Skip to main content
Wiley - PMC COVID-19 Collection logoLink to Wiley - PMC COVID-19 Collection
. 2021 Oct 22;32(3):e2309. doi: 10.1002/rmv.2309

Coronavirus disease 2019 (Covid‐19) vaccination recommendations in special populations and patients with existing comorbidities

Zeinab Mohseni Afshar 1, Arefeh Babazadeh 2, Alireza Janbakhsh 1, Feizollah Mansouri 1, Terence T Sio 3, Mark J M Sullman 4,5, Kristin Carson‐Chahhoud 6, Rezvan Hosseinzadeh 7, Mohammad Barary 7, Soheil Ebrahimpour 2,
PMCID: PMC8646697  PMID: 34677889

Summary

Vaccination against severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) is a crucial step in ending the current worldwide pandemic. However, several particularly vulnerable groups in the population were not included in sufficient numbers in coronavirus disease 2019 (Covid‐19) vaccine trials. Therefore, as science advances, the advice for vaccinating these special populations against Covid‐19 will continue to evolve. This focused review provides the latest recommendations and considerations for these special populations (i.e., patients with rheumatologic and autoimmune disorders, cancer, transplant recipients, chronic liver diseases, end‐stage renal disease, neurologic disorders, psychiatric disorders, diabetes mellitus, obesity, cardiovascular diseases, chronic obstructive pulmonary disease, human immunodeficiency virus, current smokers, pregnant and breastfeeding women, the elderly, children, and patients with allergic reactions) using the currently available research evidence.

Keywords: Covid‐19, efficacy, immunocompromise, safety, SARS‐CoV‐2, vaccination


Abbreviations

ABA

Abatacept

ACE2

Angiotensin‐converting enzyme 2

ACIP

Advisory Committee on Immunisation Practices

ACOG

American College of Obstetricians and Gynecologists

ACR

Acute cellular rejection

ASRM

American Society for Reproductive Medicine

BAFF

B cell activation factor

BMI

Body mass index

CAD

Coronary artery disease

CD

Cluster of differentiation

CDC

Centers for Disease Control and Prevention

CIDP

Chronic inflammatory demyelinating polyneuropathy

CLD

Chronic liver disorders

CNS

Central nervous system

COPD

chronic obstructive pulmonary disease

Covid‐19

Coronavirus disease 2019

CVD

Cardiovascular disorder

CVID

Common variable immunodeficiency

DMARD

Disease‐modifying antirheumatic drug

DMT1

Diabetes mellitus type 1

DMT2

Diabetes mellitus type 2

DMTs

Disease‐modifying therapies

ESRD

End‐stage renal disease

EUA

Emergency Use Authorisation

FDA

Food and Drug Administration

GBS

Guillain‐Barré syndrome

HBV

Hepatitis B virus

HCC

Hepatocellular carcinoma

HCV

Hepatitis C virus

HIV

Human immunodeficiency virus

HSCT

Haematopoietic stem cell transplant

ICI

Immune checkpoint inhibitors

ICU

Intensive care unit

IL

Interleukin

IRAE

Immune‐related adverse event

ISRR

Immunisation stress‐related response

JAK

Janus kinase

JCVI

Joint Committee on Vaccination and Immunisation

MELD

Model for End‐stage Liver Disease

MERS

Middle East respiratory syndrome

MHRA

Medicines and Healthcare Products Regulatory Agency

MIS‐C

Multisystem inflammatory syndrome in children

MTX

Methotrexate

NMD

Neuromuscular disorder

NYHA

New York Heart Association

PAD

Peripheral arterial disease

PBC

Primary biliary cholangitis

PD

Peritoneal dialysis

PEG

Polyethylene glycol

PI3K

Phosphatidylinositol 3‐kinases

PNES

Psychogenic non‐epileptic seizures

RBD

Receptor‐binding domain

SARS‐CoV‐2

Severe acute respiratory syndrome coronavirus 2

SLE

Systemic lupus erythematosus

SOT

Solid‐organ transplant

TCZ

Tocilizumab

TNF

tumour necrosis factor

WHO

World Health Organisation

1. INTRODUCTION

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) has resulted in many individuals becoming infected, more than four million deaths, and has placed an unprecedented burden on public health services worldwide. 1 , 2 , 3 Vaccinations against severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) is a crucial step in ending the current worldwide pandemic. However, several particularly vulnerable groups in the population were not included in sufficient numbers in coronavirus disease 2019 (Covid‐19) vaccine trials. 4 Table 1 summarises the current Covid‐19 vaccination recommendations in these special populations and patients with existing comorbidities. Therefore, as science advances, the advice for vaccinating these special populations against Covid‐19 will continue to evolve. This focused review provides the latest recommendations and considerations for these special populations using available research evidence.

TABLE 1.

Summary of existing Covid‐19 vaccination recommendations in special populations and in patients with existing comorbidities

Vaccine platform
mRNA Adenoviral vector
Most common side effects Fatigue, headache, chills, muscle pain, fever. Worsen after the second dose. Injection site pain, fever, muscle aches, headache, fatigue. Worsen after the second dose.
Who should not be vaccinated People with a history of allergic reactions to vaccine ingredients, including polyethylene glycol, and anyone with a history of allergic reactions to polysorbate. a Anyone with a severe allergic reaction to an ingredient in the vaccine. a
Significant side effects (rare) Pfizer/BioNTech and Moderna: Anaphylaxis, Bell's palsy, autoimmune hepatitis, myocarditis, pericarditis Janssen: VITT, demyelinatingOxford/AstraZeneca: VITT, transverse myelitis, demyelinating
Rheumatologic and autoimmune diseases • Corticosteroids: Taper to <10 mg/day prior to vaccination. • Corticosteroids: Taper to <10 mg/day prior to vaccination.
• MTX: Withhold 2 weeks before and after vaccination. • MTX: Withhold 2 weeks before and after vaccination.
• Anti‐TNF and IL‐17 medications: No specific dose reduction is required. • Anti‐TNF and IL‐17: No specific dose reduction is required.
• Anti‐IL‐6 medications: Vaccination should be 12 weeks before/after TCZ administration. • Anti‐IL‐6: Vaccination should be 12 weeks before/after TCZ administration.
• JAK inhibitors: Withhold 1–2 weeks before and after vaccination. • JAK inhibitors: Withhold 1–2 weeks before and after vaccination.
• Anti‐CD20 medications: Withhold 4 weeks before until 6 months after vaccination. • Anti‐CD20 medications: Withhold 4 weeks before until 6 months after vaccination.
• ABA: Data are not yet available. • ABA: Data are not yet available.
Cancer • Anti‐CD20 or cytotoxic therapies inactivate the mRNA vaccine.• Cytotoxic chemotherapy: Start chemotherapy courses 2 weeks after vaccination. • Cytotoxic chemotherapy: 2 weeks after vaccination• If chemotherapy has already been given, vaccination should be given between courses of chemotherapy.
• If chemotherapy is already initiated, vaccination should be given between courses of chemotherapy.• Lymphocyte or plasma cell‐depleting regimens: Vaccination should be 2 weeks before or 3 months after the end of treatment. • Lymphocyte or plasma cell‐depleting regimens: Vaccination should be 2 weeks before or 3 months after the end of treatment.
Transplant patients • Vaccination is recommended early in the course of the underlying disease. • Vaccination is recommended early in the course of the underlying disease.
• After transplantation, postpone vaccination for 3–6 months. • After transplantation, postpone vaccination for 3–6 months.
• If the first dose is received before the transplantation, the second dose should be administered at least 4 weeks after transplantation • If the first dose is received before the transplantation, the second dose should be administered at least 4 weeks after transplantation
• A third dose may be warranted for optimal immunity. • A third dose may be warranted for optimal immunity.
CLD • Recommended, with priority given to patients with higher MELD scores. • Recommended, with priority given to patients with higher MELD scores.
• Vaccination of patients with CLD undergoing treatment for HBV, HCV, PBC, and autoimmune hepatitis should be performed without discontinuing their therapy. • Vaccination of patients with CLD undergoing treatment for HBV, HCV, PBC, and autoimmune hepatitis should be performed without discontinuing their therapy.
• Vaccination is safe and recommended for patients with HCC. • Vaccination is safe and recommended for patients with HCC.
• Patients on the transplant list should receive two doses of the vaccine before the transplant. • Patients on the transplant list should receive two doses of the vaccine before the transplant.
• If the patient received the first dose before the transplant, the next dose could be given to him/her at 6 weeks to 3 months after the transplant. • If the patient received the first dose before the transplant, the next dose could be given to him/her at 6 weeks to 3 months after the transplant.
• Vaccination should be withheld in liver transplant recipients with active ACR or those receiving high‐dose corticosteroids until the condition is resolved. • Vaccination should be withheld in liver transplant recipients with active ACR or those receiving high‐dose corticosteroids until the condition is resolved.
ESRD • Taper steroid doses below 20 mg prednisone equivalent daily before vaccination. • Taper steroid doses below 20 mg prednisone equivalent daily before vaccination.
• If the patient received anti‐CD20 medication, vaccination should be delayed for at least 6 months after the last dose of the therapy. • If the patient received anti‐CD20 medication, vaccination should be delayed for at least 6 months after the last dose of the therapy.
• If an active underlying disease is present in these patients, immunosuppressive therapy is prioritised over vaccination. • If an active underlying disease is present in these patients, immunosuppressive therapy is prioritised over vaccination.
Neurologic disorders • Vaccination is recommended for MS patients. • Vaccination is recommended for MS patients.
• MS patients receiving ocrelizumab can receive the vaccine 4–6 weeks before starting the treatment or 4–6 months after ending the treatment. • MS patients receiving ocrelizumab can receive the vaccine 4–6 weeks before starting the treatment or 4–6 months after ending the treatment.
• DMTs for MS can reduce the antibody response of vaccines. • DMTs for MS can reduce the antibody response of vaccines.
• Patients receiving IRT, including alemtuzumab, rituximab, and ocrelizumab, can be vaccinated 6 months after the treatment. • Patients receiving IRT, including alemtuzumab, rituximab, and ocrelizumab, can be vaccinated 6 months after the treatment.
• In high‐dose or long‐term treatments with corticosteroids, vaccination is allowed 4–6 weeks after cessation of the treatment. • In high‐dose or long‐term treatments with corticosteroids, vaccination 4 to 6 weeks after cessation of treatment
• CDC recommended mRNA vaccines for GBS patients. • For GBS patients, data are not yet available.
Psychiatric disorders • Recommended, but no studies have been performed solely on the Covid‐19 vaccines and neuropsychiatric disorders. • Recommended, but no studies have been performed solely on the Covid‐19 vaccines and neuropsychiatric disorders.
• Antipsychotic agents suppress vaccine‐induced antibody formation. • Antipsychotic agents suppress vaccine‐induced antibody formation.
• Antidepressant therapy would normalise the vaccine‐induced immune response. • Antidepressant therapy would normalise the vaccine‐induced immune response.
DM Recommended. Patients with DMT2 are prioritised higher than patients with DMT1. Recommended. Patients with DMT2 are prioritised higher than patients with DMT1.
Obesity Recommended Recommended
CVD Recommended Recommended
HIV Recommended Data not yet available.
COPD Recommended Recommended
Current smokers Recommended Recommended
Pregnancy and breastfeeding Recommended Janssen: Individualised risk/benefit assessment should be performed before vaccination.
Oxford/AstraZeneca: Data are not yet available.
Elderly Recommended Recommended
Children Pfizer/BioNTech: FDA recommends this vaccine for adolescents 12–18 years of age. Data is not yet available.
Moderna: Data are not yet available.
Allergic diseases Recommended unless a prior history of allergy to PEG or positive skin test for this agent is present. Recommended unless a prior history of allergy to polysorbate or positive skin test for this agent is present.

Abbreviations: ABA, abatacept; ACR, acute cellular rejection; CDC, Centers for Disease Control and Prevention; CLD, chronic liver disease; COPD, chronic obstructive pulmonary disease; CVD, cardiovascular diseases; DMT1, diabetes mellitus type 1; DMT2, diabetes mellitus type 2; DMT, disease‐modifying therapy; ESRD, end‐stage renal disease; FDA, Food and Drug Administration; GBS, Guillain‐Barré syndrome; HBV, hepatitis B virus; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; HIV, human immunodeficiency virus; IRAEs, immune‐related adverse events; IRT, immune‐reconstitution therapies; MELD, model for end‐stage liver disease; MS, multiple sclerosis; PBC, primary biliary cholangitis; PEG, polyethylene glycol; SLE, systemic lupus erythematosus; VITT, vaccine‐induced immune thrombotic thrombocytopenia.

a

For more information about vaccine ingredients, please see: CDC.gov.

