cases. Available online: https://coronavirus.juy.edu/map.html (accessed on 21 August 2021).
  2. Kirchgesner, J.; Lemaitre, M.; Carrat, F.; Zureik, M.; Carbonnel, M.F.; Dray-Spira, R.; Carbonnel, F.; Dray-Spira, R. Risk of Serious and Opportunistic Infections Associated with Treatment of Inflammatory Bowel Diseases. Gastroenterology 2018, 155, 337–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zabana, Y.; Rodríguez, L.; Lobatón, T.; Gordillo, J.; Montserrat, A.; Mena, R.; Beltrán, B.; Dotti, M.; Benitez, O.; Guardiola, J.; et al. Relevant Infections in Inflammatory Bowel Disease, and Their Relationship with Immunosuppressive Therapy and Their Effects on Disease Mortality. J. Crohn’s Colitis 2019, 13, 828–837. [Google Scholar] [CrossRef] [PubMed]
  4. Tinsley, A.; Navabi, S.; Williams, E.D.; Liu, G.; Kong, L.; Coates, M.D.; Clarke, K. Increased Risk of Influenza and Influenza-Related Complications among 140,480 Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2019, 25, 369–376. [Google Scholar] [CrossRef] [PubMed]
  5. Rubin, D.T.; Feuerstein, J.D.; Wang, A.Y.; Cohen, R.D. AGA Clinical Practice Update on Management of Inflammatory Bowel Disease during the COVID-19 Pandemic: Expert Commentary. Gastroenterology 2020, 159, 350–357. [Google Scholar] [CrossRef]
  6. Kennedy, N.A.; Jones, G.-R.; Lamb, C.A.; Appleby, R.; Arnott, I.; Beattie, R.M.; Bloom, S.; Brooks, A.J.; Cooney, R.; Dart, R.J.; et al. British Society of Gastroenterology guidance for management of inflammatory bowel disease during the COVID-19 pandemic. Gut 2020, 69, 984–990. [Google Scholar] [CrossRef] [PubMed]
  7. Abreu, C. 1st Interview COVID-19 ECCO Taskforce. 2020. Available online: https://ecco-ibd.eu/images/6_Publication/6_8_Surveys/1st_interview_COVID-19%20ECCOTaskforce_published.pdf (accessed on 2 August 2021).
  8. Brenner, E.J.; Ungaro, R.C.; Gearry, R.B.; Kaplan, G.G.; Kissous-hunt, M.; Lewis, J.D.; Ng, S.C.; Rahier, J.; Reinisch, W.; Ruemmele, F.M.; et al. Corticosteroids, but Not TNF Antagonists, Are Associated with Adverse COVID-19 Outcomes in Patients with Inflammatory Bowel Diseases: Results from an International Registry. Gastroenterology 2020, 159, 481–491. [Google Scholar] [CrossRef] [PubMed]
  9. Brenner, E.; Ungaro, R.; Colombel, J.; Kappelman, M. SECURE-IBD Database Public Data. Available online: https://covidibd.org/current-data/ (accessed on 24 August 2021).
  10. Aziz, M.; Fatima, R.; Haghbin, H.; Smith, W.L.; Nawras, A. The incidence and outcomes of COVID-19 in ibd patients: A rapid review and meta-Analysis. Inflamm. Bowel Dis. 2020, 26, E132–E133. [Google Scholar] [CrossRef]
  11. Mao, R.; Liang, J.; Shen, J.; Ghosh, S.; Zhu, L.-R.; Yang, H.; Wu, K.-C.; Chen, M.-H. Implications of COVID-19 for patients with pre-existing digestive diseases. Lancet Gastroenterol. Hepatol. 2020, 5, 425–427. [Google Scholar] [CrossRef]
  12. Norsa, L.; Indriolo, A.; Sansotta, N.; Cosimo, P.; Greco, S.; D’Antiga, L. Uneventful course in IBD patients during SARS-CoV-2 outbreak in northern Italy. Gastroenterology 2020, 159, 371–372. [Google Scholar] [CrossRef]
  13. Allocca, M.; Fiorino, G.; Zallot, C.; Furfaro, F.; Radice, S.; Danese, S.; Peyrin-biroulet, L. Incidence and patterns of COVID-19 among inflammatory bowel disease patients from the Nancy and Milan cohorts. Clin. Gastroenterol. Hepatol. 2020, 18, 2134–2135. [Google Scholar] [CrossRef]
