ArticlePDF Available

Multiorgan impairment in low-risk individuals with post-COVID-19 syndrome: A prospective, community-based study

Authors:

Abstract and Figures

Objective: To assess medium-term organ impairment in symptomatic individuals following recovery from acute SARS-CoV-2 infection. Design: Baseline findings from a prospective, observational cohort study. Setting: Community-based individuals from two UK centres between 1 April and 14 September 2020. Participants: Individuals ≥18 years with persistent symptoms following recovery from acute SARS-CoV-2 infection and age-matched healthy controls. Intervention: Assessment of symptoms by standardised questionnaires (EQ-5D-5L, Dyspnoea-12) and organ-specific metrics by biochemical assessment and quantitative MRI. Main outcome measures: Severe post-COVID-19 syndrome defined as ongoing respiratory symptoms and/or moderate functional impairment in activities of daily living; single-organ and multiorgan impairment (heart, lungs, kidneys, liver, pancreas, spleen) by consensus definitions at baseline investigation. Results: 201 individuals (mean age 45, range 21-71 years, 71% female, 88% white, 32% healthcare workers) completed the baseline assessment (median of 141 days following SARS-CoV-2 infection, IQR 110-162). The study population was at low risk of COVID-19 mortality (obesity 20%, hypertension 7%, type 2 diabetes 2%, heart disease 5%), with only 19% hospitalised with COVID-19. 42% of individuals had 10 or more symptoms and 60% had severe post-COVID-19 syndrome. Fatigue (98%), muscle aches (87%), breathlessness (88%) and headaches (83%) were most frequently reported. Mild organ impairment was present in the heart (26%), lungs (11%), kidneys (4%), liver (28%), pancreas (40%) and spleen (4%), with single-organ and multiorgan impairment in 70% and 29%, respectively. Hospitalisation was associated with older age (p=0.001), non-white ethnicity (p=0.016), increased liver volume (p<0.0001), pancreatic inflammation (p<0.01), and fat accumulation in the liver (p<0.05) and pancreas (p<0.01). Severe post-COVID-19 syndrome was associated with radiological evidence of cardiac damage (myocarditis) (p<0.05). Conclusions: In individuals at low risk of COVID-19 mortality with ongoing symptoms, 70% have impairment in one or more organs 4 months after initial COVID-19 symptoms, with implications for healthcare and public health, which have assumed low risk in young people with no comorbidities. Trial registration number: NCT04369807; Pre-results.
Content may be subject to copyright.
1
DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
Multiorgan impairment in low- risk
individuals with post- COVID-19
syndrome: a prospective, community-
based study
Andrea Dennis,1 Malgorzata Wamil,2,3 Johann Alberts,4 Jude Oben,5,6
Daniel J Cuthbertson,7 Dan Wootton,8,9 Michael Crooks,10,11 Mark Gabbay,12
Michael Brady,1,13 Lyth Hishmeh,14 Emily Attree,15 Melissa Heightman,16
Rajarshi Banerjee,1 Amitava Banerjee ,16,17,18 On behalf of COVERSCAN study
investigators
To cite: DennisA, WamilM,
AlbertsJ, etal. Multiorgan
impairment in low- risk
individuals with post-
COVID-19 syndrome: a
prospective, community-
based study. BMJ Open
2021;11:e048391. doi:10.1136/
bmjopen-2020-048391
Prepublication history and
additional material for this
paper are available online. To
view these les, please visit
the journal online (http:// dx. doi.
org/ 10. 1136/ bmjopen- 2020-
048391).
RB and AB are joint senior
authors.
Received 26 December 2020
Revised 25 February 2021
Accepted 11 March 2021
For numbered afliations see
end of article.
Correspondence to
Dr Amitava Banerjee;
ami. banerjee@ ucl. ac. uk
Original research
© Author(s) (or their
employer(s)) 2021. Re- use
permitted under CC BY.
Published by BMJ.
ABSTRACT
Objective To assess medium- term organ impairment in
symptomatic individuals following recovery from acute
SARS- CoV-2 infection.
Design Baseline ndings from a prospective,
observational cohort study.
Setting Community- based individuals from two UK
centres between 1 April and 14 September 2020.
Participants Individuals ≥18 years with persistent
symptoms following recovery from acute SARS- CoV-2
infection and age- matched healthy controls.
Intervention Assessment of symptoms by standardised
questionnaires (EQ- 5D- 5L, Dyspnoea-12) and organ-
specic metrics by biochemical assessment and
quantitative MRI.
Main outcome measures Severe post- COVID-19
syndrome dened as ongoing respiratory symptoms and/or
moderate functional impairment in activities of daily living;
single- organ and multiorgan impairment (heart, lungs,
kidneys, liver, pancreas, spleen) by consensus denitions
at baseline investigation.
Results 201 individuals (mean age 45, range 21–71
years, 71% female, 88% white, 32% healthcare workers)
completed the baseline assessment (median of 141 days
following SARS- CoV-2 infection, IQR 110–162). The study
population was at low risk of COVID-19 mortality (obesity
20%, hypertension 7%, type 2 diabetes 2%, heart disease
5%), with only 19% hospitalised with COVID-19. 42%
of individuals had 10 or more symptoms and 60% had
severe post- COVID-19 syndrome. Fatigue (98%), muscle
aches (87%), breathlessness (88%) and headaches (83%)
were most frequently reported. Mild organ impairment
was present in the heart (26%), lungs (11%), kidneys
(4%), liver (28%), pancreas (40%) and spleen (4%), with
single- organ and multiorgan impairment in 70% and 29%,
respectively. Hospitalisation was associated with older age
(p=0.001), non- white ethnicity (p=0.016), increased liver
volume (p<0.0001), pancreatic inammation (p<0.01),
and fat accumulation in the liver (p<0.05) and pancreas
(p<0.01). Severe post- COVID-19 syndrome was associated
with radiological evidence of cardiac damage (myocarditis)
(p<0.05).
Conclusions In individuals at low risk of COVID-19
mortality with ongoing symptoms, 70% have impairment
in one or more organs 4 months after initial COVID-19
symptoms, with implications for healthcare and public
health, which have assumed low risk in young people with
no comorbidities.
Trial registration number NCT04369807; Pre-results.
INTRODUCTION
Early in the COVID-19 pandemic, research
and clinical practice focused on pulmonary
manifestations.1 There is increasing evidence
for direct multiorgan effects,2–7 as well as indi-
rect effects on other organ systems and disease
processes, such as cardiovascular diseases
and cancers, through changes in healthcare
delivery and patient behaviours.8–10 The
Strengths and limitations of this study
This is an ongoing, prospective, longitudinal
COVID-19 recovery study with biochemical and im-
aging characterisation of organ function, starting in
April 2020 before recognition of ‘long- COVID’, prop-
er testing availability and prospective COVID-19-
related research.
By recruiting ambulatory patients with broad inclu-
sion criteria, we focused on a real- world population
at lower risk of COVID-19 severity and mortality.
Healthy controls were included for comparison, not
individuals with postinuenza symptoms, COVID-19
without symptoms or from general clinics, which
further studies may explore.
The study population was not ethnically diverse
despite disproportionate COVID-19 impact in non-
white individuals.
To limit interaction and exposure between the trial
team and the patients, pulse oximetry, spirometry,
MRI assessment of the brain and muscle function
were not included from the outset.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
2DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
clear long- term impact on individuals and health systems
underlines the urgent need for a whole body approach
with assessment of all major organ systems following
SARS- CoV-2 infection. Quantitative MRI has recently
been used to show multiorgan impairment in individuals
post- COVID-19 hospitalisation,11 but has not been used in
non- hospitalised individuals.
COVID-19 is the convergence of an infectious disease,
undertreated non- communicable diseases and social
determinants of health, described as a ‘syndemic’.12 Pre-
existing non- communicable diseases and risk factors
predict poor COVID-19 outcomes, whether intensive
care admission or mortality.10 Research has emphasised
acute SARS- CoV-2 infection, hospitalised individuals and
COVID-19 mortality,13–15 which is likely to underestimate
the true burden of COVID-19- related disease. Among
those surviving acute infection, 10% report persistent
symptoms for 12 weeks or longer after initial infection
(‘long- COVID’, or ‘post COVID-19 syndrome’, PCS).16
However, PCS is yet to be fully defined.17–20 Neither
severity of symptoms, nor medium- term and long- term
pathophysiology across organ systems, nor the appro-
priate control populations are understood.
UK government policies have emphasised excess
mortality risk in moderate- risk and high- risk conditions,
including ‘shielding’10 and commissioning of a risk calcu-
lator to identify those at highest risk of COVID-19 severity
and mortality.21 These policies assume that younger indi-
viduals without apparent underlying conditions are at low
risk. However, unlike symptoms following critical illness22
or acute phase of other coronavirus infections,23 symp-
toms in PCS are commonly reported in individuals with
low COVID-19 mortality risk, for example, female, young
and no chronic comorbidities.14 The potential scale of
PCS in ‘lower- risk’ individuals, representing up to 80% of
the population,3 necessitates urgent policies across coun-
tries to monitor,24 treat19 and pay25 for long- term implica-
tions of COVID-19 and to mitigate impact on healthcare
utilisation and economies.