2. RHEUMATOLOGIC AND AUTOIMMUNE DISORDERS

Individuals on immunosuppressive therapies are among those most susceptible to Covid‐19‐related morbidity and mortality. Although vaccinating these populations should be a high priority for healthcare providers and governments, they have mostly been excluded from vaccine trials. 5 The most important reason behind such exclusions is that immunosuppressive therapies can impair the vaccine‐induced humoral and cellular immune responses, making it difficult to measure its effectiveness on the immune system. 6 It is important to note that there are many such diseases, and we do not fully understand the pathogenesis of any of them. Thus, it is essential to consider the diseases and the treatments for them when considering vaccination. For example, does the disease or treatment suppress T cell or B cell responses, and will this differ according to the vaccine used? In the following section, we discuss the impact of several immunosuppressive agents on vaccine response. It is essential to know that some of the information has been generalised from experiences with influenza, pneumococcal, and tetanus vaccines.

The effect of corticosteroids on vaccine‐induced antibody production is dose‐dependent. Prednisolone doses higher than 10 mg daily, or equivalent doses of other corticosteroids, impair vaccine response, and thus, tapering the dose around the time of vaccination would appear necessary. 7 Disease‐modifying antirheumatic drugs (DMARDs) are agents used for decreasing inflammation in rheumatic disorders. Methotrexate (MTX), hydroxychloroquine, sulfasalazine, leflunomide, cyclophosphamide, mycophenolate, and azathioprine are examples of these agents. 8 All DMARDs can affect antibody responses, but none of them, except MTX and cyclophosphamide, lower immunologic responses below the threshold of seroprotection. 8 In addition, MTX suppresses humoral response by interacting with the B cell activation factor (BAFF). 9 Thus, it is reasonable to withhold MTX for at least two weeks before and after vaccination. However, withholding the medication for more than two weeks may lead to a flare‐up in the underlying disease. 10 Thus, the timing of withholding medication should be carefully monitored to lower the risk of adverse events.

Anti‐tumour necrosis factors (TNFs) and interleukin (IL)‐17 blockers seem to have no significant effect on vaccine‐induced immunity unless used concurrently with MTX. 11 , 12 IL‐6 inhibitors, such as tocilizumab (TCZ), seem to have no substantial effect on vaccine‐induced seroprotection unless co‐administered with MTX. 13 However, while some studies have reported there should be at least 12 weeks between tocilizumab administration and vaccination to have the ideal antibody response, 13 other research has shown that TCZ does not impair antibody production for other types of vaccines (not Covid‐19). 14 Information about the impact of IL‐1 antagonists, including anakinra, and canakinumab, on vaccine‐induced seroprotection, is scarce, and more studies are urgently needed. 15 Janus kinase (JAK) inhibitors, such as baricitinib and tofacitinib, might trigger drug interactions with mRNA vaccines, primarily when used concurrently with MTX. 16 Therefore, it is better to withhold JAK inhibitors for 1–2 weeks on either side of the vaccination date. 16 Anti‐CD20 agents (e.g., rituximab) impair B cell production, making patients prone to severe forms of Covid‐19. These agents are also believed to profoundly affect vaccine‐induced antibody responses, even several months after their use. 17 Therefore, it is recommended that their use be limited only to essential cases or administered with a minimum gap of four weeks before and six months after vaccination. 17 The impact of T‐cell lymphocyte activation inhibitors, such as abatacept (ABA), on vaccine‐induced immunity, is controversial and more studies are needed to clarify any interactions. 18

In general, TNF inhibitors, such as TCZ, ABA, and IL‐17 antagonists, seem to impact vaccine efficacy negatively. The Centers for Disease Control and Prevention (CDC) recommends at least a two‐week spacing between administration of these agents and vaccination. 14 Thus, the decision to vaccinate rheumatologic patients who use the medications mentioned above should be individually made since it appears that low‐dose immunosuppressive agents do not significantly affect the vaccine‐induced antibody response. 14 Overall, it appears best to vaccinate these patients when their underlying disease is under control. 14 It is also noteworthy that allergic reactions in these patients may happen following vaccination, especially in patients with systemic lupus erythematosus (SLE), necessitating a more extended period (at least 2 h) of monitoring following vaccination. 14

Another important consideration is the safety profile of the Covid‐19 vaccines in rheumatology patients, which do not seem to be contraindicated since none of the treatments attenuate vaccines. 19 However, it is currently unknown whether Covid‐19 vaccines can trigger autoimmunity by direct immune‐activating or non‐specific adjuvant effects, leading to the exacerbation of rheumatologic or autoimmune disorders. Such events may follow the Pfizer/BioNTech vaccine administration. 19 Nonetheless, as the benefits of vaccinating vulnerable individuals outweigh the risk of exacerbating rheumatic disorders, the American College of Rheumatology recommends vaccination against Covid‐19 in all eligible rheumatologic patients. 20 Some have hypothesised that protein subunit‐based vaccines, such as the Novavax vaccine candidate, will have better efficacy and safety profiles for rheumatologic, autoimmune, and autoinflammatory patients. 21

3. CANCER

Patients with cancer are particularly vulnerable to adverse outcomes from moderate and severe Covid‐19 infections, which may be due to their underlying malignancy, cytotoxic chemotherapy, radiotherapy, other existing comorbidities, and advanced age. 22 We must consider the diseases and their treatments when considering vaccination. For example, some treatments may impair cellular or humoral immunity to affect the overall vaccine response. It should also be noted that these effects might differ according to the vaccine used. Leukaemia, non‐Hodgkin's lymphoma, and lung cancer are the most commonly seen malignancies related to severe Covid‐19 cases. 22 Therefore, owing to the relatively high fatality rate of Covid‐19 in active cancer patients, this group is among the most highly prioritised to be vaccinated against the disease. 22 Currently, there is no preferred Covid‐19 vaccine for these patients, and so these individuals can receive any approved vaccine under their physician's supervision.

In general, although it is believed that the natural‐ or vaccine‐induced antibody response in cancer patients is suboptimal, especially among those with haematologic malignancies, there are no absolute contraindications to the Covid‐19 vaccine in cancer patients undergoing glucocorticoid therapy, chemotherapy, radiotherapy, hormonal therapy, immunotherapy, or surgery. 23 The efficacy of mRNA vaccines in patients with solid tumours and haematological malignancies has been reported to be 83% and 72%, respectively. Anti‐CD20 or cytotoxic therapies in these patients is thought to be the reason for the lower‐than‐expected immune response in cancer patients, making mRNA vaccines less effective in these patients. 24 However, the T‐cell response induced by current vaccines is strong enough to recommend immunisation in these patients, except during the intensive phase of chemotherapy. 24 Thus, carefully considering the timing and interval between the vaccine and the last cycle of chemotherapy would be an essential factor in the adequate vaccine immune response in this population. 24 For patients planning to start cytotoxic chemotherapy, it is better to administer the first dose of the vaccine at least two weeks before initiating the first chemotherapy cycle. However, for those already on cytotoxic chemotherapy, the first dose of the vaccine can be administered between chemotherapy cycles. 25 Furthermore, since these immunocompromised patients mount insufficient antibody response after natural SARS‐CoV‐2 infection and consequently shed the virus for a more extended period, vaccinating these individuals is vital in arresting the virus cycle, 26 further validating the importance of vaccinating as many cancer patients in the community as possible.

Several potential concerns regarding using checkpoint inhibitors and targeted therapies, such as tyrosine kinase inhibitors, including erlotinib and imatinib, and its potential interference with viral vaccines exist. 27 However, there is no reported data at this time. The prevalence of immune‐related adverse events (IRAEs) following vaccination is unknown for checkpoint inhibitors, yet this side effect may occur within 2–3 days following vaccination. 27 Therefore, avoiding vaccination may be reasonable in cases of significant concern. 28 Patients receiving lymphocyte or plasma cell‐depleting regimens should delay Covid‐19 vaccination for at least 3 months following the end of their immunotherapeutic treatment to get the best antibody response. However, if they are about to start these regimens, it is reasonable to administer the first dose of the vaccine at least two weeks before starting the immunotherapy course. 24 It is also believed that the Covid‐19 vaccine is safe and effective in patients undergoing radiation therapy. 25 At the beginning of the pandemic, there were significant concerns regarding clinical resource distribution and keeping patients safe from Covid‐19 infections. 29 However, the clinical burden has substantially reduced since vaccines became available.

Another important issue in patients receiving active systemic therapies is the occurrence of post‐vaccination fever. However, any fever should not necessarily be attributed to vaccine response since other differential diagnoses, like neutropenic fever, Covid‐19 infection, post‐surgical complications, and underlying cancer relapse may also be responsible. In addition, it is possible that the vaccinated individuals, including immunocompromised patients, may acquire SARS‐CoV‐2 with or without prior infection history. Therefore, adhering to preventive measures, such as hand hygiene, face coverings, and social distancing, are still likely to be necessary for everyone in the foreseeable future.

4. TRANSPLANT PATIENTS

Transplant recipients are another at‐risk group that should be prioritised for getting vaccinated since they have an increased risk of infection and developing more severe forms of Covid‐19. 30 However, these patients have not been included in the vaccine trials to date, and therefore vaccine safety, efficacy, and durability profiles have not been measured in these patients. 31 Nevertheless, considering the consequences of severe Covid‐19 in this population and the previous experiences with other vaccines, such as influenza vaccines, in stable transplant patients, the benefits of vaccination outweigh the possible side effects. 32 Therefore, SARS‐CoV‐2 vaccination is strongly recommended for these patients. 32 Nonetheless, it is also probable that the immunosuppressed condition in these patients may cause a lower anti‐SARS‐CoV‐2 antibody response, depending on the period since the transplantation, the intensity of the immunosuppression, and the type of transplantation. 33

The conditioning and maintenance of immunosuppressive regimens and their dosing and intensity vary significantly between solid organ transplant (SOT) recipients and haematopoietic stem cell transplant (HSCT) recipients. 34 It has been suggested that antimetabolite maintenance therapy can lead to a weaker post‐vaccination antibody response than other regimens. 34 Therefore, those patients should be vaccinated early in the course of their underlying disease, as the timing of vaccination is a significant factor in determining its effectiveness. 35 Also, it is better to postpone vaccination to at least 3–6 months after transplantation when the immunosuppression is lower. 36 Nevertheless, if the first dose of the vaccine is received before transplantation, the second dose should be postponed until at least four weeks post‐transplant. 36

Some experts believe that a third dose is needed in transplant patients, considering shorter longevity and lower antibodies' efficacy. 37 A recent randomised placebo‐controlled trial showed that a third dose of the mRNA‐1273 (Moderna) vaccine could significantly increase the anti‐receptor‐binding domain (RBD) antibody levels and anti‐SARS‐CoV‐2‐specific T‐cell counts in transplant patients, indicating a higher humoral and cellular immunity triggered after a third dose of the vaccine. 37 There is also evidence of a difference in the antibody response from different vaccines, with the mRNA‐1273 (Moderna) vaccine being found to result in a more significant immune response in transplant recipients. 38 In general, vaccination is not contraindicated in stable transplant recipients, except live‐attenuated vaccines, which might lead to disseminated infection, especially when their immunosuppression condition is highest, usually occurring in the first 3–6 months after the transplant. 38 Fortunately, none of the current Covid‐19 vaccines are live‐attenuated, meaning it is possible to administer the vaccine to this vulnerable population. 38 Apart from vaccine‐related efficacy, durability, and safety issues, vaccine‐associated allograft rejection is a unique concern in this population, although this has not been reported with any Covid‐19 vaccines. 38 Nevertheless, although extremely rare, there would appear to be a slight chance of stimulating immunologic rejection reactions via the vaccination‐induced immune response. 38

5. CHRONIC LIVER DISEASES

Patients with chronic liver disorders (CLD), including cirrhosis, hepatobiliary malignancies, and transplant candidates (or recipients), are vulnerable populations at risk of more severe forms of Covid‐19 and higher mortality. 39 This population needs special attention due to their underlying disease, and many operations or treatments were delayed due to the hospitals being overwhelmed or not wanting to put patients at more risk. 39 Therefore, vaccination should also be a priority for these patients. 39 Vaccination seems to be safe in stable CLDs, such as compensated cirrhosis and viral hepatitis. 39 Moreover, individuals with decompensated cirrhosis, liver malignancies, and liver transplant patients should also be prioritised for vaccination using the Child‐Turcotte‐Pugh or the Model for End‐stage Liver Disease (MELD) scores. 40 The higher the scores, the sooner they should get the vaccine. However, the extent of the vaccine‐induced immune response is unknown and expected to be suboptimal in these patients due to their underlying disease and the medications they use. It is also noteworthy that mRNA Covid‐19 vaccines are expected to have favourable safety and efficacy profiles in these patients.