  14. Taxonera, C.; Sagastagoitia, I.; Alba, C.; Mañas, N.; Olivares, D.; Rey, E. 2019 novel coronavirus disease (COVID-19) in patients with inflammatory bowel diseases. Aliment. Pharmacol. Ther. 2020, 52, 276–283. [Google Scholar] [CrossRef]
  15. Guerra, I.; Algaba, A.; Jiménez, L.; Aller, M.M.; Garza, D.; Bonillo, D.; Esteban, L.M.M.; Bermejo, F. Incidence, Clinical Characteristics, and Evolution of SARS-CoV-2 Infection in Patients with Inflammatory Bowel Disease: A Single-Center Study in Madrid, Spain. Inflamm. Bowel Dis. 2021, 27, 25–33. [Google Scholar] [CrossRef]
  16. Rodríguez-Lago, I.; Ramírez de la Piscina, P.; Elorza, A.; Merino, O.; Ortiz de Zárate, J.; Cabriada, J.L. Characteristics and Prognosis of Patients with Inflammatory Bowel Disease during the SARS-CoV-2 Pandemic in the Basque Country (Spain). Gastroenterology 2020, 159, 781–783. [Google Scholar] [CrossRef]
  17. Zabana, Y.; Panés, J.; Nos, P.; Gomollón, F.; Esteve, M.; García-Sánchez, V.; Gisbert, J.P.; Barreiro-de-Acosta, M.; Domènech, E. The ENEIDA registry (Nationwide study on genetic and environmental determinants of inflammatory bowel disease) by GETECCU: Design, monitoring and functions. Gastroenterol. Hepatol. 2020, 43, 551–558. [Google Scholar] [CrossRef]
  18. Silverberg, M.S.; Satsangi, J.; Ahmad, T.; Arnott, I.D.; Bernstein, C.N.; Brant, S.R.; Caprilli, R.; Colombel, J.-F.; Gasche, C.; Geboes, K.; et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: Report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Can. J. Gastroenterol. 2005, 19 (Suppl. A), 5–36. [Google Scholar] [CrossRef]
  19. Huang, Y.Q.; Gou, R.; Diao, Y.S.; Yin, Q.H.; Fan, W.X.; Liang, Y.P.; Chen, Y.; Wu, M.; Zang, L.; Li, L.; et al. Charlson comorbidity index helps predict the risk of mortality for patients with type 2 diabetic nephropathy. J. Zhejiang Univ. Sci. B 2014, 15, 58–66. [Google Scholar] [CrossRef] [Green Version]
  20. Harvey, R.F.; Bradshaw, J.M. A simple index of Crohn’s-disease activity. Lancet 1980, 1, 514. [Google Scholar] [CrossRef]
  21. Lewis, J.D.; Chuai, S.; Nessel, L.; Lichtenstein, G.R.; Aberra, F.N.; Ellenberg, J.H. Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis. Inflamm. Bowel Dis. 2008, 14, 1660–1666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Guan, W.-J.; Ni, Z.-Y.; Hu, Y.; Liang, W.-H.; Ou, C.-Q.; He, J.-X.; Liu, L.; Shan, H.; Lei, C.-L.; Hui, D.S.C.; et al. Clinical Characteristics of Coronavirus Disease 2019 in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
  23. Alves-Cabratosa, L.; Comas-Cufí, M.; Blanch, J.; Martí-Lluch, R.; Ponjoan, A.; Castro-Guardiola, A.; Hurtado-Ganoza, A.; Pérez-Jaén, A.; Rexach-Fumaña, M.; Faixedas-Brunsoms, D.; et al. Persons with SARS-CoV-2 during the First and Second Waves in Catalonia (Spain): A Retrospective Observational Study Using Daily Updated Data. JMIR Public Heal. Surveill. 2021, 8, e30006. [Google Scholar] [CrossRef] [PubMed]
  24. ISC; CNE. Informe Sobre la Situación de COVID-19 en España. Informe COVID-19 no12, 20 Marzo, 2020. Available online: https://www.isciii.es/QueHacemos/Servicios/VigilanciaSaludPublicaRENAVE/EnfermedadesTransmisibles/Paginas/-COVID-19.-Informes-previos.aspx (accessed on 2 August 2021).
  25. CNE; ISC; ICI. COVID-19 en España. Available online: https://cnecovid.isciii.es/covid19/#provincias (accessed on 2 August 2021).