Therefore, in a pragmatic, prospective cohort study
of individuals with persistent symptoms at least 4 weeks
following recovery from acute SARS- CoV-2 infection and
at low risk of COVID-19 mortality, we investigated (1)
the prevalence of multiorgan impairment, compared
with healthy, age- matched controls; (2) the associations
between typical COVID-19 symptoms and multiorgan
impairment; and (3) the associations between hospital-
isation, severity of symptoms and multiorgan impairment.
METHODS
Patient population and study design
In an ongoing, prospective study, participants were
recruited to the study following expression of interest on
the study registration website. Participants learnt about
the study through advertisement on social media or via
recommendations from clinicians from four partici-
pant identification centres, the latter usually applied to
patients who had been hospitalised. Assessment took
place at two UK research imaging sites (Perspectum,
Oxford; and Mayo Clinic Healthcare, London) between
1 April 2020 and 14 September 2020, completing base-
line assessment by 14 September 2020 (figure 1). Partic-
ipants with laboratory- confirmed SARS- CoV-2 infection
(tested SARS- CoV-2- positive by oropharyngeal/naso-
pharyngeal swab by reverse- transcriptase PCR (n=62),
a positive antibody test (n=63), or with strong clinical
suspicion of infection with typical symptoms/signs and
assessed as highly likely to have COVID-19 by two inde-
pendent clinicians (n=73)) were eligible for enrolment.
Exclusion criteria were symptoms of active respiratory
viral infection (temperature >37.8°C or three or more
episodes of coughing in 24 hours), hospital discharge in
the last 7 days, and contraindications to MRI, including
implanted pacemakers, defibrillators, other metallic
implanted devices and claustrophobia. All participants
gave written informed consent.
Assessment of PCS
Assessment included patient- reported validated question-
naires (quality of life, EQ- 5D- 5L,26 and Dyspnoea-1227)
and fasting biochemical investigations (listed in online
supplemental methods). PCS was classified as ‘severe’
(defined as persistent breathlessness, score of 10 on
Dyspnoea-12, or reported moderate or greater problems
with usual activities on EQ- 5D- 5L) or ‘moderate’. These
thresholds were selected as the Dyspnoea-12 has been
correlated with the Medical Research Council (MRC)
Figure 1 Flow from recruitment to enrolment of 201 patients
with post- COVID-19 syndrome.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
3
DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
Table 1 Baseline demographics and symptoms of 201 low- risk individuals with post- COVID-19 syndrome
All patients
(N=201)
Healthy
controls (n=36)
P
value
Not hospitalised
(n=163)
Hospitalised
(n=37)
P
value
Moderate PCS
(n=77)
Severe PCS
(n=116) P value
Age (years), mean (SD) 44 (11.0) 39 (12.4) 0.013 43 (10.9) 50 (10.0) 0.001 45 (12.2) 44 (10.0) 0.419
Female, n (%) 142 (70.6) 14 (38.9) 0.032 118 (72.4) 23 (62.2) 0.302 51 (66.2) 85 (73.3) 0.374
BMI (kg/m2), median (IQR) 25.7 (22.7–28.1) 23.2 (21.4–23.1) <0.001 25.3 (22.7–27.7) 27.2 (23.1–31.0) 0.063 25.8 (22.7–27.9) 25.4 (22.5–28.2) 0.639
Ethnicity
White 176 (87.6) 33 (91.7) 148 (90.8) 28 (75.7) 67 (87.0) 106 (91.4) 0.178
Mixed 3 (1.5) 0 (0) 0.904 3 (1.8) 0 (0) 0.016 1 (1.3) 2 (1.7)
South Asian 7 (3.5) 3 (8.3) 4 (2.5) 3 (8.1) 5 (6.5) 0 (0)
Black 4 (2.0) 0 (0) 1 (0.6) 2 (5.4) 2 (2.6) 2 (1.7)
Comorbidities and risks
Smoking 0.244
Never 133 (66.2) 20 (60.6) 108 (66.3) 24 (64.9) 55 (71.4) 72 (61.7)
Current 6 (3.0) 8 (24.2) <0.001 6 (3.7) 0 (0) 0.641 3 (3.9) 3 (2.6)
Ex- smoker 62 (30.8) 5 (15.2) 49 (30.1) 13 (35.1) 19 (24.7) 41 (35.3)
Healthcare worker 64 (31.8) 4 (12.1) 0.009 50 (30.7) 13 (35.1) 0.695 33 (42.9) 28 (24.1) 0.007
Asthma 37 (18.4) 0 (0) 0.002 34 (20.9) 3 (8.1) 0.099 13 (16.9) 22 (19.0) 0.849
BMI
≥25 kg/m2113 (56.5) 7 (20) 87 (53.7) 25 (67.6) 0.144 46 (60.5) 62 (53.4) 0.374
≥30 kg/m240 (20.0) 0 (0) 28 (17.3) 12 (32.4) 0.066 16 (21.1) 24 (20.7) 1.000
Hypertension 13 (6.5) 0 (0) 0.001 11 (6.7) 2 (5.4) 1.000 6 (7.8) 7 (6.0) 0.771
Diabetes 4 (2.0) 0 (0) 0.104 4 (2.5) 0 (0.0) 1.000 4 (5.2) 0 (0.0) 0.024
Previous heart disease 9 (4.5) 0 (0) 0.001 8 (4.9) 1 (2.7) 1.000 3 (3.9) 5 (4.3) 1.000
Symptoms
Fatigue 196 (98.0) 159 (97.5) 37 (100.0) 1.000 73 (96.1) 115 (99.1) 0.302
Shortness of breath 176 (88.0) 141 (86.5) 35 (94.6) 0.262 58 (76.3) 112 (96.6) <0.0001
Muscle ache 173 (86.5) 142 (87.1) 31 (83.8) 0.597 66 (86.8) 101 (87.1) 1.000
Headache 165 (82.5) 138 (84.7) 27 (73.0) 0.098 56 (73.7) 102 (87.9) 0.019
Joint pain 156 (78.0) 127 (77.9) 29 (78.4) 1.000 56 (73.7) 94 (81.0) 0.284
Chest pain 152 (76.0) 128 (78.5) 24 (64.9) 0.090 47 (61.8) 98 (84.5) 0.001
Cough 146 (73.0) 117 (71.8) 29 (78.4) 0.539 55 (72.4) 84 (72.4) 1.000
Fever 144 (72.0) 113 (69.3) 31 (83.8) 0.104 51 (67.1) 86 (74.1) 0.329
Sore throat 143 (71.5) 120 (73.6) 23 (62.2) 0.165 50 (65.8) 86 (74.1) 0.256
Continued
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
4DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
dyspnoea grade, where level 3 warrants referral to reha-
bilitation services,27 and with EQ- 5D- 5L, less than 8% of
the general population report moderate or greater prob-
lems with usual activities.28
Multiorgan impairment in PCS compared with healthy controls
We selected MRI as the imaging modality (as in UK
Biobank) due to (1) safety (no radiation exposure, no
need for intravenous contrast and minimal contact
with the radiographer); (2) quantitative reproducibility
(>95% acquisition and image processing success rate);
(3) capacity for information sharing (digital data repos-
itory for independent analysis and research); and (4)
rapid scalability (35 min scan to phenotype lung, heart,
kidney, liver, pancreas and spleen). Multiorgan MRI data
were collected at both study sites (Oxford: MAGNETOM
Aera 1.5T; Mayo Healthcare London: MAGNETOM Vida
3T; both from Siemens Healthcare, Erlangen, Germany).
The COVERSCAN multiparametric MRI assessment typi-
cally required 35 min per patient, including the lungs,
heart, liver, pancreas, kidneys and spleen, by standardised
methodology (online supplemental file 1). In brief, we
assessed inflammation of the heart, kidneys, liver and
pancreas with quantitative T1 relaxation mapping; lung
function was characterised with a dynamic structural
T2- weighted lung scan estimating lung capacity; ectopic
fat accumulation in the liver and pancreas from proton
density fat fraction; and volume of the liver and spleen
measured from T1- weighted structural scan.
To determine impairment in each organ, we compared
MRI- derived measurements from the heart, lungs,
kidneys, liver, pancreas and spleen with reference ranges
(online supplemental table 1), which were established
as mean±2 SD from the healthy, age- matched control
subjects (n=36) and validated by scoping literature
review.11 We defined organ impairment if quantitative
T1 mapping was outside the reference ranges for the
heart, kidney, liver and pancreas, reduced estimated
lung capacity from dynamic measurements in the lungs,
or there was evidence of hepatomegaly, splenomegaly or
ectopic fat accumulation.
Symptoms and multiorgan impairment
Associations between organ impairment and symptoms
were visually assessed using a heat map, dividing those
with impairments to an organ into columns and colouring
the rows by percentage of reported symptoms.
Hospitalisation, severity and multiorgan impairment
We compared mean differences in quantitative organ
metrics for hospitalised versus not hospitalised and
moderate versus severe PCS using Kruskal- Wallis test
(Fisher’s exact test for differences in binary outcomes).