Patients with CLD who are on medical treatment for hepatitis B virus (HBV), hepatitis C virus (HCV), primary biliary cholangitis (PBC), or autoimmune hepatitis do not need to stop therapy in order to receive Covid‐19 vaccines. 41 Moreover, patients with hepatocellular carcinoma (HCC) on locoregional (i.e., imaging‐guided liver tumour‐directed procedures) or systemic therapy can also be vaccinated without pausing their treatment. 41 Nevertheless, in recent infections or fever cases, vaccination should be delayed until the condition is stable. 41 However, the use of immune checkpoint inhibitors (ICI) in patients with some liver diseases (e.g., HCC) is still a concern that should be further studied since immune‐related adverse reactions are a possible result of vaccine interactions with ICI. 41 Moreover, the timing of the vaccination in hepatobiliary cancer patients is heavily dependent on the stage of the malignancy, types of medication, and concomitant comorbidities. 41 Patients with CLD on the waiting list for transplantation should receive two doses of the vaccine, preferably before the transplant. However, they should be encouraged to receive the vaccine even if their second is scheduled after the liver transplant. 30 However, for these patients, the time interval between the two doses of the vaccine does not necessarily need to be four weeks, and the second dose should be planned after transplant (e.g., within 6 weeks). 30 Moreover, following liver transplantation, the best time to be vaccinated would be when the immunosuppression has been attenuated and other prophylactic medications are withheld, ideally six weeks to three months post‐liver transplantation. 30 In order to prevent acute cellular rejection (ACR), liver transplant recipients should not discontinue their immunosuppressive medications solely to achieve a favourable immune response after vaccination. Moreover, Covid‐19 vaccinations should be withheld in liver transplant recipients with active ACR or those receiving high‐dose corticosteroids until the condition is resolved.

6. END‐STAGE RENAL DISEASE

Patients with end‐stage renal disease (ESRD) are also more prone to infection with Covid‐19 due to their regular or occasional dialysis sessions, where they are exposed to a densely populated environment with a high possibility of SARS‐CoV‐2 transmission. 42 Moreover, these patients may present with atypical manifestations of SARS‐CoV‐2 infection, leading to a delay in diagnosing the disease. 43 In addition, patients often have multiple comorbidities and higher rates of polypharmacy. 44 Therefore, the risk of developing a severe or lethal SARS‐CoV‐2 infection is likely higher in this population, and vaccinating them early against Covid‐19 is highly recommended. 45 Moreover, although ESRD patients develop seroconversion following vaccination, they are well‐established to achieve a less robust and perhaps less durable antibody response. 46 The seropositivity rate after SARS‐CoV‐2 vaccination does not appear to differ between haemodialysis and peritoneal dialysis (PD) patients. 47 The extent of the immune response to SARS‐CoV‐2 vaccination depends on the vaccine type, the time spent since ESRD onset, and possibly age, body mass index (BMI), and nutritional status, as indicated by serum albumin and iron levels. With that in mind, several studies have suggested that a third or booster dose of vaccine would be necessary for these individuals to produce an optimal antibody response. 37 , 48 , 49 , 50

There does not appear to be a preference for one vaccine type over another, with adenoviral vector vaccines, such as the ChAdOx1 nCoV‐19 (Oxford/AstraZeneca) vaccine, and the mRNA vaccines (i.e., Pfizer/BioNTech, and Moderna) used for vaccinating ESRD patients. 49 , 51 In addition, patients with autoimmune renal diseases (e.g., IgA nephropathy) undergoing anti‐CD20 therapy (e.g., rituximab) should replace their immunosuppressive treatment with another non‐interfering regimen until a few weeks after vaccination. 52 For example, it is reasonable for these individuals to taper steroid doses below 20 mg prednisone (or equivalent) daily or wait for at least six months after the last rituximab dose before being vaccinated. 52 However, if their underlying disease is active, the immunosuppressive therapy is prioritised over‐vaccination, 53 although the activation or relapse of the underlying autoimmune kidney disease has only rarely been reported following vaccination. 54 , 55

7. NEUROLOGIC DISORDERS

Like individuals with many chronic disorders, neurological patients are at increased risk of severe Covid‐19 infection, complications, and mortality. 56 Patients with Alzheimer disease, Parkinson disease, motor neuron diseases, central nervous system (CNS) disorders, neuromuscular disorders (NMDs), and autoimmune disorders, including multiple sclerosis (MS), myasthenia gravis (MG), and Guillain‐Barré syndrome (GBS), are among the most concerning neurological disorders. 56 Thus, vaccination against Covid‐19 is vital for this population, 57 but vaccination risks and adverse events must be carefully monitored. For example, there is some concern that vaccination against SARS‐CoV‐2 may exacerbate MS by inducing immunological responses and triggering immunological reactions. 58 Nonetheless, vaccines are generally safe in MS patients, and following vaccination, there is a low probability of acute relapse, although there have been some reports of MS symptom aggravation (pseudo‐relapse). 58

Patients with a history of GBS and autoimmune conditions should receive mRNA Covid‐19 vaccines, if not contraindicated. 59 However, it is generally believed that some of the disease‐modifying therapies (DMTs) used to treat MS could reduce the antibody response following vaccination. 60 Moreover, these medications can affect the safety and efficacy of the vaccines. 61 Patients being treated with β‐interferons, glatiramer acetate, teriflunomide dimethyl fumarate, natalizumab, or sphingosine‐1‐phosphate receptor modulators fingolimod, ozanimod, and siponimod can be vaccinated at any time during their treatment, despite the likely vaccine response attenuation. 62 However, in MS patients scheduled to start ocrelizumab therapy, the two‐dose vaccine regimen should be administered at least 4–6 weeks before the initiation of their treatment course, or at least 4–6 months after the treatment course last ocrelizumab infusion. 63 In patients treated with immune‐reconstitution therapies, including alemtuzumab, and oral cladribine, it is better to delay vaccination until at least six months after the last course of treatment. 64 In patients on high‐dose or long‐term corticosteroids, vaccination should be delayed until 4–6 weeks after treatment. 65 Nonetheless, if these patients are not on DMTs, they should receive the SARS‐CoV‐2 vaccine as soon as possible.

There are theoretical concerns that mRNA‐based Covid‐19 vaccines may trigger the development of de novo neurodegenerative or neurologic disorders, such as demyelinating diseases or fever‐induced seizures. In this case, the potential vaccine‐induced adverse reaction could be even more debilitating than the viral infection. 66 , 67 The adjuvants used in vaccines, including anti‐SARS‐CoV‐2, might be responsible for potential neurologic adverse effects. 68 Another potential neurological adverse event that may result from vaccination is the immunisation stress‐related response (ISRR), which manifests itself as psychogenic non‐epileptic seizures (PNES). 69 Transverse myelitis, 70 , 71 , 72 GBS, 73 , 74 and Bell's palsy 75 are other potential neurological consequences of the Covid‐19 vaccination reported so far. Another potential adverse effect of these vaccines might be an exacerbation of MG and chronic inflammatory demyelinating polyneuropathy (CIDP). 76 The demyelinating disease has most commonly been reported following viral‐vector vaccines, which should be further investigated. 77

8. PSYCHIATRIC DISORDERS

Patients with psychiatric disorders, especially those with severe mental disorders, such as bipolar, schizophrenia, and major depressive disorders, are at increased risk of being infected with Covid‐19. Those with severe mental illnesses who were taking antipsychotics were at increased risk of mortality from Covid‐19, 78 which may be associated with patients' different lifestyles, habits, cognitive impairment, difficulties in adhering to infection control measures, and these population's socioeconomic status. 79 , 80 In addition, neuropsychiatric disorders and inflammation correlate, which could be a risk factor for more morbidity and mortality in these patients. 81 Therefore, these patients should be prioritised for Covid‐19 vaccination to minimise the risk of infection and transmission to other people. However, comorbid mental disorders have often been overlooked and underestimated in research evaluating the predictors of the severity and mortality from Covid‐19. 82 In many countries, institutionalised patients are listed high in the vaccination list, second only to healthcare personnel. 83

It must be noted that vaccinating patients with psychiatric disorders may also cause some concerns, as the efficacy, safety, and durability of the Covid‐19 vaccine are not yet known in these patients. Moreover, vaccinating these patients may also give them the false belief that they are fully protected against the disease, and thus, they may be more likely to ignore hygiene protocols. 83 Previous studies have shown a diminished antibody response to influenza and hepatitis vaccines in those with severe mental health issues. 84 , 85 Nevertheless, there are currently no published studies about the efficacy of Covid‐19 vaccines among patients with neuropsychiatric disorders. The relationship between vaccine response and psychotropic medications seems paradoxical. For example, antipsychotic agents (e.g., clozapine) might be associated with a syndrome resembling common variable immunodeficiency (CVID) in some patients that may lead to the suppression of vaccine‐induced antibody formation. 86 At the same time, antidepressant treatment might normalise the vaccine‐induced immune response. 87

9. DIABETES MELLITUS

Patients with diabetes mellitus, due to their comorbidities and acquired immunodeficiency, are at increased Covid‐19‐related morbidity and mortality. 88 Diabetes is one of the comorbidities most associated with adverse outcomes in Covid‐19 patients. However, there seems to be no difference in the severity or mortality of SARS‐CoV‐2 infection, based on whether they have diabetes type 2 (DMT2) or type 1 (DMT1). 89 Nonetheless, the CDC prioritised vaccination among patients with DMT2 over those with DMT1. 90 Therefore, vaccination is critical and necessary for this population, and endocrinologists should encourage their patients to be vaccinated as soon as possible. 91 It appears that the immune response following Covid‐19 vaccination is not affected by the serum glucose levels, as diabetic patients show an optimal antibody response. 92 Furthermore, Covid‐19 infected patients are at increased risk of developing new‐onset diabetes. Therefore, vaccination can also help to prevent an increase in diabetes mellitus in the community. 93

10. OBESITY

The association between obesity and viral infections was first demonstrated during the H1N1 epidemic in 2009, with the more body fat, the higher the risks of developing more severe illness and more extended hospitalisation in an intensive care unit (ICU). 94 , 95 , 96 The reason behind this association was thought to be the impairment of humoral and cellular immunity, along with lower vaccine‐induced immunity in these patients. 97 Another reason could be the marked rise of angiotensin‐converting enzyme 2 (ACE2) expression associated with high‐fat diets. 98 Another factor that plays a crucial role in making obese children more susceptible to infections, such as Covid‐19, is hyperinsulinism, which is due to the compensatory mechanisms of their pancreatic β cells to overcome insulin resistance in their body. 99 Thus, when higher amounts of insulin are required in intense metabolic activity, such as activating immune cells in response to the SARS‐CoV‐2 infection, their β cells cannot produce more insulin, as they are already working near their limit. 100 Moreover, SARS‐CoV‐2 can enter the pancreatic β cells via ACE2 receptors, causing virus‐triggered cell death or immune‐mediated loss of infected pancreatic β cell mass. 100 , 101 Insulin resistance in these patients can also impair the anti‐inflammatory and vasoactive characteristics of nitric oxide (NO) by reducing phosphatidylinositol 3‐kinases (PI3K). 102

Previous research has shown obesity as prevalent comorbidity among patients admitted into the ICU, especially among children and adolescents. 103 , 104 This research shows that a higher BMI may increase the likelihood of getting a severe disease. 105 Furthermore, a high BMI correlated with an increased need for mechanical ventilation in Covid‐19 patients, with about 85% of patients with a BMI > 35 kg/m2 requiring mechanical ventilation. 106 It is noteworthy that this correlation was independent of age, gender, or the presence of any other comorbidities 106 and has been confirmed elsewhere. 107 , 108 , 109 , 110 , 111 , 112 Thus, it is recommended that obese individuals, especially those with higher BMIs, be prioritised for Covid‐19 vaccination.