  26. Barreiro-de Acosta, M.; Gutiérrez, A.; Zabana, Y.; Beltrán, B.; Calvet, X.; Chaparro, M.; Domènech, E.; Esteve, M.; Panés, J.; Gisbert, J.P.; et al. Inflammatory bowel disease integral care units: Evaluation of a nationwide quality certification programme. The GETECCU experience. United Eur. Gastroenterol. J. 2021, 9, 766–772. [Google Scholar] [CrossRef]
  27. Instituto Nacional de Estadística (INE). Población por Provincias y Sexo. 2021. Available online: https://www.ine.es/jaxiT3/Datos.htm?t=2852#!tabs-tabla (accessed on 2 September 2021).
  28. Burke, K.E.; Kochar, B.; Allegretti, J.R.; Winter, R.W.; Lochhead, P.; Khalili, H.; Colizzo, F.P.; Hamilton, M.J.; Chan, W.W.; Ananthakrishnan, A.N. Immunosuppressive Therapy and Risk of COVID-19 Infection in Patients with Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2021, 27, 155–161. [Google Scholar] [CrossRef]
  29. Khan, N.; Patel, D.; Xie, D.; Lewis, J.; Trivedi, C.; Yang, Y.-X. Impact of Anti-Tumor Necrosis Factor and Thiopurine Medications on the Development of COVID-19 in Patients with Inflammatory Bowel Disease: A Nationwide Veterans Administration Cohort Study. Gastroenterology 2020, 159, 1545–1546.e1. [Google Scholar] [CrossRef]
  30. Chaparro, M.; Garre, A.; Núñez Ortiz, A.; Diz-Lois Palomares, M.T.; Rodríguez, C.; Riestra, S.; Vela, M.; Benítez, J.M.; Fernández Salgado, E.; Sánchez Rodríguez, E.; et al. Incidence, Clinical Characteristics and Management of Inflammatory Bowel Disease in Spain: Large-Scale Epidemiological Study. J. Clin. Med. 2021, 10, 2885. [Google Scholar] [CrossRef] [PubMed]
  31. Khan, N.; Mahmud, N.; Trivedi, C.; Reinisch, W.; Lewis, J.D. Risk factors for SARS-CoV-2 infection and course of COVID-19 disease in patients with IBD in the Veterans Affair Healthcare System. Gut 2021, 70, 1657–1664. [Google Scholar] [CrossRef]
  32. Ardizzone, S.; Ferretti, F.; Monico, M.C.; Carvalhas Gabrielli, A.M.; Carmagnola, S.; Bezzio, C.; Saibeni, S.; Bosani, M.; Caprioli, F.; Mazza, S.; et al. Lower incidence of COVID-19 in patients with inflammatory bowel disease treated with non-gut selective biologic therapy. J. Gastroenterol. Hepatol. 2021, 36, 3050–3055. [Google Scholar] [CrossRef]
  33. Derikx, L.A.A.P.; Lantinga, M.A.; de Jong, D.J.; van Dop, W.A.; Creemers, R.H.; Römkens, T.E.H.; Jansen, J.M.; Mahmmod, N.; West, R.L.; Tan, A.C.I.T.L.; et al. Clinical Outcomes of Covid-19 in Patients with Inflammatory Bowel Disease: A Nationwide Cohort Study. J. Crohn’s Colitis 2020, 15, 529–539. [Google Scholar] [CrossRef] [PubMed]
  34. Singh, S.; Khan, A.; Chowdhry, M.; Bilal, M.; Kochhar, G.S.; Clarke, K. Risk of Severe Coronavirus Disease 2019 in Patients with Inflammatory Bowel Disease in the United States: A Multicenter Research Network Study. Gastroenterology 2020, 159, 1575–1578.e4. [Google Scholar] [CrossRef] [PubMed]
  35. Wetwittayakhlang, P.; Albader, F.; Golovics, P.A.; Bessissow, T.; Bitton, A.; Afif, W.; Wild, G.; Lakatos, P.L. Clinical Outcomes of COVID-19 and Impact on Disease Course in Patients with Inflammatory Bowel Disease. Can. J. Gastroenterol. Hepatol. 2021, 2021, 7591141. [Google Scholar] [CrossRef]
  36. Attauabi, M.; Poulsen, A.; Theede, K.; Pedersen, N.; Larsen, L.; Jess, T.; Rosager Hansen, M.; Verner-Andersen, M.K.; Haderslev, K.V.; Berg Lødrup, A.; et al. Prevalence and Outcomes of COVID-19 Among Patients with Inflammatory Bowel Disease—A Danish Prospective Population-Based Cohort Study. J. Crohn’s Colitis 2020, 15, 540–550. [Google Scholar] [CrossRef]