We defined multiorgan impairment as 2 organs with
metrics outside the reference range. We investigated the
associations between multiorgan impairment and (1)
being hospitalised and (2) severe PCS with multivariate
All patients
(N=201)
Healthy
controls (n=36)
P
value
Not hospitalised
(n=163)
Hospitalised
(n=37)
P
value
Moderate PCS
(n=77)
Severe PCS
(n=116) P value
Diarrhoea 118 (59.0) 91 (55.8) 27 (73.0) 0.065 40 (52.6) 76 (65.5) 0.097
Abnormal pain 108 (54.0) 91 (55.8) 17 (45.9) 0.361 30 (39.5) 75 (64.7) 0.001
Wheezing 98 (49.0) 75 (46.0) 23 (62.2) 0.101 30 (39.5) 64 (55.2) 0.039
Inability to walk 80 (40.0) 58 (35.6) 22 (59.5) 0.009 24 (31.6) 50 (43.1) 0.130
Runny nose 68 (34.0) 55 (33.7) 13 (35.1) 0.85 24 (31.6) 41 (35.3) 0.642
Time interval
Initial symptoms to assessment
(days), median (IQR) 141 (110–162) 141 (112–163) 138 (97–150) 0.106 121 (89–158) 145 (121–163) 0.001
COVID-19- positive to
assessment (days), median
(IQR) 71 (41–114) 68 (35–112) 105 (59–126) 0.012 60 (43–98) 78 (34–119) 0.305
Data are presented as count (%).
Comparisons between managed at home versus hospitalised and between moderate versus PCS were conducted using Fisher’s exact test
BMI, body mass index; PCS, post- COVID-19 syndrome.
Table 1 Continued
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
5
DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
logistic regression models, adjusting for age, sex and body
mass index (BMI).
Patient and public involvement and engagement
Patients and the public have directly and indirectly
informed our research, from design to dissemination,
with regular updates and webinars, including question
and answer sessions with patients. Several clinician coau-
thors were indirectly informed by their patients in the
COVERSCAN study (RB, AB) or PCS clinics (DW, MH,
MC), who are members of organisations such as Long
Covid SOS (eg, LH) and UKDoctors#Longcovid (eg, EA).
LH and EA have been involved in the research, inter-
pretation of results, understanding implications of our
results and providing critical feedback to the manuscript.
Statistical analysis
We performed all analyses using R V.3.6.1, using descrip-
tive statistics to summarise baseline characteristics and
considering a p value less than 0.05 as statistically signif-
icant. Mean and SD were used for normally distributed
continuous variables, median with IQR for non- normally
distributed variables, and frequency and percentage for
categorical variables. For group- wise comparison for abso-
lute values between cases and healthy controls, we used
Kruskal- Wallis test.
RESULTS
Overall study population
Baseline characteristics
The study included 201 individuals (full details regarding
hospitalisation: n=199; full questionnaire data to assign
PCS severity: n=193). The mean age was 44.0 (range
21–71) years and the median BMI was 25.7 (IQR 23–28).
Of the individuals, 71% were female, 88% were white,
32% were healthcare workers and 19% had been hospi-
talised with COVID-19. Assessments (symptoms, blood
and MRI) had a median of 141 (IQR 110–162) days after
initial symptoms. Medical history included smoking
(3%), asthma (19%), obesity (20%), hypertension (7%),
diabetes (2%) and prior heart disease (5%). The healthy
control group had a mean age of 39 years (range 20–70),
40% were female, with a median BMI of 23 (IQR: 21–25)
(table 1).
Regardless of hospitalisation, the most frequently
reported symptoms were fatigue (98%), shortness of
breath (88%), muscle ache (87%) and headache (83%)
(table 1). Of the individuals, 99% had four or more and
42% had ten or more symptoms. Of individuals 70%
reported 13 weeks off paid employment. Of the inci-
dental structural findings observed on MRI (n=56), three
were cardiac (atrial septal defect, bicuspid aortic valve
and right atrial mass), one renal (hydronephrosis) and
the rest were benign cysts.
Haematological investigations, including mean corpus-
cular haemoglobin concentration (24%), and renal,
liver and lipid biochemistry, including potassium (38%),
alanine transferase (14%), lactate dehydrogenase (17%),
triglycerides (11%) and cholesterol (42%), were abnor-
mally high in 10% of individuals. Bicarbonate (10%),
phosphate (11%), uric acid (11%) and transferrin satu-
ration (19%) were abnormally low in 10% of individuals
(online supplemental table 1).
Single-organ and multiorgan impairment in PCS compared
with healthy controls
Organ impairment was more common in PCS than
healthy controls (figure 2 and online supplemental figure
Figure 2 Percentage of patients (black) and controls (grey) with individual organ measures outside of the predened normal
range. Lines represent signicant difference in the proportions between the two groups, with *p<0.05, **p<0.01, ***p<0.001. LV,
left ventricular.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
6DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
Table 2 Evidence of organ impairment in 201 low- risk individuals with post- COVID-19 syndrome
Measurement
All patients
(N=201)
Healthy
controls (n=36)
P
value
Not hospitalised
(n=163)
Hospitalised
(n=37) P value
Moderate
PCS (n=77)
Severe PCS
(n=116)
P
value
Heart
Left ventricular ejection fraction (%)
Normal (>51%) 190 (95.0) 35 (97.2) 0.699 155 (95.7) 33 (89.1) 0.124 72 (93.5) 111 (95.7) 0.353
Impaired (≤51%) 11 (5.0) 1 (2.8) 7 (4.3) 4 (10.1) 5 (6.4) 5 (4.3)
Left ventricular end diastolic volume (mL)
>264 mL in Men; >206 mL in Women 8 (4.0) 1 (2.8) 1.00 4 (2.5) 4 (10.8) 0.040 4 (5.2) 4 (3.4) 0.715
Evidence of myocarditis
≥3 segments with high T1 (≥1229 ms at 3T;
≥1015 ms at 1.5T) 39 (19.4)
2 (5.6)
0.053 30 (18.4) 8 (21.6) 0.647 9 (11.7) 29 (25.0) 0.027
Lungs
Deep breathing fractional area change (n=17 missing) (n=13 missing) (n=3 missing) (n=8 missing) (n=7 missing)
<31% 21 (11.4) 1 (2.8) 0.138 17 (11.3) 4 (11.8) 1 7 (10.1) 13 (11.9) 0.811
Kidneys
Kidney cortex T1 (n=3 missing) (n=3 missing) (n=2 missing)
Normal (<1652 ms at 3T; <1227 ms at 1.5T) 191 (96.5) 36 (100.0) 0.599 155 (96.9) 35 (94.6) 0.618 74 (98.7) 112 (96.6) 0.65
Impaired (≥1652 ms at 3T; ≥1227 ms at 1.5T) 7 (3.5) 0 (0.0) 5 (3.1) 2 (5.4) 1 (1.3) 4 (3.4)
Pancreas
Pancreatic inammation (T1 in ms) (n=11 missing) (n=13 missing) (n=7 missing) (n=4 missing) (n=4 missing) (n=6 missing)
Normal <803 ms 162 (85.3) 23 (100.0) 0.049 139 (89.1) 22 (66.7) 0.002 60 (82.2) 95 (86.4) 0.530
Impaired ≥803 ms 28 (14.7) 0 (0) 17 (10.9) 11 (33.3) 13 (17.8) 15 (13.6)
Pancreatic fat (n=4 missing)
Normal <4.6% 122 (62.2) 30 (93.8) <0.001 107 (66.9) 14 (40.0) 0.004 44 (57.9) 72 (63.7) 0.449
Impaired ≥4.6% 74 (37.8) 2 (6.2) 53 (33.1) 21 (60.0) 32 (42.1) 41 (36.3)
Liver
Liver inammation (cT1 in ms) (n=1 missing) (n=1 missing) (n=1 missing)
Normal <784 ms 177 (88.5) 36 (100) 0.030 148 (91.4) 28 (75.7) 0.018 69 (90.8) 101 (87.1) 0.494
Impaired ≥784 ms 23 (11.5) 0 (0) 14 (8.6) 9 (24.3) 7 (9.2) 15 (12.9)
Liver fat
Normal <4.8% 159 (79.1) 34 (94.4) 0.034 134 (82.2) 24 (64.9) 0.026 61 (79.2) 91 (78.4) 1
Impaired ≥4.8% 42 (20.9) 2 (5.4) 29 (17.8) 13 (35.1) 16 (20.8) 25 (21.6)
Liver volume (n=1 missing)
Continued
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
7
DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
1). Impairment was present in the heart in 26% (myocar-
ditis 19%, systolic dysfunction 9%), lung in 11% (reduced
vital capacity), kidney in 4% (inflammation), liver in 28%
(12% inflammation, 21% ectopic fat, 10% hepatomegaly),
pancreas in 40% (15% inflammation, 38% ectopic fat)
and spleen in 4% (splenomegaly) (figure 2 and table 2).
Of the individuals, 70% had impairment in at least one
organ and 29% had multiorgan impairment, with overlap
across multiple organs (figure 3). Impairment in the liver,
heart or lungs was associated with further organ impair-
ment in 63%, 62% and 48% of individuals, respectively
(figure 3).
Symptoms and multiorgan impairment
Hepatic and pulmonary impairment frequently clustered
together, with fatigue, muscle aches, fever and cough
commonly reported. Impairment in particular organs
was associated with particular symptoms—pancreas: diar-
rhoea, fever, headache and dyspnoea; heart: headache,
dyspnoea and fatigue; and kidney: wheezing, runny nose,
diarrhoea, cough, fever, headache, dyspnoea and fatigue
(figure 4).