11. CARDIOVASCULAR DISEASES

Cardiovascular disorders (CVD) and hypertension are among the comorbidities with the highest risks of adverse outcomes from SARS‐CoV‐2 since most of these patients are of advanced age and have metabolic or other underlying diseases. 113 Therefore, this group should also be prioritised for Covid‐19 vaccination. Patients who have had a recent hospitalisation, primarily in the previous six months, those with NYHA III‐IV pulmonary hypertension, high grade peripheral arterial disease (PAD), morbid obesity, stage C heart failure, 1‐ or 2‐vessel obstructive coronary artery disease (CAD) with angina, and poorly controlled diabetes, are among the most critical group of cardiovascular patients for vaccination. 114 There have been no published studies on the type of Covid‐19 vaccine most suitable for these patients.

12. CHRONIC OBSTRUCTIVE PULMONARY DISEASE

A 2020 study published in the European Respiratory Journal reported that the ACE2 receptor, a molecule found on the surface of lung cells, is the point of attachment for SARS‐CoV‐2. 115 People with Chronic Obstructive Pulmonary Disease (COPD) and current smokers have increased airway expression of this enzyme. 115 Although the up‐regulation of this receptor is essential in protecting against acute lung injury, it predisposes individuals to increased risk of SARS‐CoV‐2 infection since this receptor is used as the gateway into the epithelial cells, explaining, at least in part, the increased risk of viral respiratory tract infection in active smokers and virus‐related exacerbations in those with COPD. 116 A 2021 systematic review of 59 studies found that having COPD significantly increased the odds of poor clinical outcomes, including the risk of hospitalisation, ICU admission, and mortality. 117 Nevertheless, a review of vaccine‐related deaths by the CDC, as of January 8, identified 55 deaths, with COPD being among the most commonly reported comorbidities, alongside hypertension, dementia, diabetes, and heart failure. 118 Of these deaths, 37 were reported among residents of long‐term care facilities. 118 However, the report concluded that the benefits of the Covid‐19 vaccination outweighed the potential risks in the older frail populations. 118 Therefore, the recommendation is that individuals with COPD be prioritised for Covid‐19 vaccination, regardless of age and frailty.

13. HUMAN IMMUNODEFICIENCY VIRUS

Patients with human immunodeficiency virus (HIV), similar to other comorbidities and immunocompromising conditions, are prone to severe Covid‐19. 119 However, the risk is higher in patients with advanced immunosuppression, defined as a CD4+ T cell count of <200/μL. 120 Moreover, if they become infected with SARS‐CoV‐2, negative impacts on their antiretroviral treatments would also be expected. 121 Unfortunately, few studies have investigated the safety and efficacy of the Covid‐19 vaccines in this population. However, one study has reported the mRNA Covid‐19 vaccines, such as the BNT162b2 vaccine, to be both immunogenic and safe in patients with HIV. 122 Nevertheless, there remains some level of mistrust in these patients about Covid‐19 vaccines, and therefore, discussing this issue with these individuals to address their hesitancy is essential. 123

14. CURRENT SMOKERS

It is well established that cigarette smoking causes structural changes in the respiratory tract and decreases immune responsiveness, both systemically and locally within the lungs. 124 Therefore, smoking is a significant risk factor for the proliferation of bacterial and viral infections. Previous studies evaluating the Middle East Respiratory Syndrome—Coronavirus (MERS‐Coronavirus) outbreak found higher mortality rates among current smokers than non‐smokers and those who had never smoked. 125 , 126 , 127 A 2020 systematic review of five studies from China during the first 2 months of the SARS‐CoV‐2 pandemic found a possible association between current smokers and Covid‐19. This included negative disease progression and adverse outcomes, such as increased ICU admission, the need for mechanical ventilation, and increased mortality when compared to non‐smokers. 128 Another recent study confirms these findings, reporting that smokers are overrepresented in fatalities, especially in populations where current smoking is high. 116 The authors suggest that higher rates of Covid‐19 would be expected in countries with a higher prevalence of smoking. A more recent study reported that cumulative exposure to cigarette smoke is an independent risk factor for increased hospital admission and death from Covid‐19. 129 Given the increased likelihood of contracting SARS‐CoV‐2 and the propensity for greater disease severity, it concerns that a study from the United Kingdom (UK) found that current smokers were more likely to be undecided or unwilling to be vaccinated against Covid‐19. 130 Jackson et al. suggest that due to the disproportionately high number of current smokers among socioeconomically disadvantaged groups, lower vaccination uptake in these clusters could exacerbate the already extant health inequalities. 130 As a result, targeted interventions may be necessary to prevent the compounding of health inequalities in these populations. 130 The recommendation for Covid‐19 vaccination among otherwise healthy smokers is that vaccination should occur, and in some cases vaccination is understandably prioritised in this group.

15. PREGNANCY AND BREASTFEEDING

Covid‐19 can manifest itself in its most severe form during pregnancy. Moreover, unfavourable pregnancy outcomes, such as premature labour, myocardial injuries, preeclampsia, perinatal death, and vertical transmission to the foetus, have been reported in pregnant women with Covid‐19. 131 , 132 Therefore, the need to be vaccinated in this population is extremely important. 133 Although little is known about the efficacy and safety profile of SARS‐CoV‐2 vaccines in pregnant or lactating mothers, due to their exclusion from vaccine trials, individuals intending to become pregnant and breastfeeding women are advised to receive a Covid‐19 vaccine, mainly if they are a member of a high‐risk group (e.g., healthcare personnel). However, only the mRNA vaccines are approved in this subsection of the population. 134 , 135

Different countries have different policies for vaccinating pregnant women against Covid‐19. For example, the American College of Obstetricians and Gynecologists (ACOG) recommends vaccination in pregnant and lactating women. 136 The United States Food and Drug Administration (FDA) and the Advisory Committee on Immunisation Practices (ACIP) have left the option to vaccinate pregnant and lactating women. 137 The South African Society of Obstetricians and Gynecologists has recommended vaccination in those pregnant and breastfeeding women at higher risk of exposure. 138 Canada, Ireland, Germany, and the United Kingdom have left the decision‐making to their physician to consider the individualised risk‐to‐benefit ratio. 139 , 140 , 141 Austria has announced the contraindication of SARS‐CoV‐2 vaccination for pregnant and nursing women, 142 while the Netherlands and Japan have also recommended vaccination in pregnancy. 143

Conversely, Israel has prioritised pregnant and lactating women for vaccination, 134 while the Society for Maternal‐Foetal Medicine strongly recommends vaccination in pregnant and lactating mothers. 144 The Italian scientific societies have announced that breastfeeding does not interfere with Covid‐19 vaccination, 145 and the UK's Medicines and Healthcare Products Regulatory Agency (MHRA) has allowed SARS‐CoV‐2 vaccination during breastfeeding. 146 However, the Academy of Breastfeeding states insufficient data about the vaccines' entrance into breast milk. 147 The American Society for Reproductive Medicine (ASRM) recommends that pregnant and lactating women, and those undergoing fertility treatment, be vaccinated against SARS‐CoV‐2. 148 The World Health Organisation (WHO) states that there is currently insufficient data on this issue, and further studies are needed to recommend vaccination in this population. 149 In general, it seems that Covid‐19 vaccines with Emergency Use Authorisation (EUA), including Pfizer/BioNTech, and Moderna vaccines, are prioritised in eligible pregnant mothers, such as women older than 35 years, healthcare personnel, those with multiple gestation, cancer, chronic hypertension, chronic kidney disease, chronic obstructive pulmonary disease, cardiac diseases, immunodeficiency, autoimmune diseases, obesity, sickle cell disease, smoking, and diabetes mellitus. 150

Vaccinated pregnant mothers can pass the IgG antibodies produced to their offspring, with one case study reporting vertical transmission. 151 It was also shown that the transplacental transfer of vaccine‐induced antibodies to the newborn is more likely if the mother is vaccinated in the third trimester. 152 Similar passive transfer of IgA or IgG antibodies has not yet been observed in breastfed infants. 153 , 154 The Academy of Breastfeeding Medicine has stated that breastfeeding should not be ceased for individuals vaccinated against Covid‐19. 155 If vaccinated, it seems that pregnant and lactating women achieve comparable antibody levels to other individuals of their age. 135 Therefore, after delivery, it is essential to follow up with vaccinated mothers and their newborns to investigate possible maternal and foetal complications related to the vaccine. 156 The Joint Committee on Vaccination and Immunisation (JCVI) stated that individuals trying to conceive could also be vaccinated if they meet the eligibility criteria. These women can get pregnant even before the second dose of the vaccine. 157

16. ELDERLY

Since the beginning of the Covid‐19 pandemic, the elderly have been the most affected by the social distancing measures applied to prevent virus transmission. Government measures have caused isolation and loneliness, which have led to physical inactivity and depression in these individuals. 158 Nevertheless, older individuals (i.e., >65 years old) have a high risk of Covid‐19‐related hospitalisation, ICU admission, and mortality due to comorbidities, poor nutrition, depressed immunity, and lower organ function. 159 , 160 Moreover, the elderly are at higher risk of getting severe Covid‐19 and are less likely to have an excellent response to vaccines. Moreover, some people reside in nursing homes, which puts them at increased risk of acquiring communicable infections, such as Covid‐19. 161 Therefore, prevention of SARS‐CoV‐2 infection seems to be the most desirable approach in these patients. There is substantial concern that these people would not achieve favourable protective immunity post‐vaccination, considering this population's relatively weak antibody response. 162 However, despite the lower efficacy of the Covid‐19 vaccine in the elderly, the vaccines are still effective against preventing mortality. Therefore, vaccination is strongly recommended for this age group, 163 and all currently approved Covid‐19 vaccines are safe and effective in the geriatric population.

17. CHILDREN

Until the evolution of the most recent SARS‐CoV‐2 variants, it was believed that children did not become afflicted with Covid‐19, or at least not in its most severe forms. Thus, paediatric vaccination did not seem to be necessary. 164 Nonetheless, reports of more severe forms of the disease and increases in the hospitalisation of children due to the alpha (B.1.1.7) and delta (B.1.617.2) variants prompt the discussion of including them in the Covid‐19 vaccination program and developing a suitable vaccine for this subsection of the population. 165 , 166 Moreover, vaccinating children can decrease infection transmission to others, meaning producing herd immunity in the community. They can also be a tool to prevent postinfectious conditions, such as Kawasaki‐like and toxic shock syndrome‐like diseases, commonly referred to as multisystem inflammatory syndrome in children (MIS‐C), 167 occurring 2–4 weeks after SARS‐CoV‐2 infection. 168 However, if proved to be safe and effective, there are still significant challenges to persuade hesitant parents to accept the vaccination of their children.

Preventive measures, such as face masks, hand hygiene, and social distancing, are less applicable in pediatrics than adults since adults adhere more strictly to the health protocols. 169 Therefore, the need for a vaccine for children seems to be increasingly important. Several factors should be considered in considering vaccine responses in children, including congenital or developmental disorders, nutritional status, and maturational changes. 103 However, immunocompromised children are also likely to show lower antibody response to Covid‐19 vaccines. 103 Presently, no children younger than 12 years old have been enrolled in Covid‐19 vaccination trials since it was believed that only older children were at risk of developing severe SARS‐CoV‐2 infection, and therefore, the vaccine trials could be extended to younger children at a later date. 170 The FDA has approved Covid‐19 vaccines for those older than 12 years, but the age limit can perhaps be lowered again after further research. 171

18. ALLERGIC DISEASES

In the period December 14–23, 2020, almost 1.9 million doses of Pfizer/BioNTech Covid‐19 vaccine were administered in the US, among which 21 cases of anaphylaxis were reported to the CDC, corresponding to an estimated rate of 11.1 cases per million. 172 Allergic reactions to vaccines and medications can be caused by two primary mechanisms: IgE‐mediated and IgE‐independent pathways. 173 , 174 These reactions are mainly triggered by non‐active vaccine ingredients, such as formaldehyde, thimerosal, egg protein, and gelatin, rather than the active vaccine ingredients. 175 Other ingredients commonly used in vaccines to improve their solubility in water are polyethylene glycol (PEG) and polysorbate. 175 PEG is used in the Pfizer/BioNTech and Moderna mRNA vaccines to enhance the stability of the mRNA‐containing lipid nanoparticles, 175 , 176 while polysorbate 80 is used in the Oxford/AstraZeneca and Johnson & Johnson adenoviral vector vaccines. 177 These substances can trigger IgE formation in the body, causing mast cell degranulation. Thus, skin testing for PEG and polysorbate could be an option to prevent such catastrophic events following vaccination against Covid‐19. 178

The IgE‐independent pathway is another proposed mechanism behind vaccine‐related allergic reactions. 174 In these cases, activating complement elements, including C3a, C4a, and C5a, would trigger these inflammatory responses. 174 Hence, measuring serum tryptase and complement system elements may help diagnose vaccine‐induced reactions following Covid‐19 vaccination. 177 This vividly highlights the importance of taking a detailed history of previous severe allergic reactions to an injectable medication, vaccine, or other allergens, mainly PEG‐ and polysorbate‐containing agents, to help prevent these types of adverse events. 175 Nevertheless, CDC recommends vaccinating individuals with a prior history of allergic reactions unless a positive skin test is present. In such cases, vaccination is contraindicated under the EUA. 177

19. CONCLUSION

This article has highlighted that several high‐risk population groups should be at the top of the priority list for receiving a vaccination. It was also demonstrated that significant research gaps in this topic require many more studies to determine whether these populations should receive Covid‐19 vaccines.