  37. Ludvigsson, J.F.; Axelrad, J.; Halfvarson, J.; Khalili, H.; Larsson, E.; Lochhead, P.; Roelstraete, B.; Simon, T.G.; Söderling, J.; Olén, O. Inflammatory bowel disease and risk of severe COVID-19: A nationwide population-based cohort study in Sweden. United Eur. Gastroenterol. J. 2021, 9, 177–192. [Google Scholar] [CrossRef]
  38. Guzzetta, G.; Riccardo, F.; Marziano, V.; Poletti, P.; Trentini, F.; Bella, A.; Andrianou, X.; Del Manso, M.; Fabiani, M.; Bellino, S.; et al. Impact of a Nationwide Lockdown on SARS-CoV-2 Transmissibility, Italy. Emerg. Infect. Dis. 2021, 27, 267–270. [Google Scholar] [CrossRef] [PubMed]
  39. Pan, A.; Liu, L.; Wang, C.; Guo, H.; Hao, X.; Wang, Q.; Huang, J.; He, N.; Yu, H.; Lin, X.; et al. Association of Public Health Interventions with the Epidemiology of the COVID-19 Outbreak in Wuhan, China. JAMA 2020, 323, 1915–1923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Lodyga, M.; Maciejewska, K.; Eder, P.; Waszak, K.; Stawczyk-Eder, K.; Dobrowolska, A.; Kaczka, A.; Gąsiorowska, A.; Stępień-Wrochna, B.; Cicha, M.; et al. Social distancing during COVID-19 pandemic among inflammatory bowel disease patients. J. Clin. Med. 2021, 10, 3689. [Google Scholar] [CrossRef] [PubMed]
  41. Allocca, M.; Chaparro, M.; Gonzalez, H.A.; Bosca-Watts, M.M.; Palmela, C.; D’Amico, F.; Zacharopoulou, E.; Kopylov, U.; Ellul, P.; Bamias, G.; et al. Patients with Inflammatory Bowel Disease Are Not at Increased Risk of COVID-19: A Large Multinational Cohort Study. J. Clin. Med. 2020, 9, 3533. [Google Scholar] [CrossRef] [PubMed]
  42. Magro, F.; Rahier, J.F.; Abreu, C.; MacMahon, E.; Hart, A.; van der Woude, C.J.; Gordon, H.; Adamina, M.; Viget, N.; Vavricka, S.; et al. Inflammatory bowel disease management during the COVID-19 outbreak:The ten do’s and don’ts from the ECCO-COVID task force. J. Crohn’s Colitis 2020, 14, S798–S806. [Google Scholar] [CrossRef]
  43. Bezzio, C.; Pellegrini, L.; Manes, G.; Arena, I.; Picascia, D.; Della Corte, C.; Devani, M.; Schettino, M.; Saibeni, S. Biologic therapies may reduce the risk of COVID-19 in patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2020, 26, E107–E109. [Google Scholar] [CrossRef]
  44. Iborra, I.; Puig, M.; Marín, L.; Calafat, M.; Cañete, F.; Quiñones, C.; González-González, L.; Cardona, G.; Mañosa, M.; Domènech, E. Treatment Adherence and Clinical Outcomes of Patients with Inflammatory Bowel Disease on Biological Agents during the SARS-CoV-2 Pandemic. Dig. Dis. Sci. 2021, 66, 4191–4196. [Google Scholar] [CrossRef]
  45. The Lancet. COVID-19: Protecting health-care workers. Lancet 2020, 395, 922. [Google Scholar] [CrossRef]
  46. Bezzio, C.; Saibeni, S.; Variola, A.; Allocca, M.; Massari, A.; Gerardi, V.; Casini, V.; Ricci, C.; Zingone, F.; Amato, A.; et al. Outcomes of COVID-19 in 79 patients with IBD in Italy: An IG-IBD study. Gut 2020, 69, 1213–1217. [Google Scholar] [CrossRef]
  47. Williamson, E.J.; Walker, A.J.; Bhaskaran, K.; Bacon, S.; Bates, C.; Morton, C.E.; Curtis, H.J.; Mehrkar, A.; Evans, D.; Inglesby, P.; et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature 2020, 584, 430–436. [Google Scholar] [CrossRef]
  48. Nowak, J.K.; Lindstrøm, J.C.; Kalla, R.; Ricanek, P.; Halfvarson, J.; Satsangi, J. Age, Inflammation, and Disease Location Are Critical Determinants of Intestinal Expression of SARS-CoV-2 Receptor ACE2 and TMPRSS2 in Inflammatory Bowel Disease. Gastroenterology 2020, 159, 1151–1154.e2. [Google Scholar] [CrossRef]