Hospitalisation, severity and multiorgan impairment
The hospitalised group were older (p=0.001), had higher
BMI (p=0.063), and were more likely to be non- white
(p=0.016) and to report ‘inability to walk’ (p=0.009)
than non- hospitalised individuals. There were no other
statistically significant differences between risk factors
or symptoms between the groups. Impairment of the
liver, pancreas (eg, ectopic fat in the pancreas and liver,
hepatomegaly) and 2 organs was higher in hospitalised
individuals (all p<0.05) (figure 3 and table 2). In multi-
variate analyses, adjusting for age, sex and BMI, liver
volume remained significantly associated with hospi-
talisation (p=0.001). Hospitalised individuals had high
triglycerides (30% vs 7.2%, p=0.002), cholesterol (60%
vs 38%, p=0.04) and low- density lipoprotein- cholesterol
(57% vs 31%, p=0.01), and low transferrin saturation
(38% vs 15%, p=0.01), compared with non- hospitalised
individuals. erythrocyte sedimentation rate (ESR) (13%),
bicarbonate (12%), uric acid (16%), platelet count (13%)
and high- sensitivity C- reactive protein (CRP) (15%) were
high in 10% of hospitalised individuals.
Of the individuals, 60% (n=120) had severe PCS, with
52% reporting persistent, moderate problems under-
taking usual activities (level 3 or greater in the relevant
EQ- 5D- 5L question; 34% reported Dyspnoea-12 score
10). Of those with severe PCS, 84% were not hospitalised
and 73% were female. There were no differences in age,
BMI or ethnicity between the groups. Individuals with
severe PCS were more likely to report shortness of breath
(p<0.001), headache (p=0.019), chest pain (p=0.001),
abdominal pain (p=0.001) and wheezing (p=0.039). Of
those with ‘severe’ PCS, 25% had myocarditis compared
with 12% with moderate PCS (unadjusted: 0.023; adjust-
ment for age, sex and BMI: p=0.04; online supplemental
figure 2). Severe PCS was associated with higher mean
Measurement
All patients
(N=201)
Healthy
controls (n=36)
P
value
Not hospitalised
(n=163)
Hospitalised
(n=37) P value
Moderate
PCS (n=77)
Severe PCS
(n=116)
P
value
Normal <1935 mL 180 (89.6) 34 (97.1) 0.214 154 (94.5) 25 (67.6) <0.0001 68 (88.3) 104 (89.7) 0.816
Impaired ≥1935 mL 21 (10.4) 1 (2.9) 9 (5.5) 12 (32.4) 9 (11.7) 12 (10.3)
Spleen
Splenic volume (mL) (n=1 missing)
Normal <350 mL 194 (96.5) 32 (91.4) 0.172 160 (98.2) 33 (89.2) 0.023 74 (96.1) 112 (96.6) 1
Impaired ≥350 mL 7 (3.5) 3 (8.6) 3 (1.8) 4 (10.8) 3 (3.9) 4 (3.4)
Data are presented as count (%).
Comparisons between managed at home versus hospitalised and between moderate versus PCS were conducted using Fisher’s exact test.
PCS, post- COVID-19 syndrome.
Table 2 Continued
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
8DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
cell haemoglobin concentration (28% vs 17%), choles-
terol (46.2% vs 32.8%), CRP (10% vs 3.8%) and ESR
(10% vs 6%) than moderate PCS, but these differences
were not statistically significant (online supplemental
table 3). Muscle aches, fever and coughing were common
in severe PCS, and headache was common in individuals
with inflammation of the pancreas (figure 4).
DISCUSSION
We report three findings in the first COVID-19 recovery
study to evaluate medium- term, multiorgan impairment.
First, in low- risk individuals, there were chronic symptoms
and mild impairment in the heart, lung, liver, kidney
and pancreas 4 months post- COVID-19, compared with
healthy controls. Second, cardiac impairment was more
common in severe PCS. Third, we demonstrate feasibility
and potential utility of community- based multiorgan
assessment for PCS.
Comparison with other studies
Common symptoms were fatigue, dyspnoea, myalgia,
headache and arthralgia, despite low risk of COVID-19
mortality or hospitalisation. COVID-19 impact models
have included age, underlying conditions and mortality,
but not morbidity, multiorgan impairment and chronic
diseases.29 30 Even in non- hospitalised individuals, up
to 10% of those infected have PCS,15 31 but studies of
extrapulmonary manifestations emphasise acute illness.32
We describe mild rather than severe organ impairment,
but the pandemic’s scale and high infection rates in lower
risk individuals signal medium- term and longer- term
COVID-19 impact, which cannot be ignored in health-
care or policy spheres.
Acute myocarditis and cardiogenic shock33 are docu-
mented in hospitalised patients with COVID-19.6 In
American athletes, recent COVID-19 was associated with
myocarditis.34 Although causality cannot be attributed
and postviral syndromes have included similar find-
ings,21 we show that a quarter of low- risk individuals with
PCS have mild systolic dysfunction or myocarditis. The
significance of these findings and the associations with
contemporaneous abnormal echocardiography findings
and long- term myocardial fibrosis and impairment are
unknown. Cardiac impairment, a risk factor for severe
COVID-19, may have a role in PCS. Two further findings
that deserve investigation are pancreatic abnormalities,
given the excess diabetes risk reported in PCS,15 and the
preponderance of healthcare workers at increased PCS
risk (as observed for COVID-19 mortality), possibly due
to higher viral burden.
PCS is likely to be a syndrome rather than a single
condition. Despite an immunological basis for individual
variations in COVID-19 progression and severity,35 predic-
tion models have high rates of bias, perform poorly,36 and
focus on respiratory dysfunction and decisions for venti-
lation in acutely unwell patients, rather than multiorgan
function. Ongoing long- term studies37 exclude non-
hospitalised, low- risk individuals. During a pandemic, we
studied subclinical organ impairment in PCS, showing
low rates of incidental findings. As specialist PCS services
are rolled out,38 39 multiorgan assessment, monitoring
and community pathways have potential roles during and
beyond COVID-19, but need to be evaluated.
Implications for research, clinical practice and public health
Our findings have three research implications. First, as
countries face second waves, COVID-19 impact models
Figure 3 Multiorgan impairment in low- risk individuals with post- COVID-19 syndrome by gender and hospitalisation.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
9
DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
should include PCS, whether quality of life, healthcare util-
isation or economic effects. Second, there is urgent need
for multiorgan assessment, including blood and imaging,
as well as primary and secondary care data linkage, to
define PCS. Third, longitudinal studies of clustering of
symptoms and organ impairment will inform health
services research to plan multidisciplinary care path-
ways. There are three management implications. First,
we signal the need for multiorgan monitoring in at least
the medium term, especially extrapulmonary sequelae.
Care pathways involving MRI (with limited access in
many clinical settings) need evaluation versus other
modalities to detect organ impairment (eg, spirometry,
N- terminal pro B- type natriuretic peptide (NT- pro- BNP),
ECG, echocardiography, ultrasound and blood investi-
gations). Second, until effective vaccines and treatments
are widely available, ‘infection suppression’ (eg, social
distancing, masks, physical isolation) is the prevention
strategy. Third, whether understanding baseline risk or
multiorgan complications, PCS requires management
across specialties (eg, cardiology, gastroenterology) and
disciplines (eg, epidemiology, diagnostics, laboratory
science) (figure 5).
Limitations
There are some limitations. First, our cardiac MRI
protocol excluded gadolinium contrast due to concerns
regarding COVID-19- related renal complications, relying
on native T1 mapping to characterise myocardial inflam-
mation non- invasively (previously validated for acute
myocarditis).40 Second, for organ impairment, we show
association, not causation, and incidental findings are
possible in asymptomatic individuals41; however, our
findings are strengthened by comparison with healthy,
age- matched controls, although not matched for sex or
baseline comorbidities. Third, for pragmatic reasons, our
controls were scanned using 1.5T, but we used 3T ranges
as described in an analogous study with similar acquisi-
tion protocols. Therefore, we may be under- representing
the true proportion of impairment in those individuals
with PCS scanned at 3T. Fourth, further studies may
explore different controls, for example, individuals with
Figure 4 Percentage of reported symptoms during the acute phases of the illness within those with evidence of organ
impairment for each organ separately. Darker red indicates higher percentage of reported symptoms per impaired organ.
There are no distinct patterns of symptoms relating to each impaired organ, but a high burden of symptoms in individuals is
highlighted.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
10 DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
postinfluenza symptoms, COVID-19 without symptoms or
from general clinics. We will investigate duration, trajec-
tory, complications and recovery for specific symptoms
and organ impairment in the follow- up phase. Fifth,
our study population was not ethnically diverse, despite
disproportionate COVID-19 impact in non- white individ-
uals. Sixth, to limit interaction and exposure between the
trial team and the patients, pulse oximetry, spirometry,
MRI assessment of the brain and muscle function were
not included from the outset.
CONCLUSIONS
Our study suggests PCS has a physiological basis, with
measurable patient- reported outcomes and organ
impairment. Future research should address longer- term
follow- up of organ function beyond symptoms and blood
investigations, even in lower risk individuals; prioritisa-
tion for imaging, investigation and referral; and optimal
care pathways. Health system responses should emphasise
infection suppression and management of pre- COVID-19
and post- COVID-19 risk factors and chronic diseases.