CONFLICT OF INTEREST

Terence T. Sio reports that he provides strategic and scientific recommendations as a member of the Advisory Board and speaker for Novocure, Inc. and also as a member of the Advisory Board to Galera Therapeutics, which are not in any way associated with the content or disease site as presented in this manuscript. All other authors have no relevant financial interests to be declared.

AUTHOR CONTRIBUTIONS

  • Zeinab Mohseni Afshar: Data collection and writing the manuscript. Arefeh Babazadeh: Data collection and writing the manuscript. Alireza Janbakhsh: Data collection and helped with manuscript writing. Feizollah Mansouri: Data collection and helped with manuscript writing. Terence T. Sio: Contributed substantial revisions to the manuscript's content. Mark J. M. Sullman: Contributed substantial revisions to the manuscript's content. Kristin Carson‐Chahhoud: Helped with manuscript writing and contributed substantial revisions to the manuscript's content. Rezvan Hosseinzadeh: Data collection and helped with manuscript writing. Mohammad Barary: Data collection, helped with manuscript writing, and contributed substantial revisions to the manuscript's content. Soheil Ebrahimpour: Design of the research study and supervision.

ACKNOWLEDGEMENT

The authors would like to thank the clinical research development center of Imam Reza Hospital, Kermanshah University of Medical Sciences, and Infectious Diseases and Tropical Medicine Research Center of Health Research Institute, Babol University of Medical Sciences, for their kind support.

Mohseni Afshar Z, Babazadeh A, Janbakhsh A, et al. Coronavirus disease 2019 (Covid‐19) vaccination recommendations in special populations and patients with existing comorbidities. Rev Med Virol. 2022; 32(3):e2309. 10.1002/rmv.2309

DATA AVAILABILITY STATEMENT

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  • 1. Sadeghi‐Haddad‐Zavareh M, Bayani M, Shokri M, et al. C‐reactive protein as a prognostic indicator in COVID‐19 patients. Interdiscip Perspect Infect Dis. 2021;2021:5557582. doi: 10.1155/2021/5557582 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Eftekhar SP, Kazemi S, Barary M, et al. Effect of hydroxychloroquine and azithromycin on QT interval prolongation and other cardiac arrhythmias in COVID‐19 confirmed patients. Cardiovasc Ther. 2021;2021:6683098. doi: 10.1155/2021/6683098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Javanian M, Bayani M, Shokri M, et al. Risk factors for mortality of 557 adult patients with COVID 19 in Babol, Northern Iran: a retrospective cohort study. Bratisl Lek Listy. 2021;122(1):34‐38. doi: 10.4149/BLL_2021_003 [DOI] [PubMed] [Google Scholar]
  • 4. Incalzi RA, Trevisan C, Del Signore S, et al. Are vaccines against COVID‐19 tailored to the most vulnerable people? Vaccine. 2021;39(17):2325‐2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Sonani B, Aslam F, Goyal A, Patel J, Bansal P. COVID‐19 vaccination in immunocompromised patients. Clin Rheumatol. 2021;40(2):797‐798. doi: 10.1007/s10067-020-05547-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Arnold J, Winthrop K, Emery P. COVID‐19 vaccination and antirheumatic therapy. Rheumatology. 2021;60(8):3496‐3502. doi: 10.1093/rheumatology/keab223 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Fischer L, Gerstel PF, Poncet A, et al. Pneumococcal polysaccharide vaccination in adults undergoing immunosuppressive treatment for inflammatory diseases—a longitudinal study. Arthritis Res Ther. 2015;17(1):151. doi: 10.1186/s13075-015-0663-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Fomin I, Caspi D, Levy V, et al. Vaccination against influenza in rheumatoid arthritis: the effect of disease modifying drugs, including TNFα blockers. Ann Rheum Dis. 2006;65(2):191‐194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Park JK, Lee YJ, Bitoun S, et al. Interaction between B‐cell activation factor and methotrexate impacts immunogenicity of seasonal influenza vaccination in patients with rheumatoid arthritis. Ann Rheum Dis. 2019;78(2):282‐284. [DOI] [PubMed] [Google Scholar]
  • 10. Park JK, Choi Y, Winthrop KL, Song YW, Lee EB. Optimal time between the last methotrexate administration and seasonal influenza vaccination in rheumatoid arthritis: post hoc analysis of a randomised clinical trial. Ann Rheum Dis. 2019;78(9):1283‐1284. [DOI] [PubMed] [Google Scholar]
  • 11. Hua C, Barnetche T, Combe B, Morel J. Effect of methotrexate, anti‐tumor necrosis factor α, and rituximab on the immune response to influenza and pneumococcal vaccines in patients with rheumatoid arthritis: a systematic review and meta‐analysis. Arthritis Care Res (Hoboken). 2014;66(7):1016‐1026. doi: 10.1002/acr.22246 [DOI] [PubMed] [Google Scholar]
  • 12. MOC C. The effect of disease‐modifying antirheumatic drugs on vaccine immunogenicity in adults. Cleve Clin J Med. 2020;87(11):695. [DOI] [PubMed] [Google Scholar]
  • 13. Mori S, Ueki Y, Hirakata N, et al. Impact of tocilizumab therapy on antibody response to influenza vaccine in patients with rheumatoid arthritis. Ann Rheum Dis. 2012;71(12):2006‐2010. doi: 10.1136/annrheumdis-2012-201950 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Soy M, Keser G, Atagunduz P, et al. A practical approach for vaccinations including COVID‐19 in autoimmune/autoinflammatory rheumatic diseases: a non‐systematic review. Clin Rheumatol. 2021;40(9):3533‐3545. doi: 10.1007/s10067-021-05700-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Curtis JR, Johnson SR, Anthony DD, et al. American College of rheumatology guidance for COVID‐19 vaccination in patients with rheumatic and musculoskeletal diseases: version 1. Arthritis Rheumatol. 2021;73(7):1093‐1107. doi: 10.1002/art.41734 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Winthrop KL, Silverfield J, Racewicz A, et al. The effect of tofacitinib on pneumococcal and influenza vaccine responses in rheumatoid arthritis. Ann Rheum Dis. 2016;75(4):687‐695. doi: 10.1136/annrheumdis-2014-207191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Arad U, Tzadok S, Amir S, et al. The cellular immune response to influenza vaccination is preserved in rheumatoid arthritis patients treated with rituximab. Vaccine. 2011;29(8):1643‐1648. [DOI] [PubMed] [Google Scholar]
  • 18. Ribeiro AC, Laurindo IM, Guedes LK, et al. Abatacept and reduced immune response to pandemic 2009 influenza A/H1N1 vaccination in patients with rheumatoid arthritis. Arthritis Care Res (Hoboken). 2013;65(3):476‐480. doi: 10.1002/acr.21838 [DOI] [PubMed] [Google Scholar]
  • 19. Terracina KA, Tan FK. Flare of rheumatoid arthritis after COVID‐19 vaccination. Lancet Rheumatol. 2021;3(7):e469‐e470. doi: 10.1016/S2665-9913(21)00108-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. COVID A . Vaccine Clinical Guidance Task Force: COVID‐19 Vaccine Clinical Guidance Summary for Patients with Rheumatic and Musculoskeletal Diseases; 2021. [Google Scholar]
  • 21. Velikova T, Georgiev T. SARS‐CoV‐2 vaccines and autoimmune diseases amidst the COVID‐19 crisis. Rheumatol Int. 2021;41(3):509‐518. doi: 10.1007/s00296-021-04792-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Ribas A, Sengupta R, Locke T, et al. Priority COVID‐19 vaccination for patients with cancer while vaccine supply is limited. Cancer Discov. 2021;11(2):233‐236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Ting FI, Uy CD, Bebero KG, et al. COVID‐19 vaccine and patients with cancer. The Philippine Society of Medical Oncology (PSMO) Position Statement. 2021. https://psmo.org.ph/wp‐content/uploads/2021/04/PSMO‐Position‐Statement‐on‐COVID‐19‐vaccine‐and‐cancer‐patients‐4.pdf [Google Scholar]
  • 24. Hwang JK, Zhang T, Wang AZ, Li Z. COVID‐19 vaccines for patients with cancer: benefits likely outweigh risks. J Hematol Oncol. 2021;14(1):38. doi: 10.1186/s13045-021-01046-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Desai A, Gainor JF, Hegde A, et al. COVID‐19 vaccine guidance for patients with cancer participating in oncology clinical trials. Nat Rev Clin Oncol. 2021;18(5):313‐319. doi: 10.1038/s41571-021-00487-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Avanzato VA, Matson MJ, Seifert SN, et al. Case study: prolonged infectious SARS‐CoV‐2 shedding from an asymptomatic immunocompromised individual with cancer. Cell. 2020;183(7):1901‐1912. doi: 10.1016/j.cell.2020.10.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Waissengrin B, Agbarya A, Safadi E, Padova H, Wolf I. Short‐term safety of the BNT162b2 mRNA COVID‐19 vaccine in patients with cancer treated with immune checkpoint inhibitors. Lancet Oncol. 2021;22(5):581‐583. doi: 10.1016/S1470-2045(21)00155-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Keam B, Kang CK, Jun KI, et al. Immunogenicity of influenza vaccination in patients with cancer receiving immune checkpoint inhibitors. Clin Infect Dis. 2020;71(2):422‐425. doi: 10.1093/cid/ciz1092 [DOI] [PubMed] [Google Scholar]
  • 29. Mohindra P, Buckey CR, Chen S, Sio TT, Rong Y. Radiation therapy considerations during the COVID‐19 Pandemic: literature review and expert opinions. J Appl Clin Med Phys. 2020;21(5):6‐12. doi: 10.1002/acm2.12898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Cornberg M, Buti M, Eberhardt CS, Grossi PA, Shouval D. EASL position paper on the use of COVID‐19 vaccines in patients with chronic liver diseases, hepatobiliary cancer and liver transplant recipients. J Hepatol. 2021;74(4):944‐951. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Benotmane I, Gautier‐Vargas G, Cognard N, et al. Weak anti‐SARS‐CoV‐2 antibody response after the first injection of an mRNA COVID‐19 vaccine in kidney transplant recipients. Kidney Int. 2021;99(6):1487‐1489. doi: 10.1016/j.kint.2021.03.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Aslam S, Goldstein DR, Vos R, et al. COVID‐19 vaccination in our transplant recipients: the time is now. J Heart Lung Transplant. 2021;40(3):169‐171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Boyarsky BJ, Werbel WA, Avery RK, et al. Antibody response to 2‐dose SARS‐CoV‐2 mRNA vaccine series in solid organ transplant recipients. J Am Med Assoc. 2021;325(21):2204‐2206. doi: 10.1001/jama.2021.7489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Boyarsky BJ, Werbel WA, Avery RK, et al. Immunogenicity of a single dose of SARS‐CoV‐2 messenger RNA vaccine in solid organ transplant recipients. J Am Med Assoc. 2021;325(17):1784‐1786. doi: 10.1001/jama.2021.4385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Danziger‐Isakov L, Kumar D, Practice AICo. Vaccination of solid organ transplant candidates and recipients: guidelines from the American society of transplantation infectious diseases community of practice. Clin Transpl. 2019;33(9):e13563. doi: 10.1111/ctr.13563 [DOI] [PubMed] [Google Scholar]
  • 36. Heldman MR, Limaye AP. SARS‐CoV‐2 vaccines in kidney transplant recipients: will they be safe and effective and how will we know? J Am Soc Nephrol. 2021;32(5):1021‐1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Hall VG, Ferreira VH, Ku T, et al. Randomized trial of a third dose of mRNA‐1273 vaccine in transplant recipients. N Engl J Med. 2021;385(13):1244‐1246. doi: 10.1056/NEJMc2111462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Mulley WR, Dendle C, Ling JE, Knight SR. Does vaccination in solid‐organ transplant recipients result in adverse immunologic sequelae? A systematic review and meta‐analysis. J Heart Lung Transplant. 2018;37(7):844‐852. [DOI] [PubMed] [Google Scholar]
  • 39. Fix OK, Blumberg EA, Chang KM, et al. AASLD expert panel consensus statement: vaccines to prevent COVID‐19 infection in patients with liver disease. Hepatology. 2021;74:1049‐1064. doi: 10.1002/hep.31751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Iavarone M, D'Ambrosio R, Soria A, et al. High rates of 30‐day mortality in patients with cirrhosis and COVID‐19. J Hepatol. 2020;73(5):1063‐1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Dong Y, Dai TWY. ESMO Statements for Vaccination against COVID‐19 in Patients with Cancer; 2021. [Google Scholar]
  • 42. Hsu CM, Weiner DE, Aweh G, et al. COVID‐19 among US dialysis patients: risk factors and outcomes from a national dialysis provider. Am J Kidney Dis. 2021;77(5):748‐756.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Li SY, Tang YS, Chan YJ, Tarng DC. Impact of the COVID‐19 pandemic on the management of patients with end‐stage renal disease. J Chin Med Assoc. 2020;83(7):628‐633. doi: 10.1097/JCMA.0000000000000356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Popa C, Ipate C, Hogas S.  Covic A COVID‐19 vaccination in renal transplant patient and dialysis patients. Proc Rom Acad Ser B, Chem Life Sci Geosci. 2021:122‐5. [Google Scholar]
  • 45. Srivatana V, Wilkie C, Perl J, Watnick S. Vaccine and the need to be heard: considerations for COVID‐19 immunization in ESKD. Kidney. 2021;2(6):1048‐1050. doi: 10.34067/kid.0001932021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Stevens CE, Alter HJ, Taylor PE, et al. Hepatitis B vaccine in patients receiving hemodialysis. Immunogenicity and efficacy. N Engl J Med. 1984;311(8):496‐501. doi: 10.1056/NEJM198408233110803 [DOI] [PubMed] [Google Scholar]
  • 47. Sahin U, Muik A, Vogler I, et al. BNT162b2 vaccine Induces neutralizing antibodies and poly‐specific T cells in humans. Nature. medRxiv; 2021;595(7868):572‐577. doi: 10.1038/s41586-021-03653-6 [DOI] [PubMed] [Google Scholar]
  • 48. Anand S, Montez‐Rath ME, Han J, et al. Antibody response to COVID‐19 vaccination in patients receiving dialysis. J Am Soc Nephrol. 2021;32(10):2435‐2438. doi: 10.1681/asn.2021050611 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Grupper A, Sharon N, Finn T, et al. Humoral response to the Pfizer BNT162b2 vaccine in patients undergoing maintenance hemodialysis. Clin J Am Soc Nephrol. 2021;16:1037‐1042. doi: 10.2215/CJN.03500321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Krueger KM, Ison MG, Ghossein C. Practical guide to vaccination in all stages of CKD, including patients treated by dialysis or kidney transplantation. Am J Kidney Dis. 2020;75(3):417‐425. [DOI] [PubMed] [Google Scholar]
  • 51. Windpessl M, Bruchfeld A, Anders HJ, et al. COVID‐19 vaccines and kidney disease. Nat Rev Nephrol. 2021;17(5):291‐293. doi: 10.1038/s41581-021-00406-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Kronbichler A, Anders H‐J, Fernandez‐Juárez GM, et al. Recommendations for the use of COVID‐19 vaccines in patients with immune‐mediated kidney diseases. In Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association‐European Renal Association; 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Baker D, Roberts CAK, Pryce G, et al. COVID‐19 vaccine‐readiness for anti‐CD20‐depleting therapy in autoimmune diseases. Clin Exp Immunol. 2020;202(2):149‐161. doi: 10.1111/cei.13495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. De Serres G, Billard M‐N, Gariepy M‐C, et al. Nephrotic syndrome following four‐component meningococcal B vaccination: epidemiologic investigation of a surveillance signal. Vaccine. 2019;37(35):4996‐5002. doi: 10.1016/j.vaccine.2019.07.017 [DOI] [PubMed] [Google Scholar]
  • 55. Aydın MF, Yıldız A, Oruç A, et al. Relapse of primary membranous nephropathy after inactivated SARS‐CoV‐2 virus vaccination. Kidney Int. 2021;100(2):464‐465. doi: 10.1016/j.kint.2021.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Hartung HP, Aktas O. COVID‐19 and management of neuroimmunological disorders. Nat Rev Neurol. 2020;16(7):347‐348. doi: 10.1038/s41582-020-0368-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Sellner J, Jenkins TM, von Oertzen TJ, et al. Primary prevention of COVID‐19: advocacy for vaccination from a neurological perspective. Eur J Neurol. 2021;28(10):3226‐3229. doi: 10.1111/ene.14713 [DOI] [PubMed] [Google Scholar]
  • 58. Achiron A, Dolev M, Menascu S, et al. COVID‐19 vaccination in patients with multiple sclerosis: what we have learnt by February 2021. Mult Scler J. 2021;27(6):864‐870. doi: 10.1177/13524585211003476 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Advisory Committee on Immunization Practices (ACIP). Interim clinical considerations for use of COVID‐19 vaccines currently approved or authorized in the united states. COVID‐19 Vaccination; Vaccines & Immunizations. Centers for Disease Control and Prevention; 2021. [Google Scholar]
  • 60. Ciotti JR, Valtcheva MV, Cross AH. Effects of MS disease‐modifying therapies on responses to vaccinations: a review. Mult Scler Relat Disord. 2020;45:102439. doi: 10.1016/j.msard.2020.102439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Kelly H, Sokola B, Abboud H. Safety and efficacy of COVID‐19 vaccines in multiple sclerosis patients. J Neuroimmunol. 2021;356:577599. doi: 10.1016/j.jneuroim.2021.577599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Morales FS, Koralnik IJ, Gautam S, Samaan S, Sloane JA. Risk factors for lymphopenia in patients with relapsing‐remitting multiple sclerosis treated with dimethyl fumarate. J Neurol. 2020;267(1):125‐131. doi: 10.1007/s00415-019-09557-w [DOI] [PubMed] [Google Scholar]
  • 63. Safavi F, Nourbakhsh B, Azimi AR. B‐cell depleting therapies may affect susceptibility to acute respiratory illness among patients with multiple sclerosis during the early COVID‐19 epidemic in Iran. Mult Scler Relat Disord. 2020;43:102195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. McCarthy CL, Tuohy O, Compston DA, et al. Immune competence after alemtuzumab treatment of multiple sclerosis. Neurology. 2013;81(10):872‐876. doi: 10.1212/WNL.0b013e3182a35215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Wolf A, Alvarez E. COVID‐19 vaccination in patients with multiple sclerosis on disease‐modifying therapy. Neurol Clin Pract. 2021;11(4):358‐361. doi: 10.1212/cpj.0000000000001088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Classen JB. COVID‐19 RNA based vaccines and the risk of prion disease. Microbiol Infect Dis. 2021;5(1):1‐3. [Google Scholar]
  • 67. Lu L, Xiong W, Mu J, et al. The potential neurological effect of the COVID‐19 vaccines: a review. Acta Neurol Scand. 2021;144(1):3‐12. doi: 10.1111/ane.13417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tian M, Yang J, Li L, Li J, Lei W, Shu X. Vac‐associated neurological adverse events: a case report and literature review. Curr Pharm Des. 2019;25(43):4570‐4578. doi: 10.2174/1381612825666191119095132 [DOI] [PubMed] [Google Scholar]
  • 69. Marchetti RL, Gallucci‐Neto J, Kurcgant D, et al. Immunization stress‐related responses presenting as psychogenic non‐epileptic seizures following HPV vaccination in Rio Branco, Brazil. Vaccine. 2020;38(43):6714‐6720. [DOI] [PubMed] [Google Scholar]
  • 70. Tahir N, Koorapati G, Prasad S, et al. SARS‐CoV‐2 vaccination‐induced transverse myelitis. Cureus. 2021;13(7):e16624. doi: 10.7759/cureus.16624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Roman GC, Gracia F, Torres A, et al. Acute transverse myelitis (ATM): clinical review of 43 patients with COVID‐19‐associated ATM and 3 post‐vaccination ATM serious adverse events with the ChAdOx1 nCoV‐19 vaccine (AZD1222). Front Immunol. 2021;12:653786. doi: 10.3389/fimmu.2021.653786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Khan E, Shrestha AK, Colantonio MA, Liberio RN, Sriwastava S. Acute transverse myelitis following SARS‐CoV‐2 vaccination: a case report and review of literature. J Neurol. 2021. doi: 10.1007/s00415-021-10785-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Trimboli M, Zoleo P, Arabia G, Gambardella A. Guillain‐Barré syndrome following BNT162b2 COVID‐19 vaccine. Neurol Sci. 2021;42(11):4401‐4402. doi: 10.1007/s10072-021-05523-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Waheed S, Bayas A, Hindi F, Rizvi Z, Espinosa PS. Neurological complications of COVID‐19: Guillain‐Barre syndrome following Pfizer COVID‐19 vaccine. Cureus. 2021;13(2):e13426. doi: 10.7759/cureus.13426 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Sato K, Mano T, Niimi Y, et al. Facial nerve palsy following the administration of COVID‐19 mRNA vaccines: analysis of a self‐reporting database. Int J Infect Dis. 2021;111:310‐312. doi: 10.1016/j.ijid.2021.08.071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Pritchard J, Mukherjee R, Hughes RAC. Risk of relapse of Guillain‐Barré syndrome or chronic inflammatory demyelinating polyradiculoneuropathy following immunisation. J Neurol Neurosurg Psychiatry. 2002;73(3):348‐349. doi: 10.1136/jnnp.73.3.348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Folegatti PM, Bittaye M, Flaxman A, et al. Safety and immunogenicity of a candidate Middle East respiratory syndrome coronavirus viral‐vectored vaccine: a dose‐escalation, open‐label, non‐randomised, uncontrolled, phase 1 trial. Lancet Infect Dis. 2020;20(7):816‐826. doi: 10.1016/S1473-3099(20)30160-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Reilev M, Kristensen KB, Pottegard A, et al. Characteristics and predictors of hospitalization and death in the first 11 122 cases with a positive RT‐PCR test for SARS‐CoV‐2 in Denmark: a nationwide cohort. Int J Epidemiol. 2020;49(5):1468‐1481. doi: 10.1093/ije/dyaa140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Momen NC, Plana‐Ripoll O, Agerbo E, et al. Association between mental disorders and subsequent medical conditions. N. Engl J Med. 2020;382(18):1721‐1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. De Hert M, Mazereel V, Detraux J, Van Assche K. Prioritizing COVID‐19 vaccination for people with severe mental illness. World Psychiatr. 2021;20(1):54‐55. doi: 10.1002/wps.20826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Bauer ME, Teixeira AL. Inflammation in psychiatric disorders: what comes first? Ann N. Y Acad Sci. 2019;1437(1):57‐67. [DOI] [PubMed] [Google Scholar]
  • 82. De Picker LJ, Dias MC, Benros ME, et al. Severe mental illness and European COVID‐19 vaccination strategies. Lancet Psychiatry. 2021;8(5):356‐359. doi: 10.1016/S2215-0366(21)00046-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Yang Y, Li W, Zhang Q, et al. Should people with severe mental illness be prioritized for the COVID‐19 vaccination? Int J Biol Sci. 2021;17(6):1443‐1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Kiecolt‐Glaser JK, Glaser R, Gravenstein S, Malarkey WB, Sheridan J. Chronic stress alters the immune response to influenza virus vaccine in older adults. Proc Natl Acad Sci U. S. A. 1996;93(7):3043‐3047. doi: 10.1073/pnas.93.7.3043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Afsar B, Elsurer R, Eyileten T, Yilmaz MI, Caglar K. Antibody response following hepatitis B vaccination in dialysis patients: does depression and life quality matter? Vaccine. 2009;27(42):5865‐5869. [DOI] [PubMed] [Google Scholar]
  • 86. Ponsford MJ, Steven R, Bramhall K, et al. Clinical and laboratory characteristics of clozapine‐treated patients with schizophrenia referred to a national immunodeficiency clinic reveals a B‐cell signature resembling common variable immunodeficiency (CVID). J Clin Pathol. 2020;73(9):587‐592. doi: 10.1136/jclinpath-2019-206235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Irwin MR, Levin MJ, Laudenslager ML, et al. Varicella zoster virus‐specific immune responses to a herpes zoster vaccine in elderly recipients with major depression and the impact of antidepressant medications. Clin Infect Dis. 2013;56(8):1085‐1093. doi: 10.1093/cid/cis1208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Pal R, Bhadada SK, Misra A. COVID‐19 vaccination in patients with diabetes mellitus: current concepts, uncertainties and challenges. Diabetes Metab Syndr. 2021;15(2):505‐508. doi: 10.1016/j.dsx.2021.02.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Gregory JM, Slaughter JC, Duffus SH, et al. COVID‐19 severity is tripled in the diabetes community: a prospective analysis of the pandemic’s impact in type 1 and type 2 diabetes. Diabetes Care. 2021;44(2):526‐532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Dooling K, McClung N, Chamberland M, et al. The advisory committee on immunization Practices' interim recommendation for allocating initial supplies of COVID‐19 vaccine—United States, 2020. MMWR Morb Mortal Wkly Rep. 2020;69(49):1857‐1859. doi: 10.15585/mmwr.mm6949e1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Fernandes A, Chaudhari S, Jamil N, Gopalakrishnan G. COVID‐19 vaccine. Endocr Pract. 2021;27(2):170‐172. doi: 10.1016/j.eprac.2021.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Lampasona V, Secchi M, Scavini M, et al. Antibody response to multiple antigens of SARS‐CoV‐2 in patients with diabetes: an observational cohort study. Diabetologia. 2020;63(12):2548‐2558. doi: 10.1007/s00125-020-05284-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Abu‐Rumaileh MA, Gharaibeh AM, Gharaibeh NE. COVID‐19 vaccine and hyperosmolar hyperglycemic state. Cureus. 2021. doi: 10.7759/cureus.14125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Nave H, Beutel G, Kielstein JT. Obesity‐related immunodeficiency in patients with pandemic influenza H1N1. Lancet Infect Dis. 2011;11(1):14‐15. doi: 10.1016/S1473-3099(10)70304-2 [DOI] [PubMed] [Google Scholar]
  • 95. Diaz E, Rodriguez A, Martin‐Loeches I, et al. Impact of obesity in patients infected with 2009 influenza A(H1N1). Chest. 2011;139(2):382‐386. doi: 10.1378/chest.10-1160 [DOI] [PubMed] [Google Scholar]
  • 96. Louie JK, Acosta M, Samuel MC, et al. A novel risk factor for a novel virus: obesity and 2009 pandemic influenza A (H1N1). Clin Infect Dis. 2011;52(3):301‐312. doi: 10.1093/cid/ciq152 [DOI] [PubMed] [Google Scholar]
  • 97. Green WD, Beck MA. Obesity impairs the adaptive immune response to influenza virus. Ann Am Thorac Soc. 2017;14(Supplement_5):S406‐S409. doi: 10.1513/AnnalsATS.201706-447AW [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Al Heialy S, Hachim MY, Senok A, et al. Regulation of angiotensin‐ converting enzyme 2 in obesity: implications for COVID‐19. Front Physiol. 2020;11:555039. doi: 10.3389/fphys.2020.555039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Nogueira‐de‐Almeida CA, de Mello ED. Different criteria for the definition of insulin resistance and its relation with dyslipidemia in overweight and obese children and adolescents. Pediatr Gastroenterol Hepatol Nutr. 2018;21(1):59‐67. doi: 10.5223/pghn.2018.21.1.59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Sattar N, McInnes IB, McMurray JJV. Obesity is a risk factor for severe COVID‐19 infection: multiple potential mechanisms. Circulation. 2020;142(1):4‐6. doi: 10.1161/CIRCULATIONAHA.120.047659 [DOI] [PubMed] [Google Scholar]
  • 101. Wu CT, Lidsky PV, Xiao Y, et al. SARS‐CoV‐2 infects human pancreatic β cells and elicits β cell impairment. Cell Metab. 2021;33(8):1565‐1576.e5. doi: 10.1016/j.cmet.2021.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Korakas E, Ikonomidis I, Kousathana F, et al. Obesity and COVID‐19: immune and metabolic derangement as a possible link to adverse clinical outcomes. Am J Physiol Endocrinol Metab. 2020;319(1):E105‐E109. doi: 10.1152/ajpendo.00198.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Shekerdemian LS, Mahmood NR, Wolfe KK, et al. Characteristics and outcomes of children with coronavirus disease 2019 (COVID‐19) infection admitted to US and Canadian pediatric intensive care units. JAMA Pediatr. 2020;174(9):868‐873. doi: 10.1001/jamapediatrics.2020.1948 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Zachariah P, Johnson CL, Halabi KC, et al. Epidemiology, clinical features, and disease severity in patients with coronavirus disease 2019 (COVID‐19) in a children's hospital in New York City, New York. JAMA Pediatr. 2020;174(10):e202430. doi: 10.1001/jamapediatrics.2020.2430 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Wu J, Li W, Shi X, et al. Early antiviral treatment contributes to alleviate the severity and improve the prognosis of patients with novel coronavirus disease (COVID‐19). J Intern Med. 2020;288(1):128‐138. doi: 10.1111/joim.13063 [DOI] [PubMed] [Google Scholar]
  • 106. Simonnet A, Chetboun M, Poissy J, et al. High prevalence of obesity in severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) requiring invasive mechanical ventilation. Obesity (Silver Spring). 2020;28(7):1195‐1199. doi: 10.1002/oby.22831 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Bhatraju PK, Ghassemieh BJ, Nichols M, et al. Covid‐19 in critically ill patients in the Seattle region—case series. N Engl J Med. 2020;382(21):2012‐2022. doi: 10.1056/NEJMoa2004500 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Busetto L, Bettini S, Fabris R, et al. Obesity and COVID‐19: an Italian snapshot. Obesity (Silver Spring). 2020;28(9):1600‐1605. doi: 10.1002/oby.22918 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Hajifathalian K, Kumar S, Newberry C, et al. Obesity is associated with worse outcomes in COVID‐19: analysis of early data from New York City. Obesity (Silver Spring). 2020;28(9):1606‐1612. doi: 10.1002/oby.22923 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Popkin BM, Du S, Green WD, et al. Individuals with obesity and COVID‐19: a global perspective on the epidemiology and biological relationships. Obes Rev. 2020;21(11):e13128. doi: 10.1111/obr.13128 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Nogueira‐de‐Almeida CA, Del Ciampo LA, Ferraz IS, et al. COVID‐19 and obesity in childhood and adolescence: a clinical review. J Pediatr (Rio J). 2020;96(5):546‐558. doi: 10.1016/j.jped.2020.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Zhou Y, Chi J, Lv W, Wang Y. Obesity and diabetes as high‐risk factors for severe coronavirus disease 2019 (Covid‐19). Diabetes Metab Res Rev. 2021;37(2):e3377. doi: 10.1002/dmrr.3377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Xu PP, Tian RH, Luo S, et al. Risk factors for adverse clinical outcomes with COVID‐19 in China: a multicenter, retrospective, observational study. Theranostics. 2020;10(14):6372‐6383. doi: 10.7150/thno.46833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Driggin E, Maddox TM, Ferdinand KC, et al. ACC health policy statement on cardiovascular disease considerations for COVID‐19 vaccine prioritization: a report of the American College of cardiology solution set oversight committee. J Am Coll Cardiol. 2021;77(15):1938‐1948. doi: 10.1016/j.jacc.2021.02.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Leung JM, Yang CX, Tam A, et al. ACE‐2 expression in the small airway epithelia of smokers and COPD patients: implications for COVID‐19. Eur Respir J. 2020;55(5):2000688. doi: 10.1183/13993003.00688-2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Brake SJ, Barnsley K, Lu W, et al. Smoking upregulates angiotensin‐converting enzyme‐2 receptor: a potential adhesion site for novel coronavirus SARS‐CoV‐2 (Covid‐19). J Clin Med. 2020;9(3). doi: 10.3390/jcm9030841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Gerayeli FV, Milne S, Cheung C, et al. COPD and the risk of poor outcomes in COVID‐19: a systematic review and meta‐analysis. EClinicalMedicine. 2021;33:100789. doi: 10.1016/j.eclinm.2021.100789 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Lv G, Yuan J, Xiong X, Li M. Mortality rate and characteristics of deaths following COVID‐19 vaccination. Front Med (Lausanne). 2021;8:670370. doi: 10.3389/fmed.2021.670370 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Jiang H, Zhou Y, Tang W. Maintaining HIV care during the COVID‐19 pandemic. Lancet HIV. 2020;7(5):e308‐e309. doi: 10.1016/S2352-3018(20)30105-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Hoffmann C, Casado JL, Härter G, et al. Immune deficiency is a risk factor for severe COVID‐19 in people living with HIV. HIV Med. 2021;22(5):372‐378. [DOI] [PubMed] [Google Scholar]
  • 121. Sutton MY, Jones RL, Wolitski RJ, Cleveland JC, Dean HD, Fenton KA. A review of the Centers for Disease Control and Prevention's response to the HIV/AIDS crisis among Blacks in the United States, 1981–2009. Am J Publ Health. 2009;99(S2):S351‐S359.doi: 10.2105/ajph.2008.157958 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Levy I, Wieder‐Finesod A, Litchevsky V, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID‐19 vaccine in people living with HIV‐1. Clin Microbiol Infect. 2021;S1198-743X(21)00423-7. doi: 10.1016/j.cmi.2021.07.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Bogart LM, Ojikutu BO, Tyagi K, et al. COVID‐19 related medical mistrust, health impacts, and potential vaccine hesitancy among Black Americans living with HIV. J Acquir Immune Defic Syndr (1999). 2021;86(2):200, 207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Arcavi L, Benowitz NL. Cigarette smoking and infection. Arch Intern Med. 2004;164(20):2206‐2216. doi: 10.1001/archinte.164.20.2206 [DOI] [PubMed] [Google Scholar]
  • 125. Park JE, Jung S, Kim A, Park JE. MERS transmission and risk factors: a systematic review. BMC Publ Health. 2018;18(1):574. doi: 10.1186/s12889-018-5484-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Sherbini N, Iskandrani A, Kharaba A, et al. Middle East respiratory syndrome coronavirus in Al‐Madinah City, Saudi Arabia: demographic, clinical and survival data. J Epidemiol Glob Health. 2017;7(1):29‐36. doi: 10.1016/j.jegh.2016.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Nam HS, Park JW, Ki M, et al. High fatality rates and associated factors in two hospital outbreaks of MERS in Daejeon, the Republic of Korea. Int J Infect Dis. 2017;58:37‐42. doi: 10.1016/j.ijid.2017.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Vardavas CI, Nikitara K. COVID‐19 and smoking: a systematic review of the evidence. Tob Induc Dis. 2020;18:20. doi: 10.18332/tid/119324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Lowe KE, Zein J, Hatipoglu U, Attaway A. Association of smoking and cumulative pack‐year exposure with COVID‐19 outcomes in the Cleveland clinic COVID‐19 registry. JAMA Intern Med. 2021;181(5):709‐711. doi: 10.1001/jamainternmed.2020.8360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Jackson SE, Paul E, Brown J, Steptoe A, Fancourt D. Negative vaccine attitudes and intentions to vaccinate against Covid‐19 in relation to smoking status: a population survey of UK adults. Nicotine Tob Res. 2021;23:1623‐1628. doi: 10.1093/ntr/ntab039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Di Mascio D, Khalil A, Saccone G, et al. Outcome of coronavirus spectrum infections (SARS, MERS, COVID‐19) during pregnancy: a systematic review and meta‐analysis. Am J Obstet Gynecol MFM. 2020;2(2):100107. doi: 10.1016/j.ajogmf.2020.100107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Kotlyar AM, Grechukhina O, Chen A, et al. Vertical transmission of coronavirus disease 2019: a systematic review and meta‐analysis. Am J Obstet Gynecol. 2021;224(1):35‐53 e3. doi: 10.1016/j.ajogmf.2020.100107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Maykin MM, Heuser C, Feltovich H. With the society for maternal‐fetal medicine health policy Advocacy C. Pregnant people deserve the protection offered by SARS‐CoV‐2 vaccines. Vaccine. 2021;39(2):171‐172. doi: 10.1016/j.vaccine.2020.12.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Chervenak FA, McCullough LB, Bornstein E, et al. Professionally responsible coronavirus disease 2019 vaccination counseling of obstetrical and gynecologic patients. Am J Obstet Gynecol. 2021;224(5):470‐478. doi: 10.1016/j.ajog.2021.01.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Hughes BL, Swamy G, Eckert LON, Gyamq‐Bannerman C, Turrentine M. Vaccinating Pregnant and Lactating Patients against COVID‐19. The American College of Obstetricians and Gynecologists; 2021. [Google Scholar]
  • 136. Obstetricians ACo, Gynecologists . Vaccinating Pregnant and Lactating Patients against COVID‐19. Acog org Practice Advisory; 2020. [Google Scholar]
  • 137. Adhikari EH, Spong CY. COVID‐19 vaccination in pregnant and lactating women. JAMA. 2021;325:(11):1039. doi: 10.1001/jama.2021.1658 [DOI] [PubMed] [Google Scholar]
  • 138. Moodley J, Ngene N, Khaliq O, Hunter M. An imperative to offer pregnant and lactating women access to the COVID‐19 vaccination roll‐out programme. SAMJ S Afr Med J. 2021;111(4). [PubMed] [Google Scholar]
  • 139. Elwood C, Boucoiran I, VanSchalkwyk J, Money D, Yudin M, Poliquin V, SOGC committee opinion–COVID‐19 in pregnancy. J Obstet Gynaecol Can. 2020. doi: 10.1016/j.jogc.2020.03.012 [DOI] [Google Scholar]
  • 140. Poon LC, Yang H, Dumont S, et al. ISUOG Interim Guidance on coronavirus disease 2019 (COVID‐19) during pregnancy and puerperium: information for healthcare professionals–an update. Ultrasound Obstet Gynecol. 2020;55(6):848‐862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Royal College of Obstetricians and Gynaecologists . Updated advice on COVID‐19 vaccination in pregnancy and women who are breastfeeding. Royal College of Obstetricians and Gynaecologists Press; 2020. [Google Scholar]
  • 142. Board NV . COVID‐19 Vaccinations: Recommendations for Use by the National Vaccination Board. Federal Ministry of Republic of Austria in Social Affairs, Health, Care and Consumer Protection; 2021. https://www.sozialministerium.at/dam/jcr:36d52a19‐39fd‐4e20‐a94d‐8df4ffb50a0c/COVID‐19‐Impfungen_Anwendungsempfehlung_des_Nationalen_Impfgremiums_Version_4.2_(Stand_05.07.2021).pdf [Google Scholar]
  • 143. Machida M, Nakamura I, Kojima T, et al. Acceptance of a COVID‐19 vaccine in Japan during the COVID‐19 pandemic. Vaccines (Basel). 2021;9(3):210. doi: 10.3390/vaccines9030210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Rasmussen SA, Kelley CF, Horton JP, Jamieson DJ. Coronavirus disease 2019 (COVID‐19) vaccines and pregnancy: what obstetricians need to know. Obstet Gynecol. 2021;137(3):408‐414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Davanzo R, Agosti M, Cetin I, et al. Breastfeeding and COVID‐19 vaccination: position statement of the Italian scientific societies. Ital J Pediatr. 2021;47(1):45. doi: 10.1186/s13052-021-00998-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Rimmer A. Covid‐19: Breastfeeding Women Can Have Vaccine after Guidance Turnaround. British Medical Journal Publishing Group; 2021. [DOI] [PubMed] [Google Scholar]
  • 147. Blumberg D, Sridhar A, Lakshminrusimha S, Higgins RD, Saade G. COVID‐19 vaccine considerations during pregnancy and lactation. Am J Perinatol. 2021;38(6):523‐528. doi: 10.1055/s-0041-1726390 [DOI] [PubMed] [Google Scholar]
  • 148. Turocy JM, Robles A, Hercz D, D'Alton M, Forman EJ, Williams Z. The emotional impact of the ASRM guidelines on fertility patients during the COVID‐19 pandemic. Fertil Steril. 2020;114(3):e63. doi: 10.1016/j.fertnstert.2020.08.194 [DOI] [Google Scholar]
  • 149. Strategic Advisory Group of Experts (SAGE) . The Moderna COVID‐19 (mRNA‐1273) Vaccine: What You Need to Know. World Health Organization; 2021. https://www.who.int/news‐room/feature‐stories/detail/the‐moderna‐covid‐19‐mrna‐1273‐vaccine‐what‐you‐need‐to‐know [Google Scholar]
  • 150. Stafford IA, Parchem JG, Sibai BM. The coronavirus disease 2019 vaccine in pregnancy: risks, benefits, and recommendations. Am J Obstet Gynecol. 2021;224(5):484‐495. doi: 10.1016/j.ajog.2021.01.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Paul G, Chad R. Newborn antibodies to SARS‐CoV‐2 detected in cord blood after maternal vaccination‐a case report. BMC pediatrics. 2021;21(1). doi: 10.1186/s12887-021-02618-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Mithal LB, Otero S, Shanes ED, Goldstein JA, Miller ES. Cord blood antibodies following maternal coronavirus disease 2019 vaccination during pregnancy. Am J Obstet Gynecol. 2021;225:192‐194. doi: 10.1016/j.ajog.2021.03.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Male V. Are COVID‐19 vaccines safe in pregnancy? Nat Rev Immunol. 2021;21(4):200‐201. doi: 10.1038/s41577-021-00525-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Golan Y, Prahl M, Cassidy A, et al. Evaluation of messenger RNA from COVID‐19 BTN162b2 and mRNA‐1273 vaccines in human milk. JAMA Pediatr. 2021;175(10):1069. doi: 10.1001/jamapediatrics.2021.1929 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Zipursky JS, Greenberg RA, Maxwell C, Bogler T. Pregnancy, breastfeeding and the SARS‐CoV‐2 vaccine: an ethics‐based framework for shared decision‐making. CMAJ (Can Med Assoc J). 2021;193(9):E312‐E314. doi: 10.1503/cmaj.202833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Dashraath P, Nielsen‐Saines K, Madhi SA, Baud D. COVID‐19 vaccines and neglected pregnancy. Lancet. 2020;396(10252):e22. doi: 10.1016/S0140-6736(20)31822-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Girling J. COVID‐19 vaccination in pregnancy. Drug Ther. Bull. 2021;59(6):82‐82. doi: 10.1136/dtb.2021.000002 [DOI] [PubMed] [Google Scholar]
  • 158. Hawton A, Green C, Dickens AP, et al. The impact of social isolation on the health status and health‐related quality of life of older people. Qual Life Res. 2011;20(1):57‐67. doi: 10.1007/s11136-010-9717-2 [DOI] [PubMed] [Google Scholar]
  • 159. Kumar S, Nyodu R, Maurya VK, Saxena SK. Morphology, genome organization, replication, and pathogenesis of severe acute respiratory syndrome Coronavirus 2 (SARS‐CoV‐2). In: Saxena SK, ed. Coronavirus Disease 2019 (COVID‐19). Springer Nature; 2020. doi:10.1007%2F978‐981‐15‐4814‐7_3 [Google Scholar]
  • 160. Dhama K, Patel SK, Kumar R, et al. Geriatric population during COVID‐19 pandemic: problems, considerations, exigencies and beyond. Front Public Health. 2020;8. doi: 10.3389/fpubh.2020.574198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Rolland Y, Cesari M, Morley JE, Merchant R, Vellas B. COVID19 Vaccination in Frail People. Lots of Hope and Some Questions. Springer; 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Dhakal S, Klein SL. Host factors impact vaccine efficacy: implications for seasonal and universal influenza vaccine programs. J Virol. 2019;93(21):e00797‐19. doi: 10.1128/JVI.00797-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Sadarangani M, Abu Raya B, Conway JM, et al. Importance of COVID‐19 vaccine efficacy in older age groups. Vaccine. 2021;39(15):2020‐2023. doi: 10.1016/j.vaccine.2021.03.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Cooper DM, Afghani B, Byington CL, et al. SARS‐CoV‐2 vaccine testing and trials in the pediatric population: biologic, ethical, research, and implementation challenges. Pediatr Res. 2021. doi: 10.1038/s41390-021-01402-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Brookman S, Cook J, Zucherman M, et al. Effect of the new SARS‐CoV‐2 variant B.1.1.7 on children and young people. Lancet Child Adolesc Health. 2021;5(4):e9‐e10. doi: 10.1016/S2352-4642(21)00030-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Dougherty K, Mannell M, Naqvi O, Matson D, Stone J. SARS‐CoV‐2 B.1.617.2 (delta) variant COVID‐19 outbreak associated with a gymnastics facility—Oklahoma, April‐May 2021. MMWR Morb Mortal Wkly Rep. 2021;70(28):1004‐1007. doi: 10.15585/mmwr.mm7028e2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Klass P, Ratner AJ. Vaccinating children against covid‐19—the lessons of measles. N. Engl J Med. 2021;384(7):589‐591. [DOI] [PubMed] [Google Scholar]
  • 168. Wong BLH, Ramsay ME, Ladhani SN. Should children be vaccinated against COVID‐19 now? Arch Dis Child. 2021. [DOI] [PubMed] [Google Scholar]
  • 169. Blaisdell LL, Cohn W, Pavell JR, Rubin DS, Vergales JE. Preventing and mitigating SARS‐CoV‐2 transmission—four overnight camps, Maine, June–August 2020. MMWR (Morb Mortal Wkly Rep). 2020;69(35):1216‐1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Ladhani SN, Amin‐Chowdhury Z, Davies HG, et al. COVID‐19 in children: analysis of the first pandemic peak in England. Arch Dis Child. 2020;105(12):1180‐1185. doi: 10.1136/archdischild-2020-320042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Food and Drug Administration (FDA) . Emergency use authorization (eua) of the moderna covid‐19 vaccine to prevent coronavirus disease 2019 (covid‐19): fact sheet for healthcare providers administering vaccine (vaccination providers). Food and Drug Administration (FDA); 2021. https://www.modernatx.com/covid19vaccine‐eua/eua‐fact‐sheet‐providers.pdf [Google Scholar]
  • 172. Shimabukuro T, Nair N. Allergic reactions including anaphylaxis after receipt of the first dose of Pfizer‐BioNTech COVID‐19 vaccine. J Am Med Assoc. 2021;325(8):780‐781. doi: 10.1001/jama.2021.0600 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Nakayama T, Aizawa C. Change in gelatin content of vaccines associated with reduction in reports of allergic reactions. J Allergy Clin Immunol. 2000;106(3):591‐592. doi: 10.1067/mai.2000.108433 [DOI] [PubMed] [Google Scholar]
  • 174. Tannenbaum H, Ruddy S, Schur PH. Acute anaphylaxis associated with serum complement depletion. J Allergy Clin Immunol. 1975;56(3):226‐234. doi: 10.1016/0091-6749(75)90094-9 [DOI] [PubMed] [Google Scholar]
  • 175. Stone CA, Jr. , Liu Y, Relling MV, et al. Immediate hypersensitivity to polyethylene glycols and polysorbates: more common than we have recognized. J Allergy Clin Immunol Pract. 2019;7(5):1533‐1540. doi: 10.1016/j.jaip.2018.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Krantz MS, Liu Y, Phillips EJ, Stone CA, Jr . Anaphylaxis to PEGylated liposomal echocardiogram contrast in a patient with IgE‐mediated macrogol allergy. J Allergy Clin Immunol Pract. 2020;8(4):1416‐1419. doi: 10.1016/j.jaip.2019.12.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Banerji A, Wickner PG, Saff R, et al. mRNA vaccines to prevent COVID‐19 disease and reported allergic reactions: current evidence and suggested approach. J Allergy Clin Immunol Pract. 2021;9(4):1423‐1437. doi: 10.1016/j.jaip.2020.12.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Voysey M, Clemens SAC, Madhi SA, et al. Safety and efficacy of the ChAdOx1 nCoV‐19 vaccine (AZD1222) against SARS‐CoV‐2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet. 2021;397(10269):99‐111. doi: 10.1016/S0140-6736(20)32661-1 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.


Articles from Reviews in Medical Virology are provided here courtesy of Wiley

RESOURCES