  49. Lukin, D.J.; Kumar, A.; Hajifathalian, K.; Sharaiha, R.Z.; Scherl, E.J.; Longman, R.S. Baseline Disease Activity and Steroid Therapy Stratify Risk of COVID-19 in Patients with Inflammatory Bowel Disease. Gastroenterology 2020, 159, 1541–1544.e2. [Google Scholar] [CrossRef] [PubMed]
  50. Chan, J.F.-W.; Yuan, S.; Kok, K.-H.; To, K.K.-W.; Chu, H.; Yang, J.; Xing, F.; Liu, J.; Yip, C.C.-Y.; Poon, R.W.-S.; et al. A familial cluster of pneumonia associated with the 2019 novel coronavirus indicating person-to-person transmission: A study of a family cluster. Lancet 2020, 395, 514–523. [Google Scholar] [CrossRef] [Green Version]
  51. Axelrad, J.E.; Malter, L.; Hong, S.; Chang, S.; Bosworth, B.; Hudesman, D. From the American Epicenter: Coronavirus Disease 2019 in Patients with Inflammatory Bowel Disease in the New York City Metropolitan Area. Inflamm. Bowel Dis. 2020, 27, 662–666. [Google Scholar] [CrossRef] [PubMed]
  52. Bataille, P.; Amiot, A.; Claudepierre, P.; Paris, N.; Neuraz, A.; Lerner, I.; Garcelon, N.; Rance, B.; Grisel, O.; Moreau, T.; et al. Letter: Severe COVID-19 infection and biologic therapies—A cohort study of 7 808 patients in France. Aliment. Pharmacol. Ther. 2020, 52, 1245–1248. [Google Scholar] [PubMed]
  53. Shang, L.; Zhao, J.; Hu, Y.; Du, R.; Cao, B. On the use of corticosteroids for 2019-nCoV pneumonia. Lancet 2020, 395, 683–684. [Google Scholar] [CrossRef] [Green Version]
  54. Zhou, W.; Liu, Y.; Tian, D.; Wang, C.; Wang, S.; Cheng, J.; Hu, M.; Fang, M.; Gao, Y. Potential benefits of precise corticosteroids therapy for severe 2019-nCoV pneumonia. Signal. Transduct. Target. Ther. 2020, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  55. Agrawal, M.; Brenner, E.J.; Zhang, X.; Colombel, J.-F.; Kappelman, M.D.; Ungaro, R.C.; Gearry, R.B.; Kalpan, G.G.; Kissous-Hunt, M.; Lewis, J.D.; et al. Physician Practice Patterns in Holding Inflammatory Bowel Disease Medications due to COVID-19, in the SECURE-IBD Registry. J. Crohns. Colitis 2021, 15, 860–863. [Google Scholar] [CrossRef]
  56. Attauabi, M.; Dahlerup, J.F.; Poulsen, A.; Hansen, M.R.; Verner-Andersen, M.K.; Eraslan, S.; Prahm, A.P.; Pedersen, N.; Larsen, L.; Jess, T.; et al. Outcomes and long-term effects of COVID-19 in patients with inflammatory bowel diseases—A Danish prospective population-based cohort study with individual-level data. J. Crohn’s Colitis 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  57. Mertz, B.; Dijkstra, F.; Nielsen, J.; Kjeldsen, J. Post COVID-19 hospitalizations in patients with chronic inflammatory diseases—A nationwide cohort study. J. Autoimmun. 2021, 125, 102739. [Google Scholar]
Figure 1. Geographic distribution of COVID-19 cases in the ENEIDA registry and comparison with the general Spanish population in the first wave of the pandemic.
Figure 1. Geographic distribution of COVID-19 cases in the ENEIDA registry and comparison with the general Spanish population in the first wave of the pandemic.
Jcm 11 00421 g001
Figure 2. Inflammatory bowel disease activity at COVID-19 diagnosis and 3- and 12-month follow-up.
Figure 2. Inflammatory bowel disease activity at COVID-19 diagnosis and 3- and 12-month follow-up.
Jcm 11 00421 g002
Table 1. Clinical baseline characteristics regarding inflammatory bowel disease with COVID-19.
Table 1. Clinical baseline characteristics regarding inflammatory bowel disease with COVID-19.