Author afliations
1Perspectum, Oxford, UK
2Department of Cardiology, Great Western Hospital Foundation NHS Trust, Swindon,
UK
3Department of Cardiology, Oxford University Hospitals NHS Foundation Trust,
Oxford, UK
4Alliance Medical, Warwick, UK
5Department of Gastroenterology, Guy's and St Thomas' NHS Foundation Trust,
London, UK
6Institute for Liver and Digestive Health, University College London, London, UK
7Institute of Cardiovascular and Metabolic Medicine, University of Liverpool,
Liverpool, UK
8Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
9Department of Respiratory Research, Liverpool University Hospitals NHS
Foundation Trust, Liverpool, UK
10Department of Respiratory Medicine, Hull and East Yorkshire Hospitals NHS Trust,
Hull, UK
11Institute of Clinical and Applied Health Research, University of Hull, Hull, UK
12Institute of Population Health Sciences, University of Liverpool, Liverpool, UK
13Department of Oncology, University of Oxford, Oxford, UK
14Long COVID SOS, Oxford, UK
15UKDoctors#Longcovid, London, UK
16Department of Medicine, University College London Hospitals NHS Foundation
Trust, London, UK
17Institute of Health Informatics, University College London, London, UK
18Department of Cardiology, Barts Health NHS Trust, London, UK
Twitter Amitava Banerjee @amibanerjee1
Contributors Study design: AD, RB, JA, COVERSCAN team. Patient recruitment: RB,
COVERSCAN team. Data collection: MW, COVERSCAN team. Data analysis: AD, AB,
COVERSCAN team. Data interpretation: AB, AD, MW, RB. Initial manuscript drafting:
AB, AD, RB. Critical review of early and nal versions of the manuscript: all authors
including JO and DJC. Specialist input: MW, AB (cardiology); RB, MH, DW, MC, DJC
(general medicine); MH, MC, DW (long COVID-19); MB, RB (imaging); AD (statistics);
AB (epidemiology/public health); MG (primary care); JA (healthcare management);
LH, EA (patient and public involvement).
Funding This work was supported by the UK’s National Consortium of Intelligent
Medical Imaging (Industry Strategy Challenge Fund), Innovate UK (Grant 104688)
and the European Union’s Horizon 2020 research and innovation programme
(agreement no 719445). The research was designed, conducted, analysed and
interpreted by the authors independently of the funding sources.
Competing interests AD, RB and MB are employees of Perspectum.
Patient consent for publication Not required.
Ethics approval The study has received ethical approval (20/SC/0185).
Provenance and peer review Not commissioned; externally peer reviewed.
Data availability statement Data are available upon reasonable request from the
corresponding author.
Supplemental material This content has been supplied by the author(s). It has
not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been
peer- reviewed. Any opinions or recommendations discussed are solely those
Figure 5 Natural history of post- COVID-19 syndrome, the COVERSCAN study in low- risk individuals (N=201) and policy
recommendations.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
11
DennisA, etal. BMJ Open 2021;11:e048391. doi:10.1136/bmjopen-2020-048391
Open access
of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and
responsibility arising from any reliance placed on the content. Where the content
includes any translated material, BMJ does not warrant the accuracy and reliability
of the translations (including but not limited to local regulations, clinical guidelines,
terminology, drug names and drug dosages), and is not responsible for any error
and/or omissions arising from translation and adaptation or otherwise.
Open access This is an open access article distributed in accordance with the
Creative Commons Attribution 4.0 Unported (CC BY 4.0) license, which permits
others to copy, redistribute, remix, transform and build upon this work for any
purpose, provided the original work is properly cited, a link to the licence is given,
and indication of whether changes were made. See:https:// creativecommons. org/
licenses/ by/ 4. 0/.
ORCID iD
AmitavaBanerjee http:// orcid. org/ 0000- 0001- 8741- 3411
REFERENCES
1 World Health Organization. Clinical management of severe acute
respiratory infection (SARI) when COVID-19 disease is suspected.
interim guidance 13 March 2020, 2020. Available: https:// apps. who.
int/ iris/ handle/ 10665/ 331446
2 Pavon AG, Meier D, Samim D, etal. First documentation of persistent
SARS- Cov-2 infection presenting with late acute severe myocarditis.
Can J Cardiol 2020;36:1326.e5–1326.e7.
3 Puntmann VO, Carerj ML, Wieters I. Outcomes of cardiovascular
magnetic resonance imaging in patients recently recovered
from coronavirus disease 2019 (COVID-19). JAMA Cardiol
2019;2020:1265–73.
4 Tabary M, Khanmohammadi S, Araghi F, etal. Pathologic features of
COVID-19: a Concise review. Pathol Res Pract 2020;216:153097.
5 Alqahtani SA, Schattenberg JM. Liver injury in COVID-19: the current
evidence. United European Gastroenterol J 2020;8:509–19.
6 Somasundaram NP, Ranathunga I, Ratnasamy V, etal. The impact of
SARS- Cov-2 virus infection on the endocrine system. J Endocr Soc
2020;4:1–22.
7 Farouk SS, Fiaccadori E, Cravedi P, etal. COVID-19 and the kidney:
what we think we know so far and what we don't. J Nephrol
2020;33:1213–8.
8 Lai A, Pasea L, Banerjee A. Estimating excess mortality in people
with cancer and multimorbidity in the COVID-19 emergency. BMJ
Open 2020;10:e043828.
9 Banerjee A, Pasea L, Harris S, etal. Estimating excess 1- year
mortality associated with the COVID-19 pandemic according to
underlying conditions and age: a population- based cohort study. The
Lancet 2020;395:1715–25.
10 Banerjee A, Chen S, Pasea L. Excess deaths in people with
cardiovascular diseases during the COVID-19 pandemic. Eur J Prev
Cardiol 2020.
11 Raman B, Cassar MP, Tunnicliffe EM, etal. Medium- Term effects of
SARS- CoV-2 infection on multiple vital organs, exercise capacity,
cognition, quality of life and mental health, post- hospital discharge.
EClinicalMedicine 2021;31:100683.
12 Horton R. Ofine: COVID-19 is not a pandemic. The Lancet
2020;396:874.
13 Shovlin CL, Vizcaychipi MP. Implications for COVID-19 triage from
the ICNARC report of 2204 COVID-19 cases managed in UK adult
intensive care units. Emerg Med J 2020;37:332–3.
14 Docherty AB, Harrison EM, Green CA, etal. Features of 20 133 UK
patients in hospital with covid-19 using the ISARIC WHO Clinical
Characterisation Protocol: prospective observational cohort study.
BMJ 2020;369:m1985–12.
15 Williamson EJ, Walker AJ, Bhaskaran K, etal. Factors associated
with COVID-19- related death using OpenSAFELY. Nature
2020;584:430–6.
16 Ofce for National Statistics. The prevalence of long COVID
symptoms and COVID-19 complications, 2020. Available: https://
www. ons. gov. uk/ news/ statementsandletters/ thep reva lenc eo ongc
ovid symp toms andc ovid 19co mpli cations
17 del Rio C, Collins LF, Malani P. Long- Term health consequences of
COVID-19. JAMA 2020;324:1723.
18 Carfì A, Bernabei R, Landi F, etal. Persistent symptoms in patients
after acute COVID-19. JAMA 2020;324:603–5.
19 Nabavi N. Long covid: how to dene it and how to manage it. BMJ
2020;370:m3489.
20 Greenhalgh T, Knight M, A’Court C, etal. Management of post- acute
covid-19 in primary care. BMJ 2020;13:m3026.
21 National Institute for Health Research. New risk prediction model
could help improve guidance for people shielding from COVID-19,
2020. Available: https://www. nihr. ac. uk/ news/ new- risk- prediction-
model- could- help- improve- guidance- for- people- shielding- from-
covid- 19/ 25096
22 Hill AD, Fowler RA, Pinto R, etal. Long- term outcomes and
healthcare utilization following critical illness – a population- based
study. Crit Care 2016;20:1–10.
23 Perrin R, Riste L, Hann M, etal. Into the looking glass: post- viral
syndrome post COVID-19. Med Hypotheses 2020;144:110055.
24 George PM, Barratt SL, Condliffe R, etal. Respiratory follow- up of
patients with COVID-19 pneumonia. Thorax 2020;75:1009–16.
25 Jiang DH, McCoy RG. Planning for the Post- COVID syndrome: how
payers can mitigate long- term complications of the pandemic. J Gen
Intern Med 2020;35:3036–9.
26 Janssen MF, Pickard AS, Golicki D, etal. Measurement properties
of the EQ- 5D- 5L compared to the EQ- 5D- 3L across eight patient
groups: a multi- country study. Qual Life Res 2013;22:1717–27.
27 Yorke J, Moosavi SH, Shuldham C, etal. Quantication of
dyspnoea using descriptors: development and initial testing of the
Dyspnoea-12. Thorax 2010;65:21–6.
28 Hobbins A, Barry L, Kelleher D, etal. The health of the residents
of ireland: population norms for Ireland based on the EQ- 5D- 5L
descriptive system – a cross sectional study. HRB Open Res
2018;1:22.
29 Gupta A, Madhavan MV, Sehgal K, etal. Extrapulmonary
manifestations of COVID-19. Nat Med 2020;26:1017–32.
30 Palmer K, Monaco A, Kivipelto M, etal. The potential long-
term impact of the COVID-19 outbreak on patients with non-
communicable diseases in Europe: consequences for healthy ageing.
Aging Clin Exp Res 2020;32:1189–94.
31 Menni C, Valdes AM, Freidin MB, etal. Real- Time tracking of
self- reported symptoms to predict potential COVID-19. Nat Med
2020;26:1037–40.
32 Mandal S, Barnett J, Brill S, etal. “Long- COVID”: a cross-
sectional study of persisting symptoms, biomarker and imaging
abnormalities following hospitalisation for COVID-19. Thorax
2020:thoraxjnl-2020-215818.
33 Chau VQ, Giustino G, Mahmood K, etal. Cardiogenic shock and
hyperinammatory syndrome in young males with COVID-19. Circ
Hear Fail 2020:556–9.
34 Rajpal S, Tong MS, Borchers J, etal. Cardiovascular magnetic
resonance ndings in competitive athletes recovering from COVID-19
infection. JAMA Cardiol 2020:5–7.
35 Mathew D, Giles JR, Baxter AE, etal. Deep immune proling of
COVID-19 patients reveals distinct immunotypes with therapeutic
implications. Science 2020;369:eabc8511.
36 Wynants L, Van Calster B, Collins GS, etal. Prediction models for
diagnosis and prognosis of covid-19: systematic review and critical
appraisal. BMJ 2020;369:m1328.
37 PHOSP- COVID. Post- HOSPitalisation COVID-19 study, 2020.
Available: https://www. phosp. org/
38 NHS. NHS to offer ‘long covid’ sufferers help at specialist centres,
2020. Available: https://www. england. nhs. uk/ 2020/ 10/ nhs- to- offer-
long- covid- help/
39 National Institute for Healh and Care Excellence. COVID-19 rapid
guideline: managing the long- term effects of COVID-19, 2020.
Available: https://www. nice. org. uk/ guidance/ ng188
40 Ferreira VM, Piechnik SK, Dall’Armellina E, etal. Native T1- mapping
detects the location, extent and patterns of acute myocarditis
without the need for gadolinium contrast agents. J Cardiovasc Magn
Reson 2014;16:1–11.
41 Gibson LM, Paul L, Chappell FM, etal. Potentially serious incidental
ndings on brain and body magnetic resonance imaging of
apparently asymptomatic adults: systematic review and meta-
analysis. BMJ 2018;14:k4577.
copyright. on January 4, 2022 at India:BMJ-PG Sponsored. Protected byhttp://bmjopen.bmj.com/BMJ Open: first published as 10.1136/bmjopen-2020-048391 on 30 March 2021. Downloaded from
... 20 Current knowledge demonstrates that Long COVID can affect multiple body systems including the respiratory, cardiac, renal, endocrine and neurological systems. 15,16,19,26,28,[35][36][37][38] People present with clusters of overlapping symptoms such as fatigue or exhaustion, chest pressure or tightness, shortness of breath, headache, and cognitive dysfunction. 16,38 Long COVID can be multi-dimensional, spanning symptoms and impairments, activity limitations and social participation restrictions. ...
... The most common symptom of Long COVID is fatigue or exhaustion, 6,[16][17][18][19]28,34,77-84 a symptom which does not result from unusually difficult activity, is not easily relieved by rest or sleep, can limit functioning in day-to-day activities, and negatively impact quality of life. 85 People living with Long COVID can additionally experience post-exertional symptom exacerbation, 16 also described as postexertional malaise (often abbreviated to PEM) or post-exertional neuroimmune exhaustion. ...
... People with Long COVID can have impairments of multiple body systems including the respiratory, cardiac, renal, endocrine, and neurological systems. 15,16,19,28,36,38 Cardiac injury has been reported among people recovering from COVID-19, [137][138][139] and data from serial multi-organ MRI scans on 201 middle-aged, generally healthy individuals with Long COVID suggested evidence of mild cardiac impairment (32%). 28 COVID-19 may cause myocarditis and pericarditis. ...
Technical Report
Full-text available
In February 2021 World Physiotherapy collaborated with Long COVID Physio to develop a briefing paper on safe rehabilitation for people living with Long COVID. The purpose was to gather key opinion leaders and stakeholders from the global community in Long COVID and physiotherapy. This briefing paper brings together individuals from across the World Physiotherapy regions, community groups, organisations, interdisciplinary clinical practice, and academia to identify statements on safe rehabilitation approaches for people living with Long COVID. This paper has been produced with the helpful contributions from the following: Darren Brown, Caroline Appel, Bruno Baldi, Janet Prvu Bettger, Michelle Bull, Tracy Bury, Jefferson Cardoso, Nicola Clague-Baker, Geoff Bostick, Robert Copeland, Nnenna Chigbo, Caroline Dalton, Todd Davenport, Hannah Davis, Simon Decary, Brendan Delaney, Jessica DeMars, Sally Fowler-Davis, Michael Gabilo, Douglas Gross, Mark Hall, Jo House, Liam Humphreys, Linn Järte, Leonard Jason, Asad Khan, Ian Lahart, Kaba Dalla Lana, Amali Lokugamage, Ariane Mangar, Rebecca Martin, Joseph McVeigh, Maxi Miciak, Rachael Moses, Etienne Ngeh Ngeh, Kelly O’Brien, Shane Patman, Sue Pemberton, Sabrina Poirer, Milo Puhan, Clare Rayner, Alison Sbrana, Jaime Seltzer, Jenny Sethchell, Ondine Sherwood, Ema Singwood, Amy Small, Jake Suett, Laura Tabacof, Catherine Thomson, Jenna Tosto-Mancuso, Rosie Twomey, Marguerite Wieler, Jamie Wood.
... However, little is known of the extent to which tissue damage persists and contributes to long-term symptoms. Publications have reported multi-organ abnormalities in the short to medium term post-COVID-19 compared to controls (Dennis et al., 2021;Raman et al., 2021). ...
... However, this is unlikely to explain the greater perception of fatigue, given that the values were not clinically abnormal (Petersen et al., 2017) for the majority of patients. Other studies reporting deficits in muscle strength, evidence of myocardial inflammation and regional GM volume loss in patients recovering after a severe SARS-CoV2 infection, were mainly conducted 3-4 months after the acute infection (Dennis et al., 2021;Raman et al., 2021;Tanrıverdi et al., 2021). ...
Article
Full-text available
We evaluated the impacts of COVID‐19 on multi‐organ and metabolic function in patients following severe hospitalised infection compared to controls. Patients (n = 21) without previous diabetes, cardiovascular or cerebrovascular disease were recruited 5–7 months post‐discharge alongside controls (n = 10) with similar age, sex and body mass. Perceived fatigue was estimated (Fatigue Severity Scale) and the following were conducted: oral glucose tolerance (OGTT) alongside whole‐body fuel oxidation, validated magnetic resonance imaging and spectroscopy during resting and supine controlled exercise, dual‐energy X‐ray absorptiometry, short physical performance battery (SPPB), intra‐muscular electromyography, quadriceps strength and fatigability, and daily step‐count. There was a greater insulin response (incremental area under the curve, median (inter‐quartile range)) during the OGTT in patients [18,289 (12,497–27,448) mIU/min/L] versus controls [8655 (7948–11,040) mIU/min/L], P < 0.001. Blood glucose response and fasting and post‐prandial fuel oxidation rates were not different. This greater insulin resistance was not explained by differences in systemic inflammation or whole‐body/regional adiposity, but step‐count (P = 0.07) and SPPB scores (P = 0.004) were lower in patients. Liver volume was 28% greater in patients than controls, and fat fraction adjusted liver T1, a measure of inflammation, was raised in patients. Patients displayed greater perceived fatigue scores, though leg muscle volume, strength, force‐loss, motor unit properties and post‐exercise muscle phosphocreatine resynthesis were comparable. Further, cardiac and cerebral architecture and function (at rest and on exercise) were not different. In this cross‐sectional study, individuals without known previous morbidity who survived severe COVID‐19 exhibited greater insulin resistance, pointing to a need for physical function intervention in recovery.
... The concept of post-COVID-19 or long COVID syndrome emerged in May 2020 with initial reports of continued or developing symptoms beyond 12 weeks following acute SARS-CoV-2 infection [3,4]. Individuals with long COVID present with symptoms involving multiple organ systems, often manifesting as a cluster of symptoms, with fatigue, headache, and dyspnea being commonly reported [5,6]. ...
... Patients were classified into symptom clusters on the basis of the number of involved organs: Class I (mild) with symptoms in three organs, Class II (moderate) with symptoms in four to five organs, and Class III (severe) with symptoms in six or more organs. The clustering method was based on our previous observations, the literature, and the specifications outlined in the international Delphi consensus [1,3,6,[15][16][17]. Our classification notably aligns with the definitions provided by Ayoubkhani et al., who quantified individual symptoms in a large prospective out-of-hospital study and categorized them into three groups, similar to ours, on the basis of quantitative observations [11]. ...
