This page compiles our content related to Management of coexisting conditions in the context of COVID-19. For further information on diagnosis and treatment, follow the links below to our full BMJ Best Practice topics on the relevant conditions and symptoms.
Introduction
Considerations for perinatal care
Considerations for newborn care
Considerations for patients with dermatologic conditions receiving drugs that affect the immune response
Considerations for patients with dermatologic conditions receiving oral retinoids
Considerations for endoscopy
Considerations for patients receiving systemic anticancer therapy
Considerations for patients receiving radiation therapy
Considerations for patients with neuromuscular diseases
Use of ACE inhibitors and angiotensin-II receptor antagonists
Routine immunization
Considerations for patients who require anticoagulation
Considerations for management of patients in community psychiatry services
Considerations for mental health of adults
Mental health of children and adolescents
Considerations for the mental health of healthcare workers
Safeguarding children and young people
Use of valproate
Considerations for cardiac investigations
Considerations for patients with cardiac implantable electronic devices (CIEDs)
Considerations for patients with eating disorders
Considerations for patients with hyperthyroidism
Potential impact of COVID-19 pandemic on diagnosis and management of other conditions
Resources
Relevant conditions
go to our full topic on Coronavirus disease (COVID-19)
Our full topic on Coronavirus disease 2019 (COVID-19) includes information on diagnosis and management, as well as prevention, differential diagnosis, epidemiology, etiology, prognosis, and complications.
go to our full topic on Abnormal uterine bleeding
A Cochrane review has evaluated the effectiveness and safety of treatments for heavy menstrual bleeding that are commonly available during pandemics.[236] Treatments evaluated included nonsteroidal anti-inflammatory drugs (NSAIDs), antifibrinolytics, combined hormonal contraceptives, and progestogens. The review found that there is moderate-certainty evidence that antifibrinolytics and combined hormonal contraceptives reduce heavy menstrual bleeding compared with placebo; there is low‐certainty evidence that NSAIDs reduce heavy menstrual bleeding compared with placebo; and there is low‐certainty evidence that antifibrinolytics are more effective in reducing heavy menstrual bleeding when compared with NSAIDs and short‐cycle progestogens. The authors were unable to draw conclusions about the effects of antifibrinolytics compared with long‐cycle progestogens and no conclusions could be made about quality of life, patient satisfaction with treatment, or serious adverse events. The review’s authors suggest that within the context of a pandemic when treatment is selected remotely, antifibrinolytics (e.g., tranexamic acid), NSAIDs, and combined hormonal contraceptives can be offered. They also emphasize the importance of providing a complete face-to-face assessment (physical exam, blood tests, ultrasound) when services are available.
[ ]
Menstrual changes and abnormal bleeding are not listed as common side effects of COVID-19 vaccination; however, more than 51,000 reports of these events have been made to the UK’s Medicines and Healthcare products Regulatory Agency (MHRA) across all vaccines currently offered in the UK.[237] While evidence is currently not definitive, the Royal College of Obstetricians and Gynaecologists and the MHRA recommend that anyone presenting with menstrual disorders and/or unexpected vaginal bleeding following COVID-19 vaccination should be treated according to clinical guidelines for these conditions, as usual.[238][239] The Pharmacovigilance Risk Assessment Committee of the European Medicines Agency has concluded that there is insufficient evidence to establish a causal link between COVID-19 vaccination and absence of menstruation and are monitoring the issue.[240]
go to our full topic on Acute cholecystitis
A multisociety position statement advises that laparoscopic cholecystectomy remains the treatment of choice for acute cholecystitis during the COVID-19 pandemic. Early cholecystectomy (performed as soon as possible after the onset of symptoms) is preferred. Air exiting the peritoneum should be filtered through ultra-low particulate air filters to remove viral particles. If surgery needs to be postponed because of a COVID-19 outbreak, patients must be closely monitored for signs of sepsis and pain progression. If patients are not fit for surgery and have not improved with antibiotic therapy, percutaneous gallbladder drainage may be considered.[241]
go to our full topic on Acute kidney injury
Patients with COVID-19 may develop acute kidney injury (AKI). Age ≥60 years and severe COVID-19 are independent risk factors for mortality.[242] Patients who have severe COVID-19 are more likely to require continuous renal replacement therapy compared with patients who have nonsevere COVID-19. AKI significantly increases the risk of in-hospital death in patients with COVID-19.[242] One cohort study of patients with in-hospital AKI in the US found that COVID-19-associated AKI was associated with a greater rate of estimated glomerular filtration rate decrease after discharge compared with AKI in patients without COVID-19, and this was independent of underlying comorbidities or AKI severity.[243]
UK guidelines note that in people with COVID-19, AKI can develop at any time (before, during, or after hospital admission), and that maintaining euvolemia (optimal fluid status) is critical to reducing AKI incidence.[131]
The treatment of AKI in patients with COVID-19 appears to be the same as in other populations, including continuous renal replacement therapy if necessary.[244] Medications that can cause or worsen AKI should be stopped unless essential. Some treatments for COVID-19 may increase the risk of AKI.[131] Patients with elevated body temperature and increased respiratory rate will have greater insensible fluid losses.[131] Intravenous fluids are required in many cases, and choice should be guided by biochemistry. The goal of intravenous fluid therapy is to maintain a euvolemic state.[131] Potassium binders can be used as part of the emergency management of life-threatening hyperkalemia, alongside standard care.[131]
go to our full topic on Acute lymphocytic leukemia
Analysis of US health-insurance claims data has shown that leukemia is a significant risk factor for fatal COVID-19.[82] An international expert panel and the American Society of Hematology have made recommendations for the treatment of adult acute lymphocytic leukemia.[245][246] Clinicians should consider minimizing corticosteroid exposure and reducing doses of daunorubicin and pegaspargase (pegylated asparaginase) during treatment induction in older people and people at high risk for complications of COVID-19.[245][246] Anti-CD20 monoclonal antibodies reduce immune globulin levels; treatment with these agents should be deferred if possible. Second-generation tyrosine kinase inhibitors with reduced-dose corticosteroids should be considered in Philadelphia chromosome-positive disease.[245][246]
Clinicians should consider blinatumomab if patients are positive for minimal residual disease after two cycles of chemotherapy. If patients are negative for minimal residual disease and have received most of their chemotherapy, they may be advanced to maintenance. During maintenance, clinicians should consider reducing corticosteroids and avoiding vincristine in patients >65 years. Recommendations are given for relapsed or refractory disease and transplantation. Growth factor support should be considered in patients without COVID-19 to facilitate neutrophil count recovery and maintain absolute neutrophil count above 1000 cells per microliter during all phases of treatment. Growth factors should probably be avoided in patients with moderate-to-severe severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection because there is a potential risk of exacerbating inflammatory pulmonary injury.[245]
There are limited data on trials of SARS-CoV-2 in patients with hematological conditions. Such patients are likely to have an attenuated response to vaccination. One study of the BNT162b2 mRNA COVID-19 vaccine in patients with hematological malignancies found the antibody response was reduced compared with healthy controls, and was affected by disease treatment.[247] Additional measures, such as early vaccination of household contacts, may be considered.[248]
go to our full topic on Acute myelogenous leukemia
Analysis of US health-insurance claims data has shown that leukemia is a significant risk factor for fatal COVID-19.[82] Acute myelogenous leukemia (AML) is associated with worse survival in patients with COVID-19.[249] Patients with AML should be screened for COVID-19 before starting induction or consolidation chemotherapy.[250] Patients receiving intensive therapy should, ideally, be barrier nursed in a COVID-19-negative ward with enhanced screening and protection measures. Chemotherapy should be delayed until the resolution of symptoms and the patient has a negative polymerase chain reaction test. Cytogenetics and nucleophosmin-1 (NPM1) and fsm-related tyrosine kinase-3 (FLT3) status will guide choice of chemotherapy. Venetoclax and gilteritinib have been granted emergency approval from NHS England for use in selected patient groups.[250]
Growth factors should probably be avoided in patients with moderate-to-severe COVID-19 because there is a potential risk of exacerbating inflammatory pulmonary injury.[245]
There are limited data on trials of severe acute respiratory syndrome coronavirus 2 vaccination in patients with hematological conditions. Such patients are likely to have an attenuated response to vaccination. One study of the BNT162b2 mRNA COVID-19 vaccine in patients with hematological malignancies found the antibody response was reduced compared with healthy controls, and was affected by disease treatment.[247] Additional measures, such as early vaccination of household contacts, may be considered.[248]
go to our full topic on Addison disease
Patients with adrenal insufficiency are at an increased risk of infection, which may be complicated by developing an adrenal crisis. Guidance on prevention of adrenal crisis in patients with confirmed or suspected COVID-19 is available.[251] Patients should be given support to help them self-manage their condition safely and should be educated in the use of sick day rules. The guidelines recommend that patients with symptoms of COVID-19 should seek medical advice, and should take oral hydrocortisone or prednisone as directed. Patients are also advised to take acetaminophen for fever, and to drink regularly, monitoring how concentrated their urine appears. If there are signs of clinical deterioration (such as dizziness, intense thirst, shaking uncontrollably, drowsiness, confusion, lethargy, vomiting, severe diarrhea, increasing shortness of breath, respiratory rate >24/min, difficulty speaking) the patient or carer should inject hydrocortisone intramuscularly and call for emergency medical assistance.[251] Hospitalized patients should receive intravenous hydrocortisone and continuous intravenous fluid resuscitation with isotonic saline; fludrocortisone should be temporarily stopped.
go to our full topic on Allergic rhinitis
Statements from the British Society for Allergy and Clinical Immunology and the Italian Society of Pediatric Allergy and Immunology advise that patients with allergic rhinitis should continue usual treatment, including topical intranasal corticosteroids and antihistamines. Uncontrolled allergic rhinitis may lead to sneezing and increased hand-eye and hand-nose contact, facilitating severe acute respiratory syndrome coronavirus-2 transmission.[252][253] Post-dose observation may be decreased and intervals between doses may be increased for patients taking subcutaneous immunotherapy.[252]
go to our full topic on Aortic stenosis
A position statement from the American College of Cardiology and the Society for Cardiovascular Angiography and Interventions provides recommendations for the triage of patients referred for transcatheter aortic valve replacement (TAVR) during the pandemic.[254] For patients with severe symptomatic aortic stenosis (AS), TAVR should be considered to decrease the risk for clinical deterioration, prolonged hospital stay, or repeat hospitalization. Data are not robust enough for clear recommendations for patients with minimally symptomatic severe to critical AS; however, urgent TAVR or close outpatient virtual monitoring may be considered. Particularly high peak or mean gradient, very small calculated aortic valve area and very low dimensionless index warrant consideration of TAVR. For asymptomatic patients, consideration of TAVR may be postponed for 3 months or until elective procedures are resumed. Close outpatient monitoring should continue for all patients with severe AS.
The British Heart Valve Society has published recommendations for the outpatient management of heart valve disease following the COVID-19 pandemic. They recommend that patients with severe symptomatic AS and advanced heart failure, impaired ventricular function (left ventricular (LV) ejection fraction of <50%) or syncope are the highest clinical priority and ideally require valve intervention within 2 weeks and no more than 4 weeks. Interventions for AS include surgical aortic valve replacement (sAVR) or transcatheter aortic valve implantation (TAVI), and currently the benefits of sAVR versus TAVI should be considered for each patient, taking into account the requirement for general anesthesia and intensive care unit admission, length of stay in hospital and risk of peri-procedural exposure to COVID-19.[255]
The European Society of Cardiology advises that increased use of transcatheter aortic valve implantation via transfemoral approach performed under conscious sedation and/or local anesthesia, where feasible, may allow optimal use of resources by reducing need for intensive care admission and accelerating hospital discharge. Priority for aortic valve replacement should be given to patients with syncope and heart failure, and those with high/very high transvalvular gradients and/or impaired left ventricular function. Clinical decisions should be made by a multidisciplinary team, and telemedicine should be used to facilitate discussions if face to face meetings are not possible. The immediate and short term prognosis of individuals should be balanced against the available resources and risks of hospital acquired infection.[193]
go to our full topic on Aplastic anemia
The definitive treatment options for severe aplastic anemia (AA) are stem cell transplant or immunosuppressive therapy. The American Society of Hematology advise that for patients with absolute neutrophil count (ANC) <200/microlitre (very severe AA) the risk of delaying transplant or immunosuppressive therapy outweighs the risks of exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during hospitalization or the impact of immunosuppression on the course of infection, but that optimal management may not currently be practical.[256]
Immunosuppressive treatment options are antithymocyte globulin (ATG), cyclosporine, and eltrombopag; administration of ATG requires hospitalization. The American Society of Hematology, the European Society for Blood and Marrow Transplantation, and NHS England have released statements recommending the use of eltrombopag, with or without cyclosporine, as bridging treatment to stem cell transplant or immunosuppressive therapy with ATG for patients with severe or very severe AA during the COVID-19 pandemic.[256][257][258]
Trials of SARS-CoV-2 vaccination in patients with hematologic conditions are lacking. Such patients are likely to have an attenuated response to vaccination. One small study reported a reduced seroconversion rate and lower antibody response after a first dose of SARS-CoV-2 vaccine in people with aplastic anemia, compared with healthy controls. After a second vaccination, seropositivity and antibody levels were equivalent in people with aplastic anemia, and healthy controls.[259]
Measures, such as early vaccination of household contacts, may be considered.[248] The American Society of Hematology advises that the benefits of COVID-19 vaccination outweigh the risks in patients with AA, particularly those with additional risk factors for severe COVID-19 disease, though some patients receiving immunosuppressive therapies may not mount an appropriate immune response to the vaccine.[256]
go to our full topic on Asthma
The Global Initiative for Asthma (GINA) reports that people with asthma do not appear to be at increased risk of acquiring COVID-19 and that people with well-controlled, mild-to-moderate asthma are not at increased risk of severe COVID-19.[260] GINA also reported that people with well-controlled asthma are not at increased risk of death from COVID-19 overall; however, people who recently needed oral corticosteroids for their asthma did have an increased risk of COVID-19-related death.[260] A large cohort study of adults and children over age 12 in England found that those with poorly controlled or severe asthma were at significantly increased risk of hospitalization from COVID-19 when compared with those without asthma; however, those with mild or well-controlled asthma were not at increased risk.[261] In Scotland, a cohort study found that two or more courses of oral corticosteroids or an asthma hospital admission in the preceding 2 years was associated with an increased risk of COVID-19 hospital admission and ICU admission or death in adults.[262] Additional studies have found that patients with severe asthma (treated with 3 different classes of medication) have an increased risk of hospitalization, intensive care admission, and death from COVID-19.[263][264]
Patients should continue taking their prescribed asthma medication as usual, including inhaled and oral corticosteroids and biologic therapy.[253][260][265][266] GINA advises that all patients should have a written action plan so they know how to recognize worsening asthma, how to increase reliever and controller medications, and when to seek medical help.[260] GINA advises that nebulizers should be avoided for acute attacks due to the risk of transmitting respiratory viral particles, and that a pressurized metered-dose inhaler and spacer with mouthpiece or tightly fitting facemask can be used to deliver a short-acting beta-2 agonist instead.[260] The US Centers for Disease Control and Prevention advises that nebulizer administration may generate infectious aerosols; however, it is unclear whether association between nebulizer administration and infection is due to the generation of infectious particles or the close contact between the patient and healthcare professional administering the nebulizer.[267] GINA advises that local COVID-19 testing recommendations and infection control procedures are followed if spirometry and peak-flow measurement is needed.[260] GINA advises that use of an in-line filter can minimize risk of transmission during spirometry and that patients should be advised to stay on the mouthpiece if they feel the need to cough.[260] Canadian guidelines advise that clinical judgement is needed to determine the urgency of diagnostic and therapeutic bronchoscopy. Nonurgent procedures may be postponed in times of high community spread where the risk of viral transmission is considered unacceptably high.[268] The American Academy of Allergy, Asthma & Immunology (AAAAI) has provided recommendations on the safe practice of pulmonary procedures, including spirometry, fractional exhaled nitric oxide, nebulized treatments, and methacholine challenge.[269]
Patients should ensure they have a sufficient supply of medication at home, but should not stockpile. Patients may be reminded that they should not share inhalers or spacers with others. Clinicians should encourage smoking cessation.[253]
Advise patients that COVID-19 may present with symptoms similar to an asthma attack (e.g., cough, shortness of breath); however, additional symptoms such as fever, fatigue, and change in taste or smell are more likely to suggest COVID-19 infection.[270] A large prospective cohort study reported that 1 in 7 patients aged 16-49 years with COVID-19 presented with wheeze.[263]
In studies of patients hospitalized with COVID-19, asthma does not appear to be an independent risk factor for intubation.[271] Patients with asthma who are aged 16 years and older have more severe disease on presentation to hospital, and are more likely to require critical care, noninvasive ventilation, and oxygen compared with patients without asthma. In patients aged 50 years and older with asthma, ICS use within 2 weeks of hospital admission was associated with decreased mortality compared with patients without an underlying respiratory condition.[263]
GINA reports that in 2020, there was a reduction in asthma exacerbations in many countries.[260] Emergency department attendances and hospital admissions for asthma decreased significantly in Scotland and Wales during the first 2020 lockdown. Prescriptions for inhaled and oral corticosteroids increased significantly before lockdown in Wales, suggesting that improved adherence to preventative treatment and self-management of exacerbations may have contributed to this change. Reduced circulation of other respiratory viruses, decreased air pollution, and changes in smoking habits during lockdown may also have contributed to the decrease.[272] In England, primary care attendances for asthma exacerbations decreased during the first 2020 lockdown, but there was no significant decrease in exacerbations requiring a hospital visit.[273]
GINA recommends COVID-19 vaccination for people with asthma, following the usual vaccine precautions.[260] For people with severe asthma receiving biologic therapy, the COVID-19 vaccine and the biologic therapy should not be given on the same day.[260]
Attention deficit hyperactivity disorder (ADHD)
Signs & symptoms
Investigations
Differentials
Treatment algorithm
go to our full topic on Attention deficit hyperactivity disorder (ADHD)
The European ADHD guidelines group have published guidance for management of patients with ADHD during the COVID-19 pandemic. Parents or carers are encouraged to use behavioral parenting strategies and schools are advised to prioritize monitoring of students with ADHD. Patients should be offered the opportunity to start on medication, if indicated, following an initial assessment. Patients who are already established on medication should continue taking it as prescribed. Parents and patients should not increase medication doses above the dose prescribed to manage the stress of confinement. Routine cardiovascular examination and face-to-face monitoring can be deferred for individuals without any cardiovascular risk factors. Home blood pressure and heart rate monitoring is recommended.[274]
go to our full topic on Autoimmune hepatitis
A cohort study of 500,000 people in the UK found that autoimmune disease and history of oral corticosteroid were associated with increased risk of fatal COVID-19.[275] UK, European, and US guidelines advise against making anticipatory adjustments to immunosuppressive drugs in patients without COVID-19.[276][277] Doing so may cause a flare of autoimmune hepatitis (AIH). Budesonide may be considered to induce remission in patients without cirrhosis who have a flare of AIH, to minimize systemic corticosteroid exposure.[277]
The American Association for the Study of Liver Disease (AASLD) advises that immunosuppressive therapy should be commenced in patients with AIH, with or without COVID-19.[276]
If a patient develops COVID-19, the AASLD advises that clinicians consider reducing doses of immunosuppressants, particularly azathioprine and mycophenolate, based on general principles for managing infections in immunosuppressed patients and to decrease the risk of superinfection.[276] Adjustment should be individualized and will depend on the severity of COVID-19. If a patient is taking corticosteroids and develops COVID-19, corticosteroid dosing must be sufficient to prevent adrenal insufficiency.[277]
COVID-19 may cause abnormal liver function tests in patients with AIH; these should not be attributed to a disease flare without biopsy confirmation.[276]
A joint statement from the British Society of Gastroenterology, British Association for the Study of the Liver, NHS Blood & Transplant, and British Liver Trust advises that patients with autoimmune hepatitis should consider SARS-CoV-2 vaccination with any of the available vaccines.[278]
go to our full topic on Breast cancer
A European observational study reported an unadjusted mortality rate of 15.2% for patients with breast cancer and COVID-19. Overall mortality in patients with cancer and COVID-19 was higher in male patients, patients ≥ 65 years and patients with ≥2 comorbidities.[78] Among newly diagnosed patients, African-American patients are more likely to develop COVID-19 than white patients.[77]
The American Society of Breast Surgeons has released recommendations for the prioritization, treatment, and triage of patients with breast cancer during the COVID-19 pandemic.[279] The highest-priority conditions for treatment during the pandemic are:
- Potentially unstable breast disease (e.g., hematoma, infection): assessment and surgery
- New diagnosis of invasive breast cancer
- Surgery: revision of ischemic mastectomy flap; revascularization/revision of autologous tissue flap
- Chemotherapy: neoadjuvant/adjuvant chemotherapy for triple-negative and HER2-positive breast cancer; early chemotherapy likely to improve outcomes in metastatic disease; completion of adjuvant/neoadjuvant chemotherapy that has already started; adjuvant or metastatic endocrine therapy
- Radiation therapy for painful, inoperable breast masses; continuation of radiation therapy that has started; treatment for critical metastatic lesions (e.g., brain metastasis, spinal cord compression).