Clinical CharacteristicsCases n = 482
Gender
  Male251 (52)
  Female231 (48)
Age at COVID-19 diagnosis52 years (IQR 42–61)
IBD duration at COVID-19 diagnosis12 years (IQR 6–19)
Type of IBD, n (%)
  Crohn’s disease247 (51)
  Ulcerative colitis221 (46)
  Unclassified colitis14 (2.9)
Ulcerative colitis extent (%)
  E143 (19)
  E280 (36)
  E398 (44)
Crohn’s disease location, n (%)
  L1114 (46)
  L243 (17)
  L388 (36)
  L4 (isolated)3 (1.2)
Crohn’s disease behaviour, n (%)
  B1144 (58)
  B271 (29)
  B347 (19)
  Perianal59 (24)
  B1 + perianal29 (12)
  B2 + perianal18 (7.3)
  B3 + perianal20 (8.1)
Extraintestinal manifestation, n (%)125 (26)
Family history of IBD, n (%)64 (13)
Smoking behaviour, n (%)
  Active53 (11)
  Former smoker137 (28)
  Never smoker268 (56)
IQR: interquartile rate, IBD: inflammatory bowel disease, L1: ileal, L2: colonic, L3: ileocolonic, L4: upper gastrointestinal tract; B1: inflammatory behaviour, B2: stricturing behaviour, B3: penetrating behaviour.
Table 2. Inflammatory bowel disease activity and treatment at time of COVID-19 diagnosis.
Table 2. Inflammatory bowel disease activity and treatment at time of COVID-19 diagnosis.
IBD (Total)
n = 482
Crohn’s Disease
n = 247
Ulcerative Colitis
n = 221
p-Value *
IBD Activity at COVID-19 Diagnosis
Clinical remission385 (80)200 (81)173 (78)0.35
Active disease97 (20)47 (19)48 (22) 0.35
  Mild53 (11)26 (10.5)26 (12)
  Moderate42 (8.7)21 (8.5)20 (9)
  Severe2 (0.4)02 (0.9)
IBD treatment
None, n (%)62 (13)37 (15)23 (10.4)0.15
5-aminosalicylates, n (%)202 (42)49 (20)143 (65)<0.0001
  Oral (oral and topic)197 (41)49 (20)138 (62)
  Topical (exclusive)5 (1)05 (2.3)
  Monotherapy131 (27)31 (12)91 (41)
Systemic steroids 3 months before COVID-19 (oral or intravenous), n (%)53 (11)30 (12)21 (9.5)0.36
Systemic steroids, n (%)26 (5.4)16 (6.4)8 (3.6)0.37
Immunosuppressants
(in monotherapy), n (%)
113 (23)65 (26)56 (25)0.03
  Azathioprine90 (19)54 (22)46 (21)0.04
  Mercaptopurine8 (1.7)4 (1.6)2 (0.9)0.16
  Cyclosporine1 (0.2) 01 (0.4)0.96
  Methotrexate9 (1.9)6 (2.4)3 (1.3)0.06
  Tacrolimus1 (0.2)1 (0.4)01
  Tofacitinib4 (0.8)04 (1.8)0.10
Biologics
(in monotherapy), n (%)
117 (22)72 (29)35 (16)0.04
  Anti-TNF71 (15)42 (17)19 (8.6)<0.0001
  Vedolizumab25 (5.2)12 (4.8)13 (5.9)0.75
  Ustekinumab21 (4.3)18 (7.3)3 (1.3)0.001
Combotherapy, n (%)59 (12)45 (18)14 (6.3)0.02
  Anti-TNF plus thiopurines37 (7.7)28 (11)9 (4.1)0.02
  Anti-TNF plus methotrexate9 (1.9)6 (2.4)3 (1.3)0.62
  Vedolizumab plus thiopurines5 (1)4 (1.6)1 (0.4)0.73
  Vedolizumab plus methotrexate1 (0.2)0 1 (0.4)0.78
  Ustekinumab plus thiopurines5 (1)5 (2)00.55
  Ustekinumab plus methotrexate2 (0.4)2 (0.8)00.98
* Comparison between Crohn’s disease and ulcerative colitis. IBD: inflammatory bowel disease; TNF: tumour necrosis factor.
Table 3. Epidemiological factors of SARS-CoV-2 infection and occupational risk.
Table 3. Epidemiological factors of SARS-CoV-2 infection and occupational risk.
Route of Contagion, n (%)
Unknown242 (50)
Intrafamilial transmission108 (22)
Occupational96 (20)
Travel8 (1.7)
Occupational Risk, n (%)133 (28)
Healthcare85 (18)
Basic services (supermarket cashiers, market clerks, pharmacy)18 (3.7)
Education15 (3)
Police and fireperson5 (1)
Closed institutions2 (0.4)
Veterinary, animal control worker or conservation and forest technician4 (0.8)
Table 4. Relationship between the route of contagion and occupational risk in patients with and without a total lockdown.