Article
Full-text available
Patients with long COVID syndrome present with various symptoms affecting multiple organs. Vaccination before or after SARS-CoV-2 infection appears to reduce the incidence of long COVID or at least limit symptom deterioration. However, the impact of vaccination on the severity and extent of multi-organ long COVID symptoms and the relationship between the circulating anti-spike protein antibody levels and the severity and extent of multi-organ symptoms are unclear. This prospective cohort study included 198 patients with previous PCR-verified SARS-CoV-2 infection who met the criteria for long COVID syndrome. Patients were divided into vaccinated (n = 138, 69.7%) or unvaccinated (n = 60, 30.3%) groups. Anti-spike protein antibody levels were determined at initial clinical presentation and compared between the groups. Long COVID symptoms were quantified on the basis of the number of affected organs: Class I (mild) with symptoms in three organs, Class II (moderate) with symptoms in four to five organs, and Class III (severe) with symptoms in six or more organ systems. Associations between time to infection and vaccination with anti-spike protein antibody levels were assessed. The anti-spike protein antibody levels were 1925 ± 938 vs. 481 ± 768 BAU/mL (p < 0.001) in the vaccinated vs. unvaccinated patients. The circulating anti-spike antibody cutoff of 665.5 BAU/mL allowed us to differentiate the vaccinated from the unvaccinated patients. Vaccinated patients had fewer class II and class III multi-organ symptoms (Class II 39.9% vs. 45.0%; Class III 10.1% vs. 23.3%, p-value 0.014). Anti-spike antibody level correlated negatively with multi-organ symptom classes (p = 0.016; 95% CI −1.229 to −0.126). Anti-spike antibody levels in unvaccinated patients declined markedly with time, in contrast to the persistence of high anti-spike antibody levels in the vaccinated patients. Multi-organ symptoms were lower in vaccinated long-COVID patients, especially in those with higher anti-spike antibody levels (≥665.5 BAU/mL). Classifying the symptoms on the basis of the number of affected organs enables a more objective symptom quantification.
... Na caracterização sociodemográfica, a literatura aponta que o sexo feminino apresenta associação significativa (p<0,05) com a persistência de sintomas, (13)(14) o que não foi encontrado nesta pesquisa. Os Portuguese | Rev. enferm. ...
Article
Full-text available
Objetivo: Analisar os fatores relacionados à COVID longa na população adulta do Brasil. Métodos: Estudo transversal analítico, do tipo web-survey, com abordagem quantitativa. A amostragem foi não probabilística, do tipo intencional, e incluiu 228 adultos brasileiros que testaram positivo para COVID-19. A coleta de dados ocorreu por meio de questionário online. Para verificar a associação entre variáveis qualitativas, utilizou-se o Teste Qui-quadrado ou Teste Exato de Fisher e, nas quantitativas, aplicou-se o Teste de Mann-Whitney com significância de 0,05. Resultados: Constatou-se que houve associação de variáveis sociodemográficas com a COVID longa. Observa-se maior média de idade entre aqueles em que persistiram os sintomas (p=0,041). A renda familiar daqueles com sintomas persistentes era inferior à dos indivíduos sem persistência (p=0,005). A prática de atividade física esteve associada a não persistência dos sintomas (p=0,024). A hipertensão arterial foi a comorbidade mais prevalente naqueles com sintomas persistentes (5,9%). No quadro clínico de COVID-19 associado à persistência dos sintomas, identificam-se calafrios (p-valor=0,009), cefaleia (p-valor=0,0027), tosse (p-valor=0,000), anosmia (p-valor=0,048), ageusia (p-valor=0,013), dispneia (p-valor=0,000) e diarreia (p-valor=0,018). O sintoma de COVID longa mais prevalente foi fadiga (62,89%). Conclusão: Idade e renda estiveram associadas à COVID longa. Praticar atividade física esteve associado a não persistência de sintomas.
... Moreover, being hospitalized with COVID-19 increases the risk of subsequently suffering from cardiovascular, kidney or lung diseases, or of developing diabetes [1][2][3]. As most patients experience initial pulmonary symptoms, SARS-CoV-2 was initially considered primarily a respiratory disease; however, acute and chronic (or long) COVID-19 also affects many other organs, for example, producing cardiac dysfunction, dysautonomia and cognitive dysfunction [4][5][6][7]. The route by which COVID-19 impacts different organs is increasingly appreciated to be via the vascular system: SARS-CoV-2 infection of vascular cells produces thrombosis, endothelial dysfunction and degeneration, as well as dysregulated angiogenesis [8][9][10]. ...
Article
Full-text available
Coronavirus disease 2019 (COVID-19) was initially considered a primarily respiratory disease but is now known to affect other organs including the heart and brain. A major route by which COVID-19 impacts different organs is via the vascular system. We studied the impact of apolipoprotein E (APOE) genotype and inflammation on vascular infectivity by pseudo-typed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses in mouse and human cultured endothelial cells and pericytes. Possessing the APOE4 allele or having existing systemic inflammation is known to enhance the severity of COVID-19. Using targeted replacement human APOE3 and APOE4 mice and inflammation induced by bacterial lipopolysaccharide (LPS), we investigated infection by SARS-CoV-2. Here, we show that infectivity was higher in murine cerebrovascular pericytes compared to endothelial cells and higher in cultures expressing APOE4. Furthermore, increasing the inflammatory state of the cells by prior incubation with LPS increased infectivity into human and mouse pericytes and human endothelial cells. Our findings provide insights into the mechanisms underlying severe COVID-19 infection, highlighting how risk factors such as APOE4 genotype and prior inflammation may exacerbate disease severity by augmenting the virus’s ability to infect vascular cells.
... However, the pathological mechanism in the post-COVID phase remains poorly understood. Common to all studies is a slight increase in the percentage of patients with cardiopulmonary symptoms in the first 3 months compared to later stages around 6-9 months [30][31][32][33][34]. Although most PASC cases with cardiopulmonary symptoms do not reveal significant pathological findings requiring intervention, active follow-up, investi-gation, and diagnostic refinement of these patients are highly recommended. ...
Article
Full-text available
Post-acute sequelae of SARS-CoV-2 (PASC) is a significant health concern, particularly for patients with chronic kidney disease (CKD). This study investigates the long-term outcomes of individuals with CKD who were infected with COVID-19, focusing on their health status over a three-year period post-infection. Data were collected from both CKD and non-CKD patients who survived SARS-CoV-2 infection and were followed for three years as part of a research study on the impact, prognosis, and consequences of COVID-19 infection in CKD patients. In this prospective cohort study, we analyzed clinical records, laboratory findings, and patient-reported outcomes assessed at intervals during follow-up. The results indicated no permanent changes in renal function in any of the groups analyzed, although patients without CKD exhibited faster recovery over time. Furthermore, we examined the effect of RAAS-blocker therapy over time, finding no influence on PASC symptoms or renal function recovery. Regarding PASC symptoms, most patients recovered within a short period, but some required prolonged follow-up and specialized post-recovery management. Following up with patients in the post-COVID-19 period is crucial, as there is still insufficient information and evidence regarding the long-term effects, particularly in relation to CKD.
... Such a capillary is thus completely disabled. This impedes blood flow and, consequently, the function of the tissues and organs, including the haematopoietic organs [6]. ...
Preprint
Full-text available
My hypothesis is that the signs and symptoms of Long COVID can be explained by a shortage of blood in the body and a resulting deficient blood flow through nearly all organs. This shortage arises through damage to the blood-producing organs during the acute phase, while the breakdown of blood continues as normal, after an initial increase. In order to ensure the perfusion of organs that are directly necessary for survival, the body takes the emergency measure of diverting blood from other organs and tissues. The perfusion of the blood-producing organs is also affected by this distribution measure, which hinders the smooth recovery of the total blood volume. The body is stuck in this vicious circle: a shortage of circulating blood hinders the recovery of blood production. This explains the long duration of Long COVID. My proposed treatment of Long COVID focuses on the recovery of the correct volume of blood in the body of the right composition by the very careful administration of donor blood products under continuous expert supervision. A trial treatment can be performed in any hospital without much additional preparations, and has a lower associated risk for the patient than analysing the total blood. A diagnosis ex juvantibus, by therapeutic response, is therefore preferable, and will result in the healing process starting earlier. Indications in blood laboratory values of a shortage of blood are a high serum ferritin due to internal breakdown of blood and values for haematocrit and albumin at reciprocal extremes of the reference ranges due to a stagnation of blood production.
... In long COVID patients, these anastomoses are for a large part closed, even in rest, in order to let the small amount of blood flow through the capillary nets of the lungs. 4 A seriously ill Long COVID patient cannot afford even minimal exertion (3) because the total amount of blood is quickly insufficient to maintain an acceptable level of central circulation. Additionally, there is not enough blood available to clear the metabolic waste products after exertion. ...
Preprint
Full-text available
The signs and symptoms of Long COVID can be explained by a shortage of blood in the body and a resulting deficient blood flow through nearly all organs. This shortage arose during the acute phase of COVID19 by an increased breakdown of haematocytes, to which the liver responds by reducing the production of albumin, in order to prevent a too large decrease in haematocrit.In order to ensure the perfusion of organs that are directly necessary for survival, the body takes the emergency measure of diverting blood from other organs and tissues. The perfusion of the blood-producing organs is also affected by this distribution measure, which hinders the smooth recovery of the total blood volume. The body is stuck in this vicious circle: a shortage of circulating blood hinders the recovery of blood production. This explains the long duration of Long COVID. My proposed treatment of Long COVID focuses on the recovery of the correct volume of blood in the body of the right composition by the intravenous administration of donor blood products, starting with albumin concentrate. A trial treatment can be performed in any hospital without much additional preparations, and has a lower associated risk for the patient than analysing the total blood. A diagnosis ex juvantibus, by therapeutic response, is therefore preferable. I suspect that most Long COVID patients will exhibit a high serum ferritin level as a result of internal haemolysis, and haematocrit and albumin values at the high and low extremes of the reference range because the liver can not keep up with the recovery of the red bone marrow.