UK guidelines recommend giving highest priority to patients receiving:[86]
- Curative systemic anticancer treatment with a high (more than 50%) chance of success
- Adjuvant or neoadjuvant systemic anticancer treatment that adds at least 50% chance of cure to surgery or radiation therapy alone or treatment given at relapse.
The UK Association of Breast Surgery has published recommendations for delivering breast services during the pandemic.[280]
The Association of Breast Surgery suggests prioritizing patients in the following order if capacity for surgery is limited:[280]
- Estrogen receptor (ER) negative
- Human epidermal growth factor receptor 2 (HER2) positive
- Premenopausal patients
- Postmenopausal, ER positive patients with high risk disease (grade 3 or node positive)
- Large areas of high grade ductal carcinoma in situ (DCIS)
- Postmenopausal, ER positive patients with lower risk disease
- Remaining patients with DCIS
Neoadjuvant chemotherapy should only be given where it is clear that chemotherapy is indicated, and would be given in the adjuvant setting. Neoadjuvant chemotherapy should be routinely supported with granulocyte-colony stimulating factor during the pandemic. Multidisciplinary team discussion is recommended for all cases.[90][281]
The American College of Cardiology has made recommendations for alterations to routine cardiac surveillance for patients with breast cancer receiving systemic anticancer therapy who are at low risk for cardiotoxicity. Patients at higher risk should receive usual care.[282]
COVID-19 vaccination is associated with a higher incidence of axillary lymphadenopathy on breast MRI and mammography.[283][284] One study reported the incidence of mammographic axillary lymphadenopathy following COVID-19 vaccination as 3%.[284] Guidelines have been published to help avoid biopsies of reactive nodes. MRI-detected, unilateral, axillary lymphadenopathy ipsilateral to the vaccination arm is most likely to be COVID-19 vaccination-related if within 4 weeks of either dose. A follow-up ultrasound 6 to 8 weeks after the second vaccine dose is recommended.[283]
Further oncology resources are available at:
NCCN: COVID-19 resources for the cancer care community Opens in new window
ASCO: coronavirus resources Opens in new window
go to our full topic on Bronchiolitis
UK guidance recommends optimizing preventive treatment, including influenza vaccination and palivizumab for eligible children.[285] Key features of assessment are oxygenation, hydration, and nutrition, irrespective of the child’s potential COVID-19 status. High-flow nasal cannula oxygen is an aerosol-generating procedure; a senior clinician should be consulted if its use is being considered. Children who require admission to hospital should be tested for respiratory viruses, including severe acute respiratory syndrome coronavirus (SARS-Cov-2), influenza, and respiratory syncytial virus. Children who require intensive/high-dependency care or surgery should be prioritized for rapid SARS-CoV-2 testing.
go to our full topic on Carcinoid syndrome
Interim guidance for the management of patients with neuroendocrine tumors is available from the UK and Ireland Neuroendocrine Tumour Society.[286] Patients already receiving somatostatin analog therapy should continue on their treatment, but delivery of medication and dose intervals may change in order to minimize hospital visits; this is highly individual and decided on a patient-by-patient basis. Patients with highly functional tumors due to start treatment will need initiation of therapy to control their condition. Initiation of therapy in patients with nonfunctioning tumors may be delayed depending on patient fitness and tumor characteristics; again, this is decided on a patient-by-patient basis. Use of chemotherapy and peptide receptor radiotherapy should also be individualized, with the risks and benefits of starting or continuing considered for eligible patients. Surgery may be considered where it is life-saving or potentially curative.
go to our full topic on Cardiopulmonary resuscitation (CPR)
Giving CPR poses a high risk to healthcare workers in the context of COVID-19 due to the aerosol-generating procedures, close proximity of multiple healthcare workers and the patient, and the need to work quickly.
If cardiac arrest is recognized (patient is unresponsive and breathing abnormally), UK and international (ILCOR) guidelines advise looking for breathing, but advise against opening the airway or listening/feeling for breathing by placing the face close to the patient's mouth.[287][288]
In acute hospital settings, UK guidelines advise that full Aerosol Generating Procedure (AGP) Personal Protective Equipment (PPE) must be worn by all members of the resuscitation team before entering the room; no chest compressions or airway procedures should be started without full AGP PPE. The number of staff in the room should be restricted, and airway interventions should be done by experienced staff, minimizing aerosolization risk.[288] Updated US guidelines now advise that chest compressions or defibrillation should not be delayed for provider PPE, but that initial resuscitation personnel should be relieved by providers wearing appropriate PPE as soon as possible.[289] If the patient cannot be placed supine, cardiopulmonary resuscitation may be provided in the prone position, particularly if the patient has advanced airway and circulatory support.[193]
In first aid and community settings, lay-rescuers should perform compression-only resuscitation and defibrillation (where there is access); a cloth may be placed over the patient's mouth and nose if there is a perceived risk of infection. Pediatric cardiac arrest is more likely to be caused by a respiratory problem, and ventilation is vital; lay-rescuers may consider that the risk of not giving rescue breaths could be greater than the risk of transmission of COVID-19.[287][288][289]
In-water mouth-to-mouth resuscitation should not be performed on drowned patients. Rescuers should prioritize removal from water where PPE and first aid equipment can be used. Rescuers should wear gloves, facemask and eye protection for all resuscitations. Two-person bag-filter-mask ventilation using a high-efficiency particulate arrestance (HEPA) filter is preferred. If this is not possible, mouth-to-mask ventilation with HEPA filter is the second line technique and passive oxygenation is third line. If rescuers cannot follow this guidance, they should provide compression only CPR and cover the patient’s nose and mouth with a cloth.[290]
Brazilian guidelines recommend beginning continuous chest compressions to deliver CPR to adults. The patient’s oral cavity should be sealed with a cloth or a mask providing low flow (6-10 liters/minute) oxygen before starting chest compressions; the seal should be kept in place until an invasive airway is secured. Bag-valve-mask or bag-valve tube ventilation should be avoided if possible; if it is needed, two rescuers should provide ventilation (to allow a two-handed seal around the mask) and an oropharyngeal airway should be used. A HEPA filter should be placed between the mask and bag. If the patient is prone at the time of cardiac arrest and does not have an invasive airway, they should be repositioned supine. If the patient is intubated, chest compressions should be delivered while prone. Resuscitation of children should ideally be with chest compressions and use of a bag-valve-mask apparatus with a HEPA filter until a definitive airway is established.[291]
go to our full topic on Celiac disease
The European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) advises that a biopsy-sparing approach to diagnosis can be used in more children with suspected celiac disease during the pandemic, to mitigate potential delays in diagnosis and treatment caused by reduced access to endoscopy. A biopsy-sparing approach can be used for children who have positive endomysial antibodies and a tissue transglutaminase IgA titer between 5 and 10 times the upper limit of normal.[292]
go to our full topic on Cerebellar ataxia
Patients with cerebellar ataxia (CA) may be at higher risk from serious COVID-19 infection as they are likely to be older, and also because of the neurologic complexities of their underlying disorder and comorbid medical illnesses.[293] Recommendations suggest that patients with immune ataxias continue their therapies (including intravenous immune globulin, corticosteroids, plasma exchange), but they are considered high risk and should follow local advice on physical distancing.[293] Patients with symptoms of infection may be instructed to stop immunosuppressive therapies until they have fully recovered. Starting patients on immunotherapies may also be delayed, but balanced against the risk of not starting, particularly in patients with rapidly progressive immune-mediated ataxias.[293] Decisions for patients with CA who are hospitalized with COVID-19 should involve the ataxiologist.
go to our full topic on Cholestasis of pregnancy
Blood tests of bile acid concentrations should be performed opportunistically for symptomatic women at their regular antenatal appointments, because risks of stillbirth increase significantly with peak serum bile acid concentrations ≥100 micromol/L.[294][295] The risk of stillbirth increases particularly in the 35th gestational week. Therefore, more frequent blood testing is recommended from 34 to 37 gestational weeks.[296][295] Women with peak bile acids ≥100 micromol/L should be offered delivery from 35 gestational weeks, and those with lower levels can be offered delivery at 38 to 39 weeks’ gestation.[296]
go to our full topic on Chronic congestive heart failure
Patients with heart failure have a significantly increased risk of hospitalization and death if they develop COVID-19.[82][297][298][299]
Patients with chronic heart failure are prone to developing acute heart failure after a diagnosis of COVID-19.[300] European guidelines recommend using the same treatment strategy for acute heart failure in patients with COVID-19 as for patients without COVID-19.[193] One observational study reported that discontinuation of medical therapy for chronic heart failure (ACE inhibitors, beta-blockers, and mineralocorticoid receptor antagonists) in patients with COVID-19 was associated with increased mortality. The authors recommend that even if these medications are reduced or discontinued during the acute illness, they should be reintroduced at discharge whenever possible.[300]
go to our full topic on Chronic kidney disease
People of any age with chronic kidney disease (CKD) are at increased risk of severe illness and death from COVID-19.[301][302][303][304][305]One meta-analysis reported a hazard ratio of 3.6 for COVID-19-related mortality in patients with chronic kidney disease.[76]
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) BNT162b2 mRNA vaccine induces comparable neutralizing antibody titers in patients receiving hemodialysis and healthy controls. The adenovirus-associated vector vaccine ADZ1222 induced suboptimal neutralizing antibodies in seronaive patients receiving hemodialysis.[306] A retrospective observational study in the US found that, among patients receiving hemodialysis, vaccination with BNT162b2 or mRNA-1273 was associated with a lower risk of COVID-19 diagnosis and a lower risk of hospitalization or death among those diagnosed with COVID-19.[307] Another retrospective cohort study in the US found that among patients receiving maintenance dialysis, vaccine-induced seroresponse wanes over time across vaccine types.[308]
go to our full topic on Chronic lymphocytic leukemia
Analysis of US health-insurance claims data has shown that leukemia is a significant risk factor for fatal COVID-19.[82] Older age is associated with increased severity of COVID-19 in patients with chronic lymphocytic leukemia (CLL).[309] The American Society of Hematology recommends postponing treatment initiation for CLL in areas where COVID-19 is active. If immediate therapy is needed, treatments that can be provided in an outpatient setting with fewer clinic visits are preferred. Treatment with monoclonal antibodies should be avoided, especially in combination with targeted agents, and initiation of venetoclax should be avoided if possible.[310] Immune globulin replacement may be continued in highly selected patients where the potential benefits are outweighed by the risks of visiting a clinic for the infusion. Intravenous immune globulin can be continued in those who have COVID-19, but requires close monitoring for thromboembolic events. Patients who are already receiving treatment for CLL and have COVID-19 with mild symptoms should generally not have their treatment modified. Treatment may be modified in patients with more severe symptoms depending on the aggressiveness of CLL, history of infections, and the risk of more severe COVID complications; decisions are made on a case-by-case basis, but generally monoclonal antibodies are withheld in patients with COVID-19. Treatment with ibrutinib and acalabrutinib should be continued in patients diagnosed with CLL and COVID-19.[310]
Recommendations from Australia and New Zealand also advise delaying therapy where possible during the pandemic.[311] If therapy is considered essential, then oral therapies should be considered where available.
There are limited data on trials of severe acute respiratory syndrome coronavirus 2 vaccination in patients with hematological conditions. Such patients are likely to have an attenuated response to vaccination. Studies of the BNT162b2 mRNA COVID-19 vaccine in patients with CLL found the antibody response was reduced compared with healthy controls and affected by disease activity and treatment.[247][312] Relatively low antibody responses were seen in patients treated with Bruton's tyrosine kinase inhibitors or venetoclax alone or in combination with antiCD20 antibody. Additional measures, such as early vaccination of household contacts, may be considered.[248]
go to our full topic on Chronic myelogenous leukemia
Analysis of US health-insurance claims data has shown that leukemia is a significant risk factor for fatal COVID-19.[82] The American Society of Hematology has published recommendations on the management of chronic myelogenous leukemia (CML) during the COVID-19 pandemic.[313] Treatment with tyrosine kinase inhibitors (TKI) is not immunosuppressive and should not be interrupted in patients without COVID-19 infection receiving treatment, or delayed in those with newly diagnosed CML. Some TKIs may have pulmonary side effects and stopping therapy may be considered in affected patients who have COVID-19. Patients in remission should be monitored as normal where possible. Remote monitoring or less-frequent monitoring may be considered where local conditions change. Patients in the chronic phase of CML are not at higher risk of COVID-19 infection, and those that do have COVID-19 infection may not be at higher risk of more severe disease. Possible drug-drug interactions should be reviewed in patients being treated for COVID-19.
There are limited data on trials of severe acute respiratory syndrome coronavirus 2 vaccination in patients with hematological conditions. Such patients are likely to have an attenuated response to vaccination. One study of the BNT162b2 mRNA COVID-19 vaccine in patients with hematological malignancies found the antibody response was reduced compared with healthy controls, and was affected by disease treatment.[247] Additional measures, such as early vaccination of household contacts, may be considered.[248]
Chronic obstructive pulmonary disease (COPD)
Signs & symptoms
Investigations
Differentials
Treatment algorithm
go to our full topic on Chronic obstructive pulmonary disease (COPD)
Patients with COPD are at higher risk for severe COVID-19 illness and death and should carefully follow public health advice.[263][264][302][314] One meta-analysis reported a 3.18-fold increased risk of mortality and a 3.63-fold risk of severe disease in people with COVID-19 and pre-existing COPD.[315] The Global Initiative for Chronic Obstructive Lung Disease (GOLD) recommends that patients with COPD who have new or worsening respiratory symptoms, fever, and/or any other possible COVID-19-related symptoms should be tested for severe acute respiratory disease coronavirus 2 (SARS-CoV-2), even if the symptoms are mild.[316] A large prospective cohort study found that patients aged 50 years and older with COPD, with or without asthma, were more likely to require supplemental oxygen and noninvasive ventilation, but less likely to receive invasive mechanical ventilation or critical care compared with patients without a respiratory condition. Mortality in this group exceeded 40%.[263]
GOLD advises that patients should maintain their regular treatment and that there is currently no evidence to recommend avoiding corticosteroids (inhaled or oral) in patients with COPD during the COVID-19 pandemic.[316]
Exacerbations of COPD should be managed by the patient following their individualized plan, and there should be no change to advance prescribing of rescue antibiotics and corticosteroids. Patients should not start rescue antibiotics and corticosteroids to treat symptoms of COVID-19, and should not start prophylactic antibiotics to reduce risk.[317] Canadian guidelines recommend that patients with COPD who develop COVID-19 should continue their usual inhaled maintenance therapy and that acute exacerbations of COPD should be treated with prednisone if needed, irrespective of whether the exacerbation is triggered by SARS-CoV-2.[265]
To reduce the risk of acute exacerbations, and a poorer outcome from COVID-19 infection, strongly encourage patients who are still smoking to stop.[317]
GOLD considers nebulization to have an increased risk of droplet generation and disease transmission.[316] GOLD advises that where possible, dry powder inhalers, pressurized metered dose inhalers, and soft mist inhalers are used instead of nebulizers for drug delivery. The US Centers for Disease Control and Prevention advises that nebulizer administration may generate infectious aerosols, however it is unclear whether association between nebulizer administration and infection is due to the generation of infectious particles or the close contact between the patient and healthcare professional administering the nebulizer.[267]
GOLD recommends that during periods of high COVID-19 prevalence, spirometry is restricted to patients requiring urgent diagnosis of COPD, and/or to assess lung function prior to interventional procedures or surgery.[316]
The British Thoracic Society has developed online pulmonary rehabilitation resources for patients to use while face-to-face meetings are not possible and a resource pack for patients who survive COVID-19.[318]
One study in Singapore reported a significant and sustained decrease in hospital admissions for acute exacerbations of COPD, including respiratory virus infection-associated acute exacerbations, during the pandemic, which coincided with the introduction of public health measures such as social distancing and universal mask-wearing.[319]
go to our full topic on Chronic pain
A panel of experts from North America and Europe has developed recommendations to guide management of chronic pain during the pandemic.[320] Opioids may continue to be prescribed or initiated and patients should be informed of the potential risks and impact of long-term opioid use on the immune system. Patients who regularly use nonsteroidal anti-inflammatory drugs (NSAIDs) should continue, while being monitored for adverse effects, and patients should promptly report any mild fever or new myalgia. Use of corticosteroids increases the potential for adrenal insufficiency and altered immune response; clinicians should consider the risks and benefits of corticosteroid injections and use a decreased dose.[320] Insertion of new intrathecal pumps should be avoided, except for highly selected cancer patients where the benefit outweighs the risk. New neurostimulator trials or implants should also be avoided.
Studies have found no association between use of NSAIDs and worse outcomes in patients with COVID-19; one prospective multicentre study in the UK in patients hospitalized with COVID-19 found that those taking NSAIDs before admission had the same outcomes as those who did not.[321] Results from one retrospective cohort study suggest that long-term opioid therapy may be associated with an increased risk of severe illness and complications, including death, in adults with COVID-19 infection; prospective studies are needed.[322]
A retrospective, population-based cohort study conducted in England reported that rates of death involving COVID-19 were higher for disabled people than for nondisabled people during the first two waves of the COVID-19 pandemic. Relative risks were particularly high among people with greater levels of activity limitation, disabled women, and younger disabled people. Adverse socioeconomic, demographic, and health-related risk factors accounted for some, but not all, of the elevated risk among disabled people.[323]
go to our full topic on Chronic sinusitis
The European Academy of Allergy and Clinical Immunology advise that patients with chronic rhinosinusitis should continue to take intranasal corticosteroids at their individually prescribed dose and should not modify or stop treatment without consulting their physician.[324] There is no evidence that the use of intranasal corticosteroids in the approved dose and indications leads to increased risk of COVID-19 infection or a more severe disease course. Discontinuation of intranasal corticosteroids can lead to worsening of chronic sinusitis. Systemic corticosteroids should be used with caution during the pandemic and in patients with severe symptoms and no therapeutic alternative. Patients with severe chronic rhinosinusitis with nasal polyps should be considered for systemic corticosteroids if surgery is not possible; patients should quarantine themselves during the course of treatment, and for one week afterwards.[324]
Physicians should assess the risks and benefits of initiating biologics in low-risk patients; biologic therapy may be deferred in high-risk patients during the pandemic. Patients who do not have COVID-19 and are already receiving biologic therapy may continue following a risk/benefit assessment; there is insufficient evidence to recommend discontinuation. Physicians should discontinue or postpone biologic therapy in patients who test positive for COVID-19.[325][324]
go to our full topic on Cirrhosis
Patients with chronic liver disease have a higher mortality rate from COVID-19 infection, and mortality is associated with liver disease severity.[276][277] COVID-19 infection may precipitate acute-on-chronic liver failure in patients with decompensated cirrhosis.[326]
European guidelines advise that patients who have cirrhosis and COVID-19 should be admitted to the hospital for inpatient care. Such patients are at high risk of new or worsening hepatic decompensation, severe COVID-19, and death.[277] Patients with cirrhosis and portal hypertension should avoid nonsteroidal anti-inflammatory drugs. Care should be taken to avoid acetaminophen overdosing in patients with cirrhosis.[327] Guidelines to prevent complications should be followed closely for patients with decompensated cirrhosis. Vaccination against Streptococcus pneumoniae and influenza is recommended. Treatment for complications (e.g., spontaneous bacterial peritonitis, hepatic encephalopathy, ascites) should be continued.[277] Organ donations and transplants are likely to be reduced in many countries. Liver transplant services should be restored following the peak of the pandemic, wherever possible. Where resources remain limited, listing for transplantation should be restricted to patients with poor short-term prognosis, including those with acute liver failure, acute-on-chronic liver failure, a high model for end-stage liver disease (MELD) score, and hepatocellular carcinoma at the upper limits of the Milan criteria.[277]
Transient elastography may reduce the need for endoscopic screening for varices in some patients with cirrhosis. Non-invasive assessments including the Baveno VI criteria, platelet-to-liver stiffness measurement ratio, liver stiffness measurement and spleen stiffness measurement have good predictive value for clinically significant varices and to identify patients at risk of bleeding. Screening for varices should balance the risks of severe acute respiratory disease coronavirus 2 (SARS-CoV-2) transmission from endoscopy against the risk of bleeding. Testing for SARS-CoV-2 is recommended before endoscopy. Endoscopy should not be delayed if the patient is negative for SARS-CoV-2 and the burden of COVID-19 in the local area is low.[277] Endoscopic eradication of esophageal varices should be performed following a variceal bleed.[328]
Patients with cirrhosis may have an attenuated response to SARS-CoV-2 vaccination; however, experts recommend priority vaccination for these patients because of the high COVID-19-related mortality in patients with decompensated cirrhosis.[329] A joint statement from the British Society of Gastroenterology, British Association for the Study of the Liver, NHS Blood & Transplant, and British Liver Trust advises that patients with chronic liver disease should consider SARS-CoV-2 vaccination with any of the available vaccines.[278] One cohort study of individuals with cirrhosis found that use of COVID-19 mRNA vaccines were associated with a reduction in COVID-19 infection and a reduction in hospitalization or death due to COVID-19 infection after 28 days.[330]
Clostridium difficile-associated disease
Signs & symptoms
Investigations
Differentials
Treatment algorithm
go to our full topic on Clostridium difficile-associated disease
As there is a potential risk of transmission of severe acute respiratory disease coronavirus 2 (SARS-CoV-2) via fecal microbiota transplantation (FMT), the US Food and Drug Administration (FDA) has made the following new recommendations for stool donated after 1 December 2019:[331]
- Screen donors to identify those who may be currently or recently infected with SARS-CoV-2.
- Test donors and/or donor stool for SARS-CoV-2, if possible.
- Patients should give informed consent after being advised about the potential risk of transmission of SARS-CoV-2 via FMT.
Stool used for FMT should have been donated before 1 December 2019 if these criteria are not met.
go to our full topic on Community-acquired pneumonia
Antibiotics should be started as soon as possible for patients in the community with suspected or confirmed secondary bacterial pneumonia. Patients should be advised to seek urgent medical help if their symptoms do not improve as expected, or rapidly or significantly worsen, regardless of whether they are taking an antibiotic. Antibiotics should not be used to prevent secondary bacterial pneumonia.[131]
Tests including respiratory and blood cultures, severe acute respiratory disease coronavirus 2 (SARS-CoV-2) polymerase chain reaction (PCR), throat samples for respiratory viral and atypical pathogen PCR, chest imaging, full blood count, and urinary legionella and pneumococcal antigen tests are recommended for patients admitted to hospital (or other acute care settings), to help identify secondary bacterial pneumonia and guide decisions about antibiotic use.[131] High C-reactive protein (CRP) levels do not distinguish between bacterial and COVID-19 pneumonia, but low CRP levels make secondary bacterial pneumonia less likely.[131]
Empiric antibiotics should be started if there is clinical suspicion of secondary bacterial infection in patients with COVID-19 in hospital.[131] Antibiotic treatment should be started within 4 hours of diagnosis and within 1 hour if the patient has suspected sepsis.[131] Choice of antibiotic will depend on local resistance data and availability. Specialist advice on antibiotic choice is recommended for patients who are immunocompromised, pregnant, in critical care, or who have a history of infection with resistant organisms or repeated infective exacerbations of lung disease.
Use of antibiotics should be reviewed at 24-48 hours, or as soon as test results are available. A narrower spectrum antibiotic should be used, if appropriate. Antibiotic treatment should stop after five days, unless there is an indication to continue.[131] Patients should be reassessed if they do not improve as expected, or if symptoms become significantly or rapidly worse. Specialist advice should be sought if there is clinical or microbiological evidence of infection and the person’s condition does not improve after 48-72 hours antibiotic treatment, or if an infection with multidrug-resistant bacteria is suspected.[131]
Where possible, clinicians should discuss the benefits, risks, and likely outcomes of any treatment with the patients, their relatives, and caregivers. The patient's preference about treatment and escalation plans should be sought, and clinicians should enquire about any advance care plans, advance decisions to refuse treatment, or "do not attempt resuscitation" decisions.[131]
World Health Organization guidelines advise that antibiotics should not be prescribed for patients with mild COVID-19 and should only be prescribed for patients moderate COVID-19 if there is clinical suspicion of a bacterial infection.[55]
Fever, cough, and shortness of breath may be symptoms of fungal pneumonias, including coccidioidomycosis, histoplasmosis, and blastomycosis. Fungal pneumonias may be indistinguishable from COVID-19 and bacterial pneumonias. The diagnosis should be considered if SARS-CoV-2 testing is negative, although coinfection with fungi and SARS-CoV-2 may occur.[332]
go to our full topic on Congenital heart disease
People with congenital heart disease (CHD) may be at increased risk for more severe COVID-19 infection, particularly those with more severe anatomic and physiologic features of CHD.[333][334] Simple biventricular defects, complex ventricular defects, and heart surgery are associated with severe COVID-19.[335] Additional considerations for management have been recommended during the current pandemic with strategies for prevention and management of COVID-19 in adults with CHD based on risk stratification.[334][336] For example, patients in the low risk category (e.g., those with normal ventricular function, normal exercise capacity, no relevant arrhythmia, no pulmonary hypertension) can be advised to take general prevention measures against COVID-19. Low risk patients with mild COVID-19 infection may be cared for at home with remote follow-up, but there should still be a low threshold for hospital admission if there is deterioration/progression or dyspnea. Adults with CHD in the high risk category (e.g., those with cyanotic conditions, univentricular palliated conditions, severe stenosis or regurgitation, severe ventricular dysfunction, or pulmonary arterial hypertension) are advised to follow stricter prevention measures, such as physical distancing. High risk patients with COVID-19 infection generally require hospital admission and involvement of a CHD specialist.
It is recommended that cardiac medications, including aspirin, ACE inhibitors, angiotensin-II receptor antagonists, beta-blockers, diuretics, and antiarrhythmic medications are continued during COVID-19 illness, unless there is a clear contraindication.[333] Clinicians should be aware of the QT-prolonging effects of some COVID-19 medications (e.g., chloroquine or hydroxychloroquine, azithromycin, lopinavir/ritonavir).
Adults with CHD at high risk are advised to follow meticulous physical distancing.[334]
In the UK, people aged 12 years or above with specific underlying health conditions that put them at risk of severe COVID-19 should be offered vaccination; this currently includes those with congenital heart disease.[28]
go to our full topic on Contraception - existing users
UK guidelines advise that a 6- to 12-month course of combined hormonal contraception can be provided without rechecking body mass index and blood pressure.
A 12-month course of a progestogen-only pill can be issued without a face-to-face review.[337]
Users of depot medroxyprogesterone may be offered ongoing contraception with desogestrel (if it is available as a progestogen-only pill).
Routine removals of long-acting contraception should be postponed and users counseled on contraceptive efficacy past the duration of licensed use.[337]
go to our full topic on Contraception - new users
Many new patients can be safely screened and offered a prescription for contraception remotely.[337]
UK guidance recommends that patients who wish to start contraception may be assessed remotely and offered a 6- to 12-month course of desogestrel (as a progestogen-only pill). If desogestrel is not suitable, complete remote assessment of medical eligibility and accurate self-reported blood pressure and body mass index is needed to prescribe combined hormonal contraception.[337]
Provision of long-acting reversible contraception for women who cannot tolerate oral contraception or who take teratogenic drugs should adhere to local infection control protocols.[337]
go to our full topic on Contraception - emergency
The Faculty of Sexual and Reproductive Healthcare in the UK recommends that a copper intrauterine device (Cu-IUD) should continue to be offered as first-line emergency contraception, where possible, to eligible patients. If Cu-IUD provision is delayed, additional oral emergency contraception should be offered. If a Cu-IUD is unsuitable or declined, clinicians should perform a remote assessment to determine the most suitable oral emergency contraception. In addition to this, clinicians should prescribe 3 months' supply of desogestrel (as a progestogen-only pill) and provide clear instructions about starting contraception and taking a pregnancy test.[337]
go to our full topic on Crohn disease
People with inflammatory bowel disease are at increased risk of COVID-19-related critical care admission or death, compared with the general population, after adjusting for confounding and mediating factors (hazard ratio [HR] 1.08, 95% confidence interval [CI] 1.01 to 1.16). South Asian, black, and mixed ethnicity people with inflammatory bowel disease have a higher risk of COVID-19-related death, compared with white people with inflammatory bowel disease.[64]
People taking tumor necrosis factor inhibitors or interleukin (IL)-12/IL-23 inhibitors do not have an increased risk of COVID-19-related critical care admission or death, compared with people taking standard systemic therapy (e.g., azathioprine).[64]
Patients should be advised to continue their current medications. Medication should only be stopped or reduced in discussion with a specialist. Preventing disease flares is a priority to reduce the risk of corticosteroid use and hospitalization.[338]
Blood tests to monitor response to therapy should be performed at the minimum safe frequency.[338]
International guidelines recommend that patients should stop taking methotrexate, thiopurines, or tofacitinib if they develop COVID-19. Detailed recommendations for stopping and restarting medications are given depending on the level of inflammatory bowel disease activity and severity of COVID-19 infection.[339][340] Decisions on surgery should be individualized for each patient with a multidisciplinary team.[341]
The British Society of Gastroenterology has published a position statement strongly supporting SARS-CoV-2 vaccination for patients with IBD.[342] A third dose (or booster dose) of SARS-CoV-2 vaccine is also recommended for all patients with IBD receiving immunosuppressive treatment and all patients with IBD who are extremely clinically vulnerable. A third dose of SARS-CoV-2 vaccine results in a serologic response in 84% of patients with immune-mediated inflammatory diseases who had a weak response to the standard 2-dose regimen.[343] Some immunosuppressive therapies may impact efficacy of COVID-19 vaccines in patients with IBD, and this should be considered in the timing of vaccinations.[343][344]
Prognostic models for predicting the probability of adverse COVID-19 outcomes in patients with IBD are being developed.[345]
go to our full topic on Cushing syndrome
Management of Cushing syndrome is complex and recommendations for clinical practice during the COVID-19 pandemic have been developed by an international group of experts.[346] They advise that patients with active Cushing syndrome are immunosuppressed, and should follow public health advice to minimize their risk of infection.[346] Patients with active Cushing syndrome who develop COVID-19 may not develop fever, and symptoms of dyspnea may be more severe.[347] Cushing syndrome is a hypercoagulable state and anticoagulation with low molecular weight heparin is recommended for hospitalized patients with COVID-19 and Cushing syndrome.[347] Patients with active Cushing syndrome are at risk of prolonged viral infection and secondary bacterial and fungal infection. Prolonged antiviral treatment and empirical antibiotic prophylaxis should be considered for hospitalized patients.[347]
Patients with severe Cushing syndrome should receive prophylaxis for Pneumocystis jirovecii; symptoms of COVID-19 may be similar to infections such asPneumocystis jirovecii pneumonia, and differentiation is needed to ensure appropriate treatment.[346]
go to our full topic on Cutaneous T-cell lymphoma
The European Organisation for Research and Treatment of Cancer has published guidelines on the management of patients with primary cutaneous lymphoma.[348] Patients with indolent or low-risk lymphomas who are on topical or systemic treatment should be consulted remotely to avoid trips to hospital. In some centers, phototherapy may be postponed.[348]
Patients with aggressive or late-stage lymphoma should be treated, because patients with controlled disease have fewer infections. Systemic therapy and radiation should be started if indicated. Treatment with medications including brentuximab vedotin, rituximab, gemcitabine, and extracorporeal photopheresis should be continued. Treatment should not be stopped prophylactically and risk/benefit discussion should take place for each patient. Patients in partial remission, particularly older patients or those with comorbidities, may consider stopping treatment or reducing its frequency.[348]
go to our full topic on Dementia
Patients with dementia, especially vascular dementia, have increased odds of developing COVID-19 compared with patients without dementia. Black patients with dementia are more likely to develop COVID-19 than white patients with dementia.[349] A US case control study reported that, for patients with COVID-19 and dementia, the risk of hospitalization is almost 60% and 6-month mortality is almost 21%.[349]
The European Academy of Neurology has published advice for healthcare professionals who look after patients with dementia.[350] Infection with COVID-19 may cause worsening confusion and precipitate delirium or acute cognitive decline.[350][351] A significant change in daily routine during the pandemic may trigger behavioral disturbances, and patients with dementia may be less able to comply with infection prevention measures such as washing hands or wearing a face covering. The following measures may be helpful: looking at old photographs, objects, or newspaper clippings, singing old songs, keeping to a regular schedule, simple exercise such as climbing a flight of stairs, using lighting appropriate to the time of day, going outside to orient a person to the time of day, assisting with hand hygiene, facilitating telephone and video calls from relatives, asking directly about symptoms of infection, and accounting for an individual's cognitive impairment when explaining the pandemic.[350]
Further resources are available at:
Alzheimer's Association: coronavirus (COVID-19) - tips for dementia caregivers Opens in new window
Alzheimer Europe: COVID-19 Opens in new window
Alzheimer’s Disease International. ADI offers advice and support during COVID-19 Opens in new window
NHS: Dementia wellbeing in the COVID-19 pandemic Opens in new window
go to our full topic on Diabetes (type 1)
Patients with type 1 diabetes are at higher risk for severe illness.[301][352] They are more likely to need hospitalization, intensive care, and mechanical ventilation if they develop COVID-19, compared with patients who do not have diabetes, and have a higher case fatality rate and increased odds of in-hospital death with COVID-19.[76][304][353][354][355][356] Poor glycemic control, hypertension, recent diabetic ketoacidosis, previous stroke, previous heart failure, renal impairment, body mass index <20 kg/m² or ≥40 kg/m², male sex, older age, nonwhite ethnicity, and socioeconomic deprivation are associated with increased mortality from COVID-19.[353][356][357][358][359] One meta-analysis found that diabetes was associated with a 14% increase in absolute risk of COVID-19 death.[360] Although prevalence of diabetes is lower in patients <50 years, one study found that younger patients with diabetes have an increased relative risk of fatal outcomes compared with older patients.[361] Patients with COVID-19 infection are at greater risk of hyperglycemia with ketones, including patients with newly diagnosed diabetes. Patients with type 1 diabetes are more likely to require intensive care admission than patients with type 2 diabetes due to the presence of diabetic ketoacidosis.[355] COVID-19 disease can precipitate atypical presentations of diabetes emergencies (e.g., mixed diabetic ketoacidosis and hyperosmolar states).[362] At admission hyperglycemia may also be an independent factor associated with poor prognosis for those hospitalized with COVID-19.[363][364]
UK guidance advises checking blood glucose and ketones in all patients with diabetes who are admitted to the hospital.[362] Out of the hospital, patients should follow their usual sick day rules, taking care to continue insulin, remain hydrated, and monitor blood glucose and ketones as appropriate.[365][366] Clinicians may need to prescribe additional blood glucose and ketone testing equipment to support increased monitoring. Patients admitted to intensive care may have insulin resistance and increased insulin requirements. There is a risk of hypoglycemia if feeding is interrupted (e.g., if the patient is nursed prone).[362] Specialist advice may be needed, particularly for patients who have severe illness on admission or if infusion pumps for insulin are not available.[362]
Studies suggest that SARS-CoV-2 infection might also be associated with new-onset diabetes.[367] In the US, one study found that people aged <18 years with COVID-19 were more likely to receive a new diabetes diagnosis more than 30 days after infection compared with those without COVID-19 and those with prepandemic acute respiratory infections. Non-SARS-CoV-2 respiratory infection was not associated with an increased risk for diabetes.[368]
A panel of international experts has published practical recommendations for the management of diabetes in patients with and without COVID-19.[369] They advise that those with diabetes who have not been infected with severe acute respiratory disease coronavirus 2 (SARS-CoV-2) should intensify their metabolic control as a measure to prevent COVID-19 infection, including blood pressure and lipid control, and patients should reduce their risk of exposure by having remote healthcare consultations where possible and following public health advice on hand hygiene and physical distancing. The panel recommends that patients with diabetes and COVID-19 require continuous and reliable glycemic control and that they continue antihypertensive and lipid-lowering treatments. The panel also advises that patients without diabetes are monitored for new-onset diabetes triggered by SARS-CoV-2 infection, particularly those at high risk for metabolic disease. People with type 1 diabetes are more susceptible to infection and require more intensive monitoring and supportive therapy to reduce the risk of metabolic decompensation, including diabetic ketoacidosis; the panel advised that patients are made aware of this and reminded about typical symptoms, home-measurement of urine or blood ketones, sick day rules, and seeking medical advice early if concerned.[369] Guidelines for the management of dexamethasone-related hyperglycemia in patients with diabetes and COVID-19 have also been published.[370]
In the UK, people aged 12 years or above with specific underlying health conditions that put them at risk of severe COVID-19 should be offered vaccination; this currently includes those with type 1 diabetes.[28]
Further diabetes resources are available at:
American Diabetes Association: COVID-19 professional resources Opens in new window
go to our full topic on Diabetes (type 2)
Patients with type 2 diabetes are at higher risk for severe illness.[301][352] They are more likely to need intensive care and mechanical ventilation if they develop COVID-19, compared with patients who do not have diabetes, and have a higher case fatality rate and increased odds of in-hospital death with COVID-19.[76][303][304][354][356][371] Poor glycemic control, hypertension, previous stroke, previous heart failure, renal impairment, cancer, body mass index <20 kg/m² or ≥40 kg/m², male sex, older age, nonwhite ethnicity, socioeconomic deprivation, and elevated C-reactive protein are associated with increased mortality from COVID-19.[357][358][359][372][373][374] One meta-analysis found that diabetes was associated with a 14% increase in absolute risk of COVID-19 death.[360] Although prevalence of diabetes is lower in patients <50 years, one meta-analysis found that younger patients with diabetes have an increased relative risk of fatal outcomes compared with older patients.[361]
Medications for diabetes may also impact the course of COVID-19 illness. Use of insulin is associated with increased risk of hospitalization, intensive care admission, and death.[373][374][375] Metformin use is associated with decreased mortality and reduced infection severity in people with diabetes and COVID-19.[374][376][377] In one study, use of beta-blockers was associated with increased mortality, and use of dipeptidyl peptidase-4 (DPP4) inhibitors was associated with decreased mortality.[373] In a meta-analysis of observational studies, DPP4 inhibitors were associated with a nonsignificant decrease (of 3%) in the risk for COVID-19-related death, but when given in an inpatient setting, the decrease in risk for COVID-19-related death was 50%.[378] Glucagon-like peptide-1 receptor agonist (GLP-1RA) use and sodium-glucose co-transporter-2 (SGLT2) inhibitor use have been associated with lower odds of mortality and other adverse outcomes compared with DPP4 inhibitors.[379] A large cohort study of US veterans reported that SGLT2 inhibitor use, GLP-1RA use, and angiotensin receptor blocker use are associated with decreased odds of hospitalization.[374] SGLT2 inhibitor use was associated with decreased mortality.[374] Another meta-analysis suggested that pre-admission use of GLP-1RA may be associated with reduced COVID-19 mortality in patients with type 2 diabetes.[380] Randomized controlled trials are needed.
Patients with severe COVID-19 who are taking metformin should be monitored for development of acidosis, lactic acidosis, and reduced kidney function. Patients with COVID-19 infection appear to have a greater risk of hyperglycemia with ketones, including patients with type 2 diabetes and those with newly diagnosed diabetes. COVID-19 disease can precipitate atypical presentations of diabetes emergencies (e.g., mixed diabetic ketoacidosis and hyperosmolar states).[362] At admission hyperglycemia may also be an independent factor associated with poor prognosis for those hospitalized with and without COVID-19.[363]
Patients taking SGLT2 inhibitors should be advised to stop these if they become unwell, to reduce their risk of developing diabetic ketoacidosis.[369] Metformin may need to be temporarily stopped if patients are at risk of dehydration.[366][369] UK guidance advises temporarily stopping SGLT2 inhibitors and metformin in all patients admitted to the hospital.[362] Blood glucose and ketones should be checked in all patients with diabetes who are admitted to the hospital.[362]
Patients should follow their usual sick day rules, taking care to continue insulin, remain hydrated, and monitor blood glucose and ketones as appropriate.[365][366] Clinicians may need to prescribe additional blood glucose and ketone testing equipment to support increased monitoring. Patients admitted to intensive care may have insulin resistance and increased insulin requirements. There is a risk of hypoglycemia if feeding is interrupted (e.g., if the patient is nursed prone).[362] Specialist advice may be needed, particularly for patients who have severe illness on admission or if infusion pumps for insulin are not available.[362]
Studies suggest that SARS-CoV-2 infection might also be associated with new-onset diabetes.[367] In the US, one study found that people aged <18 years with COVID-19 were more likely to receive a new diabetes diagnosis more than 30 days after infection compared with those without COVID-19 and those with prepandemic acute respiratory infections. Non-SARS-CoV-2 respiratory infection was not associated with an increased risk for diabetes.[368]
A panel of international experts has published practical recommendations for the management of diabetes in patients with or without COVID-19.[369] They advise that those with diabetes who have not been infected with severe acute respiratory disease coronavirus 2 (SARS-CoV-2) should intensify their metabolic control as a measure to prevent COVID-19 infection, including blood pressure and lipid control, and patients should reduce their risk of exposure by having remote healthcare consultations where possible and following public health advice on hand hygiene and physical distancing. The panel recommends that patients with diabetes and COVID-19 require continuous and reliable glycemic control and that they continue antihypertensive and lipid-lowering treatments. The panel also advises that patients without diabetes are monitored for new-onset diabetes triggered by SARS-CoV-2 infection, particularly those at high risk for metabolic disease. People with type 2 diabetes and comorbid conditions such as obesity and fatty liver disease may be at increased risk for more severe COVID-19 disease, and those with fatty liver disease may be screened for hyperinflammation using trends in laboratory tests to determine where immunosuppression might improve the outcome.[369] Recommendations for use of antidiabetic drugs in patients with COVID-19 have been published.[381] Guidelines for the management of dexamethasone-related hyperglycemia in patients with diabetes and COVID-19 have also been published.[370]
One study in the UK looking at the indirect effects of the COVID-19 pandemic on type 2 diabetes between March and December 2020 found reductions in diagnosis and monitoring of type 2 diabetes. Those aged 65 years and older, men, and people from deprived areas had the greatest reductions in diagnosis rates.[219]
In the UK, people aged 12 years or above with specific underlying health conditions that put them at risk of severe COVID-19 should be offered vaccination; this currently includes those with type 2 diabetes.[28]
Further diabetes resources are available at:
American Diabetes Association: COVID-19 professional resources Opens in new window
go to our full topic on Eczema
The British Association of Dermatologists has issued advice to patients with eczema affecting the hands. Patients should adhere to national advice to wash hands with soap and water. Patients should be advised to pat the skin dry and apply emollient generously after handwashing and when the skin feels dry. Patients should be advised that applying emollient before sleep and covering the hands with cotton gloves may help their condition. Patients should protect their hands using gloves if they need to handle detergent for purposes other than handwashing (e.g., washing a child's hair, washing dishes, or cleaning).[382] Patients with facial dermatitis are advised to apply a barrier cream before wearing a face mask and to avoid masks containing metal wires in case of nickel allergy.[253]
For information on managing patients with eczema who take drugs that affect the immune response, please see the section "considerations for patients with dermatologic conditions receiving drugs that affect the immune response" in the introduction to this topic.
go to our full topic on Epilepsy
The European Academy of Neurology has published advice on the management of epilepsy during the COVID-19 pandemic.[383] Patients with epilepsy should be advised to continue taking their medication, and regular follow-up should continue. Fever can trigger seizures in some people with epilepsy, and experts recommend using antipyretics if people with epilepsy develop COVID-19. Coronavirus infection per se is not known to trigger seizures.[384]
go to our full topic on Essential hypertension
Hypertension is associated with an increased risk of severe COVID-19 and death; this association is stronger in younger populations.[76][303][304] One meta-analysis found that hypertension was associated with an 11% increase in absolute risk of COVID-19 death.[360]
Meta-analysis found no association between ACE inhibitor or angiotensin-II receptor antagonist use and mortality from COVID-19, need for mechanical ventilation, or adverse cardiac events.[105] Use of ACE inhibitors or angiotensin-II receptor antagonists has been associated with increased survival in patients with COVID-19.[108][109] Calcium-channel blocker use was associated with decreased COVID-19 mortality in patients with hypertension.[385]
British, European, American, and Asian heart groups advise that patients should continue to take ACE inhibitors and angiotensin-II receptor antagonists as prescribed.[110][111][112][113] Any change in medication should be based on individual patient risk assessment.
go to our full topic on Food allergy
The British Society for Allergy and Clinical Immunology has recommended modifications to pediatric allergy services during the pandemic. Most new patient and follow-up visits can be performed using telehealth. Allergy testing, and most food challenges, can be deferred. Priority for hospital testing should be given to food challenges where there is a critical nutritional need and it would be unsafe for the parent or caregiver to perform the food challenges: for example, in infants with milk/soya/hydrolysate food protein enterocolitis syndrome. Where possible, dietitians should contact patients on multiple food exclusions to establish whether food shortages are a concern; additional vitamins, supplements, or formulas may be needed. Initiation and updosing of food immunotherapy should be deferred.[386] Sublingual and subcutaneous immunotherapy should be continued as usual in patients who have no symptoms of COVID-19 and have not been exposed to infected individuals within the last 14 days, in patients who have had a negative reverse-transcriptase polymerase chain reaction (RT-PCR) test and in patients who have serum IgG antibodies to severe acute respiratory disease coronavirus 2 (SARS-CoV-2) without virus-specific IgM. Patients who develop COVID-19, have been exposed to infected individuals, or have a positive RT-PCR test should discontinue allergen immunotherapy, independent of disease severity, until symptoms have resolved or adequate quarantine has been performed.[387]
A consensus statement from the Italian Society of Pediatric Allergy and Immunology advises that it may be more difficult for children to access specialty allergy foods during the pandemic, and the potential requirement to try new products increases the risk of an allergic reaction. The society recommends that children have a written action plan with emergency drug doses and have two available epinephrine autoinjectors.[253]
go to our full topic on Giant cell arteritis
A cohort study of 500,000 people in the UK found that autoimmune disease and history of oral corticosteroid were associated with increased risk of fatal COVID-19.[275] Experts recommend that patients taking long-term glucocorticoids for giant cell arteritis (GCA) should titrate the dose down to the lowest possible dose that achieves symptom control. Patients at risk of adrenal insufficiency who develop COVID-19 may require additional doses of glucocorticoid to prevent an adrenal crisis. Adjuvant immunosuppression should be considered at an early stage, since major relapse of GCA might require in-person consultation and/or significant glucocorticoid dose increases.[388]
Clinicians are advised to engage patients with rheumatic disease in a shared decision-making process to discuss receiving a severe acute respiratory disease coronavirus 2 (SARS-CoV-2) vaccine. The response to vaccination is likely to be blunted in patients receiving systemic immunomodulatory therapies compared with the general population. The American College of Rheumatology advises that, beyond known allergies to vaccine components, there are no additional contraindications to SARS-CoV-2 vaccination for patients with autoimmune and inflammatory rheumatic diseases. Vaccination should ideally occur when rheumatologic disease is well controlled.[389] EULAR recommends that patients with rheumatic and musculoskeletal diseases are strongly advised to receive a SARS-CoV-2 vaccination with any of the vaccines approved in their country.[390]
Hematopoietic stem cell transplantation
UK guidelines advise that for at least 2 weeks before receiving hematopoietic stem cell transplantation (HSCT), patients should follow professional advice on how to minimize their risk of respiratory infection, including COVID-19.[391] All patients receiving HSCT should be tested for severe acute respiratory disease coronavirus 2 (SARS-CoV-2) and have a negative test before starting conditioning.[391][392] Polymerase chain reaction testing is preferred.[392] Patients should also be tested if they have any symptoms of COVID-19.
If COVID-19 is confirmed, HSCT should be deferred. The length of deferral is guided by the patient’s risk of disease progression, morbidity and mortality, and their symptom burden from COVID-19.[392] Patients who have been in close contact with someone with COVID-19 in the last week may be considered for deferral; individual risks and benefits should be assessed.[391]
UK guidelines advise that before HSCT, donors should follow clinical advice on reducing risk of exposure to COVID-19.[391] Donors should be tested at the initial assessment, before stem cells or donor lymphocytes are harvested, and 72 hours before starting conditioning if fresh cell donations are needed. Donors who test positive should defer donations for 2 weeks after their symptoms resolve; however, if less than 2 weeks has passed and donation is urgent, this should be referred for risk assessment.[391]
HSCT should be deferred if possible, particularly for myeloma, low-grade lymphoproliferative conditions, chronic hematologic conditions, and nonmalignant indications.[393]
Following transplantation, patients are at high risk of severe sickness and should follow the national recommendations for protecting themselves.[301][391][392][394]
Clinicians in Italy have reported assessing patients within 3 months of transplantation and without symptoms of COVID-19 in-person. Patients who are 3 to 24 months post-transplantation may be screened for symptoms of infection or graft-versus-host disease and triaged to in-person or telehealth consultations as appropriate.[395]
Patients who are profoundly immunosuppressed following HSCT may shed viable SARS-CoV-2 virus for up to 78 days after the onset of symptoms.[396]
Further hematology resources are available at:
go to our full topic on Hepatic steatosis
Patients with non-alcoholic fatty liver disease (NAFLD) are at higher risk of disease progression if they contract COVID-19, compared with people without NAFLD.[397] European guidelines recommend that early hospital admission should be considered for patients with NAFLD.[277] It is unclear whether NAFLD is an independent risk factor for poor COVID-19 prognosis or whether observations are confounded by the presence of obesity, hypertension, or type 2 diabetes. Treatment for arterial hypertension should be continued.[277] Clinicians should also optimize diabetes management, promote weight loss, and give nutritional guidance, which may prevent a severe disease course in future severe acute respiratory syndrome coronavirus 2 infection. People with type 2 diabetes and fatty liver disease may be screened for hyperinflammation using trends in laboratory tests to determine where immunosuppression might improve the outcome.[369]
go to our full topic on Hepatitis B
Usual treatment should be continued for patients with chronic hepatitis B (HBV) infection who are established on treatment. In patients without COVID-19, treatment should be initiated according to local or national guidelines.[277] The impact of interferon alfa on systemic inflammation in patients with COVID-19 is not known. European Association for the Study of the Liver (EASL) guidelines recommend that alternative agents should be considered when starting treatment for HBV during the pandemic.[277]
Patients who have chronic, occult, or resolved HBV infection and COVID-19 should be considered for antiviral therapy to avoid HBV reactivation and hepatitis flare if they receive immunosuppressive agents for treatment of COVID-19.[277][328] EASL guidelines advise that initiation of antiviral therapy is usually not warranted in patients with HBV and COVID-19 and may be deferred until recovery. In patients with suspected severe acute HBV hepatitis or a flare of disease activity, a specialist should be consulted to decide whether to initiate antiviral therapy.[277] Asia-Pacific guidelines recommend that patients who are newly diagnosed with HBV infection at the time of presentation with COVID-19 should be started on antiviral therapy.[328] Use of tenofovir with lopinavir/ritonavir is relatively contraindicated as the concentration of tenofovir might be increased when these drugs are used together.[328]
In the UK, people aged 16 years or above with specific underlying health conditions that put them at risk of severe COVID-19 should be offered vaccination; this currently includes those with chronic hepatitis.[28]
go to our full topic on Hepatocellular carcinoma
European and Asia-Pacific position papers recognize that screening for hepatocellular carcinoma (HCC) using ultrasound may need to be deferred during the COVID-19 pandemic, depending on local resources (including availability of treatment options) and individual patient risk assessment. Patients at highest risk should be prioritized for screening, including patients with: elevated alfa-fetoprotein levels, chronic hepatitis B, advanced cirrhosis, and nonalcoholic steatohepatosis/diabetes.[277][328] HCC surveillance can be deferred until after recovery in patients who develop COVID-19.[277] The European Association for the Study of the Liver recommends that guidelines for treatment of patients with HCC should be followed, including multidisciplinary board review to make treatment recommendations, systemic treatments, and evaluation for liver transplant.[277] The Asia-Pacific Working Group for Liver Derangement during the COVID-19 Pandemic recommends that patients with HCC who have COVID-19 should have treatment for HCC deferred until after recovery from COVID-19. For those who have had surgical resection deferred, bridging transarterial chemoembolization, radiofrequency ablation, or systemic chemotherapy might be considered in selected patients.[328]
Organ donations and transplants are likely to be reduced in many countries. Listing for transplantation should be restricted to patients at the upper limit of the Milan criteria.[277] Guidelines emphasize the importance of vaccination againstStreptococcus pneumoniae and influenza.[277]
Recommendations for the management of patients with primary hepatic malignancies during the COVID-19 pandemic have also been developed by an international group of experts.[398] They propose treatment recommendations for different stages of HCC (according to the Barcelona Clinic Liver Cancer classification system), specifically surgery, locoregional, and systemic therapy, and suggest strategies to modify risk and assist with multidisciplinary treatment decision-making.
go to our full topic on HIV infection
There is little evidence that the COVID-19 infection rate is different in people living with HIV compared with those without HIV infection in European and North American populations, after accounting for demographic factors.[399][400][401][402][403] One cohort study conducted in the US reported lower prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin G (IgG) amongst people living with HIV compared with people without HIV (3.7% and 7.4% respectively), which may reflect increased shielding precautions taken by people living with HIV. SARS-CoV-2 IgG titers were lower in people living with HIV with a history of PCR-confirmed SARS-CoV-2 infection compared with people without HIV with previous SARS-CoV-2 infection.[404]
People living with HIV may be at increased risk for more severe disease. Data from the National COVID Cohort Collaborative (N3C) in the US found that after adjusting for all covariates, people with HIV had higher odds of COVID-19-related death and hospitalization, but lower odds of mild or moderate COVID-19 than people without HIV.[405] The association between HIV and COVID-19-related death and hospitalization was stronger among older age groups, male, Black, African-American, Hispanic, or Latinx adults. People with lower CD4 cell count (<200 cells per microliter) were at higher risk of poor outcomes.[405]
One systematic review reported that two-thirds of people with HIV who developed COVID-19 had mild or moderate symptoms, most commonly fever and cough.[406] Age >50 years, male sex, and multimorbidity are associated with increased risk of death from COVID-19.[406] A cohort study in the US found that people living with HIV who are diagnosed with COVID-19 have an increased risk of hospitalization compared with people without HIV, and hospitalization is associated with progression of HIV disease stage.[407] Another cohort study in Spain found that people living with HIV with immunosuppression, detectable HIV viremia, chronic comorbidities, and some subpopulations (such as older people and migrants) may be at increased risk of severe outcomes from COVID-19.[403]
The clinical course of COVID-19 in the African population is also under investigation; one study reports that patients with HIV in South Africa had a higher risk of death compared with people without HIV, irrespective of viral suppression.[408] A study conducted in Zambia reported that HIV status alone was not associated with COVID-19 infection severity or mortality. However, patients with severe HIV disease (hemoglobin <8.0 g/dL, CD2 <200 cells/microliter, active TB, or body mass index <18.5 kg/m²) were more likely to develop severe COVID-19 and were at increased risk of death from COVID-19.[409]
Guidance from the US, UK, and Europe recognizes that many people living with HIV are older and have comorbid chronic medical conditions such as cardiovascular disease or lung disease, which increase the risk for severe COVID-19 infection. The guidelines recommend that until more is known, additional caution is advised for all people with HIV, especially if advanced (i.e., CD4 cell count <200/microliter) or poorly controlled. Influenza and pneumococcal vaccinations should be kept up to date.[410][411] Advice from the Infectious Diseases Society of America and HIV Medicine Association states that people with HIV have a normal life expectancy and a readily treatable infection, therefore HIV status and current HIV control should not be factors in decision-making regarding potentially life-saving interventions or enrollment into clinical trials. Antiretroviral therapy should be continued in the hospital without interruption. Changes in antiretroviral therapy are generally not recommended. Routine viral load monitoring in patients with suppressed HIV and no adherence concerns can be delayed for up to 6 months to reduce the burden on testing laboratories. Viral load testing should be prioritized for patients with adherence concerns, patients whose HIV is not fully suppressed, patients starting a new drug regimen, and pregnant patients.[412] Pre-exposure prophylaxis to prevent HIV infection should be taken as directed; there is no evidence that it is effective against COVID-19.[413] The Centers for Disease Control and Prevention recommends that people with HIV can receive the Pfizer-BioNTech and Moderna vaccines if they have no contraindications. It is not yet known whether the level of protection is as strong for people with HIV as people without HIV.[412] One prospective cohort study of CoronaVac found that immunogenicity in people with HIV was reduced compared with people with no known immunosuppression.[414]
Further resources are available at:
WHO: Q&A - HIV, antiretrovirals and COVID-19 Opens in new window
Guidance for COVID-19 and people with HIV Opens in new window
HIVinfo: HIV and opportunistic infections, coinfections, and conditions Opens in new window
go to our full topic on Hodgkin lymphoma
Analysis of US health-insurance claims data has shown that lymphoma is a significant risk factor for fatal COVID-19.[82] Interim treatment guidelines for the management of adult patients during the pandemic have been provided by experts from the UK and also from Australia and New Zealand.[311][415] Hodgkin lymphoma is curable in most patients and delivery of dose- and time-intensive treatment remains a high priority; recommendations are given for patients with early-stage and advanced-stage disease, elderly Hodgkin, relapsed Hodgkin, and nodular lymphocyte-predominant Hodgkin.
The American Society of Hematology has also published advice on the treatment of Hodgkin lymphoma.[416] Chemotherapy followed by interim staging positron emission tomography/computed tomography (PET/CT) is usually preferred to chemotherapy plus radiation therapy for early and advanced-stage disease because fewer hospital visits are needed. The International Lymphoma Radiation Oncology Group has published emergency guidelines for radiation therapy in hematologic malignancies, should radiation therapy be necessary. Alternative dose fractionations may be given.[101] Bleomycin should be omitted following a negative PET/CT to reduce the risk of bleomycin pneumonitis. Many experts recommend increased use of granulocyte-colony stimulating factor to reduce neutropenia and use of prophylactic antibiotics when neutropenia is expected. Recommendations are also given for older adult and pediatric patients and those with relapsed or refractory disease.[416] In general, it is considered safe and appropriate for people with Hodgkin lymphoma to be vaccinated against severe acute respiratory syndrome coronavirus 2.[416]
go to our full topic on Idiopathic pulmonary fibrosis
Patients with idiopathic pulmonary fibrosis are at increased risk of hospitalization and death due to COVID-19.[264] The Canadian Thoracic Society has published guidelines for the management of patients with interstitial lung disease, including idiopathic pulmonary fibrosis, during the pandemic.[417]
Decisions about stopping, adjusting, and restarting treatment in patients who develop COVID-19 should be made in conjunction with the patient's specialist team.[417]
Initiation of immunotherapy should be deferred in patients with newly diagnosed or suspected COVID-19.[417] Clinicians should use the lowest effective dose of immunotherapy in patients without COVID-19.[417] Antifibrotic drugs may be continued if the patient's blood parameters are in the acceptable range and there is no other reason to stop (e.g., significant adverse effects).[417]
Antifibrotic therapy does not increase the risk of getting COVID-19 or make severe disease more likely. Patients who are already taking antifibrotic therapy should continue. Patients with a new diagnosis of idiopathic pulmonary fibrosis may start antifibrotic therapy if a multidisciplinary team confirms the diagnosis, usual eligibility criteria are satisfied, and the appropriate blood monitoring can be performed.[417]
Clinical judgment is needed to determine the urgency of diagnostic and therapeutic bronchoscopy. Elective bronchoscopy or lung biopsy should be deferred in patients with SARS-CoV-2 infection.[417]
Patients with persistent respiratory symptoms following recovery from COVID-19 should be evaluated for post-COVID pulmonary fibrosis and/or exacerbation of pre-existing interstitial lung disease.[417]
Further resources are available at:
British Thoracic Society. COVID-19: information for the respiratory community. Opens in new window
go to our full topic on Immune thrombocytopenia
The American Society for Hematology has published advice on management of immune thrombocytopenia (immune thrombocytopenic purpura [ITP]) during the pandemic. Hospital visits should be minimized and treatment guided by symptom management rather than frequent platelet counts. Viral infections can trigger acute ITP or trigger exacerbations in patients with stable disease, so testing for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is recommended in these patients.[418]
Treatment of new patients should be individualized depending on: urgency of need to increase the platelet count, amount of bleeding, comorbidities, minimizing exposure to SARS-CoV-2, and usual practice. Most patients with immune thrombocytopenia do not experience severe bleeding with platelet counts above 20,000/microliter, in the absence of comorbidities. Intravenous immune globulin (IVIG) or oral thrombopoietic agents (e.g., eltrombopag or avatrombopag) are first line because they are not immunosuppressive. No change to treatment is recommended for patients who are stable on low doses of immunosuppressive drugs. Treatment change may be considered for patients taking higher doses of immunosuppressive drugs or corticosteroids; however, this must be balanced against the increased monitoring requirements and risk of relapse. If indicated, IVIG or oral thrombopoietic agents may allow dose reduction or cessation of immunosuppressive medication or corticosteroids. Rituximab should be avoided if possible.[418]
If a patient with immune thrombocytopenia develops COVID-19, IVIG should be given to maintain the platelet count above 10,000-20,000/microliter; platelet transfusion should be reserved to treat bleeding or cover procedures with a high bleeding risk. If the patient already takes a thrombopoietic agent, the dose can be increased or a second agent may be started. Treatments for COVID-19 illness should be given as appropriate. If the patient has had a splenectomy, intravenous antibiotics should be administered until bacterial cultures are documented negative, even if COVID-19 is strongly suspected as the cause.[418]
The guidance advises that vaccine administration in general can occasionally result in a drop in the platelet count in stable ITP patients; however, current knowledge suggests that the expected benefits of receiving the SARS-CoV-2 vaccine likely outweigh the risks.[418] Taking baseline and postvaccination platelet counts may be considered, particularly in those with ongoing thrombocytopenia or a history of unstable platelet counts.
go to our full topic on Influenza
Signs and symptoms of influenza infection and COVID-19 infection are similar and can be difficult to distinguish clinically; only testing can distinguish between them. The US National Institutes of Health (NIH) COVID-19 Treatment Guidelines recommend testing for both SARS-CoV-2 and influenza viruses in all hospitalized patients with acute respiratory illness when both viruses are co-circulating.[91] The guidelines advise that treatment of influenza is the same in all patients, regardless of SARS-CoV-2 coinfection, and that hospitalized patients should be started on empiric treatment for influenza as soon as possible without waiting for influenza testing results (antiviral treatment for influenza can be stopped when influenza has been ruled out by nucleic acid detection assay in upper respiratory tract specimens for nonintubated patients, and in both upper and lower respiratory tract specimens for intubated patients).[91]
The US Centers for Disease Control and Prevention recommend influenza vaccination of persons aged ≥6 months. Influenza vaccine and COVID-19 vaccine may be administered at the same visit. Influenza vaccinations should be deferred for patients with suspected or confirmed COVID-19 until they have recovered from the acute illness and no longer need to isolate.[123] The UK 2022-2023 influenza immunization program includes all children ages 2-3 years on 31 August 2022, all primary school aged children (from reception to Year 6), secondary school-aged children focusing on Years 7, 8 and 9, people ages 6 months to under 50 years in clinical risk groups, pregnant women, people ages 50 years and older, people living in long-stay residential homes, close contacts of immunocompromised people, carers, and frontline health and social care staff.[124][125] Influenza activity levels were very low globally in 2020-2021, and a resurgence in influenza activity to prepandemic levels or higher is expected in winter 2022-2023. Influenza vaccination is important to reduce morbidity and mortality from influenza, and to reduce hospitalizations during a time when there may be co-circulation of influenza, SARS-CoV-2, and other respiratory viruses.[124]
One study found that influenza infection was associated with a lower risk of SARS-CoV-2 infection, indicating that there may be pathogenic competition between them.[385] Coinfection with influenza and SARS-CoV-2 has been associated with an increased risk of death or severe disease.[419][420]
go to our full topic on Learning disability
People with learning disabilities (LDs) are at higher risk of death from COVID-19 than the general population. Analysis of US health-insurance claims data found that intellectual disability was associated with a 2.75-fold increase in the odds of death from COVID-19.[82] A study of nearly 65,000,000 patients across 547 healthcare organizations in the US found that having an intellectual disability was the strongest independent risk factor for a COVID-19 diagnosis and the strongest independent risk factor for COVID-19 mortality other than age.[421]
In England, one study found that adults with learning disability had a 5-fold increased risk of hospitalization with COVID-19, and an 8-fold increased risk of death from COVID-19, during the first and second waves of the pandemic. People who have severe or profound learning difficulty, Down syndrome, cerebral palsy, or live in residential care are at particularly high risk.[422] Another study found that in the UK, compared with the general population, people living with intellectual disabilities were already at a higher risk of all-cause mortality before the pandemic and that during the pandemic there was a further steep increase in mortality risk relative to the general population.[158]
Guidance recommends ensuring that patients with LDs have a handheld summary of their medical information, activities, and preferences to support staff in caring for them if admitted to hospital. Community LD teams are encouraged to work with people in their care and their families to develop COVID-19 care plans, which should include any issues associated with diagnostic overshadowing, views of parents, carers and family members, any reasonable adjustments required, communication needs, specialist mental health support, anticipatory care plans, and any end-of-life or do not attempt cardiopulmonary resuscitation discussions.[423]
In the UK, people aged 12 years or above with specific underlying health conditions that put them at risk of severe COVID-19 should be offered vaccination; this currently includes those with learning disability.[28]
go to our full topic on Liver dysfunction
Patients with COVID-19 may have abnormal liver function tests, including elevated aminotransferases and mildly elevated bilirubin. Low serum albumin on admission to the hospital is a marker of COVID-19 severity. Recommendations from the American Association for Study of Liver Diseases (AASLD), the American Gastroenterological Association (AGA), and the Asia-Pacific Working Group for Liver Derangement during the COVID-19 Pandemic all advise regular monitoring of liver biochemistries in all hospitalized patients with COVID-19, particularly those treated with remdesivir or tocilizumab, regardless of baseline values.[276][328][424] The Asia-Pacific Working Group advises that while the optimal interval for liver tests is uncertain, it would be reasonable to monitor liver tests twice weekly in patients on potentially hepatotoxic medication and patients with pre-existing liver disease, and more frequently in any patients with abnormal liver function.[328] The AASLD also advises that abnormal liver biochemistries should not be a contraindication to using investigational or off-label therapeutics for COVID-19, although aspartate aminotransferase (AST) or alanine aminotransferase (ALT) levels >5 times the upper limit of normal (ULN) may exclude patients from consideration of some investigational agents.[276] The Asia-Pacific Working Group recommends that off-label COVID-19 therapies may be used with caution and close monitoring in those with abnormal liver function; the treatment should be stopped in those with moderate-to-severe liver injury (ie, ALT >5 times ULN or alkaline phosphatase >2 times ULN, and total bilirubin >2 times ULN or presence of coagulopathy or clinical decompensation).[328] Other causes of abnormal liver function tests, including viral hepatitides, should be considered in patients with COVID-19 and abnormal liver biochemistries.[328][424] In patients with autoimmune hepatitis or liver transplant recipients who develop COVID-19, suspected disease flare or acute cellular rejection should be confirmed on biopsy.[276] The Asia-Pacific Working Group recommends screening for hepatitis B surface antigen (HBsAg) in patients who are receiving systemic corticosteroids or other potent immunosuppressants for 7 days or longer as COVID-19 therapy. Tocilizumab has been associated with HBV reactivation. European guidelines recommend screening with HBV serology before initiating treatment with tocilizumab.[277] In patients with hepatitis C virus (HCV) infection, concomitant use of a protease inhibitor-containing direct-acting antiviral regimen with lopinavir/ritonavir is contraindicated, as protease inhibitor concentrations may increase when these drugs are used together, risking ALT elevations.[328]
A joint statement from the British Society of Gastroenterology, British Association for the Study of the Liver, NHS Blood & Transplant, and British Liver Trust advises that patients with chronic liver disease, autoimmune hepatitis, and those post-liver transplant should consider SARS-CoV-2 vaccination with any of the available vaccines.[278]
go to our full topic on Migraine
The European Academy of Neurology has published advice on the management of migraine during the COVID-19 pandemic.[425] Patients with migraine should be encouraged to continue managing lifestyle and dietary triggers: for example, stress, diet, alcohol consumption, and sleep. Social isolation, anxiety, and depression may negatively affect medication overuse, and medications for treatment of acute migraine should be limited to less than two times per week. Nonsteroidal anti-inflammatory drugs should be used as needed: they have established efficacy in the treatment of acute migraine, and there is no evidence that they can exacerbate symptoms of COVID-19. Acetaminophen and triptans may also be used as required for acute attacks.
go to our full topic on Mitral regurgitation
A position statement from the American College of Cardiology and the Society for Cardiovascular Angiography and Interventions provides recommendations for the triage of patients referred for structural heart disease interventions during the pandemic.[254] The recommendations suggest that percutaneous mitral valve repair (edge-to-edge repair) can be safely deferred in the majority of patients with mitral regurgitation (MR), but some high risk patients should be considered for edge-to-edge repair during the pandemic. Patients who are deferred should be contacted on a weekly basis to monitor for decompensation.Valve-in-valve transcatheter mitral valve replacement (TMVR) is resource-intensive and should be deferred if the patient can be sufficiently managed on medical therapy in the interim. Valve-in-valve TMVR should be considered for patients with severe bioprosthetic mitral stenosis or mitral regurgitation who are inpatients with congestive heart failure or outpatients who have had hospitalizations for congestive heart failure within 30 days despite optimized guideline-directed medical therapy.
The British Heart Valve Society has published recommendations for the outpatient management of heart valve disease following the COVID-19 pandemic. They recommend that patients with severe symptomatic MR should be considered for urgent surgical repair; transcatheter mitral valve therapies may be considered in decompensated MR when timely access to surgical repair/replacement is not possible.[255]
The European Society of Cardiology advises that priority should be given to the treatment of patients with acute primary MR complicating acute myocardial infarction or infective endocarditis, and people with severe symptomatic MR who remain symptomatic despite guideline-directed treatment and seem likely to require hospital admission. The choice of intervention should be guided by the multidisciplinary team.[193]
go to our full topic on Mucormycosis
Mucormycosis has been increasingly reported in patients with coronavirus disease 2019 (COVID-19), particularly in patients who have diabetes and have also received corticosteroids.[426][427][428][429][430][431] Clinicians should have a low threshold of suspicion for diagnosis. Warning signs and symptoms include: nasal congestion; blackish/bloody nasal discharge; sinus or facial pain; toothache or loosening of teeth; vision disturbances; hemoptysis; and necrotic eschar on skin, palate, or nasal turbinates. Management strategies in the context of COVID-19 include, but are not limited to: controlling hyperglycemia, diabetes, or diabetic ketoacidosis; reducing corticosteroid dose with the aim to rapidly discontinue; discontinuing immunomodulating drugs; extensive surgical debridement to remove all necrotic material; antifungal therapy (e.g., amphotericin-B) for 4 to 6 weeks; and appropriate supportive care and monitoring.[432][431] Prevention involves: controlling hyperglycemia; monitoring blood glucose level in COVID-19 patients after discharge (whether or not they are diabetic); and judicious use of corticosteroids, antibiotics, and antifungals.[432]
go to our full topic on Multiple myeloma
The American Society of Hematology (ASH) and European Myeloma Network (EMN) advise that patients who have multiple myeloma with active disease need treatment during the COVID-19 pandemic, but this can be adapted for each patient to reduce additional COVID-19 exposure.[393][433] For patients who require treatment, ASH advises giving 6-12 cycles of bortezomib, lenalidomide, and dexamethasone (RVD), followed by lenalidomide maintenance (with the addition of bortezomib every 2 weeks for high-risk patients). Older myeloma patients may start treatment with RVD or daratumumab, lenalidomide, and dexamethasone (DRd) depending on cytogenetic risk and other comorbidities, and if necessary can continue on lenalidomide and dexamethasone (Rd) only after achieving best response.[393]
Patients should continue on maintenance therapy to reduce the risk of relapse. Lenalidomide can be provided for up to 2 months, with telemedicine visits and home phlebotomy as needed. Higher-risk patients on RVD should continue taking RVD, although if appropriate this could be changed to Rd. If a patient develops COVID-19, maintenance therapy should be interrupted until the infection resolves. Hematopoietic stem cell transplantation may be delayed if resources are limited; otherwise eligible patients should proceed to stem cell based transplant, particularly if they have high-risk myeloma.[393]
The EMN provides recommendations for transplant-eligible and transplant ineligible patients. Autologous stem cell transplantation should be postponed in patients with standard-risk disease and may be considered in patients with high-risk disease after 6-8 cycles of induction treatment. Either RVD, bortezomib with thalidomide and dexamethasone, (VTD), or daratumumab with VTD are the preferred induction therapies.[433] Patients not eligible for transplant should be given all-oral regimens (e.g., Rd), with the addition of bortezomib or daratumumab considered for patients with high-risk disease or for those without sufficient response to Rd.[433]
Guidelines from Australia and New Zealand advise that treatment during the pandemic may depend on available resources, but that management should be guided by the need for disease control in high-risk patients and avoiding unnecessary immune suppression in low-risk patients.[311] Treatment decisions should be individualized, taking into account factors such as newly diagnosed versus relapsed disease, stage, disease burden and rate of progression, and patient factors such as age, frailty, and comorbidities.[311]
The UK Medicines and Healthcare products Regulatory Agency has agreed temporary modifications to the pregnancy prevention programmes for patients taking thalidomide, lenalidomide, and pomalidomide. A home pregnancy test is sufficient, provided the patient has adequate support and instruction, the test meets the minimum sensitivity requirements and the result is verified by the prescriber. If the clinician deems it appropriate, these medications can be initiated during a remote consultation.[434]
There are limited data on trials of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination in patients with hematological conditions. However, available data in patients with multiple myeloma suggest an attenuated response to vaccination.[435] The European Myeloma Network has published a consensus for vaccination against SARS-CoV-2, recommending that all patients with monoclonal gammopathy of unknown significance, smouldering multiple myeloma, multiple myeloma, and monoclonal gammopathies of clinical significance are vaccinated with a COVID vaccine.[436] Recommendations are also given on when patients should preferably be vaccinated, the risk factors that should be considered for a poor response to vaccination, and actions that can be considered in the case of immune impairment.[436]
go to our full topic on Multiple sclerosis
The Association of British Neurologists (ABN) has produced guidance on the use of disease-modifying therapies in patients with multiple sclerosis (MS) during the pandemic.[437] The ABN advises that the effect of disease-modifying therapies on the risk of COVID-19 remains uncertain. Disease-modifying treatments should be offered to patients, provided that the benefits of treatment outweigh the risks. The local rate of COVID-19 infection, the patient’s general health, and their exposure to the virus all influence this risk/benefit decision. Patients should be advised if their disease-modifying therapy might affect the efficacy of a vaccine or the severity of COVID-19. The guidance provides information on considered level of risk for specific disease-modifying therapies.[437] The US National MS Society also recommends that decisions on the use of disease-modifying therapies are individualized and should consider disease factors, risks and benefits of therapies, and risks associated with COVID-19.[438] The National MS Society recommends that people currently taking disease-modifying therapies should continue, and if they develop symptoms of COVID-19 or test positive, their therapies should be reviewed with someone familiar with their care.
Observational studies of patients with MS have found that risk factors for severe forms of COVID-19 are older age, increased levels of disability (Expanded Disability Severity Scale [EDSS] score ≥6, requiring assistance to walk, or being unable to walk), black race, cardiovascular disease, diabetes, obesity, and corticosteroid treatment within the previous 2 months.[439][440] One study found no association between use of disease-modifying therapies and severity of COVID-19.[439] Another study found that patients taking rituximab had an increased risk of hospitalization compared with patients who did not take any disease-modifying therapy, but there was no significantly increased risk of intensive care unit admission, need for mechanical ventilation, or death among the patients taking rituximab.[440]
Some disease-modifying therapies may impact efficacy of COVID-19 vaccines, and this should be considered in the timing of treatment courses and vaccinations.[441][442][443][444][445] Effective cytotoxic T-cell responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been reported in patients with MS who were treated with ocrelizumab and received two or three doses of SARS-CoV-2 vaccine.[446]
go to our full topic on Non-Hodgkin lymphoma
Analysis of US health-insurance claims data has shown that lymphoma is a significant risk factor for fatal COVID-19.[82] Indolent and aggressive non-Hodgkin lymphomas are associated with worse survival in patients with COVID-19.[77][249] Interim treatment guidelines for the management of adult patients during the pandemic have been provided by experts from the UK and also from Australia and New Zealand.[311][415] For most patients with aggressive non-Hodgkin lymphoma subtypes, treatment is delivered with curative intent and this remains the clinical priority. Recommendations are given for patients with Burkitt lymphoma, diffuse large B-cell lymphoma (DLBCL), primary mediastinal B cell lymphoma, central nervous system (CNS) lymphoma, peripheral T cell lymphoma, and relapsed/refractory aggressive lymphoma. For patients with low-grade non-Hodgkin lymphoma and not requiring immediate treatment, watchful waiting may be considered; initiation of treatment should be based on a risk–benefit discussion between the patient and physician.
The American Society for Hematology has published advice for the management of aggressive lymphomas. R-CHOP (rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone) remains the standard of care for diffuse large B cell lymphoma. For older patients, R-mini-CHOP (a reduced dose regimen) with growth factor support is recommended. Subcutaneous rituximab may be considered for patients who have tolerated a first intravenous dose. Recommendations are also given for double-hit and primary mediastinal B cell lymphomas, patients at higher risk of CNS involvement, and patients with relapsed or refractory disease.[447] The International Lymphoma Radiation Oncology Group has published emergency guidelines for radiation therapy in hematologic malignancies, should radiation therapy be necessary. Alternative dose fractionations may be given.[101]
There are limited data on trials of severe acute respiratory syndrome coronavirus 2 vaccination in patients with hematological conditions. Such patients are likely to have an attenuated response to vaccination. One study of the BNT162b2 mRNA COVID-19 vaccine in patients with hematological malignancies found the antibody response was reduced compared with healthy controls, and was affected by disease treatment.[247] Additional measures, such as early vaccination of household contacts, may be considered.[248]
Non-ST-elevation myocardial infarction (NSTEMI)
Signs & symptoms
Investigations
Differentials
Treatment algorithm
go to our full topic on Non-ST-elevation myocardial infarction (NSTEMI)
COVID-19 infection is associated with an increased risk of acute myocardial infarction.[448]
The European Society of Cardiology has published guidance on the diagnosis and management of cardiovascular disease during the COVID-19 pandemic, and the European Association of Percutaneous Cardiovascular Interventions (EAPCI) has issued a position statement on invasive management of acute coronary syndromes.[193][449]
Patients presenting with non-ST-elevation acute coronary syndrome should be risk stratified into four groups: very high, high, intermediate, and low risk. Very high-risk patients include patients with cardiogenic shock, hemodynamic instability, recurrent or persistent chest pain refractory to medical therapy, life-threatening arrhythmias, cardiac arrest, mechanical complications of myocardial infarction, acute heart failure, and recurrent intermittent ST-elevation. High-risk patients are those with an established diagnosis of NSTEMI based on cardiac troponins and at least one of: dynamic ST/T changes, or recurrent symptoms.
Testing for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) should be performed as soon as possible after first medical contact. However, patients who are very high risk require immediate invasive management as per ST-elevation myocardial infarction (STEMI) protocols. High-risk patients should have early intervention (ideally within 24 hours) after their SARS-CoV-2 test results are known. Intermediate- and low-risk patients should initially be managed with noninvasive testing once their SARS-CoV-2 test results are known. Coronary computed tomography angiography (CCTA) is the favored investigation for intermediate-risk patients where equipment and expertise are available. Noninvasive imaging using CCTA may speed up risk stratification, avoid an invasive approach, and allow early discharge.[193][449]
UK guidance advises using ECG and measuring high-sensitivity troponin and N-terminal pro B-type natriuretic peptide in patients hospitalized with COVID-19 who have signs or symptoms of acute myocardial injury. In patients with severe COVID-19, elevated troponin levels may result from cardiac inflammatory response rather than acute coronary syndrome. Patients with suspected or confirmed acute myocardial injury should receive continuous ECG monitoring, as well as blood pressure, heart rate, and fluid monitoring, in a setting where deterioration can be quickly identified. Where diagnosis is not clear, ECG should be monitored daily and high-sensitivity troponin measurements should be repeated, because dynamic change may help establish a clear diagnosis and monitor the course of illness.[131]
Guidelines from Australia and New Zealand state that reliance on troponin measurements to diagnose acute coronary syndrome in patients with COVID-19 can be misleading, and greater emphasis should be given to high risk clinical features: recurrent chest pain, dynamic ECG changes, heart failure, hemodynamic instability, major arrhythmias, and the presence of regional wall motion abnormalities on echocardiography. Invasive investigations should be deferred in stable patients, particularly if they are COVID-19 positive.[450]
A cohort study from the UK reported that black and minority ethnic (BAME) patients had higher rates of hospitalization for acute myocardial infarction and longer time to coronary angiography for NSTEMI than white patients during the pandemic, compared with the same pre-pandemic period. BAME patients were more likely than white patients to present with out-of-hospital cardiac arrest and cardiogenic shock during the COVID-19 pandemic compared with the period before the pandemic. In-hospital and 7-day mortality was higher in BAME patients (relative to white patients) during the COVID-19 pandemic compared with the pre-pandemic period.[451]
go to our full topic on Obesity in adults
Obesity (body mass index [BMI] ≥30kg/m²) is associated with increased risk of a positive COVID-19 test, hospitalization, severe COVID-19 requiring admission to intensive care, mechanical ventilation, and death.[301][303][354][452][453][454][455][456][457][458] The association is strongest in patients <65 years.[303][455] One meta-analysis found that obesity was associated with a 12% increase in absolute risk of COVID-19 death.[360] Patients with obesity who develop COVID-19 are also at higher risk for venous thromboembolism and dialysis.[453]
Being overweight (BMI >25kg/m² and <30kg/m²) might increase the risk of severe illness from COVID-19, and is associated with increased risk of mechanical ventilation.[301][354][455][457]
A study in the US found a nonlinear (J-shaped) relationship between BMI and risk of hospitalization, intensive care admission, and death.[455] A study in the UK also found nonlinear associations between BMI and hospital admission and death due to COVID-19, and a linear association between BMI and intensive care admission due to COVID-19.[457] In the UK study, the relative risk due to increasing BMI was particularly notable in people younger than 40 years and of black ethnicity.
There is currently no high-quality research on the effects of weight loss on COVID-19, but that the role of excess weight as a risk factor for serious COVID-19 complications warrants further consideration.[452] For those living with obesity, weight loss has been shown to bring general long-term health benefits.
Patients with obesity who recover from COVID-19 may be at increased risk of postacute sequelae. One study found that patients with obesity who did not require ICU admission and survived the acute phase of COVID-19 had an increased risk of hospital admission and needing diagnostic tests over the subsequent 8 months compared with patients with normal BMI.[459]
Vaccination against COVID-19 is effective in people with overweight or obesity. Vaccination reduces the risk of COVID-19-related hospitalization 14 days or more after a second dose of vaccine by 68% in people with overweight and obesity, compared with 66% in people of healthy weight.[460]
go to our full topic on Obesity in children
The American Academy of Pediatrics advises that children and adolescents with obesity are at increased risk for COVID-19 and worsening of their obesity during the pandemic. Disruption to routine, sleep dysregulation, decreased physical activity, food insecurity, and increased screen time may all contribute.[195] A cohort study of over 430,000 children and young people aged 2-19 years reported that the rate of body mass index increase approximately doubled during the pandemic,compared with the prepandemic period. Children with prepandemic overweight or obesity and younger school-aged children experienced the largest increases.[461] Clinicians should continue to provide obesity treatment and should identify and treat obesity-related comorbidities during the pandemic. Clinicians should counsel patients and their families about the risk of COVID-19, screen for disordered eating (e.g., binge eating, purging, and restrictive eating), and connect families to resources to address social determinants of health.[195]
One study in the US found that among children aged 12 to 17 years hospitalized with COVID-19, approximately two-thirds had obesity.[462] The study also found that compared with patients without obesity, those with obesity required higher levels and longer duration of care.
go to our full topic on Obstructive sleep apnea
Obstructive sleep apnea (OSA) may increase the risk of developing COVID-19 infection, and may also increase the risk of having more severe COVID-19 infection.[463] In a study of people with diabetes who were hospitalized for COVID-19, treated OSA was independently associated with an increased risk of death.[464] One systematic review noted that many of the risk factors for OSA (such as age, hypertension, cardiovascular disease, lung disease, diabetes, and obesity) are associated with worse outcomes for COVID-19 infection.[465] The review also found that the COVID-19 pandemic has had a large effect on the diagnosis and treatment of OSA, and that new diagnosis and treatment pathways may be necessary (e.g., using disposable sleep study kits).
go to our full topic on Olfactory loss
Olfactory loss (anosmia) may be a presenting symptom of COVID-19. The European Rhinologic Society advises against prescribing intranasal or systemic corticosteroids for patients with sudden olfactory loss. Patients should be advised to continue their usual medications, including intranasal corticosteroids prescribed for other indications.[466]
go to our full topic on Osteoporosis
Guidelines from an international group of experts suggest altering the approach to management of osteoporosis during the current pandemic:[467]
- Zoledronic acid can be delayed for 6 to 9 months during the pandemic.
- Patients established on 6-monthly denosumab should continue without any delay and self-administration can be considered where appropriate. Pre-treatment checking of serum vitamin D and calcium levels can be waived and empiric treatment with cholecalciferol (vitamin D3) can be considered for all patients.
- Patients established on teriparatide, abaloparatide, or romosozumab should continue; however, periods of discontinuation for many weeks are unlikely to affect the long-term beneficial effects on fracture risk reduction.
- No new patients should be started on zoledronic acid, teriparatide, abaloparatide, or romosozumab due to the risk of confusion from potential adverse effects of the therapies and symptoms of COVID-19.
- If not contraindicated, alternative treatment, such as continuing with an oral bisphosphonate, should be considered.
The American Society of Bone and Mineral Research (ASBMR) has also published recommendations for the management of osteoporosis during the pandemic.[468]
The American College of Rheumatology advises that the denosumab dosing interval can be extended if necessary to minimize healthcare encounters, but should not exceed 8 months.[469]
Patients should be educated on the importance of continuing with calcium and vitamin D through supplements or diet, and lifestyle measures such as regular exercise and healthy diet.
go to our full topic on Palliative care
Patients with COVID-19 can deteriorate rapidly; treatment escalation plans should be put in place as soon as possible.[131]
Cough should initially be managed with nonpharmacologic measures if possible. Patients should be discouraged from lying on their back because this makes coughing ineffective. Simple measures, such as honey, may be used.[470]
A combination of opioid and benzodiazepine may be considered for people with COVID-19 and who have moderate to severe breathlessness, are distressed, and are at the end of life. Consider concomitant use of an antiemetic and regular stimulant laxative.[131]
Consider whether the sublingual, rectal, or subcutaneous route is appropriate for administration for medication; this may be easier for relatives or caregivers to administer if there are fewer healthcare staff.[131]
Implantable cardiac defibrillators (ICDs) cannot be deactivated remotely. If a patient with an ICD is receiving end of life care, the treating clinical team should secure a magnet to the skin over the ICD where possible, rather than using the programmer.[192]
Further resources are available at:
The World Hospice and Palliative Care Alliance: COVID-19 resources Opens in new window
go to our full topic on Pancreatic cancer
A UK consensus statement recommends that endoscopic therapy for malignant biliary obstruction, with biopsy or cytology specimen collection if indicated, should continue during the COVID-19 pandemic. Urgent (2 week wait) cancer referrals and endoscopic ultrasound requests should be considered on a case-by-case basis.[471]
Surgery for resectable pancreatic cancer remains the standard of care and should be performed whenever possible. If surgery is not available, systemic anticancer therapy (SACT) or hypofractionated chemoradiotherapy should be offered. Fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) is the preferred SACT regimen. Radiation therapy may be given as 35-45 Gy in 5 fractions, depending on center expertise, or 36 Gy in 15 fractions with concurrent capecitabine.[471] FOLFIRINOX is most appropriate in patients with a good performance status without significant comorbidities. Dose modification, use of prophylactic antibiotics and growth factors and physical distancing measures should be used to reduce the risk of severe COVID-19 infection.
Patients with locally advanced pancreatic cancer are usually treated with upfront SACT, with or without radiation therapy. Hypofractionated radiation therapy or chemoradiotherapy may reduce risk of severe COVID-19 infection and allow a deferral or break from SACT; this should be balanced against the risk of metastasis without upfront chemotherapy. The risks of treatment in patients aged over 80 years are likely to outweigh the benefits. For fit patients with no significant comorbidities, treatment options include four cycles of modified FOLFIRINOX with or without hypofractionated radiation therapy or five cycles of radiation therapy alone.[471]
The median improvement in survival with palliative chemotherapy for metastatic disease is <6 months so the risks of treatment are likely to outweigh the benefits for many patients. The decision to treat should be taken on a case-by-case basis. Early response assessment should be considered, depending on radiology capacity, because this may allow a shorter duration of chemotherapy. A break from chemotherapy may be appropriate for patients with low volume disease or good disease control. Second-line palliative chemotherapy should not be offered.[471]
go to our full topic on Parkinson disease
Patients with Parkinson disease who are treated with deep brain stimulation (DBS) require ongoing outpatient visits and surgical care and may not tolerate interruption or cessation of therapy, with some experiencing life-threatening DBS-withdrawal syndrome.[472] In the current pandemic, many elective procedures are being deferred; however, practical recommendations are available to guide management of DBS device complications or battery replacement. Patients who are at high risk for severe or life-threatening symptoms or hospitalization with DBS cessation would be considered the highest priority for DBS replacement; patients at lower risk may be able to have replacement postponed.[472]
Pediatric rheumatic diseases
The American College of Rheumatology (ACR) has published guidance for the management of pediatric rheumatic disease (PRD) during the pandemic.[473] Currently, the evidence does not suggest that children with PRD and children receiving immunomodulatory therapies for PRD have a higher risk of severe COVID-19 infection, and general preventative measures are advised. The ACR guidance provides recommendations for ongoing treatment of patients during the pandemic, including for those who have been exposed to severe acute respiratory syndrome coronavirus (SARS-CoV-2), and for those with probable or confirmed infection.
go to our full topic on Prostate cancer
Radiation oncologists from the US and the UK have agreed upon recommendations to safely manage patients with prostate cancer during the COVID-19 pandemic.[474]
Androgen deprivation therapy may allow radiation therapy to be deferred. If androgen deprivation therapy cannot be delivered, the benefits of radiation therapy should be weighed against the risk of COVID-19, taking into account the patient's age, comorbidities, and immunosuppression.[474]
If radiation therapy is deemed necessary and the benefits outweigh the risks, the shortest fractionation schedule that has evidence of efficacy and safety should be followed.[474]
go to our full topic on Psoriasis
People with inflammatory skin diseases have a higher risk of COVID-19 related death than the general population, after adjusting for confounding and mediating factors (HR 1.07, 96% CI 1.02 to 1.11). They are also at higher risk of hospital and critical care admission. South Asian people and mixed ethnicity people with inflammatory skin diseases are at higher risk of COVID-19-related death, compared with white people with inflammatory skin diseases.[64]
People taking tumor necrosis factor inhibitors, interleukin (IL)-12/IL-23 inhibitors, or IL-17 inhibitors do not have an increased risk of COVID-19-related critical care admission or death, compared with people taking standard systemic therapy (e.g., methotrexate).[64]
The National Psoriasis Foundation has published guidance for the management of psoriatic disease during the COVID-19 pandemic.[66] Patients and clinicians should make shared decisions regarding use of systemic therapies during the pandemic. In most cases, it is recommended that patients who do not have SARS-CoV-2 infection should continue biological or oral therapies.[66] Chronic systemic corticosteroids should be avoided if possible. If used, the dose should be tapered to the lowest therapeutic dose.[66] Initiation of systemic therapies should be discussed with patients who are newly diagnosed with psoriasis, because untreated psoriatic disease may lead to permanent joint damage and disability. Newly diagnosed patients, patients with unstable disease or flares, patients requiring a thorough skin and/or joint examination, and patients at risk for skin cancer should receive in-person care where pandemic conditions allow.[66]
The International Psoriasis Council recommends discontinuing or postponing the use of immunosuppressive medications in patients diagnosed with COVID-19.[65] Patients with psoriasis who become infected with SARS-CoV-2 should receive evidence-based therapies and supportive care. Systemic corticosteroids should not be withheld due to concern about a psoriasis flare following withdrawal. Dermatology and/or rheumatology consultation may be needed for hospitalized patients. Decisions about restarting treatments for psoriasis and psoriatic arthritis after recovery form COVID-19 should be made on a case-by-case basis.[66]
The National Psoriasis Foundation recommends that in most cases, patients should take the first mRNA-based vaccine for which they are eligible and they are offered. Systemic medication for psoriasis or psoriatic arthritis is not a contraindication to any of the available SARS-CoV-2 vaccines.[66] A 2-week interruption of methotrexate following a SARS-CoV-2 vaccine booster has been shown to boost antibody response.[475]
go to our full topic on Pulmonary embolism
The American Society of Hematology advises that normal D-dimer can be used to effectively rule out pulmonary embolism (PE) in patients with COVID-19. Radiologic imaging is not necessary if the D-dimer is normal in the context of low pretest probability. D-dimer is elevated in patients with COVID-19, particularly those with severe or critical disease, independent of the presence of venous thromboembolism (VTE). D-dimer cannot be used to diagnose VTE/PE in patients with COVID-19. If objective imaging to confirm or refute a diagnosis of VTE/PE is not feasible, clinicians should seek other evidence for VTE/PE. Clinical features that increase the likelihood of PE include symptoms or signs of deep vein thrombosis, unexplained hypotension or tachycardia, unexplained worsening respiratory status, and risk factors for thrombosis (e.g., history of thrombosis, cancer, hormonal therapy).[476]
Patients with COVID-19 who experience an incident thromboembolic event or who are highly suspected to have thromboembolic disease be managed with therapeutic anticoagulation.[91]
All hospitalized adults with COVID-19 should receive pharmacologic thromboprophylaxis, unless the risk of bleeding outweighs the risk of thrombosis.[476] Consult your local guidance for more information.
Renal transplant
Guidelines from the British Transplantation Society and Renal Association recommend stopping mycophenolate and azathioprine in renal transplant recipients who develop COVID-19, until the patient has fully recovered. If the patient has severe or progressive disease, clinicians should consider stopping or reducing calcineurin inhibitors.[477]
Studies have found that kidney transplant recipients may have an inadequate antibody response to vaccination against COVID-19 after two and three doses.[478][479][480][481][482] One meta-analysis of prospective observational studies found that 6% of organ transplant recipients seroconverted after one dose of SARS-CoV-2 vaccine and 39% of organ transplant recipients seroconverted after two doses of SARS-CoV-2 vaccine. Antibody titers were lower in organ transplant recipients who seroconverted compared with healthy controls.[482] Additional strategies to improve the immune response to vaccination in renal transplant recipients should be considered. One retrospective registry-based cohort study of renal transplant recipients found that compared with those who had not been vaccinated, receiving two doses of mRNA vaccine did result in a lower risk for SARS-CoV-2 infection.[483]
The British Transplant Society and the Centre of Evidence in Transplantation have provided an open-access repository of guidance for solid organ donation and transplantation in the context of COVID-19. British Transplantation Society: COVID-19 information Opens in new window
go to our full topic on Rheumatoid arthritis
People with inflammatory joint disease are at increased risk of COVID-19-related death, critical care admission, and hospital admission.[64][275][484][485] Black and South Asian patients with inflammatory joint disease are at higher risk of COVID-19-related death and critical care admission, compared with white patients with inflammatory joint disease.[64] Black, Asian and Hispanic patients with rheumatoid arthritis are at higher risk of hospitalization with COVID-19, compared with white patients.[485][486] Older age, comorbid hypertension, comorbid COPD, and glucocorticoid use have also been associated with an increased risk of hospitalization.[487]
Patients taking TNF inhibitors, IL-12/IL-23 inhibitors, IL-17 inhibitors, JAK inhibitors, or IL-6 inhibitors have no increased risk of COVID-19 related death, COVID-19-related critical care admission or death, or COVID-19-related hospital admission, compared with patients taking standard systemic therapy (e.g., methotrexate). Compared with people on standard systemic therapy, people receiving rituximab have an increased risk of COVID-19-related death, critical care admission, and hospital admission. Patients taking JAK inhibitors have an increased risk of COVID-19 related hospital admission, compared with patients on standard systemic therapy.[64]
Patients should continue their usual medication and observe recommended infection prevention and control precautions.[390][488][489] If it is possible and clinically safe, corticosteroid dose may be tapered. Clinicians should consider alternatives to corticosteroids where possible, and if corticosteroids are needed, prescribe the lowest effective dose for the shortest possible time. Corticosteroid injections should only be given when a patient has significant disease activity and/or intrusive and persistent symptoms, and there are no suitable alternatives.[490]
Conventional synthetic disease-modifying antirheumatic drugs (DMARDS) may be started or switched in patients with newly diagnosed or active inflammatory arthritis.[469]
Patients receiving immunosuppressive medication may develop atypical symptoms of COVID-19 (e.g., patients who take an oral corticosteroid may not develop fever). Patients who take an oral corticosteroid should not stop suddenly if they develop COVID-19.[469] American College of Rheumatology guidance recommends that patients should stop hydroxychloroquine and chloroquine if they become infected with severe acute respiratory disease coronavirus 2 (SARS-CoV-2) because of the potential for cardiotoxicity, which could be heightened by COVID-19 infection and receipt of other drugs in hospital.[469] Interleukin-6 receptor inhibitors may be continued in select circumstances as part of a shared decision-making process.[469]
Patients may continue taking nonsteroidal anti-inflammatory drugs (NSAIDs); NSAIDs have not been associated with increased risk of severe COVID-19.[321][491][492][493][494]
The American College of Rheumatology advises stopping NSAIDs in patients who develop severe manifestations of COVID-19, such as kidney, cardiac, or gastrointestinal injury.[469]
Following recovery from COVID-19, US guidelines recommend that rheumatic disease treatments can be restarted within 7-14 days of symptom resolution in patients with mild or no pneumonia who were treated in the ambulatory setting or with self quarantine. Decisions regarding restarting rheumatic disease therapies in patients who had more severe COVID-19 should be taken on an individual basis. If a patient had a positive polymerase chain reaction test for SARS-CoV-2 but has remained asymptomatic, rheumatic disease treatments may be restarted 10-17 days after the positive test result.[469]
Patients are advised to have influenza, whooping cough, and pertussis vaccinations.[495]
Clinicians are advised to engage patients with rheumatic disease in a shared decision-making process to discuss receiving a SARS-CoV-2 vaccine.[389][496] The response to vaccination is likely to be blunted in patients receiving systemic immunomodulatory therapies compared with the general population.[343][389][496] A 2-week interruption of methotrexate following a SARS-CoV-2 vaccine booster has been shown to boost antibody response.[475] A third dose of SARS-CoV-2 vaccine results in a serologic response in 84% of patients with immune-mediated inflammatory diseases who had a weak response to the standard 2-dose regimen.[343] The American College of Rheumatology advises that, beyond known allergies to vaccine components, there are no additional contraindications to SARS-CoV-2 vaccination for patients with autoimmune and inflammatory rheumatic diseases. Vaccination should ideally occur when rheumatologic disease is well controlled.[389] EULAR recommends that patients with rheumatic and musculoskeletal diseases are strongly advised to receive a SARS-CoV-2 vaccination with any of the vaccines approved in their country.[390]
go to our full topic on Sarcoidosis
Guidance on the management of patients with sarcoidosis during the COVID-19 pandemic is available.[497] De-escalation of therapy can be considered for patients who are clinically stable, with few or no symptoms and no severe organ manifestations. Glucocorticoid doses may be gradually reduced in most patients who are receiving glucocorticoid monotherapy. Dose reduction, prolongation of dosing interval, or a drug holiday may be considered for stable patients taking disease-modifying drugs such as methotrexate, mycophenolate, and azathioprine. Dose reduction, or prolongation of the dosing interval, may be considered for stable patients taking immunosuppressive biological agents. Patients should be advised how to recognize and report symptoms suggestive of active disease following de-escalation of therapy.[497]
De-escalation of therapy may be contraindicated in patients with clinically active, organ- or life-threatening sarcoidosis. Such patients include those with uveitis, neurosarcoidosis, progressive pulmonary disease, and cardiac disease. In these circumstances, patients receiving glucocorticoid monotherapy should take the lowest possible dose to achieve disease control. If a patient requires a glucocorticoid dose equivalent to 40 mg/day to 60 mg/day of prednisone, clinicians should consider adding a disease-modifying drug in order to reduce glucocorticoid requirement. Patients who are stabilized on disease-modifying drugs should continue current doses, provided the expected benefits of treatment outweigh the risks. Patients who are receiving immunosuppressive biological therapy should continue. Home administration may be required if infusion units are closed.[497]
Investigations such as pulmonary function tests and chest imaging should be used judiciously, to avoid unnecessary hospital visits.[497]
go to our full topic on Sickle cell disease
Patients with sickle cell disease are at higher risk of severe disease and death if they become infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A registry of patients with sickle cell disease and COVID-19 in the US reported that the rate of hospitalization amongst adults with sickle cell disease was 69%, the intensive care admission rate was 11% and mortality was 7%.[498] A cohort study conducted in the UK reported a 4-fold increased risk of hospitalization with COVID-19, and a 2.5-fold increased risk of mortality from COVID-19, in people with sickle cell disease, compared with the general population.[499] Sickle cell trait is associated with increased risk of COVID-19 mortality and an increased incidence of acute kidney failure in the 60 days following COVID-19 diagnosis.[500]
The Sickle Cell Disease Association of America has published advice on reducing sickle cell disease morbidity during the COVID-19 pandemic. Patients should be advised to adhere carefully to their usual medication, to use a thermometer at home, and to seek prompt medical advice if they develop fever. Clinicians should ensure that patients have an adequate supply of medication to manage acute and chronic pain, and consider starting or optimizing therapies known to reduce acute sickle cell pain frequency to reduce the need for hospital attendance.[501]
Patients who have acute sickle cell pain without fever or signs of infection should be encouraged to manage pain with oral medication at home. Patients should be closely monitored, with a low threshold for arranging a face-to-face evaluation and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing.[501]
Patients with fever, cough, or shortness of breath require immediate evaluation for COVID-19. Care should include an assessment for other sources of infection with culture of blood (and other specimens as indicated), testing for typical viral infections, administering broad-spectrum antibiotics to cover encapsulated organisms, and assessing for acute chest syndrome. If the patient tests negative for SARS-CoV-2, home treatment with oral antibiotics and close monitoring may be appropriate. If possible, patients should be given an incentive spirometer to use at home.[501]
Patients with confirmed COVID-19 should be monitored closely for signs of rapidly progressive acute chest syndrome (thrombocytopenia, acute kidney injury, hepatic dysfunction, altered mental status, and multiorgan failure). The symptoms of acute chest syndrome may overlap significantly with symptoms of COVID-19. Standard care for acute chest syndrome should be given, including supplemental oxygen, empiric antibiotics, oseltamivir until influenza is excluded, incentive spirometry, and good pain control. Patients with worsening anemia, evidence of hypoxia, and chest x-ray changes should receive a transfusion of red blood cells. Clinicians should consider the possibility of undiagnosed pulmonary hypertension in acutely ill patients and be alert for signs of fat emboli syndrome. Signs of fat emboli syndrome include worsening anemia and mental status, hemolysis, thrombocytopenia, hypoalbuminemia, respiratory distress, and petechial rash; it may progress quickly and carries a high mortality. Patients who have COVID-19 and are discharged from the hospital remain at high risk of secondary bacterial infection and acute chest syndrome; they should be monitored daily.[501]
If availability of blood products is limited, the highest priority indications for chronic transfusion are: stroke prevention, progressive or critical neurovascular disease, recurrent acute chest syndrome unresponsive to hydroxyurea, and cardiovascular or respiratory comorbidity. Clinicians should assess whether patients can switch to hydroxyurea or whether transfusion strategy can be temporarily altered.[501]
go to our full topic on Smoking cessation
In patients with COVID-19, evidence suggests that smoking is associated with an increased risk of symptomatic COVID-19 infection, more severe disease, and death.[502][503][504] One meta-analysis found that smoking was associated with a 7% increase in absolute risk of COVID-19 death.[360] People who smoke tobacco may also have an increased risk of contracting COVID-19. It is well-established that smoking damages the lungs and airways, and weakens the immune response; people exposed to second-hand smoke are also at increased risk.
Vaping/use of e-cigarettes is often used as nicotine-replacement therapy; however, the evidence on benefits and harms is still developing. People should not share vaping devices or cigarettes. Smoking cessation is strongly encouraged.[502]
ST-elevation myocardial infarction (STEMI)
Signs & symptoms
Investigations
Differentials
Treatment algorithm
go to our full topic on ST-elevation myocardial infarction (STEMI)
COVID-19 infection is associated with an increased risk of acute myocardial infarction.[448] Among patients with STEMI, a concomitant diagnosis of COVID-19 has been associated with significantly higher rates of in-hospital mortality.[505]
The European Society of Cardiology has published guidance on the diagnosis and management of cardiovascular disease during the COVID-19 pandemic, and the European Association of Percutaneous Cardiovascular Interventions (EAPCI) has issued a position statement on invasive management of acute coronary syndromes.[193][449]
The guidance emphasizes that the pandemic should not compromise the timely reperfusion of patients with STEMI, therefore in the absence of previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) testing all patients should be managed as if they are COVID-19 positive. Primary percutaneous coronary intervention (PCI) is the reperfusion treatment of choice if it can be performed within 120 minutes in appropriate facilities while ensuring the safety of healthcare professionals and other patients. Experience suggests that a delay of up to 60 minutes may occur due to implementing protective measures, and clinicians should take this into account when assessing whether timely primary PCI is possible. If primary PCI cannot be performed within the target time, fibrinolysis is the intervention of choice provided there are no contraindications. All patients should undergo testing for SARS-CoV-2 as soon as possible following first medical contact irrespective of reperfusion strategy, at the latest upon admission to the intensive care unit after primary PCI. Clinicians should consider immediate complete revascularization, if indicated and appropriate, in order to avoid staged procedures and reduce hospital stay.[193][449]
UK guidance advises using ECG and measuring high-sensitivity troponin and N-terminal pro B-type natriuretic peptide in patients hospitalized with COVID-19 who have signs or symptoms of acute myocardial injury. The same ECG diagnostic criteria for cardiac conditions apply in patients with COVID-19 and the general population.[193] Patients with ST-elevation should be referred immediately to a cardiologist for consideration of percutaneous coronary intervention. In patients with severe COVID-19, elevated troponin levels may result from cardiac inflammatory response rather than acute coronary syndrome. Patients with suspected or confirmed acute myocardial injury should receive continuous ECG monitoring, as well as blood pressure, heart rate, and fluid monitoring, in a setting where deterioration can be quickly identified. Where diagnosis is not clear, ECG should be monitored daily and high-sensitivity troponin measurements should be repeated, because dynamic change may help to monitor the course of illness and establish a diagnosis.[131]
A cohort study from the UK reported that black and minority ethnic (BAME) patients had higher rates of hospitalization for acute myocardial infarction and longer time to reperfusion therapy for STEMI than white patients during the pandemic, compared with the same pre-pandemic period. BAME patients were more likely than white patients to present with STEMI, out-of-hospital cardiac arrest, and cardiogenic shock during the COVID-19 pandemic compared with the period before the pandemic. In-hospital and 7-day mortality was higher in BAME patients (relative to white patients) during the COVID-19 pandemic compared with the pre-pandemic period.[451]
go to our full topic on Stroke
COVID-19 infection is associated with an increased risk of ischemic stroke.[448] Cerebrovascular disease is associated with increased mortality from COVID-19.[304]
Guidance from the American Heart Association/American Stroke Association advises that patients with COVID-19 may present with neurologic symptoms (such as dizziness, headache, or encephalopathy) at the same time as, or even preceding, the development of respiratory symptoms and fever. Patients affected by stroke may be unable to give a history of COVID-19 symptoms or exposure owing to confusion or aphasia. Patients with stroke frequently develop a fever from stroke complications, including aspiration pneumonia and urinary tract infection; these patients require rapid evaluation for COVID-19.[506]
All stroke teams should endeavor to adhere to guidelines for patient selection for therapy, treatment timeframes, and post-recanalization monitoring. Teams should use their judgment, guided by local circumstances, to treat as many patients with acute stroke as possible. Patients with large intracerebral bleeds, subarachnoid hemorrhage, or large ischemic strokes at risk of herniation should be monitored in an intensive care setting, with appropriately trained personnel, where possible. Stable patients may be moved out of intensive care to step-down facilities during the 24-hour post-thrombolysis or thrombectomy follow-up period, if an intensive care bed is needed. Stroke physicians should provide guidance to staff if patients with acute stroke have suspected or confirmed COVID-19 and require admission to a COVID-19 unit.[506]
go to our full topic on Strongyloides infection
Experts have made recommendations to reduce the risk ofStrongyloides hyperinfection or dissemination in people at moderate to high risk of Strongyloides infection. There is a risk of hyperinfection following exposure to immunosuppressive drugs. Chronic strongyloidiasis is often asymptomatic; suspicion should be based on risk factors including residence in an endemic area, rural residence, and exposure to soil during labour. A screen-and-treat strategy is recommended for patients at moderate to high risk ofStrongyloides infection without confirmed COVID-19, asymptomatic patients with a positive polymerase chain reaction test, and patients with mild COVID-19 who are not candidates for dexamethasone. Serologic testing is preferred. Patients in the hospital setting who are at moderate to high risk ofStrongyloides infection, are SARS-CoV-2 positive, and are initiating or are likely candidates for dexamethasone should be treated presumptively with ivermectin. Patients at moderate to high risk ofStrongyloides infection who have unexplained gram-negative rod infections after receiving dexamethasone or other immunosuppressive agents should have diagnostic testing forStrongyloides infection. Ivermectin should be given while awaiting results.[507]
go to our full topic on Substance use disorders
Positive urine drug tests for cocaine, fentanyl, heroin, and methamphetamine in patients diagnosed with, or at risk of, substance abuse disorders increased significantly during the COVID-19 pandemic, compared with the preceding 4 months.[508] An urban emergency department in the US has reported a large increase in visits for nonfatal, unintentional opioid overdoses in March to June 2020, compared with the same period in 2019.[509] Fatal opioid overdoses also reportedly increased in some areas during lockdown periods.[510] Adults with disabilities were twice as likely to experience new or increased substance misuse during the period from February to March 2021 compared with adults without disabilities.[149]
A retrospective analysis of US emergency medical services registry data observed a sharp rise in overdose-related cardiac arrests during April 2020, with peak rates in May 2020, more than double than in the same period in 2018 and 2019.[511] The increase in overdose-related deaths in the US during the pandemic was primarily driven by synthetic opioids.[512] The Substance Abuse and Mental Health Services Administration in the US has introduced flexibility in the Opioid Treatment Program, depending on stability of the patient.[135]
A survey comparing self-reported alcohol consumption in 2019 and 2020 found that frequency of alcohol consumption increased overall, and particularly in women, adults age 30 to 59 years, and non-Hispanic white individuals.[513] Alcohol-related deaths increased by 25% in 2020 compared with the prepandemic period in 2019. The largest increase was observed in people aged 25 to 44 years.[514]
go to our full topic on Syphilis
The US Food and Drug Administration has published an alert advising that false-positive Rapid Plasma Reagin (RPR) test results can occur when using the Bio-Rad Laboratories BioPlex 2200 Syphilis Total & RPR kit, in some people who have received a COVID-19 vaccine. Based on information provided by the manufacturer, Bio-Rad Laboratories, RPR false reactivity was observed in some individuals for at least five months following a COVID-19 vaccination.[515] Treponemal testing for syphilis such as Treponema pallidum particle agglutination (TP-PA) and treponemal immunoassays do not appear to be impacted by this issue and patients who received a reactive RPR result using the Bio-Rad BioPlex 2200 Syphilis Total & RPR test kit should be aware that they may need to be retested for syphilis with another test to confirm results.
go to our full topic on Systemic lupus erythematosus (SLE)
Chloroquine and hydroxychloroquine should be started at full dose for patients with newly diagnosed SLE. Chloroquine and hydroxychloroquine should be continued at the same dose during pregnancy.[469]
Belimumab, ACE inhibitors, angiotensin-II receptor antagonists, and glucocorticoids may be initiated if indicated. High-dose glucocorticoids or immunosuppressants may be initiated for patients with lupus nephritis. Glucocorticoids should not be stopped abruptly and should be used at the lowest possible dose to control disease.[469]
Nonsteroidal anti-inflammatory drugs (NSAIDs) may be continued following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure, but should be stopped if the patient develops severe COVID-19.[469] Chloroquine and hydroxychloroquine should be stopped temporarily if the patient develops COVID-19.[469]
Following recovery from COVID-19, US guidelines recommend that rheumatic disease treatments can be restarted within 7-14 days of symptom resolution in patients with mild or no pneumonia who were treated in the ambulatory setting or with self quarantine. Decisions regarding restarting rheumatic disease therapies in patients who had more severe COVID-19 should be taken on an individual basis. If a patient had a positive polymerase chain reaction test for SARS-CoV-2 but has remained asymptomatic, rheumatic disease treatments may be restarted 10-17 days after the positive test result.[469]
Race, presence of at least one comorbidity, and body mass index have been identified as predictors of hospitalization in patients with SLE and COVID-19.[516] One study in the US found that, similar to findings in the general US population, black, Asian, and Latin American patients with rheumatic disease and COVID-19 had an increased risk of hospitalization and ventilatory support compared with white patients, even after adjustment for demographic, rheumatic disease activity, and comorbidities.[486] Another cohort study conducted in the US found that systemic autoimmune rheumatic disease was associated with increased risk of hospitalization, intensive care admission, acute kidney injury, and venous thromboembolism. The risk was largely due to comorbidities, except for the risk of venous thromboembolism. Risk of mechanical ventilation and death was not increased in the patients with systemic autoimmune rheumatic disease.[484]
Clinicians are advised to engage patients with rheumatic disease in a shared decision-making process to discuss receiving a SARS-CoV-2 vaccine. The response to vaccination is likely to be blunted in patients receiving systemic immunomodulatory therapies compared with the general population. The American College of Rheumatology advises that, beyond known allergies to vaccine components, there are no additional contraindications to SARS-CoV-2 vaccination for patients with autoimmune and inflammatory rheumatic diseases. Vaccination should ideally occur when rheumatologic disease is well controlled.[389] EULAR recommends that patients with rheumatic and musculoskeletal diseases are strongly advised to receive a SARS-CoV-2 vaccination with any of the vaccines approved in their country.[390]
go to our full topic on Systemic vasculitis
A cohort study conducted in the UK reported that the most common symptoms of COVID-19 in patients with systemic vasculitis were dyspnea, fever, and cough. The most common complications were respiratory failure (54%), acute kidney injury (18%), and secondary infection (15%). Over 90% of patients were admitted to hospital, and 28% died. Comorbid respiratory disease and glucocorticoid exposure were associated with severe outcomes; subtype of vasculitis and vasculitis disease activity were not.[517]
Patients should follow general infection prevention precautions, e.g., hand hygiene and social distancing. Some measures to reduce healthcare encounters (and potential severe acute respiratory syndrome coronavirus 2 exposure) may be appropriate; clinicians and patients should make a shared decision. Glucocorticoids should be used at the lowest dose to control disease and should not be stopped abruptly, regardless of infection or exposure status.[518]
High-dose glucocorticoids or other immunosuppressants may be initiated if indicated for patients with systemic inflammatory or vital organ-threatening disease.[518]
Clinicians are advised to engage patients with rheumatic disease in a shared decision-making process to discuss receiving a SARS-CoV-2 vaccine. The response to vaccination is likely to be blunted in patients receiving systemic immunomodulatory therapies compared with the general population. The American College of Rheumatology advises that, beyond known allergies to vaccine components, there are no additional contraindications to SARS-CoV-2 vaccination for patients with autoimmune and inflammatory rheumatic diseases. Vaccination should ideally occur when rheumatological disease is well controlled.[389] EULAR recommends that patients with rheumatic and musculoskeletal diseases are strongly advised to receive a SARS-CoV-2 vaccination with any of the vaccines approved in their country.[390]
go to our full topic on Thalassemia
The American Society of Hematology has published recommendations for the treatment of thalassemia during the pandemic.[519] They advise that blood transfusion and luspatercept should be continued as usual. Iron chelation should be continued in well patients. If a patient develops COVID-19 then interruption of iron chelation is usually advisable; the case should be discussed with the patient's hematologist. Febrile, splenectomized patients should be investigated for bacterial infection and receive empiric antibiotics to cover secondary bacterial infections.[519]
The Thalassaemia International Federation (TIF) has published a position statement suggesting management strategies during the pandemic, covering patients’ risk level, adaptation of hemoglobinopathy care, safety of blood transfusions, blood supply challenges, and lifestyle and nutritional considerations.[520]
go to our full topic on Thrombotic thrombocytopenic purpura
Plasma exchange remains the recommended initial treatment for immune-mediated thrombotic thrombocytopenic purpura (iTTP). Corticosteroids and rituximab should still be used in treatment of acute iTTP. Patients with severely deficient ADAMTS13 activity may still receive rituximab to prevent relapse; the potential increased risk of COVID-19 complications should be balanced against the benefit of delaying or preventing relapses of iTTP. If access to plasma exchange is limited, the patient should ideally be transferred to a facility that can offer plasma exchange; otherwise, caplacizumab and immunosuppressive therapy alone may be considered.[521]
If a patient develops COVID-19, plasma exchange should be used in the same way as for other patients; the risks and benefits of corticosteroids and rituximab should be carefully considered. Caplacizumab may be used in conjunction with plasma exchange as a temporizing measure to protect from exacerbations and relapses until recovery from COVID-19; after recovery, corticosteroids and/or rituximab may be used to increase ADAMTS13 activity.[521]
Trials of severe acute respiratory syndrome coronavirus 2 vaccination in patients with hematological conditions are lacking. Such patients are likely to have an attenuated response to vaccination. Additional measures, such as early vaccination of household contacts, may be considered.[248]
go to our full topic on Tuberculosis
Coinfection with tuberculosis (TB) and COVID-19 is common globally, and is associated with higher mortality than infection with COVID-19 alone. The mean in-hospital fatality rates for TB and COVID-19 coinfection are 6.5% in high-income countries, and 22.5% in low/middle-income countries.[522]
Symptoms of tuberculosis may mimic those of COVID-19, and testing for tuberculosis may be considered in selected patients presenting with COVID-19.[523]
The Centers for Disease Control and Prevention advise that administration of mRNA COVID-19 vaccines should not be delayed because of testing for tuberculosis (TB) infection. Immune-based testing (tuberculin skin testing or interferon-release assay) may be done before or at the same time as COVID-19 vaccination. If this is not possible, immune-based testing should be delayed for at least 4 weeks after completion of COVID-19 vaccination, but generally should not be cancelled. Patients who have active TB, or an illness that is being evaluated as active TB, may receive COVID-19 vaccination. Severe or acute illness is a precaution to administration of all vaccines. Patients who have symptoms or other diagnostic findings consistent with active TB should receive further evaluation, regardless of immune based testing results.[524]
The UK National Health Service advises that an interferon-release assay blood test may be used instead of tuberculin skin testing for contact tracing to reduce person-to-person contact during the pandemic. Testing for latent tuberculosis and neonatal Bacillus Calmette-Guérin vaccination should continue.[525]
go to our full topic on Ulcerative colitis
People with inflammatory bowel disease are at increased risk of COVID-19-related critical care admission or death, compared with the general population, after adjusting for confounding and mediating factors (HR 1.08, 95% CI 1.01 to 1.16). South Asian, black, and mixed ethnicity people with inflammatory bowel disease have a higher risk of COVID-19-related death, compared with white people with inflammatory bowel disease.[64]
People taking tumor necrosis factor inhibitors, interleukin (IL)-12/IL-23 inhibitors, or Janus kinase (JAK) inhibitors do not have an increased risk of COVID-19-related critical care admission or death, compared with people taking standard systemic therapy (e.g., azathioprine). Patients taking JAK inhibitors have an increased risk of COVID-19 related hospital admission, compared with patients on standard systemic therapy.[64]
Blood tests to monitor response to therapy should be performed at the minimum safe frequency.[338] International guidelines recommend that patients stop taking methotrexate, thiopurines, or tofacitinib if they develop COVID-19. Detailed recommendations for stopping and restarting medications are given depending on the level of IBD activity and severity of COVID-19 infection.[339][340] Decisions on surgery should be individualized for each patient with a multidisciplinary team.[341]
The British Society of Gastroenterology has published a position statement strongly supporting SARS-CoV-2 vaccination for patients with UC.[342] A third dose (or booster dose) of SARS-CoV-2 vaccine is also recommended for all patients with IBD receiving immunosuppressive treatment and all patients with IBD who are extremely clinically vulnerable. A third dose of SARS-CoV-2 vaccine results in a serologic response in 84% of patients with immune-mediated inflammatory diseases who had a weak response to the standard 2-dose regimen.[343] Some immunosuppressive therapies may impact efficacy of COVID-19 vaccines in patients with IBD, and this should be considered in the timing of vaccinations.[343][344]
Prognostic models for predicting the probability of adverse COVID-19 outcomes in patients with IBD are being developed.[345]
go to our full topic on Uveitis
The International Uveitis Study Group has published consensus recommendations on the management of patients with uveitis during the COVID-19 pandemic. Patients without symptoms of COVID-19 should continue on their usual immunosuppressive treatment. Patients with symptoms or signs of COVID-19 should be tested as soon as possible to confirm the diagnosis. Immunosuppressive treatment, except tocilizumab or interferon, should be stopped. Slow reduction of systemic corticosteroids should be discussed with the treating medical team. Patients who are asymptomatic and test positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) should continue immunosuppressive therapy. The dose should be reduced if their white blood cell count falls below 4000 cells/microlitre.[526]
Patients who are due to start immunosuppressive treatment should be tested for SARS-CoV-2 in addition to the usual screen for infectious diseases. Consideration may be given to using local or regional corticosteroids as bridging therapy to delay the start of systemic immunosuppression. Patients with Behcet’s disease may require first-line systemic therapy; if so, self administered injections to reduce hospital outpatient visits could be considered.[527]
go to our full topic on Viral gastroenteritis
COVID-19 may present with gastrointestinal (GI) symptoms that mimic viral gastroenteritis. The estimated pooled prevalence of GI symptoms in patients with COVID-19 varies from less than 10% to 15%.[424][528] Nausea or vomiting, anorexia, and diarrhea are the most common manifestations.[528] Patients with severe COVID-19 had higher rates of GI symptoms than those with less severe disease. Most patients with GI symptoms and COVID-19 have concomitant respiratory symptoms or fever; 3% of patients reported GI symptoms only.[529] Patients may present with nausea or diarrhea 1 to 2 days prior to onset of fever and breathing difficulties.[530] A retrospective cohort study found that median duration of viral shedding in stool samples was 22 days, compared with 18 days in respiratory samples and 16 days in serum samples. The median duration of shedding was lower in mild illness (14 days) compared to severe illness (21 days).[531]
Guidelines from the American Gastroenterological Association (AGA) recommend that outpatients with new-onset diarrhea are asked about high risk contact exposure, whether they have a history of COVID-19-associated symptoms, and whether they have other GI symptoms (nausea, vomiting, abdominal pain).[424] Patients with new-onset GI symptoms should be monitored for symptoms of COVID-19, as GI symptoms may precede other COVID-related symptoms by a few days. Currently, there is not enough evidence to support stool testing for diagnosis or monitoring of COVID-19 as part of routine clinical practice.[424] In hospitalized patients with known or suspected COVID-19, the AGA recommends obtaining a thorough history of GI symptoms, including onset, characteristics, duration, and severity.
go to our full topic on Vitamin B12 deficiency
The British Society for Haematology has published guidance on vitamin B12 (cyanocobalamin) supplementation during the COVID-19 pandemic.
Patients who have B12 deficiency that is not related to diet (e.g., pernicious anemia, gastrectomy, inflammatory bowel disease, achlorhydria) should be screened for symptoms of COVID-19 before injections. The ongoing need for B12 injections should be assessed for each patient; oral vitamin B12 may be offered until intramuscular injections can be resumed, aiming to have the shortest possible break between injections.[532]
Patients who have B12 deficiency related to diet should be offered advice on dietary sources of B12. Patients may suspend B12 supplementation during the pandemic because they are B12 replete; patients may also be offered oral B12 supplementation.[532]
Further hematology resources are available at:
go to our full topic on Vitamin D deficiency
A link between vitamin D deficiency and COVID-19 infection is emerging. Studies have found that lower levels of 25-hydroxy-vitamin D (25(OH)D) among patients admitted to hospital with COVID-19 may be associated with more severe illness.[533][534] One prospective cohort study found that a lower 25(OH)D concentration was associated with increased risk of invasive mechanical ventilation and in-hospital mortality, even after adjustment for age, sex, comorbidities, and day of illness.[533] Vitamin D deficiency persisted after recovery from critical illness.
In the UK, people are advised to consider taking a daily vitamin D supplement to maintain bone and muscle health during the COVID‑19 pandemic. The guidance advises:[535]
Adults (including women who are pregnant or breast-feeding), young people, and children over 4 years should consider taking a daily supplement of vitamin D between October and early March.
Adults, young people, and children over 4 years should consider taking a daily supplement of vitamin D throughout the year if they have little or no sunshine exposure, or if they have dark skin.
Babies from birth to 1 year should have a daily supplement of vitamin D throughout the year if they are breastfed or formula-fed and are having less than 500 mL of infant formula a day.
Children aged 1-4 years should have a daily supplement of vitamin D throughout the year.
Randomized trials are needed to determine whether a causal relationship exists between vitamin D concentrations early in the course of the disease and development of critical illness.
Contributors
Authors
Editorial Team
BMJ Publishing Group
Disclosures
BMJ staff and contractors follow BMJ company policy on declarations of interests. None have interests to declare.
Evidence
Can interventions commonly available during pandemics control heavy menstrual bleeding?
More EvidenceLog in or subscribe to access all of BMJ Best Practice
Use of this content is subject to our disclaimer