Table 4. Relationship between the route of contagion and occupational risk in patients with and without a total lockdown.
Risk VariablePatients with Total Lockdown
(n = 229)
Patients without Total Lockdown
(n = 225)
p-Value
Route of contagion, n (%)
Intrafamilial transmission70 (31)38 (17)0.001
  Infection on March 202047 (20)26 (12)0.007
  Infection on April–July 202023 (10)12 (5.3)0.034
Occupational19 (8.3)77 (34)<0.0001
  Infection on March 202019 (8.2)48 (21)<0.0001
  Infection on April–July 2020029 (13)<0.0001
Travel3 (1.3)5 (2.2)0.69
  Infection on March 20203 (1.3)5 (2.2)0.69
  Infection on April–July 2020--
Unknown137 (60)105 (47)0.005
  Infection on March 202083 (36)69 (31)0.167
  Infection on April–July 202054 (24)36 (16)0.003
Occupational risk, n (%)
Occupational risk (all)38 (17)92 (41)<0.0001
  Healthcare18 (7.9)65 (29)<0.0001
Table 5. Outcome of the COVID-19-EII cohort and treatment administered. The results are compared with those reported in the SECURE-IBD cohort (as for 24 August 2021) [9] .
Table 5. Outcome of the COVID-19-EII cohort and treatment administered. The results are compared with those reported in the SECURE-IBD cohort (as for 24 August 2021) [9] .
A.
Outcomes
OutcomeHospitalisedICU AdmissionSevere COVID-19Death
COVID-19-EII
n = 482
SECURE-IBD
n = 6438
COVID-19-EII
n = 482
SECURE-IBD
n = 6438
COVID-19-EII
n = 482
SECURE-IBD
n = 6438
COVID-19-EII
n = 482
SECURE-IBD
n = 6438
n = 168
(35%)
n = 977
(15%)
n = 13
(2.6%)
n = 184 (2.8%)n = 38 *
(7.8%)
n = 257 **
(3.9%)
n = 18
(3.7%)
n = 104
(1.6%)
B.
Treatment taking into account each specific outcome
DRUGTotalOUTCOMES
HospitalisedICUSevere COVID-19Death
CohortCOVID-19-EII
n = 482
SECURE-IBD
n = 6438
COVID-19-EII
n = 168
SECURE-IBD
n = 977
COVID-19-EII
n = 13
SECURE-IBD
n = 184
COVID-19-EII *
n = 38
SECURE-IBD **
n = 257
COVID-19-EII
n = 18
SECURE-IBD
n = 104
5-aminosalicylates, 202 (42)1924 (30)79 (47)411 (42)10 (77)81 (44)24 (63)118 (46)9 (50)53 (51)
Alone131 (27)-52 (31)-5 (38)-16 (42)-6 (33)-
With other IBD drugs71 (15)-27 (16)-5 (28)-8 (21)-3 (17)-
Systemic steroids26 (5.4)414 (6.4)11 (6.5)146 (15)1 (7.7)41 (22)4 (10.5)53 (21)1 (5.6)28 (27)
Thiopurines (monotherapy)108 (22)551 (8.6)38 (23)114 (12)3 (23)26 (14)5 (13)33 (13)1 (5.6)11 (10.6)
Methotrexate (monotherapy)9 (1.9)49 (0.8)4 (2.4)13 (1.3)1 (7.7)1 (0.5)2 (5.3)3 (1.2)1 (5.6)2 (1.9)
Anti-TNF (monotherapy)71 (15)2082 (32)11 (6.5)178 (18)2 (15)24 (13)3 (7.9)31 (12)2 (11)10 (9.6)
Anti-TNF in combotherapy46 (9.5)636 (9.9)20 (12)91 (9.3)017 (9)2 (5.3)21 (8.2)2 (11)6 (5.8)
Vedolizumab30 (6.2)706 (11)15 (8.9)94 (9.6)021 (11)4 (10.5)28 (10.9)2 (11)9 (8.6)
Ustekinumab28 (5.8)602 (9)6 (3.6)50 (5.1)1 (7.7)9 (4.9)1 (2.6)11 (4.3)1 (5.6)5 (4.8)
Tofacitinib4 (0.8)103 (1.6)2 (1.2)12 (1.2)04 (2.2)04 (1.5)01 (0.9)
ICU: intensive care unit, TNF: tumour necrosis factor. Brenner EJ, Ungaro RC, Colombel JF, Kappelman MD. SECURE-IBD Database Public Data, https://covidibd.org/current-data/ accessed on 24 August 2021. * Severe COVID-19 for the COVID-19-EII study: composite of intensive care unit admission and/or use of active amines and/or respiratory distress and/or invasive oxygen therapy and/or death. ** Severe COVID-19 for the SECURE-IBD registry: composite of intensive care unit admission and/or use of ventilator and/or death.