... Longer follow-up studies have demonstrated that the most frequent Long COVID-19 symptoms are fatigue (78%), post-exertional malaise (72%), and cognitive dysfunction (55%) [5]. Debilitating sequels with organ damage involving the pulmonary, cardiovascular, musculoskeletal and autonomic nervous systems have also been reported, though in much lower incidence [7]. This constellation of symptoms may overlap with those experienced by patients suffering from fibromyalgia (FM) and chronic fatigue syndrome (CFS) [8,9]. ...
Article
Full-text available
Background: The COVID-19 (Coronavirus disease 2019) pandemic has prompted extensive research into lingering effects, especially in ‘Long COVID’ patients. Despite exploration, contributing factors remain elusive; Objective: This study explores the potential link between distinctive personality profiles, particularly type D personality, and an increased risk of Long COVID; Methods: A retrospective cross-sectional study at Tel-Aviv Sourasky Medical Center’s Post-COVID clinic analyzed data from 373 Long COVID patients through comprehensive questionnaires covering Long COVID syndrome, Fibromyalgia criteria, personality assessments, social support, and subjective evaluations of cognitive decline, health and life quality. In total, 116 out of 373 patients completed the questionnaire, yielding a 31% participation rate; Results: Cluster analysis revealed two groups, with Cluster 1 (N = 58) exhibiting Type D personality traits while Cluster 2 (N = 56) not meeting criteria for Type D personality. In comparison to Cluster 2, Cluster 1 patients reported heightened anxiety, depression, reduced social support, increased pain symptoms, manifestations of fibromyalgia, cognitive decline, and poor sleep quality, contributing to a diminished quality-of-life perception; Conclusions: findings highlight diverse personality profiles among Long COVID patients, emphasizing the need for tailored care. This approach shows potential for improving Long COVID patient care, aligning with the evolving personalized medicine paradigm.
Article
Full-text available
Aims Cardiovascular diseases (CVDs) increase mortality risk from coronavirus infection (COVID-19). There are also concerns that the pandemic has affected supply and demand of acute cardiovascular care. We estimated excess mortality in specific CVDs, both ‘direct’, through infection, and ‘indirect’, through changes in healthcare. Methods and results We used (i) national mortality data for England and Wales to investigate trends in non-COVID-19 and CVD excess deaths; (ii) routine data from hospitals in England (n = 2), Italy (n = 1), and China (n = 5) to assess indirect pandemic effects on referral, diagnosis, and treatment services for CVD; and (iii) population-based electronic health records from 3 862 012 individuals in England to investigate pre- and post-COVID-19 mortality for people with incident and prevalent CVD. We incorporated pre-COVID-19 risk (by age, sex, and comorbidities), estimated population COVID-19 prevalence, and estimated relative risk (RR) of mortality in those with CVD and COVID-19 compared with CVD and non-infected (RR: 1.2, 1.5, 2.0, and 3.0). Mortality data suggest indirect effects on CVD will be delayed rather than contemporaneous (peak RR 1.14). CVD service activity decreased by 60–100% compared with pre-pandemic levels in eight hospitals across China, Italy, and England. In China, activity remained below pre-COVID-19 levels for 2–3 months even after easing lockdown and is still reduced in Italy and England. For total CVD (incident and prevalent), at 10% COVID-19 prevalence, we estimated direct impact of 31 205 and 62 410 excess deaths in England (RR 1.5 and 2.0, respectively), and indirect effect of 49 932 to 99 865 deaths. Conclusion Supply and demand for CVD services have dramatically reduced across countries with potential for substantial, but avoidable, excess mortality during and after the pandemic.
Article
Full-text available
Background The medium-term effects of Coronavirus disease (COVID-19) on organ health, exercise capacity, cognition, quality of life and mental health are poorly understood. Methods Fifty-eight COVID-19 patients post-hospital discharge and 30 age, sex, body mass index comorbidity-matched controls were enrolled for multiorgan (brain, lungs, heart, liver and kidneys) magnetic resonance imaging (MRI), spirometry, six-minute walk test, cardiopulmonary exercise test (CPET), quality of life, cognitive and mental health assessments. Findings At 2–3 months from disease-onset, 64% of patients experienced breathlessness and 55% reported fatigue. On MRI, abnormalities were seen in lungs (60%), heart (26%), liver (10%) and kidneys (29%). Patients exhibited changes in the thalamus, posterior thalamic radiations and sagittal stratum on brain MRI and demonstrated impaired cognitive performance, specifically in the executive and visuospatial domains. Exercise tolerance (maximal oxygen consumption and ventilatory efficiency on CPET) and six-minute walk distance were significantly reduced. The extent of extra-pulmonary MRI abnormalities and exercise intolerance correlated with serum markers of inflammation and acute illness severity. Patients had a higher burden of self-reported symptoms of depression and experienced significant impairment in all domains of quality of life compared to controls (p<0.0001 to 0.044). Interpretation A significant proportion of patients discharged from hospital reported symptoms of breathlessness, fatigue, depression and had limited exercise capacity. Persistent lung and extra-pulmonary organ MRI findings are common in patients and linked to inflammation and severity of acute illness. Funding NIHR Oxford and Oxford Health Biomedical Research Centres, British Heart Foundation Centre for Research Excellence, UKRI, Wellcome Trust, British Heart Foundation.
Article
Full-text available
Large numbers of people are being discharged from hospital following COVID-19 without assessment of recovery. In 384 patients (mean age 59.9 years; 62% male) followed a median 54 days post discharge, 53% reported persistent breathlessness, 34% cough and 69% fatigue. 14.6% had depression. In those discharged with elevated biomarkers, 30.1% and 9.5% had persistently elevated d-dimer and C reactive protein, respectively. 38% of chest radiographs remained abnormal with 9% deteriorating. Systematic follow-up after hospitalisation with COVID-19 identifies the trajectory of physical and psychological symptom burden, recovery of blood biomarkers and imaging which could be used to inform the need for rehabilitation and/or further investigation.
Article
Full-text available
Objectives: To estimate the impact of the COVID-19 pandemic on cancer care services and overall (direct and indirect) excess deaths in people with cancer. Methods: We employed near real-time weekly data on cancer care to determine the adverse effect of the pandemic on cancer services. We also used these data, together with national death registrations until June 2020 to model deaths, in excess of background (pre-COVID-19) mortality, in people with cancer. Background mortality risks for 24 cancers with and without COVID-19-relevant comorbidities were obtained from population-based primary care cohort (Clinical Practice Research Datalink) on 3 862 012 adults in England. Results: Declines in urgent referrals (median=-70.4%) and chemotherapy attendances (median=-41.5%) to a nadir (lowest point) in the pandemic were observed. By 31 May, these declines have only partially recovered; urgent referrals (median=-44.5%) and chemotherapy attendances (median=-31.2%). There were short-term excess death registrations for cancer (without COVID-19), with peak relative risk (RR) of 1.17 at week ending on 3 April. The peak RR for all-cause deaths was 2.1 from week ending on 17 April. Based on these findings and recent literature, we modelled 40% and 80% of cancer patients being affected by the pandemic in the long-term. At 40% affected, we estimated 1-year total (direct and indirect) excess deaths in people with cancer as between 7165 and 17 910, using RRs of 1.2 and 1.5, respectively, where 78% of excess deaths occured in patients with ≥1 comorbidity. Conclusions: Dramatic reductions were detected in the demand for, and supply of, cancer services which have not fully recovered with lockdown easing. These may contribute, over a 1-year time horizon, to substantial excess mortality among people with cancer and multimorbidity. It is urgent to understand how the recovery of general practitioner, oncology and other hospital services might best mitigate these long-term excess mortality risks.
Article
The COVID-19 pandemic has led to an unprecedented surge in hospitalised patients with viral pneumonia. The most severely affected patients are older men, individuals of black and Asian minority ethnicity and those with comorbidities. COVID-19 is also associated with an increased risk of hypercoagulability and venous thromboembolism. The overwhelming majority of patients admitted to hospital have respiratory failure and while most are managed on general wards, a sizeable proportion require intensive care support. The long-term complications of COVID-19 pneumonia are starting to emerge but data from previous coronavirus outbreaks such as severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS) suggest that some patients will experience long-term respiratory complications of the infection. With the pattern of thoracic imaging abnormalities and growing clinical experience, it is envisaged that interstitial lung disease and pulmonary vascular disease are likely to be the most important respiratory complications. There is a need for a unified pathway for the respiratory follow-up of patients with COVID-19 balancing the delivery of high-quality clinical care with stretched National Health Service (NHS) resources. In this guidance document, we provide a suggested structure for the respiratory follow-up of patients with clinicoradiological confirmation of COVID-19 pneumonia. We define two separate algorithms integrating disease severity, likelihood of long-term respiratory complications and functional capacity on discharge. To mitigate NHS pressures, virtual solutions have been embedded within the pathway as has safety netting of patients whose clinical trajectory deviates from the pathway. For all patients, we suggest a holistic package of care to address breathlessness, anxiety, oxygen requirement, palliative care and rehabilitation.