Table 6. Sequelae at 3- and 12-months of COVID-19 and the impact of the infection on changes in therapeutic regimens for IBD.
Table 6. Sequelae at 3- and 12-months of COVID-19 and the impact of the infection on changes in therapeutic regimens for IBD.
3 Months Follow-Up
(n = 462)
12 Months Follow-Up
(n = 451)
COVID-19 sequelae, n (%)65 (14)72 (15)
Psychologic sequelae, n (%)20 (4.3)15 (3.3)
Physical sequelae, n (%) 55 (12)67 (15)
  Asthenia21 (4.5)22 (4.8)
  Myalgia/Arthralgia 7 (1.5)3 (0.7)
  Anosmia4 (0.9)7 (1.5)
  Dyspnoea4 (0.9)6 (1.3)
  Odynophagia 2 (0.4)2 (0.4)
  Dysgeusia2 (0.4)2 (0.4)
  Hair loss1 (0.2)1 (0.2)
  Pulmonary fibrosis1 (0.2)3 (0.6)
  Bronchial hyperreactivity1 (0.2)1 (0.2)
  Deep venous thrombosis/pulmonary thrombosis1 (0.2)1 (0.2)
  Headache1 (0.2)6 (1.3)
  Obstructive pulmonary disease1 (0.2)3 (0.6)
  Paraesthesia1 (0.2)3 (0.6)
  Wegener’s vasculitis-1 (0.2)
Immunosuppression withdrawal, n (%)65 (14)6 (1.3)
  Transient58 (13)1 (0.2)
  Definitive7 (1.5)5 (1.1)
  De-escalation from combo to monotherapy13 (2.9)1 (0.2)
Patients requiring Immunosuppression initiation or modification, n (%)12 (2.6)43 * (9.5)
  Systemic corticosteroids1 (0.2)7 (1.6)
  Thiopurines2 (0.4)5 (1.1)
  Methotrexate01 (0.2)
  Anti-TNF5 (1)18 (4)
  Vedolizumab1 (0.2)7 (1.6)
  Ustekinumab2 (0.4)12 (2.7)
  Tofacitinib1 (0.2)6 (1.3)
* Some patients required more than one new immunosuppressant (combined or sequential); therefore, this number expresses the total number of patients that required immunosuppression initiation/modification from 3 to 12 months after COVID-19. IBD = inflammatory bowel disease; TNF: tumour necrosis factor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zabana, Y.; Marín-Jiménez, I.; Rodríguez-Lago, I.; Vera, I.; Martín-Arranz, M.D.; Guerra, I.; Gisbert, J.P.; Mesonero, F.; Benítez, O.; Taxonera, C.; et al. Nationwide COVID-19-EII Study: Incidence, Environmental Risk Factors and Long-Term Follow-Up of Patients with Inflammatory Bowel Disease and COVID-19 of the ENEIDA Registry. J. Clin. Med. 2022, 11, 421. https://doi.org/10.3390/jcm11020421

AMA Style

Zabana Y, Marín-Jiménez I, Rodríguez-Lago I, Vera I, Martín-Arranz MD, Guerra I, Gisbert JP, Mesonero F, Benítez O, Taxonera C, et al. Nationwide COVID-19-EII Study: Incidence, Environmental Risk Factors and Long-Term Follow-Up of Patients with Inflammatory Bowel Disease and COVID-19 of the ENEIDA Registry. Journal of Clinical Medicine. 2022; 11(2):421. https://doi.org/10.3390/jcm11020421

Chicago/Turabian Style

Zabana, Yamile, Ignacio Marín-Jiménez, Iago Rodríguez-Lago, Isabel Vera, María Dolores Martín-Arranz, Iván Guerra, Javier P. Gisbert, Francisco Mesonero, Olga Benítez, Carlos Taxonera, and et al. 2022. "Nationwide COVID-19-EII Study: Incidence, Environmental Risk Factors and Long-Term Follow-Up of Patients with Inflammatory Bowel Disease and COVID-19 of the ENEIDA Registry" Journal of Clinical Medicine 11, no. 2: 421. https://doi.org/10.3390/jcm11020421

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop