Summary

In addition to its pulmonary effects, coronavirus disease 2019 (COVID-19) has also been found to cause acute kidney injury (AKI), which has been linked to high mortality rates. In this review, we collected data from 20 clinical studies on post-COVID-19-related AKI and 97 cases of AKI associated with COVID-19 vaccination. Acute tubular injury was by far the most common finding in the kidneys of patients with COVID-19-related AKI. Among patients hospitalized for COVID-19, 34.0% developed AKI, of which 59.0%, 19.1% and 21.9% were Stages 1, 2 and 3, respectively. Though kidney disease and other adverse effects after COVID-19 vaccination overall appear rare, case reports have accumulated suggesting that COVID-19 vaccination may be associated with a risk of subsequent kidney disease. Among the patients with post-vaccination AKI, the most common pathologic findings include crescentic glomerulonephritis (29.9%), acute tubular injury (23.7%), IgA nephropathy (18.6%), antineutrophil cytoplasmic autoantibody-associated vasculitis (17.5%), minimal change disease (17.5%) and thrombotic microangiopathy (10.3%). It is important to note that crescentic glomerulonephritis appears to be more prevalent in patients who have newly diagnosed renal involvement. The proportions of patients with AKI Stages 1, 2 and 3 after COVID-19 vaccination in case reports were 30.9%, 22.7% and 46.4%, respectively. In general, clinical cases of new-onset and recurrent nephropathy with AKI after COVID-19 vaccination have a positive prognosis. In this article, we also explore the underlying pathophysiological mechanisms of AKI associated with COVID-19 infection and its vaccination by describing key renal morphological and clinical features and prognostic findings.

Introduction

Since the discovery of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019, there have been over 500 million confirmed cases of coronavirus disease 2019 (COVID-19) reported to the World Health Organization (WHO) as of May 25, 2022, with 6 million deaths. Mass vaccination has been a key strategy in containing the COVID-19 pandemic, particularly against severe COVID-19. According to multiple phase III and IV trials, the safety profile of these vaccines is good, with minimal occurrences of serious reactions.1–3

The widespread use of vaccinations has raised concerns about adverse effects such as acute kidney injury (AKI) and the emergence of new glomerulonephritis on a global scale.4,5 Despite this, the mechanisms, risk factors and long-term consequences of postvaccination AKI are not yet well established.6 This article presents a comprehensive review of the clinical features, treatment and prognosis of new-onset AKI following COVID-19 infection and vaccination. Additionally, potential mechanisms are summarized to further understand this phenomenon.

Search strategy

This narrative review was conducted following a thorough literature search of various databases, including PubMed/Medline, Embase and Web of Science (WOS), up until May 2023. Keywords are ‘glomerulonephritis’, ‘acute kidney disease’, ‘acute kidney injury’, ‘COVAN’, ‘SARS-CoV-2’, ‘COVID-19’, ‘coronavirus’, ‘novel corona virus’, ‘vaccination’ and ‘vaccine’. The definition of AKI according to the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines is as follows: Stage 1 is characterized by an increase in serum creatinine (Scr) by 0.3 mg/dl within 48 h or a 1.5- to 1.9-fold increase in Scr from baseline within 7 days; Stage 2 is defined as a 2- to 2.9-fold increase in Scr within 7 days and Stage 3 is marked by a 3-fold or more increase in Scr within 7 days or the initiation of renal replacement therapy (RRT).7,8

New-onset AKI post-SARS-CoV-2 infection

Pathological types and clinical features

A literature review on natural kidney biopsy findings in the minority of patients with COVID-19 showed the following findings with their corresponding percentages in parentheses: collapsing glomerulopathy (CG) (28.4%), acute tubular injury (ATI) (13.9%), diabetic nephropathy (DN) (9.7%), focal segmental glomerulosclerosis (FSGS) (7.6%), minimal change disease (MCD) (5.4%) and membranous nephropathy (MN) (4.5%).9 However, most patients with AKI after COVID-19 do not undergo biopsy. Of course, most patients with suspected acute tubular necrosis (ATN) do not undergo biopsy, and therefore, any biopsy series will be enriched with patients with hematuria and/or proteinuria and thus patients with non-ATN diagnoses. Autopsy studies have demonstrated that ATI is by far the most common finding in the kidneys of patients with COVID-19-related AKI.10 Virtually all experts in the field believe that ATN is the most common renal diagnosis after COVID-19 infection.11,12

We conducted a thorough search of various databases to identify recent large cohort studies on AKI in patients with COVID-19, as outlined in Supplementary Table S1.13–32 Among patients hospitalized for COVID-19, 34.0% developed AKI, and among these cases of AKI, 59.0%, 19.1% and 21.9% were Stages 1, 2 and 3, respectively (Table 1). Proteinuria and hematuria were more prominent in patients with COVID-19 and ATN than in most patients with ATN.32,33 In addition, low levels of hematuria and proteinuria were common in patients with COVID-19 without AKI, suggesting that they may not predict AKI. The most common comorbidities associated with COVID-19-related AKI were diabetes mellitus (DM) (7.4–72.1%), hypertension (HT) (15–66.8%), heart disease (30%) and chronic kidney disease (CKD) (0.7–22.6%).13,34,35 In addition, it was found that patients in the AKI group were significantly older than those in the non-AKI group,36 and patients with COVID-19 and AKI were more commonly male (67.9%) than female. Several studies have identified various factors that independently predict severe COVID-19-related AKI. These include older age, male sex, black race, CKD, DM, cardiovascular disease, an immunosuppressed state, the severity of COVID-19, apolipoprotein L1 (APOL1) variants, angiotensin-converting enzyme 2 (ACE2) polymorphism, elevated albuminuria, serum cystatin C, potassium, kidney injury molecule-1 (KIM-1) and D-dimer levels at hospital admission.17,32,37,38

Table 1.

Comparing the characteristics of post-COVID-19 and post-vaccination acute kidney injury

AKI associated with COVID-19 infectionAKI associated with COVID-19 vaccine
Possibly associated risk factorsOlder age, male sex, black race, chronic kidney disease, diabetes mellitus, cardiovascular disease, immunosuppressed state, severity of COVID-19, APOL1 variants, ACE2 polymorphism, elevated albuminuria, serum cystatin C, potassium, kidney injury molecule-1 and D-dimer levels at hospital admission17,32,37,38Coexisting active illnesses, older age and past disease histories168
Clinical manifestationsProteinuria, hematuriaNephrotic syndrome, gastrointestinal discomfort, hematuria, fever and headache
Comorbidities/medical history
  • Hypertension (15–66.8%)

  • diabetes mellitus (7.4–72.1%)

  • heart disease (30%)

  • pre-existing CKD (0.7–22.6%)13,33,34

  • Hypertension (35.4%)

  • diabetes mellitus (20.7%)

  • thyroid dysfunction (13.4%)

  • dyslipidemia (12.2%)

Pathological type
  • Collapsing glomerulopathy (28.4%)

  • acute tubular injury (13.9%)

  • diabetic nephropathy (9.7%)

  • focal segmental glomerulosclerosis (7.6%)

  • minimal change disease (5.4%)

  • membranous nephropathy (4.5%)9

  • Crescentic glomerulonephritis (29.9%)

  • acute tubular injury (23.7%)

  • IgA nephropathy (18.6%)

  • ANCA-associated vasculitis (17.5%)

  • minimal change disease (17.5%)

  • thrombotic microangiopathy (10.3%)

Treatment
  • Control of the potential source of infection

  • proper isolation of a patient

  • hemodynamic support

  • electrolyte and acid–base control

  • fluid balance

  • avoidance of potentially nephrotoxic drugs

  • renal replacement therapy

  • asymptomatic supportive care

  • Steroids

  • renal replacement therapy

  • rituximab

  • plasma exchange

  • cyclophosphamide

  • mycophenolate mofetil

  • intravenous immunoglobulins

Mean age (years)71.0 (n = 20 416)55.4 (n = 96)
Male sex (%)67.9 (n = 26 921)56.7 (n = 97)
AKI incidence (%)
 Overall34.0 (n = 89 216)
 Race
  • White: 41

  • Black: 20.8

  • Asians: 8.1

  • Other/multiracial: 25.426

 ICU44.3 (n = 6124)
 AKI Stage 159.0 (n = 29 931)30.9 (n = 97)
 AKI Stage 219.1 (n = 29 931)22.7 (n = 97)
 AKI Stage 321.9 (n = 29 931)46.4 (n = 97)
Outcome (%)
 Mortality rate in 30 days40.5 (n = 30 210)0 (n = 97)
 Complete remission in 3 months49.4 (n = 85)
 Partial remission in 3 months28.2 (n = 85)
 No response in 3 months22.4 (n = 85)
 Dependent on hemodialysis22.3 (n = 462)28,1698.2 (n = 85)
Potential mechanisms
 Common
  • a. Renin–angiotensin–aldosterone system abnormalities:

  • proinflammatory properties of upregulated AGII, polymorphism in ACE2 and upregulation/downregulation of ACE2

  • b. Rhabdomyolysis:

  • medullary vasoconstriction, intrarenal hypoxia and damage of renal cells by reactive oxygen species

  • c. Microthrombi:

  • inflammatory infiltrations, obstruction and subsequent hypoxia

  • d. Hyperinflammation:

  • cytokine storm and complement activation

  • e. Dysregulation of neutrophil extracellular traps

  • f. Genetic susceptibility:

  • APOL1 high-risk variants

  • g. Cardiovascular comorbidity

 Difference
  • a. Cytopathic effect of replicating virus:

  • immune-mediated tissue damage,

  • glomerulonephritis,

  • tubular injury

  • b. Septic shock and inflammation

  • c. Mechanical ventilation

  • d. Drugs with a potential nephrotoxic effect

  • e. Organ crosstalk:

  • between injured lungs, heart and kidneys

  • a. Spike protein induced cross immune response:

  • tubular injury,

  • glomerulonephritis,

  • tissue damage

  • b. Underlying disease:

  • hypertension,

  • diabetes mellitus,

  • autoimmune disease

  • c. Adjuvants: inducing an enhanced immune response

AKI associated with COVID-19 infectionAKI associated with COVID-19 vaccine
Possibly associated risk factorsOlder age, male sex, black race, chronic kidney disease, diabetes mellitus, cardiovascular disease, immunosuppressed state, severity of COVID-19, APOL1 variants, ACE2 polymorphism, elevated albuminuria, serum cystatin C, potassium, kidney injury molecule-1 and D-dimer levels at hospital admission17,32,37,38Coexisting active illnesses, older age and past disease histories168
Clinical manifestationsProteinuria, hematuriaNephrotic syndrome, gastrointestinal discomfort, hematuria, fever and headache
Comorbidities/medical history
  • Hypertension (15–66.8%)

  • diabetes mellitus (7.4–72.1%)

  • heart disease (30%)

  • pre-existing CKD (0.7–22.6%)13,33,34

  • Hypertension (35.4%)

  • diabetes mellitus (20.7%)

  • thyroid dysfunction (13.4%)

  • dyslipidemia (12.2%)

Pathological type
  • Collapsing glomerulopathy (28.4%)

  • acute tubular injury (13.9%)

  • diabetic nephropathy (9.7%)

  • focal segmental glomerulosclerosis (7.6%)

  • minimal change disease (5.4%)

  • membranous nephropathy (4.5%)9

  • Crescentic glomerulonephritis (29.9%)

  • acute tubular injury (23.7%)

  • IgA nephropathy (18.6%)

  • ANCA-associated vasculitis (17.5%)

  • minimal change disease (17.5%)

  • thrombotic microangiopathy (10.3%)

Treatment
  • Control of the potential source of infection

  • proper isolation of a patient

  • hemodynamic support

  • electrolyte and acid–base control

  • fluid balance

  • avoidance of potentially nephrotoxic drugs

  • renal replacement therapy

  • asymptomatic supportive care

  • Steroids

  • renal replacement therapy

  • rituximab

  • plasma exchange

  • cyclophosphamide

  • mycophenolate mofetil

  • intravenous immunoglobulins

Mean age (years)71.0 (n = 20 416)55.4 (n = 96)
Male sex (%)67.9 (n = 26 921)56.7 (n = 97)
AKI incidence (%)
 Overall34.0 (n = 89 216)
 Race
  • White: 41

  • Black: 20.8

  • Asians: 8.1

  • Other/multiracial: 25.426

 ICU44.3 (n = 6124)
 AKI Stage 159.0 (n = 29 931)30.9 (n = 97)
 AKI Stage 219.1 (n = 29 931)22.7 (n = 97)
 AKI Stage 321.9 (n = 29 931)46.4 (n = 97)
Outcome (%)
 Mortality rate in 30 days40.5 (n = 30 210)0 (n = 97)
 Complete remission in 3 months49.4 (n = 85)
 Partial remission in 3 months28.2 (n = 85)
 No response in 3 months22.4 (n = 85)
 Dependent on hemodialysis22.3 (n = 462)28,1698.2 (n = 85)
Potential mechanisms
 Common
  • a. Renin–angiotensin–aldosterone system abnormalities:

  • proinflammatory properties of upregulated AGII, polymorphism in ACE2 and upregulation/downregulation of ACE2

  • b. Rhabdomyolysis:

  • medullary vasoconstriction, intrarenal hypoxia and damage of renal cells by reactive oxygen species

  • c. Microthrombi:

  • inflammatory infiltrations, obstruction and subsequent hypoxia

  • d. Hyperinflammation:

  • cytokine storm and complement activation

  • e. Dysregulation of neutrophil extracellular traps

  • f. Genetic susceptibility:

  • APOL1 high-risk variants

  • g. Cardiovascular comorbidity

 Difference
  • a. Cytopathic effect of replicating virus:

  • immune-mediated tissue damage,

  • glomerulonephritis,

  • tubular injury

  • b. Septic shock and inflammation

  • c. Mechanical ventilation

  • d. Drugs with a potential nephrotoxic effect

  • e. Organ crosstalk:

  • between injured lungs, heart and kidneys

  • a. Spike protein induced cross immune response:

  • tubular injury,

  • glomerulonephritis,

  • tissue damage

  • b. Underlying disease:

  • hypertension,

  • diabetes mellitus,

  • autoimmune disease

  • c. Adjuvants: inducing an enhanced immune response

ACE2, angiotensin-converting enzyme 2; AKI, acute kidney injury; CKD, chronic kidney disease; ICU, intensive care unit.

Table 1.

Comparing the characteristics of post-COVID-19 and post-vaccination acute kidney injury

AKI associated with COVID-19 infectionAKI associated with COVID-19 vaccine
Possibly associated risk factorsOlder age, male sex, black race, chronic kidney disease, diabetes mellitus, cardiovascular disease, immunosuppressed state, severity of COVID-19, APOL1 variants, ACE2 polymorphism, elevated albuminuria, serum cystatin C, potassium, kidney injury molecule-1 and D-dimer levels at hospital admission17,32,37,38Coexisting active illnesses, older age and past disease histories168
Clinical manifestationsProteinuria, hematuriaNephrotic syndrome, gastrointestinal discomfort, hematuria, fever and headache
Comorbidities/medical history
  • Hypertension (15–66.8%)

  • diabetes mellitus (7.4–72.1%)

  • heart disease (30%)

  • pre-existing CKD (0.7–22.6%)13,33,34

  • Hypertension (35.4%)

  • diabetes mellitus (20.7%)

  • thyroid dysfunction (13.4%)

  • dyslipidemia (12.2%)

Pathological type
  • Collapsing glomerulopathy (28.4%)

  • acute tubular injury (13.9%)

  • diabetic nephropathy (9.7%)

  • focal segmental glomerulosclerosis (7.6%)

  • minimal change disease (5.4%)

  • membranous nephropathy (4.5%)9

  • Crescentic glomerulonephritis (29.9%)

  • acute tubular injury (23.7%)

  • IgA nephropathy (18.6%)

  • ANCA-associated vasculitis (17.5%)

  • minimal change disease (17.5%)

  • thrombotic microangiopathy (10.3%)

Treatment
  • Control of the potential source of infection

  • proper isolation of a patient

  • hemodynamic support

  • electrolyte and acid–base control

  • fluid balance

  • avoidance of potentially nephrotoxic drugs

  • renal replacement therapy

  • asymptomatic supportive care

  • Steroids

  • renal replacement therapy

  • rituximab

  • plasma exchange

  • cyclophosphamide

  • mycophenolate mofetil

  • intravenous immunoglobulins

Mean age (years)71.0 (n = 20 416)55.4 (n = 96)
Male sex (%)67.9 (n = 26 921)56.7 (n = 97)
AKI incidence (%)
 Overall34.0 (n = 89 216)
 Race
  • White: 41

  • Black: 20.8

  • Asians: 8.1

  • Other/multiracial: 25.426

 ICU44.3 (n = 6124)
 AKI Stage 159.0 (n = 29 931)30.9 (n = 97)
 AKI Stage 219.1 (n = 29 931)22.7 (n = 97)
 AKI Stage 321.9 (n = 29 931)46.4 (n = 97)
Outcome (%)
 Mortality rate in 30 days40.5 (n = 30 210)0 (n = 97)
 Complete remission in 3 months49.4 (n = 85)
 Partial remission in 3 months28.2 (n = 85)
 No response in 3 months22.4 (n = 85)
 Dependent on hemodialysis22.3 (n = 462)28,1698.2 (n = 85)
Potential mechanisms
 Common
  • a. Renin–angiotensin–aldosterone system abnormalities:

  • proinflammatory properties of upregulated AGII, polymorphism in ACE2 and upregulation/downregulation of ACE2

  • b. Rhabdomyolysis:

  • medullary vasoconstriction, intrarenal hypoxia and damage of renal cells by reactive oxygen species

  • c. Microthrombi:

  • inflammatory infiltrations, obstruction and subsequent hypoxia

  • d. Hyperinflammation:

  • cytokine storm and complement activation

  • e. Dysregulation of neutrophil extracellular traps

  • f. Genetic susceptibility:

  • APOL1 high-risk variants

  • g. Cardiovascular comorbidity

 Difference
  • a. Cytopathic effect of replicating virus:

  • immune-mediated tissue damage,

  • glomerulonephritis,

  • tubular injury

  • b. Septic shock and inflammation

  • c. Mechanical ventilation

  • d. Drugs with a potential nephrotoxic effect

  • e. Organ crosstalk:

  • between injured lungs, heart and kidneys

  • a. Spike protein induced cross immune response:

  • tubular injury,

  • glomerulonephritis,

  • tissue damage

  • b. Underlying disease:

  • hypertension,

  • diabetes mellitus,

  • autoimmune disease

  • c. Adjuvants: inducing an enhanced immune response

AKI associated with COVID-19 infectionAKI associated with COVID-19 vaccine
Possibly associated risk factorsOlder age, male sex, black race, chronic kidney disease, diabetes mellitus, cardiovascular disease, immunosuppressed state, severity of COVID-19, APOL1 variants, ACE2 polymorphism, elevated albuminuria, serum cystatin C, potassium, kidney injury molecule-1 and D-dimer levels at hospital admission17,32,37,38Coexisting active illnesses, older age and past disease histories168
Clinical manifestationsProteinuria, hematuriaNephrotic syndrome, gastrointestinal discomfort, hematuria, fever and headache
Comorbidities/medical history
  • Hypertension (15–66.8%)

  • diabetes mellitus (7.4–72.1%)

  • heart disease (30%)

  • pre-existing CKD (0.7–22.6%)13,33,34

  • Hypertension (35.4%)

  • diabetes mellitus (20.7%)

  • thyroid dysfunction (13.4%)

  • dyslipidemia (12.2%)

Pathological type
  • Collapsing glomerulopathy (28.4%)

  • acute tubular injury (13.9%)

  • diabetic nephropathy (9.7%)

  • focal segmental glomerulosclerosis (7.6%)

  • minimal change disease (5.4%)

  • membranous nephropathy (4.5%)9

  • Crescentic glomerulonephritis (29.9%)

  • acute tubular injury (23.7%)

  • IgA nephropathy (18.6%)

  • ANCA-associated vasculitis (17.5%)

  • minimal change disease (17.5%)

  • thrombotic microangiopathy (10.3%)

Treatment
  • Control of the potential source of infection

  • proper isolation of a patient

  • hemodynamic support

  • electrolyte and acid–base control

  • fluid balance

  • avoidance of potentially nephrotoxic drugs

  • renal replacement therapy

  • asymptomatic supportive care

  • Steroids

  • renal replacement therapy

  • rituximab

  • plasma exchange

  • cyclophosphamide

  • mycophenolate mofetil

  • intravenous immunoglobulins

Mean age (years)71.0 (n = 20 416)55.4 (n = 96)
Male sex (%)67.9 (n = 26 921)56.7 (n = 97)
AKI incidence (%)
 Overall34.0 (n = 89 216)
 Race
  • White: 41

  • Black: 20.8

  • Asians: 8.1

  • Other/multiracial: 25.426

 ICU44.3 (n = 6124)
 AKI Stage 159.0 (n = 29 931)30.9 (n = 97)
 AKI Stage 219.1 (n = 29 931)22.7 (n = 97)
 AKI Stage 321.9 (n = 29 931)46.4 (n = 97)
Outcome (%)
 Mortality rate in 30 days40.5 (n = 30 210)0 (n = 97)
 Complete remission in 3 months49.4 (n = 85)
 Partial remission in 3 months28.2 (n = 85)
 No response in 3 months22.4 (n = 85)
 Dependent on hemodialysis22.3 (n = 462)28,1698.2 (n = 85)
Potential mechanisms
 Common
  • a. Renin–angiotensin–aldosterone system abnormalities:

  • proinflammatory properties of upregulated AGII, polymorphism in ACE2 and upregulation/downregulation of ACE2

  • b. Rhabdomyolysis:

  • medullary vasoconstriction, intrarenal hypoxia and damage of renal cells by reactive oxygen species

  • c. Microthrombi:

  • inflammatory infiltrations, obstruction and subsequent hypoxia

  • d. Hyperinflammation:

  • cytokine storm and complement activation

  • e. Dysregulation of neutrophil extracellular traps

  • f. Genetic susceptibility:

  • APOL1 high-risk variants

  • g. Cardiovascular comorbidity

 Difference
  • a. Cytopathic effect of replicating virus:

  • immune-mediated tissue damage,

  • glomerulonephritis,

  • tubular injury

  • b. Septic shock and inflammation

  • c. Mechanical ventilation

  • d. Drugs with a potential nephrotoxic effect

  • e. Organ crosstalk:

  • between injured lungs, heart and kidneys

  • a. Spike protein induced cross immune response:

  • tubular injury,

  • glomerulonephritis,

  • tissue damage

  • b. Underlying disease:

  • hypertension,

  • diabetes mellitus,

  • autoimmune disease

  • c. Adjuvants: inducing an enhanced immune response

ACE2, angiotensin-converting enzyme 2; AKI, acute kidney injury; CKD, chronic kidney disease; ICU, intensive care unit.

Treatment and prognosis

To prevent the patient’s condition from deteriorating rapidly, it is important to develop a comprehensive treatment plan for those with COVID-19-related AKI as soon as possible. It is recommended to follow the supportive care guidelines outlined in KDIGO for managing COVID-19-related AKI in patients.39 The management of COVID-19-associated AKI is similar to that of AKI caused by septic shock and is primarily supportive. Implementing lung-protective ventilation may help reduce the risk of developing or worsening AKI by mitigating volutrauma and barotrauma.40 Li et al.41 found that patients with COVID-19 and AKI had a 5.3-fold higher mortality risk than those without AKI, which was higher than that of patients with other chronic comorbidities. Furthermore, Xiao et al.161 reported that mortality rates for 7.3% AKI Stage 1 but significantly increased to 64.3% for Stages 2 and 3 after 28 days.

New-onset and relapsed kidney disease with AKI post-COVID-19 vaccination

Pathological types and clinical features

The cases of a total of 97 patients are summarized in Table 2, including 86 (88.7%) patients with newly diagnosed renal involvement and 11 (11.3%) patients with recurrent kidney disease with AKI.4–6,42–113 Major types of renal pathology in post-COVID-19 vaccination AKI data collected from the literature included crescentic glomerulonephritis (29.9%, 29 of 97 patients), ATI (23.7%, 23 of 97 patients), IgA nephropathy (IgAN) (18.6%, 18 of 97 patients), antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis (17.5%, 17 of 97 patients), minimal change disease (17.5%, 17 of 97 patients), thrombotic microangiopathy (10.3%, 10 of 97 patients) and antiglomerular basement membrane (anti-GBM) glomerulonephritis (5.2%, 5 of 97 patients). Some other renal pathologies included anti-GBM glomerulonephritis in five patients, MN in three patients, lupus nephritis (LN) in three patients, ANCA-negative vasculitis in two patients, FSGS in one patient, podocytopathy in one patient and focal proliferative glomerulonephritis in one patient. There were 29 patients with crescentic glomerulonephritis in the new kidney involvement group and none in the relapsed group (Table 3). The mean age was 55.4 (range 12–84) years, and 56.7% (55 of 97 patients) of patients were male.

Table 2.

Summary of published cases of newly diagnosed acute kidney injury following SARS-CoV-2 vaccination

CaseAuthorsAge/sexCountry (race)Medical historyVaccine
Onset (day)Baseline-Scr (mg/dl)After vaccine-Scr (mg/dl) (day)Newly HT/worseSymptomsDiagnosisTreatmentsOutcomes
TypeManufacturerOnset after which dose
New-onset kidney disease with AKI
1.Leclerc et al.4271/MCanadaDyslipidemia treated with rosuvastatinVectorAstraZenecaFirstD10.710.6 (D14)IIIEdema, oligoanuricMCDHD, mPSL 1 g/d 1–3 days, PSL 60 mg/dCR Scr was 1.2 mg/dl, UPCR 28 mg/mmol at D81
2.Lim et al.4351/MKoreaNoneVectorJanssenFirstD7NA1.96 (D34) 2.37 (D36)IEdema, foamy urineMCDmPSL 64 mg/dCR Scr was 0.95 mg/dl, UPCR was 0.2 g/g at D57
3.Lebedev et al.450/MIsraelNonemRNAPfizer-BioNTechFirstD40.782.31 (D10)IIIEdema, diarrhea, abdominal painMCD with ATIPSL 80 mg/dCR Scr was 0.97 mg/dl, UACR was 155 mg/g at D37
4.Maas et al.4480s/MNetherlandsVTEmRNAPfizer-BioNTechFirstD7NA1.43 (D7)IIEdemaMCD with ATIPSL 80 mg/dCR UPCR was 0.68 g/g after 10 days of PSL
5.D’Agati et al.577/MUSA (Caucasian)T2DMmRNAPfizer-BioNTechFirstD71.0–1.32.33 (D14)IEdemaMCD with ATImPSL 1 g/d 1–3 days, PSL 60 mg/dNR Scr was 3.74 mg/dl, UTP was 18.8 g/d at D35
6.Holzworth et al.4563/FUSAHTmRNAModernaFirst<D70.71.48 (>D28)IIIEdema, dyspneaMCD with ATI and AINmPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dNA
7.Weijers et al.4661/FNetherlandsAIH, hypothyroidismmRNAPfizer-BioNTechFirstD10.7–0.81.47 (D4)NAEdemaMCDHD, steroids 1 mg/kg/dCR Scr was <1 mg/dl at D77, UTP was 0 g/d at D58
8.Lim et al.4751/MKoreaNoneVectorJanssenFirstD7Normal1.54 (D21) 1.96 (D33)NAEdemaMCDHigh-dose steroidCR was achieved after 3 weeks of treatment
9.Park et al.4834/MKoreaNAmRNAModernaSecondD2NA0.86 (D3) 5.38 (D7)NAPeriocular edemaMCDPSL 60 mg/dCR Scr was 0.89 mg/dl, UPCR was 0.28 g/g at D77
10.Gulumsek et al.9258/MTurkeyDMmRNAPfizer-BioNTechSecondD7Normal2.4 (D7)+EdemaMCDmPSL 80 mg/dCR Scr was 0.64 mg/dl, UTP was 0.067 g/d within 30 days
11.Annicchiarico Petruzzelli et al.4912/MItalyNonemRNAPfizer-BioNTechFirstD70.781.3 (D9) 4 (D13)IEdema, headacheMCD, tubular obstructionRRT, pulse mPSL,CR was achieved at 12 weeks
12.Baskaran et al.5055/MAustraliaNAVectorAstraZenecaSecondD7NA7.16NAEdemaMCD with ATI and AINHigh-dose steroidNA
13.Hanna et al.5160/MCanadaNonemRNAPfizer-BioNTechFirstD100.891.34 (D45)IIEdema, dyspneaMCD with ATIPSL 80 mg/dCR Scr was 1.03 mg/dl at 11 weeks
14.Psyllaki et al.5265/MGreece (Caucasian)NAmRNAPfizer-BioNTechFirstD10NAeGFR 52 mL/min/1.73 m2NAEdemaMCD with ATIPulse mPSL, PSL, cyclosporineCR was achieved within 10 days
15.Klomjit et al.683/MUSA (Caucasian)NAmRNAModernaSecondD281.192.19NAAKIMCD, ATNHigh-dose steroidPR Scr was 1.2 mg/dl during last follow-up
16.Da et al.5470/MSingaporeEdema after first vaccinemRNAPfizer-BioNTechSecondD1NA1.28IEdemaMN (anti-PLA2R-)Irbesartan, frusemide, warfarinNR within D60
17.Thammathiwat et al.5553/MThailandEdema and foamy urine after first vaccineInactivatedSinovac Life ScienceSecondD2NA1.5 2.4 (peak)IIEdemaSecondary MN (anti-PLA2R-)mPSL, PSL, CyC, ACEiCR
18.Schaubschlager et al.5863/MUSAHypothyroid, valvular heart disease, aortic valve replacement, SLE without renal involvementmRNAPfizer-BioNTechSecondD140.9–1.22.1 (peak)NANephrotic proteinuria, edemaSevere podocytopathyNANA
19.Kudose et al.5950/FUSA (Caucasian)HT, obesity, APSmRNAModernaSecondD21.31.7NAGross hematuria, fever, body achesIgAN (M1E0S1T1C1)ConservativeCR hematuria resolved within D5
20.Tan et al.6041/FChineseGDMmRNAPfizer-BioNTechSecondD1Normal1.73 (D2)IGross hematuria, headache, myalgiaIgAN with fibrocellular and fibrous crescentsPulse mPSL, PSL, CyCNA
21.Hanna et al.6117/MUSA (Caucasian)Foamy urinemRNAPfizer-BioNTechSecond<D1Normal1.78 (D6)IGross hematuriaIgAN (M1E1S1T1C1)Pulse mPSLPR Scr improved (duration not reported)
22.Anderegg et al.6239/MSwitzerlandHTmRNAModernaSecondimmediatelyNAAKINAFlu-like symptoms, fever, macrohematuriaSevere crescentic IgANHigh-dose glucocorticoids, CyCCR Scr was normalized within several weeks
23.Klomjit et al.638/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD141.31.6NAGross hematuriaIgANConservativeNA
24.Klomjit et al.644/MUSA (Caucasian)NAmRNAModernaFirstD141.12.5NAAKIIgAN, AINHigh-dose steroidNR Scr was 3.6 mg/dl during last follow-up
25.Klomjit et al.666/MUSA (Caucasian)NAmRNAModernaFirstD141.11.5NAGross hematuriaIgANPSLPR Scr was 1.4 mg/dl during last follow-up
26.Klomjit et al.662/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD421.02.2NAAKIIgANConservativePR Scr was 2.0 mg/dl during last follow-up
27.Ritter et al.6320/MSwitzerland (Caucasian)Allergic rhinoconjunctivitsmRNAModernaSecond1NormalAKINAFever, chills, body aches, gross hematuria, dizzinessIgANRASBCR Scr normalized at D49
28.Morimoto et al.9377/MJapanese (Asian)HT, atrial fibrillationmRNAModernaThirdD901.371.67 (D60) 16.29 (D90)IIEvaluation of anorexia, pruritus, lower-extremity edemaIgAN with MPGN-like changes, grade III (A/C) M1E1S1T1C1HD, 3-day pulse mPSL, PSL 50 mg/dNR dependent on HD
29.Chen et al.6455/MChina (Asian)Chronic hepatitis B infectionmRNAModernaFirstD14Normal1.94NANausea, vomiting, general malaiseATN, IgAN (M1E0S0T0C0)HDNR Scr was 4.0 mg/dl at D104
30.Ritter et al.6369/MSwitzerland (Caucasian)Hypertensive cardiomyopathy, BPH, depressionmRNAPfizer-BioNTechSecondD33NAAKI+Gross hematuria, fatigue, loss of appetiteIgAN, ANCASteroids, CyC, HDNR dependent on HD at D78
31.Fernández et al.5738/FArgentinaObesityinactivatedSinopharmFirstD7NA1.74IIAsthenia, foamy urineIgAN with crescent, IFTACorticosteroids, CyCPR
32.Fernández et al.5753/MArgentinaHT, overweightVectorAstraZenecaFirstD10NA3.07IAsthenia, foamy urine, edemaIgAN with fibrocellular crescents, ATI, IFTACorticosteroids, CyCPR
33.Park et al.6550/MUSAHT, CKDmRNAModernaSecondD11.171.54NAHematuria, foamy urineIgAN with 13% active crescentsRAASiPR Scr was 1.24 mg/dl during last follow-up
34.Park et al.6567/MUSAHT, CKDmRNAModernaFirstD301.22.9NAHematuria, foamy urineIgA vasculitisPrednisone 40 mg/d for 1 weekPR Scr was 1.4 mg/dl during last follow-up
35.Sugita et al.6667/FJapanHTmRNAPfizer-BioNTechSecondD1Normal0.83 (D7) 2.2 (D15)NARash, edema, gross hematuriaIgA vasculitismPSL 0.5 g 1–3 days, PSL, CyC, azathioprinePR Scr was 0.65 mg/dl, UTP was 1 g/d at D180
36.Tan et al.6060/FMalayHyperlipidemiamRNAPfizer-BioNTechSecondD1Normal6.11 (D39)IIIGross hematuriaAnti-GBM nephritisPulse mPSL, PSL, CyC, PLEXNA
37.Sacke et al.71Older/FUSANonemRNAModernaSecondD14Normal7.8NAFever, gross hematuria, anorexia, nauseaAnti-GBM with mesangial IgA depositsmPSL, CyC, PLEXNR remained HD dependent
38.Coorey et al.7269/FAustraliaHT, hypercholesterolemiaVectorAstraZenecaFirstD560.5613.3NALethargy, anuriaAnti-GBM nephritisHD, pulse mPSL, PSL, CyC, PLEXNR remained HD dependent
39.Coorey et al.7272/FAustraliaColorectal cancerVectorAstraZenecaSecondD21Normal13.6NAMalaise, diarrheaAnti-GBM nephritisHD, high-dose steroids, CyC, PLEX, RTXNR remained HD dependent
40.Klomjit et al.677/MUSA (Caucasian)NAmRNAPfizer-BioNTechFirstD711.8+HTAtypical anti-GBM nephritisPSL, MMFNR Scr was 2.9 mg/dl during last follow-up
41.Zamoner et al.9858/FBrazilHyperthyroidism (2006)VectorAstraZenecaFirstD21.02.2 3.3 (peak)+Fatigue, paleness, arthralgia, foamy urineANCA-associated pauci-immune CrGNmPSL, CyC, azathioprinePR Scr was 1.87 mg/dl, UTP was 0.5 g during last follow-up
42.Bansal et al.9967/FIndia (South-Asian)HTInactivatedBBV152SecondD141.22.8 (D21) 6.4 (D28)IILassitude, swellingANCA-associated pauci-immune CrGNHD, mPSL, CyCNR Scr was 3.8 mg/dl during last follow-up
43.Sekar et al.10052/MUSA (Caucasian)HTmRNAModernaSecondD11.118.41 (D14)NAHeadache, weaknessPR3-ANCA vasculitisRTX, CyC, PSL, HDNR remained HD dependent
44.Anderegg et al.6281/MSwitzerlandSustained flu-like symptoms after first vaccinemRNAModernaSecond<D1NAAKINAFlu-like symptoms worsenedPR3-ANCA vasculitisHigh-dose glucocorticoids, CyC, PLEXPR renal function improved within D21
45.Feghali et al.10158/MUSA (Caucasian)NonemRNAModernaSecondD4NA4.1NAHematuria, proteinuriaPR3-ANCA vasculitismPSL 1 g 1–3 days, PSL 60 mg/kg/d, RTX, CyC, PLEXPR Scr was 1.5 mg/dl after 10 weeks of diagnosis
46.Villa et al.10263/MSpainNoneVectorAstraZenecaFirstD2Normal2.9 (D7)NAFlu-like syndrome, hemoptysisMPO-ANCA vasculitisHigh-dose glucocorticoids, CyCNR Scr was 2.08 mg/dl at D49
47.Noel et al.10362/FUSASystemic sclerosis, T2DMmRNAPfizer-BioNTechSecondD28<15.18NAEdemaMPO-ANCA vasculitismPSL, CyC, MMFNR Scr was 2.6 mg/dl at 8 months
48.Hakroush et al.10479/FGermany (Caucasian)HT, degenerative disc diseasemRNAPfizer-BioNTechSecondD140.711.38 (D14) 6.57 (D24)NAWeakness, upper thigh painMPO-ANCA vasculitis, ATImPSL 250 mg/d 1–3 days, PSL 1 mg/kg/d, CyCCR Scr was normalized within D47
49.Klomjit et al.682/FUSA (Caucasian)NAmRNAModernaSecondD280.82.5NAAKI, hematuria, proteinuriaMPO-ANCA vasculitisHigh-dose steroid, RTXPR Scr was 2.3 mg/dl during last follow-up
50.Avalos et al.9474/FUSAHT, T2DM, hypothyroidismmRNAPfizer-BioNTechSecondD14Normal4.78 (D14)NAMalaise, fatigue, chest pain, lower extremity rashMPO-ANCA vasculitismPSL 1 g/d 1–5 days, PSL 1 mg/kg, RTXNA
51.Shakoor et al.10578/FUSAT2DM, HT, atrial fibrillationmRNAPfizer-BioNTechFirst<D70.771.31 (D16)NANausea, vomiting, diarrheaAKINoneCR improved spontaneously
mRNAPfizer-BioNTechSecondD6Normal3.54 (D6)NALethargy, nausea, vomiting, diarrheaMPO-ANCA vasculitismPSL 1 g 1–3 days, PSL 1 mg/kg/d, RTXR Scr was 1.71 mg/dl at 1-month follow-up
52.So et al.10642/MKoreaTuberculosismRNAPfizer-BioNTechSecond<D70.973.05 (D21)NAGeneral weakness, shortness of breath, edema, gross hematuriaMPO-ANCA vasculitismPSL 500 mg 1–3 days, PLEX, RTX, PSL 1 mg/kg/dPR Scr was 1.51 mg/dl at D51
53.Dube et al.10729/FUSACongenital diffuse cystic lung diseasemRNAPfizer-BioNTechSecondD160.81.25 (D16) 1.91 (D49)NormalNAMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL 1 mg/kg/d, RTX, CyCCR Scr was 1.01 mg/dl at D133
54.Ma et al.10870/FChina (Asian)HT, hyperlipidemia, kidney stonesInactivatedSinovac Life ScienceFirstD10.545.43 (D20) 6 (D22)NAPoor appetite, nausea, fatigue, foamy urineMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL, CyCCR Scr was 1.1 mg/dl at D322
55.Kim et al.10972/FKoreaNAmRNAModernaThirdD180.811.25 (D18) 4.71 (peak)IAnorexia, abdominal pain, febrile sensationMPO-ANCA vasculitismPSL, CyCPR Scr was 1.34 mg/dl after 2 months of diagnosis
56.Suzuki et al.11072/MJapanProstatic hypertrophymRNAPfizer-BioNTechSecondD1Normal5.0 (D14) 8.51 (D17)NormalFever, progressive fatigue, loss of appetiteMPO-ANCA vasculitismPSL, PSL, HD, RTXPR Scr was 2.2 mg/dl at D65
57.Schaubschlager et al.5877/FUASHT, fibrillation, monoclonal gammopathymRNAModernaSecondD600.92.9 (peak)NASymptomatic bradycardia, hematuriaMPO-ANCA vasculitisNANA
58.Gillion et al.6977/MBelgiumNoneVectorAstraZenecaFirstD281.22.7NAFever, night sweatANCA-negative CrGNmPSLCR Scr was normalized within D56
59.Kim et al.7016/FKoreaNonemRNAPfizer-BioNTechSecondD14Normal9.57IDyspnea, headache, edemaANCA-negative CrGNHD, mPSL, MMFNR remained in CKD stage
60.Wisnik-Rainville et al.9524/MHispanic-AmericanHT, sleep apneaVectorJanssenFirstD71.06.5 (D14)IIChest pain, headache, dizziness, vision changesFocal CrGNHD, ACEi, corticosteroids, RTX, CycNR Scr was not improvement in several months
61.Tan et al.7370s/FUKT2DM (2004), HTVectorAstraZenecaFirstD20.94.7 (D8) 6.99 (peak)NAWeakness, headacheAINPSLPR Scr was 2.07 mg/dl at D56
62.Soma et al.9654/MJapanT2DMmRNAPfizer-BioNTechFourthD31–1.34.72 (D5) 9 (peak)IFatigueATIPSLCR Scr was 1 mg/dl at D19
63.Mira et al.7445/FPortugal (Caucasian)Total thyroidectomymRNAPfizer-BioNTechSecondD10.8518.4 (D8)NormalAnorexia, nausea, vomiting, urine output reductionAIN, ATIHD, mPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dCR Scr was 1.02 mg/dl at D37
64.Unver et al.7567/FTurkeyT2DM, MCD in PRInactivatedSinovac Life ScienceSecondD100.84.2 (D26)IIIEdema, headacheAIN, ATImPSL 500 mg/d 1–3 days, PSL 1 mg/kg/d, cyclosporine APR Scr was 1.12 mg/dl at D60, UTP was 3 g/d at D115
65.Lim et al.4744/MKoreaT2DM, chronic hepatitis B infection, hyperlipidemiamRNAModernaFirstD10.914.13 (D7) 4.94 (D21)NAGastrointestinal discomfort, anorexiaATNHigh-dose steroidPR Scr was 1.89 mg/dl, UPCR was 0.3 g/g at D42
66.Choi et al.7617/MKoreaNAmRNAPfizer-BioNTechSecondD1Normal3.1 (D3)NAEpigastric pain, poor oral intake, feverATNSupportive careCR Scr was 1.0 mg/dl at D12
67.Choi et al.7612/MKoreaNonemRNAPfizer-BioNTechSecondD2Normal2.28 (D21)NormalAnorexia, vomitingATNOral steroidCR Scr was 1.1 mg/dl at D38
68.Schaubschlager et al.5860/FUSAHT, hypothyroidism, diffuse large B-cell lymphomamRNAModernaSecondD280.94.9 (peak)NAFatigue, shortness of breathATIN, ICGN with endocapillary hypercellularityNANA
69.Lim et al.4777/FKoreaT2DM, chronic hepatitis B, hepatocellular carcinomamRNAPfizer-BioNTechSecondD10.9810.67 (D7) 11.15 (D14)NASevere nausea and vomitingATN with myoglobin tubular castsHDPR Scr was 2.12 mg/dl, within 4 months
70.Missoum et al.9058/MAlgeriaHTInactivatedSinovac Life ScienceFirstD9Normal8.9NAFever, arthralgias, purpuraLeukocytoclastic vasculitis ATNHD, prednisonePR Scr was 2.8 mg/dl at D90
71.Al-Rasbi et al.8737/MOmanNonemRNAPfizer-BioNTechFirstD12Normal3.05 (D15) 5.75 (D18)NAArm pain, swelling and paresthesiaNon-oliguric AKI, myositis, thrombocytopenia, myocarditisIVIG 1 g/kg/d, mPSL 1 g/dCR
72.Unger et al.8869/FUSAT2DM, HT, morbid obesity, hyperlipidemia, hypothyroidismmRNAPfizer-BioNTechThirdD20.81.77 (D8) 2.73 (D10)NADark brown and frothy urineRhabdomyolysisIntravenous and oral hydrationCR Scr was 1.16 mg/dl at D77
73.Banamah et al.8958/FKingdom of Saudi ArabiaSchizophrenia (>10 years)mRNAPfizer-BioNTechThirdD1NA2.7 (D1) 8.5 (D8)NAWeakness, lethargy, dark brown urineRhabdomyolysisRRTCR Scr was 1.32 mg/dl at D42
74.Al Rawahi et al.7764/MSultanate of OmanHT, hyperlipidemiaVectorAstraZenecaFirstD7NA1.18 (D7)IFever, lethargy, abdominal painaTTP, VITTArgatroban, fondaparinux, hydrocortisone, IVIGCR renal function improved at D15
75.Yocum et al.7862/FUSAHyperlipidemia, GERD, hypothyroidism, HTVectorJanssenFirstD37NA2.19 (D37) 6 (D38)IIIAltered mental statusaTTP, VITT,PLEX, HD, mPSL, packed RBCsNA
76.Osmanodja et al.7925/MGermanyNonemRNAModernaFirstD2NA1.5 (D13)NAFever, headache, petechiaeaTTPPLEX, PSL 250 mg 1–3 days, caplacizumabCR Scr was 1 mg/dl at D27
77.Alislambouli et al.8061/MKorean-AmericanNAmRNAPfizer-BioNTechFirstD5NA1.57 (D5)NAFever, confusion, headache, emesis, ecchymosisaTTPPLEX, mPSL 1 g 1–3 days, RTXCR rapid and excellent response
78.Yoshida et al.8157/MJapanNonemRNAPfizer-BioNTechFirstD7NA1.57 (D14)NAFatigue, loss of appetite, jaundiceaTTPPLEX, PSL, RTXCR in good condition at D48
79.Ruhe et al.8284/FGermanyNAmRNAPfizer-BioNTechFirstD16NA1.95 (D16)IIIPartial hemiplegia, petechiaeaTTPPLEX, RTX, corticosteroidCR Scr was 0.6 mg/dl at D34
80.Chamarti et al.8380/MHispanicHT, T2DM, hyperlipidemia, gout, IDAmRNAPfizer-BioNTechSecondD12NA2.4 (D14)IGeneralized weakness, malaiseaTTPPLEX, packed RBCs, platelets, prednisoneCR Scr was 1 mg/dl at D30
81.Lim et al.4769/FKoreaT2DMVectorAstraZenecaFirstD20.83.69 (D14)NAGeneral weakness, gastrointestinal discomfortaTTPNoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
82.Claes et al.8438/FBelgiumNAmRNAModernaThirdD10.866.4 (D9)IIIHeadache, general malaiseaHUSHD, PLEX, eculizumabCR Scr was 1.04 mg/dl at D90
83.Tawhari et al.9738/MSaudi ArabiaNoneVectorAstraZenecaFirstD7Normal10.8 (D7)IIShortness of breath, body weakness, fatigabilityaHUSHD, PLEX, RTX, prednisoneNR dependent on HD
84.Kim et al.8666/FKoreaNoneVectorAstraZenecaSecondD500.661.81NAFoamy urine, edemaClass III LNmPSL, CyC, hydroxychloroquine, prednisoneCR Scr was 0.93 mg/dl after 10-day treatment
85.Vnučá et al.9125/FSlovakiaT1DM, HT, KT, autoimmune thyroiditis, COVID-19 infectionVectorAstraZenecaFirstD140.9510.74 (D24)NormalFatigue, general weakness, vomiting, with the inability to eat or drinkAcute kidney rejectionmPSL, PLEX, IVIG, HD, RTXNR Scr was 5.4 mg/dl at D98
86.Nune et al.11144/FUKMild asthmamRNAPfizer-BioNTechFirstD2Normal2.18 (D2)HypotensiveLeft arm pain, vomiting, loose stools, chest tightness, skin rashMultisystem inflammatory syndromemPSL 1 g/d 1–3 days, antibioticsPR
Relapsed kidney disease with AKI
1.Mancianti et al.5339/MItaly Caucasian)MCD in remission for 37 yearsmRNAPfizer-BioNTechFirstD30.91.8 (D8)NAEdemaMCDPSL 1 mg/kg/dCR
2.Klomjit et al.667/FUSA (Caucasian)MCDmRNAModernaSecondD2111.6NAEdemaMCDHigh-dose steroid, RTXCR Scr was 1.1 mg/dl at D40
3.Aydin et al.11266/FTurkeyHyperlipidemia, DM, HT, MN in CR for 8 yearsInactivatedSinovac Life ScienceFirstD14Normal2.78 (D14)NAEdemaMN (anti-PLA2R 1:120.53)NANA
4.Fernández et al.5759/FArgentinaHT, obesity, MPG (2015, CR)VectorGamaleyaFirstD14NA1.24IFoamy urine, edemaFocal proliferative GNCorticosteroidsPR
5.Huang et al.5654/MChinaAsthma, FSGS (2014, CR)VectorAstraZenecaSecondD3029.6 16.42 (peak)NAEdema, fatigue, poor appetiteFSGS, AINPSL, HDNR
6.Hanna et al.6113/MUSA (Caucasian)IgAN, T1DMmRNAPfizer-BioNTechSecond<D10.541.31 (D2)NAGross hematuria, vomitingIgAN (M0E0S0T0C0)ConservativeCR hematuria and Scr resolved within D6
7.Perrin et al.6741/FFranceIgAN, KTmRNAPfizer-BioNTechFirstD2NAScr transiently increasedNAGross hematuriaIgANConservativeCR spontaneously resolved
8.Watanabe et al.6854/FUSA (Caucasian)IgANmRNAModernaSecondD21.23.04 (D7)NormalGross hematuriaIgANPrednisone 60 mg/dCR Scr was 1.07 mg/dl at D90
9.Sekar et al.8530/MAmerican (African)LN (WHO Class 2 and 5 in 2019), COVID-19 infection 6 months priormRNAModernaFirstD30.792.81 (D3) 3.19 (D4)NAFevers, weakness, headacheLNMMF, hydroxychloroquine, prednisoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
10.Schaubschlager et al.5839/FUSASLE, hashimoto thyroiditis, LN class V (2015)mRNAModernaSecondD230.9–1.15.6 (peak)NALower extremity cellulitis, deep vein thrombosisATIN, ICGN with class II LNNANA
11.Martinez-Zayas et al.11353/MUSAANCA-associated vasculitis (2008), HIVmRNAPfizer-BioNTechSecondFew weeks0.92.49IFever, fatigue, shortness of breathICGNmPSL 1 g/d 1–3 days, RTX, prednisonePR
CaseAuthorsAge/sexCountry (race)Medical historyVaccine
Onset (day)Baseline-Scr (mg/dl)After vaccine-Scr (mg/dl) (day)Newly HT/worseSymptomsDiagnosisTreatmentsOutcomes
TypeManufacturerOnset after which dose
New-onset kidney disease with AKI
1.Leclerc et al.4271/MCanadaDyslipidemia treated with rosuvastatinVectorAstraZenecaFirstD10.710.6 (D14)IIIEdema, oligoanuricMCDHD, mPSL 1 g/d 1–3 days, PSL 60 mg/dCR Scr was 1.2 mg/dl, UPCR 28 mg/mmol at D81
2.Lim et al.4351/MKoreaNoneVectorJanssenFirstD7NA1.96 (D34) 2.37 (D36)IEdema, foamy urineMCDmPSL 64 mg/dCR Scr was 0.95 mg/dl, UPCR was 0.2 g/g at D57
3.Lebedev et al.450/MIsraelNonemRNAPfizer-BioNTechFirstD40.782.31 (D10)IIIEdema, diarrhea, abdominal painMCD with ATIPSL 80 mg/dCR Scr was 0.97 mg/dl, UACR was 155 mg/g at D37
4.Maas et al.4480s/MNetherlandsVTEmRNAPfizer-BioNTechFirstD7NA1.43 (D7)IIEdemaMCD with ATIPSL 80 mg/dCR UPCR was 0.68 g/g after 10 days of PSL
5.D’Agati et al.577/MUSA (Caucasian)T2DMmRNAPfizer-BioNTechFirstD71.0–1.32.33 (D14)IEdemaMCD with ATImPSL 1 g/d 1–3 days, PSL 60 mg/dNR Scr was 3.74 mg/dl, UTP was 18.8 g/d at D35
6.Holzworth et al.4563/FUSAHTmRNAModernaFirst<D70.71.48 (>D28)IIIEdema, dyspneaMCD with ATI and AINmPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dNA
7.Weijers et al.4661/FNetherlandsAIH, hypothyroidismmRNAPfizer-BioNTechFirstD10.7–0.81.47 (D4)NAEdemaMCDHD, steroids 1 mg/kg/dCR Scr was <1 mg/dl at D77, UTP was 0 g/d at D58
8.Lim et al.4751/MKoreaNoneVectorJanssenFirstD7Normal1.54 (D21) 1.96 (D33)NAEdemaMCDHigh-dose steroidCR was achieved after 3 weeks of treatment
9.Park et al.4834/MKoreaNAmRNAModernaSecondD2NA0.86 (D3) 5.38 (D7)NAPeriocular edemaMCDPSL 60 mg/dCR Scr was 0.89 mg/dl, UPCR was 0.28 g/g at D77
10.Gulumsek et al.9258/MTurkeyDMmRNAPfizer-BioNTechSecondD7Normal2.4 (D7)+EdemaMCDmPSL 80 mg/dCR Scr was 0.64 mg/dl, UTP was 0.067 g/d within 30 days
11.Annicchiarico Petruzzelli et al.4912/MItalyNonemRNAPfizer-BioNTechFirstD70.781.3 (D9) 4 (D13)IEdema, headacheMCD, tubular obstructionRRT, pulse mPSL,CR was achieved at 12 weeks
12.Baskaran et al.5055/MAustraliaNAVectorAstraZenecaSecondD7NA7.16NAEdemaMCD with ATI and AINHigh-dose steroidNA
13.Hanna et al.5160/MCanadaNonemRNAPfizer-BioNTechFirstD100.891.34 (D45)IIEdema, dyspneaMCD with ATIPSL 80 mg/dCR Scr was 1.03 mg/dl at 11 weeks
14.Psyllaki et al.5265/MGreece (Caucasian)NAmRNAPfizer-BioNTechFirstD10NAeGFR 52 mL/min/1.73 m2NAEdemaMCD with ATIPulse mPSL, PSL, cyclosporineCR was achieved within 10 days
15.Klomjit et al.683/MUSA (Caucasian)NAmRNAModernaSecondD281.192.19NAAKIMCD, ATNHigh-dose steroidPR Scr was 1.2 mg/dl during last follow-up
16.Da et al.5470/MSingaporeEdema after first vaccinemRNAPfizer-BioNTechSecondD1NA1.28IEdemaMN (anti-PLA2R-)Irbesartan, frusemide, warfarinNR within D60
17.Thammathiwat et al.5553/MThailandEdema and foamy urine after first vaccineInactivatedSinovac Life ScienceSecondD2NA1.5 2.4 (peak)IIEdemaSecondary MN (anti-PLA2R-)mPSL, PSL, CyC, ACEiCR
18.Schaubschlager et al.5863/MUSAHypothyroid, valvular heart disease, aortic valve replacement, SLE without renal involvementmRNAPfizer-BioNTechSecondD140.9–1.22.1 (peak)NANephrotic proteinuria, edemaSevere podocytopathyNANA
19.Kudose et al.5950/FUSA (Caucasian)HT, obesity, APSmRNAModernaSecondD21.31.7NAGross hematuria, fever, body achesIgAN (M1E0S1T1C1)ConservativeCR hematuria resolved within D5
20.Tan et al.6041/FChineseGDMmRNAPfizer-BioNTechSecondD1Normal1.73 (D2)IGross hematuria, headache, myalgiaIgAN with fibrocellular and fibrous crescentsPulse mPSL, PSL, CyCNA
21.Hanna et al.6117/MUSA (Caucasian)Foamy urinemRNAPfizer-BioNTechSecond<D1Normal1.78 (D6)IGross hematuriaIgAN (M1E1S1T1C1)Pulse mPSLPR Scr improved (duration not reported)
22.Anderegg et al.6239/MSwitzerlandHTmRNAModernaSecondimmediatelyNAAKINAFlu-like symptoms, fever, macrohematuriaSevere crescentic IgANHigh-dose glucocorticoids, CyCCR Scr was normalized within several weeks
23.Klomjit et al.638/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD141.31.6NAGross hematuriaIgANConservativeNA
24.Klomjit et al.644/MUSA (Caucasian)NAmRNAModernaFirstD141.12.5NAAKIIgAN, AINHigh-dose steroidNR Scr was 3.6 mg/dl during last follow-up
25.Klomjit et al.666/MUSA (Caucasian)NAmRNAModernaFirstD141.11.5NAGross hematuriaIgANPSLPR Scr was 1.4 mg/dl during last follow-up
26.Klomjit et al.662/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD421.02.2NAAKIIgANConservativePR Scr was 2.0 mg/dl during last follow-up
27.Ritter et al.6320/MSwitzerland (Caucasian)Allergic rhinoconjunctivitsmRNAModernaSecond1NormalAKINAFever, chills, body aches, gross hematuria, dizzinessIgANRASBCR Scr normalized at D49
28.Morimoto et al.9377/MJapanese (Asian)HT, atrial fibrillationmRNAModernaThirdD901.371.67 (D60) 16.29 (D90)IIEvaluation of anorexia, pruritus, lower-extremity edemaIgAN with MPGN-like changes, grade III (A/C) M1E1S1T1C1HD, 3-day pulse mPSL, PSL 50 mg/dNR dependent on HD
29.Chen et al.6455/MChina (Asian)Chronic hepatitis B infectionmRNAModernaFirstD14Normal1.94NANausea, vomiting, general malaiseATN, IgAN (M1E0S0T0C0)HDNR Scr was 4.0 mg/dl at D104
30.Ritter et al.6369/MSwitzerland (Caucasian)Hypertensive cardiomyopathy, BPH, depressionmRNAPfizer-BioNTechSecondD33NAAKI+Gross hematuria, fatigue, loss of appetiteIgAN, ANCASteroids, CyC, HDNR dependent on HD at D78
31.Fernández et al.5738/FArgentinaObesityinactivatedSinopharmFirstD7NA1.74IIAsthenia, foamy urineIgAN with crescent, IFTACorticosteroids, CyCPR
32.Fernández et al.5753/MArgentinaHT, overweightVectorAstraZenecaFirstD10NA3.07IAsthenia, foamy urine, edemaIgAN with fibrocellular crescents, ATI, IFTACorticosteroids, CyCPR
33.Park et al.6550/MUSAHT, CKDmRNAModernaSecondD11.171.54NAHematuria, foamy urineIgAN with 13% active crescentsRAASiPR Scr was 1.24 mg/dl during last follow-up
34.Park et al.6567/MUSAHT, CKDmRNAModernaFirstD301.22.9NAHematuria, foamy urineIgA vasculitisPrednisone 40 mg/d for 1 weekPR Scr was 1.4 mg/dl during last follow-up
35.Sugita et al.6667/FJapanHTmRNAPfizer-BioNTechSecondD1Normal0.83 (D7) 2.2 (D15)NARash, edema, gross hematuriaIgA vasculitismPSL 0.5 g 1–3 days, PSL, CyC, azathioprinePR Scr was 0.65 mg/dl, UTP was 1 g/d at D180
36.Tan et al.6060/FMalayHyperlipidemiamRNAPfizer-BioNTechSecondD1Normal6.11 (D39)IIIGross hematuriaAnti-GBM nephritisPulse mPSL, PSL, CyC, PLEXNA
37.Sacke et al.71Older/FUSANonemRNAModernaSecondD14Normal7.8NAFever, gross hematuria, anorexia, nauseaAnti-GBM with mesangial IgA depositsmPSL, CyC, PLEXNR remained HD dependent
38.Coorey et al.7269/FAustraliaHT, hypercholesterolemiaVectorAstraZenecaFirstD560.5613.3NALethargy, anuriaAnti-GBM nephritisHD, pulse mPSL, PSL, CyC, PLEXNR remained HD dependent
39.Coorey et al.7272/FAustraliaColorectal cancerVectorAstraZenecaSecondD21Normal13.6NAMalaise, diarrheaAnti-GBM nephritisHD, high-dose steroids, CyC, PLEX, RTXNR remained HD dependent
40.Klomjit et al.677/MUSA (Caucasian)NAmRNAPfizer-BioNTechFirstD711.8+HTAtypical anti-GBM nephritisPSL, MMFNR Scr was 2.9 mg/dl during last follow-up
41.Zamoner et al.9858/FBrazilHyperthyroidism (2006)VectorAstraZenecaFirstD21.02.2 3.3 (peak)+Fatigue, paleness, arthralgia, foamy urineANCA-associated pauci-immune CrGNmPSL, CyC, azathioprinePR Scr was 1.87 mg/dl, UTP was 0.5 g during last follow-up
42.Bansal et al.9967/FIndia (South-Asian)HTInactivatedBBV152SecondD141.22.8 (D21) 6.4 (D28)IILassitude, swellingANCA-associated pauci-immune CrGNHD, mPSL, CyCNR Scr was 3.8 mg/dl during last follow-up
43.Sekar et al.10052/MUSA (Caucasian)HTmRNAModernaSecondD11.118.41 (D14)NAHeadache, weaknessPR3-ANCA vasculitisRTX, CyC, PSL, HDNR remained HD dependent
44.Anderegg et al.6281/MSwitzerlandSustained flu-like symptoms after first vaccinemRNAModernaSecond<D1NAAKINAFlu-like symptoms worsenedPR3-ANCA vasculitisHigh-dose glucocorticoids, CyC, PLEXPR renal function improved within D21
45.Feghali et al.10158/MUSA (Caucasian)NonemRNAModernaSecondD4NA4.1NAHematuria, proteinuriaPR3-ANCA vasculitismPSL 1 g 1–3 days, PSL 60 mg/kg/d, RTX, CyC, PLEXPR Scr was 1.5 mg/dl after 10 weeks of diagnosis
46.Villa et al.10263/MSpainNoneVectorAstraZenecaFirstD2Normal2.9 (D7)NAFlu-like syndrome, hemoptysisMPO-ANCA vasculitisHigh-dose glucocorticoids, CyCNR Scr was 2.08 mg/dl at D49
47.Noel et al.10362/FUSASystemic sclerosis, T2DMmRNAPfizer-BioNTechSecondD28<15.18NAEdemaMPO-ANCA vasculitismPSL, CyC, MMFNR Scr was 2.6 mg/dl at 8 months
48.Hakroush et al.10479/FGermany (Caucasian)HT, degenerative disc diseasemRNAPfizer-BioNTechSecondD140.711.38 (D14) 6.57 (D24)NAWeakness, upper thigh painMPO-ANCA vasculitis, ATImPSL 250 mg/d 1–3 days, PSL 1 mg/kg/d, CyCCR Scr was normalized within D47
49.Klomjit et al.682/FUSA (Caucasian)NAmRNAModernaSecondD280.82.5NAAKI, hematuria, proteinuriaMPO-ANCA vasculitisHigh-dose steroid, RTXPR Scr was 2.3 mg/dl during last follow-up
50.Avalos et al.9474/FUSAHT, T2DM, hypothyroidismmRNAPfizer-BioNTechSecondD14Normal4.78 (D14)NAMalaise, fatigue, chest pain, lower extremity rashMPO-ANCA vasculitismPSL 1 g/d 1–5 days, PSL 1 mg/kg, RTXNA
51.Shakoor et al.10578/FUSAT2DM, HT, atrial fibrillationmRNAPfizer-BioNTechFirst<D70.771.31 (D16)NANausea, vomiting, diarrheaAKINoneCR improved spontaneously
mRNAPfizer-BioNTechSecondD6Normal3.54 (D6)NALethargy, nausea, vomiting, diarrheaMPO-ANCA vasculitismPSL 1 g 1–3 days, PSL 1 mg/kg/d, RTXR Scr was 1.71 mg/dl at 1-month follow-up
52.So et al.10642/MKoreaTuberculosismRNAPfizer-BioNTechSecond<D70.973.05 (D21)NAGeneral weakness, shortness of breath, edema, gross hematuriaMPO-ANCA vasculitismPSL 500 mg 1–3 days, PLEX, RTX, PSL 1 mg/kg/dPR Scr was 1.51 mg/dl at D51
53.Dube et al.10729/FUSACongenital diffuse cystic lung diseasemRNAPfizer-BioNTechSecondD160.81.25 (D16) 1.91 (D49)NormalNAMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL 1 mg/kg/d, RTX, CyCCR Scr was 1.01 mg/dl at D133
54.Ma et al.10870/FChina (Asian)HT, hyperlipidemia, kidney stonesInactivatedSinovac Life ScienceFirstD10.545.43 (D20) 6 (D22)NAPoor appetite, nausea, fatigue, foamy urineMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL, CyCCR Scr was 1.1 mg/dl at D322
55.Kim et al.10972/FKoreaNAmRNAModernaThirdD180.811.25 (D18) 4.71 (peak)IAnorexia, abdominal pain, febrile sensationMPO-ANCA vasculitismPSL, CyCPR Scr was 1.34 mg/dl after 2 months of diagnosis
56.Suzuki et al.11072/MJapanProstatic hypertrophymRNAPfizer-BioNTechSecondD1Normal5.0 (D14) 8.51 (D17)NormalFever, progressive fatigue, loss of appetiteMPO-ANCA vasculitismPSL, PSL, HD, RTXPR Scr was 2.2 mg/dl at D65
57.Schaubschlager et al.5877/FUASHT, fibrillation, monoclonal gammopathymRNAModernaSecondD600.92.9 (peak)NASymptomatic bradycardia, hematuriaMPO-ANCA vasculitisNANA
58.Gillion et al.6977/MBelgiumNoneVectorAstraZenecaFirstD281.22.7NAFever, night sweatANCA-negative CrGNmPSLCR Scr was normalized within D56
59.Kim et al.7016/FKoreaNonemRNAPfizer-BioNTechSecondD14Normal9.57IDyspnea, headache, edemaANCA-negative CrGNHD, mPSL, MMFNR remained in CKD stage
60.Wisnik-Rainville et al.9524/MHispanic-AmericanHT, sleep apneaVectorJanssenFirstD71.06.5 (D14)IIChest pain, headache, dizziness, vision changesFocal CrGNHD, ACEi, corticosteroids, RTX, CycNR Scr was not improvement in several months
61.Tan et al.7370s/FUKT2DM (2004), HTVectorAstraZenecaFirstD20.94.7 (D8) 6.99 (peak)NAWeakness, headacheAINPSLPR Scr was 2.07 mg/dl at D56
62.Soma et al.9654/MJapanT2DMmRNAPfizer-BioNTechFourthD31–1.34.72 (D5) 9 (peak)IFatigueATIPSLCR Scr was 1 mg/dl at D19
63.Mira et al.7445/FPortugal (Caucasian)Total thyroidectomymRNAPfizer-BioNTechSecondD10.8518.4 (D8)NormalAnorexia, nausea, vomiting, urine output reductionAIN, ATIHD, mPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dCR Scr was 1.02 mg/dl at D37
64.Unver et al.7567/FTurkeyT2DM, MCD in PRInactivatedSinovac Life ScienceSecondD100.84.2 (D26)IIIEdema, headacheAIN, ATImPSL 500 mg/d 1–3 days, PSL 1 mg/kg/d, cyclosporine APR Scr was 1.12 mg/dl at D60, UTP was 3 g/d at D115
65.Lim et al.4744/MKoreaT2DM, chronic hepatitis B infection, hyperlipidemiamRNAModernaFirstD10.914.13 (D7) 4.94 (D21)NAGastrointestinal discomfort, anorexiaATNHigh-dose steroidPR Scr was 1.89 mg/dl, UPCR was 0.3 g/g at D42
66.Choi et al.7617/MKoreaNAmRNAPfizer-BioNTechSecondD1Normal3.1 (D3)NAEpigastric pain, poor oral intake, feverATNSupportive careCR Scr was 1.0 mg/dl at D12
67.Choi et al.7612/MKoreaNonemRNAPfizer-BioNTechSecondD2Normal2.28 (D21)NormalAnorexia, vomitingATNOral steroidCR Scr was 1.1 mg/dl at D38
68.Schaubschlager et al.5860/FUSAHT, hypothyroidism, diffuse large B-cell lymphomamRNAModernaSecondD280.94.9 (peak)NAFatigue, shortness of breathATIN, ICGN with endocapillary hypercellularityNANA
69.Lim et al.4777/FKoreaT2DM, chronic hepatitis B, hepatocellular carcinomamRNAPfizer-BioNTechSecondD10.9810.67 (D7) 11.15 (D14)NASevere nausea and vomitingATN with myoglobin tubular castsHDPR Scr was 2.12 mg/dl, within 4 months
70.Missoum et al.9058/MAlgeriaHTInactivatedSinovac Life ScienceFirstD9Normal8.9NAFever, arthralgias, purpuraLeukocytoclastic vasculitis ATNHD, prednisonePR Scr was 2.8 mg/dl at D90
71.Al-Rasbi et al.8737/MOmanNonemRNAPfizer-BioNTechFirstD12Normal3.05 (D15) 5.75 (D18)NAArm pain, swelling and paresthesiaNon-oliguric AKI, myositis, thrombocytopenia, myocarditisIVIG 1 g/kg/d, mPSL 1 g/dCR
72.Unger et al.8869/FUSAT2DM, HT, morbid obesity, hyperlipidemia, hypothyroidismmRNAPfizer-BioNTechThirdD20.81.77 (D8) 2.73 (D10)NADark brown and frothy urineRhabdomyolysisIntravenous and oral hydrationCR Scr was 1.16 mg/dl at D77
73.Banamah et al.8958/FKingdom of Saudi ArabiaSchizophrenia (>10 years)mRNAPfizer-BioNTechThirdD1NA2.7 (D1) 8.5 (D8)NAWeakness, lethargy, dark brown urineRhabdomyolysisRRTCR Scr was 1.32 mg/dl at D42
74.Al Rawahi et al.7764/MSultanate of OmanHT, hyperlipidemiaVectorAstraZenecaFirstD7NA1.18 (D7)IFever, lethargy, abdominal painaTTP, VITTArgatroban, fondaparinux, hydrocortisone, IVIGCR renal function improved at D15
75.Yocum et al.7862/FUSAHyperlipidemia, GERD, hypothyroidism, HTVectorJanssenFirstD37NA2.19 (D37) 6 (D38)IIIAltered mental statusaTTP, VITT,PLEX, HD, mPSL, packed RBCsNA
76.Osmanodja et al.7925/MGermanyNonemRNAModernaFirstD2NA1.5 (D13)NAFever, headache, petechiaeaTTPPLEX, PSL 250 mg 1–3 days, caplacizumabCR Scr was 1 mg/dl at D27
77.Alislambouli et al.8061/MKorean-AmericanNAmRNAPfizer-BioNTechFirstD5NA1.57 (D5)NAFever, confusion, headache, emesis, ecchymosisaTTPPLEX, mPSL 1 g 1–3 days, RTXCR rapid and excellent response
78.Yoshida et al.8157/MJapanNonemRNAPfizer-BioNTechFirstD7NA1.57 (D14)NAFatigue, loss of appetite, jaundiceaTTPPLEX, PSL, RTXCR in good condition at D48
79.Ruhe et al.8284/FGermanyNAmRNAPfizer-BioNTechFirstD16NA1.95 (D16)IIIPartial hemiplegia, petechiaeaTTPPLEX, RTX, corticosteroidCR Scr was 0.6 mg/dl at D34
80.Chamarti et al.8380/MHispanicHT, T2DM, hyperlipidemia, gout, IDAmRNAPfizer-BioNTechSecondD12NA2.4 (D14)IGeneralized weakness, malaiseaTTPPLEX, packed RBCs, platelets, prednisoneCR Scr was 1 mg/dl at D30
81.Lim et al.4769/FKoreaT2DMVectorAstraZenecaFirstD20.83.69 (D14)NAGeneral weakness, gastrointestinal discomfortaTTPNoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
82.Claes et al.8438/FBelgiumNAmRNAModernaThirdD10.866.4 (D9)IIIHeadache, general malaiseaHUSHD, PLEX, eculizumabCR Scr was 1.04 mg/dl at D90
83.Tawhari et al.9738/MSaudi ArabiaNoneVectorAstraZenecaFirstD7Normal10.8 (D7)IIShortness of breath, body weakness, fatigabilityaHUSHD, PLEX, RTX, prednisoneNR dependent on HD
84.Kim et al.8666/FKoreaNoneVectorAstraZenecaSecondD500.661.81NAFoamy urine, edemaClass III LNmPSL, CyC, hydroxychloroquine, prednisoneCR Scr was 0.93 mg/dl after 10-day treatment
85.Vnučá et al.9125/FSlovakiaT1DM, HT, KT, autoimmune thyroiditis, COVID-19 infectionVectorAstraZenecaFirstD140.9510.74 (D24)NormalFatigue, general weakness, vomiting, with the inability to eat or drinkAcute kidney rejectionmPSL, PLEX, IVIG, HD, RTXNR Scr was 5.4 mg/dl at D98
86.Nune et al.11144/FUKMild asthmamRNAPfizer-BioNTechFirstD2Normal2.18 (D2)HypotensiveLeft arm pain, vomiting, loose stools, chest tightness, skin rashMultisystem inflammatory syndromemPSL 1 g/d 1–3 days, antibioticsPR
Relapsed kidney disease with AKI
1.Mancianti et al.5339/MItaly Caucasian)MCD in remission for 37 yearsmRNAPfizer-BioNTechFirstD30.91.8 (D8)NAEdemaMCDPSL 1 mg/kg/dCR
2.Klomjit et al.667/FUSA (Caucasian)MCDmRNAModernaSecondD2111.6NAEdemaMCDHigh-dose steroid, RTXCR Scr was 1.1 mg/dl at D40
3.Aydin et al.11266/FTurkeyHyperlipidemia, DM, HT, MN in CR for 8 yearsInactivatedSinovac Life ScienceFirstD14Normal2.78 (D14)NAEdemaMN (anti-PLA2R 1:120.53)NANA
4.Fernández et al.5759/FArgentinaHT, obesity, MPG (2015, CR)VectorGamaleyaFirstD14NA1.24IFoamy urine, edemaFocal proliferative GNCorticosteroidsPR
5.Huang et al.5654/MChinaAsthma, FSGS (2014, CR)VectorAstraZenecaSecondD3029.6 16.42 (peak)NAEdema, fatigue, poor appetiteFSGS, AINPSL, HDNR
6.Hanna et al.6113/MUSA (Caucasian)IgAN, T1DMmRNAPfizer-BioNTechSecond<D10.541.31 (D2)NAGross hematuria, vomitingIgAN (M0E0S0T0C0)ConservativeCR hematuria and Scr resolved within D6
7.Perrin et al.6741/FFranceIgAN, KTmRNAPfizer-BioNTechFirstD2NAScr transiently increasedNAGross hematuriaIgANConservativeCR spontaneously resolved
8.Watanabe et al.6854/FUSA (Caucasian)IgANmRNAModernaSecondD21.23.04 (D7)NormalGross hematuriaIgANPrednisone 60 mg/dCR Scr was 1.07 mg/dl at D90
9.Sekar et al.8530/MAmerican (African)LN (WHO Class 2 and 5 in 2019), COVID-19 infection 6 months priormRNAModernaFirstD30.792.81 (D3) 3.19 (D4)NAFevers, weakness, headacheLNMMF, hydroxychloroquine, prednisoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
10.Schaubschlager et al.5839/FUSASLE, hashimoto thyroiditis, LN class V (2015)mRNAModernaSecondD230.9–1.15.6 (peak)NALower extremity cellulitis, deep vein thrombosisATIN, ICGN with class II LNNANA
11.Martinez-Zayas et al.11353/MUSAANCA-associated vasculitis (2008), HIVmRNAPfizer-BioNTechSecondFew weeks0.92.49IFever, fatigue, shortness of breathICGNmPSL 1 g/d 1–3 days, RTX, prednisonePR

ACEi, angiotensin-converting enzyme inhibitor; AKI, acute kidney injury; AIH, autoimmune hepatitis; AIN, acute interstitial nephritis; ATN, acute tubular necrosis; ATIN, acute tubulointerstitial nephritis; ATI, acute tubular injury; APS, antiphospholipid syndrome; ANCA, antineutrophil cytoplasmic autoantibodies; ANCA GN, antineutrophil cytoplasmic autoantibody-associated glomerulonephritis; anti-PLA2R, antiphospholipase A2 receptor; aHUS, atypical hemolytic uremic syndrome; aTTP, acquired thrombotic thrombocytopenic purpura; BPH, benign prostatic hyperplasia; CKD, chronic kidney disease; CyC, cyclophosphamide; CR, complete remission; CRF, chronic renal failure; CrGN, crescentic glomerulonephritis; DM, diabetes mellitus; eGFR, estimated glomerular filtration rate; F, female; FGN, fibrillary glomerulonephritis; FSGS, focal segmental glomerulosclerosis; GBM, glomerular basement membrane; GDM, gestational diabetes; GERD, gastroesophageal reflux disease; HD, hemodialysis; HIV, human immunodeficiency virus; HT, hypertension; ICGN, immune-complex glomerulonephritis; IDA, iron deficiency anemia; IFTA, interstitial fibrosis and tubular atrophy; IgAN, IgA nephropathy; ITT, vaccine-immune thrombotic thrombocytopenia; IVIG, intravenous immunoglobulins; KT, kidney transplantation; LN, lupus nephritis; M, male; MCD, minimal change disease; mPSL, methylprednisolone; MMF, mycophenolate mofetil; MN, membranous nephropathy; MPA, mycophenolic acid; NA, not applicable; NR, no response; PR, partial remission; PLEX, plasma exchange; PSL, prednisolone; R, response; RAAS, renin–angiotensin system; RASB, renin–angiotensin system blockade; RBCs, red blood cells; RTX, rituximab; RRT, renal replacement therapy; Scr, serum creatinine; TAC, tacrolimus; T1DM, Type 1 diabetes mellitus; T2DM, Type 2 diabetes mellitus; UACR, urinary albumin-creatinine ratio; UTP, 24-h urine protein; UPCR, urine protein-to-creatinine ratio; VITT, vaccine-induced immune thrombotic thrombocytopenia; VTE, venous thromboembolism.

Table 2.

Summary of published cases of newly diagnosed acute kidney injury following SARS-CoV-2 vaccination

CaseAuthorsAge/sexCountry (race)Medical historyVaccine
Onset (day)Baseline-Scr (mg/dl)After vaccine-Scr (mg/dl) (day)Newly HT/worseSymptomsDiagnosisTreatmentsOutcomes
TypeManufacturerOnset after which dose
New-onset kidney disease with AKI
1.Leclerc et al.4271/MCanadaDyslipidemia treated with rosuvastatinVectorAstraZenecaFirstD10.710.6 (D14)IIIEdema, oligoanuricMCDHD, mPSL 1 g/d 1–3 days, PSL 60 mg/dCR Scr was 1.2 mg/dl, UPCR 28 mg/mmol at D81
2.Lim et al.4351/MKoreaNoneVectorJanssenFirstD7NA1.96 (D34) 2.37 (D36)IEdema, foamy urineMCDmPSL 64 mg/dCR Scr was 0.95 mg/dl, UPCR was 0.2 g/g at D57
3.Lebedev et al.450/MIsraelNonemRNAPfizer-BioNTechFirstD40.782.31 (D10)IIIEdema, diarrhea, abdominal painMCD with ATIPSL 80 mg/dCR Scr was 0.97 mg/dl, UACR was 155 mg/g at D37
4.Maas et al.4480s/MNetherlandsVTEmRNAPfizer-BioNTechFirstD7NA1.43 (D7)IIEdemaMCD with ATIPSL 80 mg/dCR UPCR was 0.68 g/g after 10 days of PSL
5.D’Agati et al.577/MUSA (Caucasian)T2DMmRNAPfizer-BioNTechFirstD71.0–1.32.33 (D14)IEdemaMCD with ATImPSL 1 g/d 1–3 days, PSL 60 mg/dNR Scr was 3.74 mg/dl, UTP was 18.8 g/d at D35
6.Holzworth et al.4563/FUSAHTmRNAModernaFirst<D70.71.48 (>D28)IIIEdema, dyspneaMCD with ATI and AINmPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dNA
7.Weijers et al.4661/FNetherlandsAIH, hypothyroidismmRNAPfizer-BioNTechFirstD10.7–0.81.47 (D4)NAEdemaMCDHD, steroids 1 mg/kg/dCR Scr was <1 mg/dl at D77, UTP was 0 g/d at D58
8.Lim et al.4751/MKoreaNoneVectorJanssenFirstD7Normal1.54 (D21) 1.96 (D33)NAEdemaMCDHigh-dose steroidCR was achieved after 3 weeks of treatment
9.Park et al.4834/MKoreaNAmRNAModernaSecondD2NA0.86 (D3) 5.38 (D7)NAPeriocular edemaMCDPSL 60 mg/dCR Scr was 0.89 mg/dl, UPCR was 0.28 g/g at D77
10.Gulumsek et al.9258/MTurkeyDMmRNAPfizer-BioNTechSecondD7Normal2.4 (D7)+EdemaMCDmPSL 80 mg/dCR Scr was 0.64 mg/dl, UTP was 0.067 g/d within 30 days
11.Annicchiarico Petruzzelli et al.4912/MItalyNonemRNAPfizer-BioNTechFirstD70.781.3 (D9) 4 (D13)IEdema, headacheMCD, tubular obstructionRRT, pulse mPSL,CR was achieved at 12 weeks
12.Baskaran et al.5055/MAustraliaNAVectorAstraZenecaSecondD7NA7.16NAEdemaMCD with ATI and AINHigh-dose steroidNA
13.Hanna et al.5160/MCanadaNonemRNAPfizer-BioNTechFirstD100.891.34 (D45)IIEdema, dyspneaMCD with ATIPSL 80 mg/dCR Scr was 1.03 mg/dl at 11 weeks
14.Psyllaki et al.5265/MGreece (Caucasian)NAmRNAPfizer-BioNTechFirstD10NAeGFR 52 mL/min/1.73 m2NAEdemaMCD with ATIPulse mPSL, PSL, cyclosporineCR was achieved within 10 days
15.Klomjit et al.683/MUSA (Caucasian)NAmRNAModernaSecondD281.192.19NAAKIMCD, ATNHigh-dose steroidPR Scr was 1.2 mg/dl during last follow-up
16.Da et al.5470/MSingaporeEdema after first vaccinemRNAPfizer-BioNTechSecondD1NA1.28IEdemaMN (anti-PLA2R-)Irbesartan, frusemide, warfarinNR within D60
17.Thammathiwat et al.5553/MThailandEdema and foamy urine after first vaccineInactivatedSinovac Life ScienceSecondD2NA1.5 2.4 (peak)IIEdemaSecondary MN (anti-PLA2R-)mPSL, PSL, CyC, ACEiCR
18.Schaubschlager et al.5863/MUSAHypothyroid, valvular heart disease, aortic valve replacement, SLE without renal involvementmRNAPfizer-BioNTechSecondD140.9–1.22.1 (peak)NANephrotic proteinuria, edemaSevere podocytopathyNANA
19.Kudose et al.5950/FUSA (Caucasian)HT, obesity, APSmRNAModernaSecondD21.31.7NAGross hematuria, fever, body achesIgAN (M1E0S1T1C1)ConservativeCR hematuria resolved within D5
20.Tan et al.6041/FChineseGDMmRNAPfizer-BioNTechSecondD1Normal1.73 (D2)IGross hematuria, headache, myalgiaIgAN with fibrocellular and fibrous crescentsPulse mPSL, PSL, CyCNA
21.Hanna et al.6117/MUSA (Caucasian)Foamy urinemRNAPfizer-BioNTechSecond<D1Normal1.78 (D6)IGross hematuriaIgAN (M1E1S1T1C1)Pulse mPSLPR Scr improved (duration not reported)
22.Anderegg et al.6239/MSwitzerlandHTmRNAModernaSecondimmediatelyNAAKINAFlu-like symptoms, fever, macrohematuriaSevere crescentic IgANHigh-dose glucocorticoids, CyCCR Scr was normalized within several weeks
23.Klomjit et al.638/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD141.31.6NAGross hematuriaIgANConservativeNA
24.Klomjit et al.644/MUSA (Caucasian)NAmRNAModernaFirstD141.12.5NAAKIIgAN, AINHigh-dose steroidNR Scr was 3.6 mg/dl during last follow-up
25.Klomjit et al.666/MUSA (Caucasian)NAmRNAModernaFirstD141.11.5NAGross hematuriaIgANPSLPR Scr was 1.4 mg/dl during last follow-up
26.Klomjit et al.662/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD421.02.2NAAKIIgANConservativePR Scr was 2.0 mg/dl during last follow-up
27.Ritter et al.6320/MSwitzerland (Caucasian)Allergic rhinoconjunctivitsmRNAModernaSecond1NormalAKINAFever, chills, body aches, gross hematuria, dizzinessIgANRASBCR Scr normalized at D49
28.Morimoto et al.9377/MJapanese (Asian)HT, atrial fibrillationmRNAModernaThirdD901.371.67 (D60) 16.29 (D90)IIEvaluation of anorexia, pruritus, lower-extremity edemaIgAN with MPGN-like changes, grade III (A/C) M1E1S1T1C1HD, 3-day pulse mPSL, PSL 50 mg/dNR dependent on HD
29.Chen et al.6455/MChina (Asian)Chronic hepatitis B infectionmRNAModernaFirstD14Normal1.94NANausea, vomiting, general malaiseATN, IgAN (M1E0S0T0C0)HDNR Scr was 4.0 mg/dl at D104
30.Ritter et al.6369/MSwitzerland (Caucasian)Hypertensive cardiomyopathy, BPH, depressionmRNAPfizer-BioNTechSecondD33NAAKI+Gross hematuria, fatigue, loss of appetiteIgAN, ANCASteroids, CyC, HDNR dependent on HD at D78
31.Fernández et al.5738/FArgentinaObesityinactivatedSinopharmFirstD7NA1.74IIAsthenia, foamy urineIgAN with crescent, IFTACorticosteroids, CyCPR
32.Fernández et al.5753/MArgentinaHT, overweightVectorAstraZenecaFirstD10NA3.07IAsthenia, foamy urine, edemaIgAN with fibrocellular crescents, ATI, IFTACorticosteroids, CyCPR
33.Park et al.6550/MUSAHT, CKDmRNAModernaSecondD11.171.54NAHematuria, foamy urineIgAN with 13% active crescentsRAASiPR Scr was 1.24 mg/dl during last follow-up
34.Park et al.6567/MUSAHT, CKDmRNAModernaFirstD301.22.9NAHematuria, foamy urineIgA vasculitisPrednisone 40 mg/d for 1 weekPR Scr was 1.4 mg/dl during last follow-up
35.Sugita et al.6667/FJapanHTmRNAPfizer-BioNTechSecondD1Normal0.83 (D7) 2.2 (D15)NARash, edema, gross hematuriaIgA vasculitismPSL 0.5 g 1–3 days, PSL, CyC, azathioprinePR Scr was 0.65 mg/dl, UTP was 1 g/d at D180
36.Tan et al.6060/FMalayHyperlipidemiamRNAPfizer-BioNTechSecondD1Normal6.11 (D39)IIIGross hematuriaAnti-GBM nephritisPulse mPSL, PSL, CyC, PLEXNA
37.Sacke et al.71Older/FUSANonemRNAModernaSecondD14Normal7.8NAFever, gross hematuria, anorexia, nauseaAnti-GBM with mesangial IgA depositsmPSL, CyC, PLEXNR remained HD dependent
38.Coorey et al.7269/FAustraliaHT, hypercholesterolemiaVectorAstraZenecaFirstD560.5613.3NALethargy, anuriaAnti-GBM nephritisHD, pulse mPSL, PSL, CyC, PLEXNR remained HD dependent
39.Coorey et al.7272/FAustraliaColorectal cancerVectorAstraZenecaSecondD21Normal13.6NAMalaise, diarrheaAnti-GBM nephritisHD, high-dose steroids, CyC, PLEX, RTXNR remained HD dependent
40.Klomjit et al.677/MUSA (Caucasian)NAmRNAPfizer-BioNTechFirstD711.8+HTAtypical anti-GBM nephritisPSL, MMFNR Scr was 2.9 mg/dl during last follow-up
41.Zamoner et al.9858/FBrazilHyperthyroidism (2006)VectorAstraZenecaFirstD21.02.2 3.3 (peak)+Fatigue, paleness, arthralgia, foamy urineANCA-associated pauci-immune CrGNmPSL, CyC, azathioprinePR Scr was 1.87 mg/dl, UTP was 0.5 g during last follow-up
42.Bansal et al.9967/FIndia (South-Asian)HTInactivatedBBV152SecondD141.22.8 (D21) 6.4 (D28)IILassitude, swellingANCA-associated pauci-immune CrGNHD, mPSL, CyCNR Scr was 3.8 mg/dl during last follow-up
43.Sekar et al.10052/MUSA (Caucasian)HTmRNAModernaSecondD11.118.41 (D14)NAHeadache, weaknessPR3-ANCA vasculitisRTX, CyC, PSL, HDNR remained HD dependent
44.Anderegg et al.6281/MSwitzerlandSustained flu-like symptoms after first vaccinemRNAModernaSecond<D1NAAKINAFlu-like symptoms worsenedPR3-ANCA vasculitisHigh-dose glucocorticoids, CyC, PLEXPR renal function improved within D21
45.Feghali et al.10158/MUSA (Caucasian)NonemRNAModernaSecondD4NA4.1NAHematuria, proteinuriaPR3-ANCA vasculitismPSL 1 g 1–3 days, PSL 60 mg/kg/d, RTX, CyC, PLEXPR Scr was 1.5 mg/dl after 10 weeks of diagnosis
46.Villa et al.10263/MSpainNoneVectorAstraZenecaFirstD2Normal2.9 (D7)NAFlu-like syndrome, hemoptysisMPO-ANCA vasculitisHigh-dose glucocorticoids, CyCNR Scr was 2.08 mg/dl at D49
47.Noel et al.10362/FUSASystemic sclerosis, T2DMmRNAPfizer-BioNTechSecondD28<15.18NAEdemaMPO-ANCA vasculitismPSL, CyC, MMFNR Scr was 2.6 mg/dl at 8 months
48.Hakroush et al.10479/FGermany (Caucasian)HT, degenerative disc diseasemRNAPfizer-BioNTechSecondD140.711.38 (D14) 6.57 (D24)NAWeakness, upper thigh painMPO-ANCA vasculitis, ATImPSL 250 mg/d 1–3 days, PSL 1 mg/kg/d, CyCCR Scr was normalized within D47
49.Klomjit et al.682/FUSA (Caucasian)NAmRNAModernaSecondD280.82.5NAAKI, hematuria, proteinuriaMPO-ANCA vasculitisHigh-dose steroid, RTXPR Scr was 2.3 mg/dl during last follow-up
50.Avalos et al.9474/FUSAHT, T2DM, hypothyroidismmRNAPfizer-BioNTechSecondD14Normal4.78 (D14)NAMalaise, fatigue, chest pain, lower extremity rashMPO-ANCA vasculitismPSL 1 g/d 1–5 days, PSL 1 mg/kg, RTXNA
51.Shakoor et al.10578/FUSAT2DM, HT, atrial fibrillationmRNAPfizer-BioNTechFirst<D70.771.31 (D16)NANausea, vomiting, diarrheaAKINoneCR improved spontaneously
mRNAPfizer-BioNTechSecondD6Normal3.54 (D6)NALethargy, nausea, vomiting, diarrheaMPO-ANCA vasculitismPSL 1 g 1–3 days, PSL 1 mg/kg/d, RTXR Scr was 1.71 mg/dl at 1-month follow-up
52.So et al.10642/MKoreaTuberculosismRNAPfizer-BioNTechSecond<D70.973.05 (D21)NAGeneral weakness, shortness of breath, edema, gross hematuriaMPO-ANCA vasculitismPSL 500 mg 1–3 days, PLEX, RTX, PSL 1 mg/kg/dPR Scr was 1.51 mg/dl at D51
53.Dube et al.10729/FUSACongenital diffuse cystic lung diseasemRNAPfizer-BioNTechSecondD160.81.25 (D16) 1.91 (D49)NormalNAMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL 1 mg/kg/d, RTX, CyCCR Scr was 1.01 mg/dl at D133
54.Ma et al.10870/FChina (Asian)HT, hyperlipidemia, kidney stonesInactivatedSinovac Life ScienceFirstD10.545.43 (D20) 6 (D22)NAPoor appetite, nausea, fatigue, foamy urineMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL, CyCCR Scr was 1.1 mg/dl at D322
55.Kim et al.10972/FKoreaNAmRNAModernaThirdD180.811.25 (D18) 4.71 (peak)IAnorexia, abdominal pain, febrile sensationMPO-ANCA vasculitismPSL, CyCPR Scr was 1.34 mg/dl after 2 months of diagnosis
56.Suzuki et al.11072/MJapanProstatic hypertrophymRNAPfizer-BioNTechSecondD1Normal5.0 (D14) 8.51 (D17)NormalFever, progressive fatigue, loss of appetiteMPO-ANCA vasculitismPSL, PSL, HD, RTXPR Scr was 2.2 mg/dl at D65
57.Schaubschlager et al.5877/FUASHT, fibrillation, monoclonal gammopathymRNAModernaSecondD600.92.9 (peak)NASymptomatic bradycardia, hematuriaMPO-ANCA vasculitisNANA
58.Gillion et al.6977/MBelgiumNoneVectorAstraZenecaFirstD281.22.7NAFever, night sweatANCA-negative CrGNmPSLCR Scr was normalized within D56
59.Kim et al.7016/FKoreaNonemRNAPfizer-BioNTechSecondD14Normal9.57IDyspnea, headache, edemaANCA-negative CrGNHD, mPSL, MMFNR remained in CKD stage
60.Wisnik-Rainville et al.9524/MHispanic-AmericanHT, sleep apneaVectorJanssenFirstD71.06.5 (D14)IIChest pain, headache, dizziness, vision changesFocal CrGNHD, ACEi, corticosteroids, RTX, CycNR Scr was not improvement in several months
61.Tan et al.7370s/FUKT2DM (2004), HTVectorAstraZenecaFirstD20.94.7 (D8) 6.99 (peak)NAWeakness, headacheAINPSLPR Scr was 2.07 mg/dl at D56
62.Soma et al.9654/MJapanT2DMmRNAPfizer-BioNTechFourthD31–1.34.72 (D5) 9 (peak)IFatigueATIPSLCR Scr was 1 mg/dl at D19
63.Mira et al.7445/FPortugal (Caucasian)Total thyroidectomymRNAPfizer-BioNTechSecondD10.8518.4 (D8)NormalAnorexia, nausea, vomiting, urine output reductionAIN, ATIHD, mPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dCR Scr was 1.02 mg/dl at D37
64.Unver et al.7567/FTurkeyT2DM, MCD in PRInactivatedSinovac Life ScienceSecondD100.84.2 (D26)IIIEdema, headacheAIN, ATImPSL 500 mg/d 1–3 days, PSL 1 mg/kg/d, cyclosporine APR Scr was 1.12 mg/dl at D60, UTP was 3 g/d at D115
65.Lim et al.4744/MKoreaT2DM, chronic hepatitis B infection, hyperlipidemiamRNAModernaFirstD10.914.13 (D7) 4.94 (D21)NAGastrointestinal discomfort, anorexiaATNHigh-dose steroidPR Scr was 1.89 mg/dl, UPCR was 0.3 g/g at D42
66.Choi et al.7617/MKoreaNAmRNAPfizer-BioNTechSecondD1Normal3.1 (D3)NAEpigastric pain, poor oral intake, feverATNSupportive careCR Scr was 1.0 mg/dl at D12
67.Choi et al.7612/MKoreaNonemRNAPfizer-BioNTechSecondD2Normal2.28 (D21)NormalAnorexia, vomitingATNOral steroidCR Scr was 1.1 mg/dl at D38
68.Schaubschlager et al.5860/FUSAHT, hypothyroidism, diffuse large B-cell lymphomamRNAModernaSecondD280.94.9 (peak)NAFatigue, shortness of breathATIN, ICGN with endocapillary hypercellularityNANA
69.Lim et al.4777/FKoreaT2DM, chronic hepatitis B, hepatocellular carcinomamRNAPfizer-BioNTechSecondD10.9810.67 (D7) 11.15 (D14)NASevere nausea and vomitingATN with myoglobin tubular castsHDPR Scr was 2.12 mg/dl, within 4 months
70.Missoum et al.9058/MAlgeriaHTInactivatedSinovac Life ScienceFirstD9Normal8.9NAFever, arthralgias, purpuraLeukocytoclastic vasculitis ATNHD, prednisonePR Scr was 2.8 mg/dl at D90
71.Al-Rasbi et al.8737/MOmanNonemRNAPfizer-BioNTechFirstD12Normal3.05 (D15) 5.75 (D18)NAArm pain, swelling and paresthesiaNon-oliguric AKI, myositis, thrombocytopenia, myocarditisIVIG 1 g/kg/d, mPSL 1 g/dCR
72.Unger et al.8869/FUSAT2DM, HT, morbid obesity, hyperlipidemia, hypothyroidismmRNAPfizer-BioNTechThirdD20.81.77 (D8) 2.73 (D10)NADark brown and frothy urineRhabdomyolysisIntravenous and oral hydrationCR Scr was 1.16 mg/dl at D77
73.Banamah et al.8958/FKingdom of Saudi ArabiaSchizophrenia (>10 years)mRNAPfizer-BioNTechThirdD1NA2.7 (D1) 8.5 (D8)NAWeakness, lethargy, dark brown urineRhabdomyolysisRRTCR Scr was 1.32 mg/dl at D42
74.Al Rawahi et al.7764/MSultanate of OmanHT, hyperlipidemiaVectorAstraZenecaFirstD7NA1.18 (D7)IFever, lethargy, abdominal painaTTP, VITTArgatroban, fondaparinux, hydrocortisone, IVIGCR renal function improved at D15
75.Yocum et al.7862/FUSAHyperlipidemia, GERD, hypothyroidism, HTVectorJanssenFirstD37NA2.19 (D37) 6 (D38)IIIAltered mental statusaTTP, VITT,PLEX, HD, mPSL, packed RBCsNA
76.Osmanodja et al.7925/MGermanyNonemRNAModernaFirstD2NA1.5 (D13)NAFever, headache, petechiaeaTTPPLEX, PSL 250 mg 1–3 days, caplacizumabCR Scr was 1 mg/dl at D27
77.Alislambouli et al.8061/MKorean-AmericanNAmRNAPfizer-BioNTechFirstD5NA1.57 (D5)NAFever, confusion, headache, emesis, ecchymosisaTTPPLEX, mPSL 1 g 1–3 days, RTXCR rapid and excellent response
78.Yoshida et al.8157/MJapanNonemRNAPfizer-BioNTechFirstD7NA1.57 (D14)NAFatigue, loss of appetite, jaundiceaTTPPLEX, PSL, RTXCR in good condition at D48
79.Ruhe et al.8284/FGermanyNAmRNAPfizer-BioNTechFirstD16NA1.95 (D16)IIIPartial hemiplegia, petechiaeaTTPPLEX, RTX, corticosteroidCR Scr was 0.6 mg/dl at D34
80.Chamarti et al.8380/MHispanicHT, T2DM, hyperlipidemia, gout, IDAmRNAPfizer-BioNTechSecondD12NA2.4 (D14)IGeneralized weakness, malaiseaTTPPLEX, packed RBCs, platelets, prednisoneCR Scr was 1 mg/dl at D30
81.Lim et al.4769/FKoreaT2DMVectorAstraZenecaFirstD20.83.69 (D14)NAGeneral weakness, gastrointestinal discomfortaTTPNoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
82.Claes et al.8438/FBelgiumNAmRNAModernaThirdD10.866.4 (D9)IIIHeadache, general malaiseaHUSHD, PLEX, eculizumabCR Scr was 1.04 mg/dl at D90
83.Tawhari et al.9738/MSaudi ArabiaNoneVectorAstraZenecaFirstD7Normal10.8 (D7)IIShortness of breath, body weakness, fatigabilityaHUSHD, PLEX, RTX, prednisoneNR dependent on HD
84.Kim et al.8666/FKoreaNoneVectorAstraZenecaSecondD500.661.81NAFoamy urine, edemaClass III LNmPSL, CyC, hydroxychloroquine, prednisoneCR Scr was 0.93 mg/dl after 10-day treatment
85.Vnučá et al.9125/FSlovakiaT1DM, HT, KT, autoimmune thyroiditis, COVID-19 infectionVectorAstraZenecaFirstD140.9510.74 (D24)NormalFatigue, general weakness, vomiting, with the inability to eat or drinkAcute kidney rejectionmPSL, PLEX, IVIG, HD, RTXNR Scr was 5.4 mg/dl at D98
86.Nune et al.11144/FUKMild asthmamRNAPfizer-BioNTechFirstD2Normal2.18 (D2)HypotensiveLeft arm pain, vomiting, loose stools, chest tightness, skin rashMultisystem inflammatory syndromemPSL 1 g/d 1–3 days, antibioticsPR
Relapsed kidney disease with AKI
1.Mancianti et al.5339/MItaly Caucasian)MCD in remission for 37 yearsmRNAPfizer-BioNTechFirstD30.91.8 (D8)NAEdemaMCDPSL 1 mg/kg/dCR
2.Klomjit et al.667/FUSA (Caucasian)MCDmRNAModernaSecondD2111.6NAEdemaMCDHigh-dose steroid, RTXCR Scr was 1.1 mg/dl at D40
3.Aydin et al.11266/FTurkeyHyperlipidemia, DM, HT, MN in CR for 8 yearsInactivatedSinovac Life ScienceFirstD14Normal2.78 (D14)NAEdemaMN (anti-PLA2R 1:120.53)NANA
4.Fernández et al.5759/FArgentinaHT, obesity, MPG (2015, CR)VectorGamaleyaFirstD14NA1.24IFoamy urine, edemaFocal proliferative GNCorticosteroidsPR
5.Huang et al.5654/MChinaAsthma, FSGS (2014, CR)VectorAstraZenecaSecondD3029.6 16.42 (peak)NAEdema, fatigue, poor appetiteFSGS, AINPSL, HDNR
6.Hanna et al.6113/MUSA (Caucasian)IgAN, T1DMmRNAPfizer-BioNTechSecond<D10.541.31 (D2)NAGross hematuria, vomitingIgAN (M0E0S0T0C0)ConservativeCR hematuria and Scr resolved within D6
7.Perrin et al.6741/FFranceIgAN, KTmRNAPfizer-BioNTechFirstD2NAScr transiently increasedNAGross hematuriaIgANConservativeCR spontaneously resolved
8.Watanabe et al.6854/FUSA (Caucasian)IgANmRNAModernaSecondD21.23.04 (D7)NormalGross hematuriaIgANPrednisone 60 mg/dCR Scr was 1.07 mg/dl at D90
9.Sekar et al.8530/MAmerican (African)LN (WHO Class 2 and 5 in 2019), COVID-19 infection 6 months priormRNAModernaFirstD30.792.81 (D3) 3.19 (D4)NAFevers, weakness, headacheLNMMF, hydroxychloroquine, prednisoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
10.Schaubschlager et al.5839/FUSASLE, hashimoto thyroiditis, LN class V (2015)mRNAModernaSecondD230.9–1.15.6 (peak)NALower extremity cellulitis, deep vein thrombosisATIN, ICGN with class II LNNANA
11.Martinez-Zayas et al.11353/MUSAANCA-associated vasculitis (2008), HIVmRNAPfizer-BioNTechSecondFew weeks0.92.49IFever, fatigue, shortness of breathICGNmPSL 1 g/d 1–3 days, RTX, prednisonePR
CaseAuthorsAge/sexCountry (race)Medical historyVaccine
Onset (day)Baseline-Scr (mg/dl)After vaccine-Scr (mg/dl) (day)Newly HT/worseSymptomsDiagnosisTreatmentsOutcomes
TypeManufacturerOnset after which dose
New-onset kidney disease with AKI
1.Leclerc et al.4271/MCanadaDyslipidemia treated with rosuvastatinVectorAstraZenecaFirstD10.710.6 (D14)IIIEdema, oligoanuricMCDHD, mPSL 1 g/d 1–3 days, PSL 60 mg/dCR Scr was 1.2 mg/dl, UPCR 28 mg/mmol at D81
2.Lim et al.4351/MKoreaNoneVectorJanssenFirstD7NA1.96 (D34) 2.37 (D36)IEdema, foamy urineMCDmPSL 64 mg/dCR Scr was 0.95 mg/dl, UPCR was 0.2 g/g at D57
3.Lebedev et al.450/MIsraelNonemRNAPfizer-BioNTechFirstD40.782.31 (D10)IIIEdema, diarrhea, abdominal painMCD with ATIPSL 80 mg/dCR Scr was 0.97 mg/dl, UACR was 155 mg/g at D37
4.Maas et al.4480s/MNetherlandsVTEmRNAPfizer-BioNTechFirstD7NA1.43 (D7)IIEdemaMCD with ATIPSL 80 mg/dCR UPCR was 0.68 g/g after 10 days of PSL
5.D’Agati et al.577/MUSA (Caucasian)T2DMmRNAPfizer-BioNTechFirstD71.0–1.32.33 (D14)IEdemaMCD with ATImPSL 1 g/d 1–3 days, PSL 60 mg/dNR Scr was 3.74 mg/dl, UTP was 18.8 g/d at D35
6.Holzworth et al.4563/FUSAHTmRNAModernaFirst<D70.71.48 (>D28)IIIEdema, dyspneaMCD with ATI and AINmPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dNA
7.Weijers et al.4661/FNetherlandsAIH, hypothyroidismmRNAPfizer-BioNTechFirstD10.7–0.81.47 (D4)NAEdemaMCDHD, steroids 1 mg/kg/dCR Scr was <1 mg/dl at D77, UTP was 0 g/d at D58
8.Lim et al.4751/MKoreaNoneVectorJanssenFirstD7Normal1.54 (D21) 1.96 (D33)NAEdemaMCDHigh-dose steroidCR was achieved after 3 weeks of treatment
9.Park et al.4834/MKoreaNAmRNAModernaSecondD2NA0.86 (D3) 5.38 (D7)NAPeriocular edemaMCDPSL 60 mg/dCR Scr was 0.89 mg/dl, UPCR was 0.28 g/g at D77
10.Gulumsek et al.9258/MTurkeyDMmRNAPfizer-BioNTechSecondD7Normal2.4 (D7)+EdemaMCDmPSL 80 mg/dCR Scr was 0.64 mg/dl, UTP was 0.067 g/d within 30 days
11.Annicchiarico Petruzzelli et al.4912/MItalyNonemRNAPfizer-BioNTechFirstD70.781.3 (D9) 4 (D13)IEdema, headacheMCD, tubular obstructionRRT, pulse mPSL,CR was achieved at 12 weeks
12.Baskaran et al.5055/MAustraliaNAVectorAstraZenecaSecondD7NA7.16NAEdemaMCD with ATI and AINHigh-dose steroidNA
13.Hanna et al.5160/MCanadaNonemRNAPfizer-BioNTechFirstD100.891.34 (D45)IIEdema, dyspneaMCD with ATIPSL 80 mg/dCR Scr was 1.03 mg/dl at 11 weeks
14.Psyllaki et al.5265/MGreece (Caucasian)NAmRNAPfizer-BioNTechFirstD10NAeGFR 52 mL/min/1.73 m2NAEdemaMCD with ATIPulse mPSL, PSL, cyclosporineCR was achieved within 10 days
15.Klomjit et al.683/MUSA (Caucasian)NAmRNAModernaSecondD281.192.19NAAKIMCD, ATNHigh-dose steroidPR Scr was 1.2 mg/dl during last follow-up
16.Da et al.5470/MSingaporeEdema after first vaccinemRNAPfizer-BioNTechSecondD1NA1.28IEdemaMN (anti-PLA2R-)Irbesartan, frusemide, warfarinNR within D60
17.Thammathiwat et al.5553/MThailandEdema and foamy urine after first vaccineInactivatedSinovac Life ScienceSecondD2NA1.5 2.4 (peak)IIEdemaSecondary MN (anti-PLA2R-)mPSL, PSL, CyC, ACEiCR
18.Schaubschlager et al.5863/MUSAHypothyroid, valvular heart disease, aortic valve replacement, SLE without renal involvementmRNAPfizer-BioNTechSecondD140.9–1.22.1 (peak)NANephrotic proteinuria, edemaSevere podocytopathyNANA
19.Kudose et al.5950/FUSA (Caucasian)HT, obesity, APSmRNAModernaSecondD21.31.7NAGross hematuria, fever, body achesIgAN (M1E0S1T1C1)ConservativeCR hematuria resolved within D5
20.Tan et al.6041/FChineseGDMmRNAPfizer-BioNTechSecondD1Normal1.73 (D2)IGross hematuria, headache, myalgiaIgAN with fibrocellular and fibrous crescentsPulse mPSL, PSL, CyCNA
21.Hanna et al.6117/MUSA (Caucasian)Foamy urinemRNAPfizer-BioNTechSecond<D1Normal1.78 (D6)IGross hematuriaIgAN (M1E1S1T1C1)Pulse mPSLPR Scr improved (duration not reported)
22.Anderegg et al.6239/MSwitzerlandHTmRNAModernaSecondimmediatelyNAAKINAFlu-like symptoms, fever, macrohematuriaSevere crescentic IgANHigh-dose glucocorticoids, CyCCR Scr was normalized within several weeks
23.Klomjit et al.638/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD141.31.6NAGross hematuriaIgANConservativeNA
24.Klomjit et al.644/MUSA (Caucasian)NAmRNAModernaFirstD141.12.5NAAKIIgAN, AINHigh-dose steroidNR Scr was 3.6 mg/dl during last follow-up
25.Klomjit et al.666/MUSA (Caucasian)NAmRNAModernaFirstD141.11.5NAGross hematuriaIgANPSLPR Scr was 1.4 mg/dl during last follow-up
26.Klomjit et al.662/MUSA (Caucasian)NAmRNAPfizer-BioNTechSecondD421.02.2NAAKIIgANConservativePR Scr was 2.0 mg/dl during last follow-up
27.Ritter et al.6320/MSwitzerland (Caucasian)Allergic rhinoconjunctivitsmRNAModernaSecond1NormalAKINAFever, chills, body aches, gross hematuria, dizzinessIgANRASBCR Scr normalized at D49
28.Morimoto et al.9377/MJapanese (Asian)HT, atrial fibrillationmRNAModernaThirdD901.371.67 (D60) 16.29 (D90)IIEvaluation of anorexia, pruritus, lower-extremity edemaIgAN with MPGN-like changes, grade III (A/C) M1E1S1T1C1HD, 3-day pulse mPSL, PSL 50 mg/dNR dependent on HD
29.Chen et al.6455/MChina (Asian)Chronic hepatitis B infectionmRNAModernaFirstD14Normal1.94NANausea, vomiting, general malaiseATN, IgAN (M1E0S0T0C0)HDNR Scr was 4.0 mg/dl at D104
30.Ritter et al.6369/MSwitzerland (Caucasian)Hypertensive cardiomyopathy, BPH, depressionmRNAPfizer-BioNTechSecondD33NAAKI+Gross hematuria, fatigue, loss of appetiteIgAN, ANCASteroids, CyC, HDNR dependent on HD at D78
31.Fernández et al.5738/FArgentinaObesityinactivatedSinopharmFirstD7NA1.74IIAsthenia, foamy urineIgAN with crescent, IFTACorticosteroids, CyCPR
32.Fernández et al.5753/MArgentinaHT, overweightVectorAstraZenecaFirstD10NA3.07IAsthenia, foamy urine, edemaIgAN with fibrocellular crescents, ATI, IFTACorticosteroids, CyCPR
33.Park et al.6550/MUSAHT, CKDmRNAModernaSecondD11.171.54NAHematuria, foamy urineIgAN with 13% active crescentsRAASiPR Scr was 1.24 mg/dl during last follow-up
34.Park et al.6567/MUSAHT, CKDmRNAModernaFirstD301.22.9NAHematuria, foamy urineIgA vasculitisPrednisone 40 mg/d for 1 weekPR Scr was 1.4 mg/dl during last follow-up
35.Sugita et al.6667/FJapanHTmRNAPfizer-BioNTechSecondD1Normal0.83 (D7) 2.2 (D15)NARash, edema, gross hematuriaIgA vasculitismPSL 0.5 g 1–3 days, PSL, CyC, azathioprinePR Scr was 0.65 mg/dl, UTP was 1 g/d at D180
36.Tan et al.6060/FMalayHyperlipidemiamRNAPfizer-BioNTechSecondD1Normal6.11 (D39)IIIGross hematuriaAnti-GBM nephritisPulse mPSL, PSL, CyC, PLEXNA
37.Sacke et al.71Older/FUSANonemRNAModernaSecondD14Normal7.8NAFever, gross hematuria, anorexia, nauseaAnti-GBM with mesangial IgA depositsmPSL, CyC, PLEXNR remained HD dependent
38.Coorey et al.7269/FAustraliaHT, hypercholesterolemiaVectorAstraZenecaFirstD560.5613.3NALethargy, anuriaAnti-GBM nephritisHD, pulse mPSL, PSL, CyC, PLEXNR remained HD dependent
39.Coorey et al.7272/FAustraliaColorectal cancerVectorAstraZenecaSecondD21Normal13.6NAMalaise, diarrheaAnti-GBM nephritisHD, high-dose steroids, CyC, PLEX, RTXNR remained HD dependent
40.Klomjit et al.677/MUSA (Caucasian)NAmRNAPfizer-BioNTechFirstD711.8+HTAtypical anti-GBM nephritisPSL, MMFNR Scr was 2.9 mg/dl during last follow-up
41.Zamoner et al.9858/FBrazilHyperthyroidism (2006)VectorAstraZenecaFirstD21.02.2 3.3 (peak)+Fatigue, paleness, arthralgia, foamy urineANCA-associated pauci-immune CrGNmPSL, CyC, azathioprinePR Scr was 1.87 mg/dl, UTP was 0.5 g during last follow-up
42.Bansal et al.9967/FIndia (South-Asian)HTInactivatedBBV152SecondD141.22.8 (D21) 6.4 (D28)IILassitude, swellingANCA-associated pauci-immune CrGNHD, mPSL, CyCNR Scr was 3.8 mg/dl during last follow-up
43.Sekar et al.10052/MUSA (Caucasian)HTmRNAModernaSecondD11.118.41 (D14)NAHeadache, weaknessPR3-ANCA vasculitisRTX, CyC, PSL, HDNR remained HD dependent
44.Anderegg et al.6281/MSwitzerlandSustained flu-like symptoms after first vaccinemRNAModernaSecond<D1NAAKINAFlu-like symptoms worsenedPR3-ANCA vasculitisHigh-dose glucocorticoids, CyC, PLEXPR renal function improved within D21
45.Feghali et al.10158/MUSA (Caucasian)NonemRNAModernaSecondD4NA4.1NAHematuria, proteinuriaPR3-ANCA vasculitismPSL 1 g 1–3 days, PSL 60 mg/kg/d, RTX, CyC, PLEXPR Scr was 1.5 mg/dl after 10 weeks of diagnosis
46.Villa et al.10263/MSpainNoneVectorAstraZenecaFirstD2Normal2.9 (D7)NAFlu-like syndrome, hemoptysisMPO-ANCA vasculitisHigh-dose glucocorticoids, CyCNR Scr was 2.08 mg/dl at D49
47.Noel et al.10362/FUSASystemic sclerosis, T2DMmRNAPfizer-BioNTechSecondD28<15.18NAEdemaMPO-ANCA vasculitismPSL, CyC, MMFNR Scr was 2.6 mg/dl at 8 months
48.Hakroush et al.10479/FGermany (Caucasian)HT, degenerative disc diseasemRNAPfizer-BioNTechSecondD140.711.38 (D14) 6.57 (D24)NAWeakness, upper thigh painMPO-ANCA vasculitis, ATImPSL 250 mg/d 1–3 days, PSL 1 mg/kg/d, CyCCR Scr was normalized within D47
49.Klomjit et al.682/FUSA (Caucasian)NAmRNAModernaSecondD280.82.5NAAKI, hematuria, proteinuriaMPO-ANCA vasculitisHigh-dose steroid, RTXPR Scr was 2.3 mg/dl during last follow-up
50.Avalos et al.9474/FUSAHT, T2DM, hypothyroidismmRNAPfizer-BioNTechSecondD14Normal4.78 (D14)NAMalaise, fatigue, chest pain, lower extremity rashMPO-ANCA vasculitismPSL 1 g/d 1–5 days, PSL 1 mg/kg, RTXNA
51.Shakoor et al.10578/FUSAT2DM, HT, atrial fibrillationmRNAPfizer-BioNTechFirst<D70.771.31 (D16)NANausea, vomiting, diarrheaAKINoneCR improved spontaneously
mRNAPfizer-BioNTechSecondD6Normal3.54 (D6)NALethargy, nausea, vomiting, diarrheaMPO-ANCA vasculitismPSL 1 g 1–3 days, PSL 1 mg/kg/d, RTXR Scr was 1.71 mg/dl at 1-month follow-up
52.So et al.10642/MKoreaTuberculosismRNAPfizer-BioNTechSecond<D70.973.05 (D21)NAGeneral weakness, shortness of breath, edema, gross hematuriaMPO-ANCA vasculitismPSL 500 mg 1–3 days, PLEX, RTX, PSL 1 mg/kg/dPR Scr was 1.51 mg/dl at D51
53.Dube et al.10729/FUSACongenital diffuse cystic lung diseasemRNAPfizer-BioNTechSecondD160.81.25 (D16) 1.91 (D49)NormalNAMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL 1 mg/kg/d, RTX, CyCCR Scr was 1.01 mg/dl at D133
54.Ma et al.10870/FChina (Asian)HT, hyperlipidemia, kidney stonesInactivatedSinovac Life ScienceFirstD10.545.43 (D20) 6 (D22)NAPoor appetite, nausea, fatigue, foamy urineMPO-ANCA vasculitismPSL 500 mg 1–3 days, PSL, CyCCR Scr was 1.1 mg/dl at D322
55.Kim et al.10972/FKoreaNAmRNAModernaThirdD180.811.25 (D18) 4.71 (peak)IAnorexia, abdominal pain, febrile sensationMPO-ANCA vasculitismPSL, CyCPR Scr was 1.34 mg/dl after 2 months of diagnosis
56.Suzuki et al.11072/MJapanProstatic hypertrophymRNAPfizer-BioNTechSecondD1Normal5.0 (D14) 8.51 (D17)NormalFever, progressive fatigue, loss of appetiteMPO-ANCA vasculitismPSL, PSL, HD, RTXPR Scr was 2.2 mg/dl at D65
57.Schaubschlager et al.5877/FUASHT, fibrillation, monoclonal gammopathymRNAModernaSecondD600.92.9 (peak)NASymptomatic bradycardia, hematuriaMPO-ANCA vasculitisNANA
58.Gillion et al.6977/MBelgiumNoneVectorAstraZenecaFirstD281.22.7NAFever, night sweatANCA-negative CrGNmPSLCR Scr was normalized within D56
59.Kim et al.7016/FKoreaNonemRNAPfizer-BioNTechSecondD14Normal9.57IDyspnea, headache, edemaANCA-negative CrGNHD, mPSL, MMFNR remained in CKD stage
60.Wisnik-Rainville et al.9524/MHispanic-AmericanHT, sleep apneaVectorJanssenFirstD71.06.5 (D14)IIChest pain, headache, dizziness, vision changesFocal CrGNHD, ACEi, corticosteroids, RTX, CycNR Scr was not improvement in several months
61.Tan et al.7370s/FUKT2DM (2004), HTVectorAstraZenecaFirstD20.94.7 (D8) 6.99 (peak)NAWeakness, headacheAINPSLPR Scr was 2.07 mg/dl at D56
62.Soma et al.9654/MJapanT2DMmRNAPfizer-BioNTechFourthD31–1.34.72 (D5) 9 (peak)IFatigueATIPSLCR Scr was 1 mg/dl at D19
63.Mira et al.7445/FPortugal (Caucasian)Total thyroidectomymRNAPfizer-BioNTechSecondD10.8518.4 (D8)NormalAnorexia, nausea, vomiting, urine output reductionAIN, ATIHD, mPSL 500 mg/d 1–3 days, PSL 1 mg/kg/dCR Scr was 1.02 mg/dl at D37
64.Unver et al.7567/FTurkeyT2DM, MCD in PRInactivatedSinovac Life ScienceSecondD100.84.2 (D26)IIIEdema, headacheAIN, ATImPSL 500 mg/d 1–3 days, PSL 1 mg/kg/d, cyclosporine APR Scr was 1.12 mg/dl at D60, UTP was 3 g/d at D115
65.Lim et al.4744/MKoreaT2DM, chronic hepatitis B infection, hyperlipidemiamRNAModernaFirstD10.914.13 (D7) 4.94 (D21)NAGastrointestinal discomfort, anorexiaATNHigh-dose steroidPR Scr was 1.89 mg/dl, UPCR was 0.3 g/g at D42
66.Choi et al.7617/MKoreaNAmRNAPfizer-BioNTechSecondD1Normal3.1 (D3)NAEpigastric pain, poor oral intake, feverATNSupportive careCR Scr was 1.0 mg/dl at D12
67.Choi et al.7612/MKoreaNonemRNAPfizer-BioNTechSecondD2Normal2.28 (D21)NormalAnorexia, vomitingATNOral steroidCR Scr was 1.1 mg/dl at D38
68.Schaubschlager et al.5860/FUSAHT, hypothyroidism, diffuse large B-cell lymphomamRNAModernaSecondD280.94.9 (peak)NAFatigue, shortness of breathATIN, ICGN with endocapillary hypercellularityNANA
69.Lim et al.4777/FKoreaT2DM, chronic hepatitis B, hepatocellular carcinomamRNAPfizer-BioNTechSecondD10.9810.67 (D7) 11.15 (D14)NASevere nausea and vomitingATN with myoglobin tubular castsHDPR Scr was 2.12 mg/dl, within 4 months
70.Missoum et al.9058/MAlgeriaHTInactivatedSinovac Life ScienceFirstD9Normal8.9NAFever, arthralgias, purpuraLeukocytoclastic vasculitis ATNHD, prednisonePR Scr was 2.8 mg/dl at D90
71.Al-Rasbi et al.8737/MOmanNonemRNAPfizer-BioNTechFirstD12Normal3.05 (D15) 5.75 (D18)NAArm pain, swelling and paresthesiaNon-oliguric AKI, myositis, thrombocytopenia, myocarditisIVIG 1 g/kg/d, mPSL 1 g/dCR
72.Unger et al.8869/FUSAT2DM, HT, morbid obesity, hyperlipidemia, hypothyroidismmRNAPfizer-BioNTechThirdD20.81.77 (D8) 2.73 (D10)NADark brown and frothy urineRhabdomyolysisIntravenous and oral hydrationCR Scr was 1.16 mg/dl at D77
73.Banamah et al.8958/FKingdom of Saudi ArabiaSchizophrenia (>10 years)mRNAPfizer-BioNTechThirdD1NA2.7 (D1) 8.5 (D8)NAWeakness, lethargy, dark brown urineRhabdomyolysisRRTCR Scr was 1.32 mg/dl at D42
74.Al Rawahi et al.7764/MSultanate of OmanHT, hyperlipidemiaVectorAstraZenecaFirstD7NA1.18 (D7)IFever, lethargy, abdominal painaTTP, VITTArgatroban, fondaparinux, hydrocortisone, IVIGCR renal function improved at D15
75.Yocum et al.7862/FUSAHyperlipidemia, GERD, hypothyroidism, HTVectorJanssenFirstD37NA2.19 (D37) 6 (D38)IIIAltered mental statusaTTP, VITT,PLEX, HD, mPSL, packed RBCsNA
76.Osmanodja et al.7925/MGermanyNonemRNAModernaFirstD2NA1.5 (D13)NAFever, headache, petechiaeaTTPPLEX, PSL 250 mg 1–3 days, caplacizumabCR Scr was 1 mg/dl at D27
77.Alislambouli et al.8061/MKorean-AmericanNAmRNAPfizer-BioNTechFirstD5NA1.57 (D5)NAFever, confusion, headache, emesis, ecchymosisaTTPPLEX, mPSL 1 g 1–3 days, RTXCR rapid and excellent response
78.Yoshida et al.8157/MJapanNonemRNAPfizer-BioNTechFirstD7NA1.57 (D14)NAFatigue, loss of appetite, jaundiceaTTPPLEX, PSL, RTXCR in good condition at D48
79.Ruhe et al.8284/FGermanyNAmRNAPfizer-BioNTechFirstD16NA1.95 (D16)IIIPartial hemiplegia, petechiaeaTTPPLEX, RTX, corticosteroidCR Scr was 0.6 mg/dl at D34
80.Chamarti et al.8380/MHispanicHT, T2DM, hyperlipidemia, gout, IDAmRNAPfizer-BioNTechSecondD12NA2.4 (D14)IGeneralized weakness, malaiseaTTPPLEX, packed RBCs, platelets, prednisoneCR Scr was 1 mg/dl at D30
81.Lim et al.4769/FKoreaT2DMVectorAstraZenecaFirstD20.83.69 (D14)NAGeneral weakness, gastrointestinal discomfortaTTPNoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
82.Claes et al.8438/FBelgiumNAmRNAModernaThirdD10.866.4 (D9)IIIHeadache, general malaiseaHUSHD, PLEX, eculizumabCR Scr was 1.04 mg/dl at D90
83.Tawhari et al.9738/MSaudi ArabiaNoneVectorAstraZenecaFirstD7Normal10.8 (D7)IIShortness of breath, body weakness, fatigabilityaHUSHD, PLEX, RTX, prednisoneNR dependent on HD
84.Kim et al.8666/FKoreaNoneVectorAstraZenecaSecondD500.661.81NAFoamy urine, edemaClass III LNmPSL, CyC, hydroxychloroquine, prednisoneCR Scr was 0.93 mg/dl after 10-day treatment
85.Vnučá et al.9125/FSlovakiaT1DM, HT, KT, autoimmune thyroiditis, COVID-19 infectionVectorAstraZenecaFirstD140.9510.74 (D24)NormalFatigue, general weakness, vomiting, with the inability to eat or drinkAcute kidney rejectionmPSL, PLEX, IVIG, HD, RTXNR Scr was 5.4 mg/dl at D98
86.Nune et al.11144/FUKMild asthmamRNAPfizer-BioNTechFirstD2Normal2.18 (D2)HypotensiveLeft arm pain, vomiting, loose stools, chest tightness, skin rashMultisystem inflammatory syndromemPSL 1 g/d 1–3 days, antibioticsPR
Relapsed kidney disease with AKI
1.Mancianti et al.5339/MItaly Caucasian)MCD in remission for 37 yearsmRNAPfizer-BioNTechFirstD30.91.8 (D8)NAEdemaMCDPSL 1 mg/kg/dCR
2.Klomjit et al.667/FUSA (Caucasian)MCDmRNAModernaSecondD2111.6NAEdemaMCDHigh-dose steroid, RTXCR Scr was 1.1 mg/dl at D40
3.Aydin et al.11266/FTurkeyHyperlipidemia, DM, HT, MN in CR for 8 yearsInactivatedSinovac Life ScienceFirstD14Normal2.78 (D14)NAEdemaMN (anti-PLA2R 1:120.53)NANA
4.Fernández et al.5759/FArgentinaHT, obesity, MPG (2015, CR)VectorGamaleyaFirstD14NA1.24IFoamy urine, edemaFocal proliferative GNCorticosteroidsPR
5.Huang et al.5654/MChinaAsthma, FSGS (2014, CR)VectorAstraZenecaSecondD3029.6 16.42 (peak)NAEdema, fatigue, poor appetiteFSGS, AINPSL, HDNR
6.Hanna et al.6113/MUSA (Caucasian)IgAN, T1DMmRNAPfizer-BioNTechSecond<D10.541.31 (D2)NAGross hematuria, vomitingIgAN (M0E0S0T0C0)ConservativeCR hematuria and Scr resolved within D6
7.Perrin et al.6741/FFranceIgAN, KTmRNAPfizer-BioNTechFirstD2NAScr transiently increasedNAGross hematuriaIgANConservativeCR spontaneously resolved
8.Watanabe et al.6854/FUSA (Caucasian)IgANmRNAModernaSecondD21.23.04 (D7)NormalGross hematuriaIgANPrednisone 60 mg/dCR Scr was 1.07 mg/dl at D90
9.Sekar et al.8530/MAmerican (African)LN (WHO Class 2 and 5 in 2019), COVID-19 infection 6 months priormRNAModernaFirstD30.792.81 (D3) 3.19 (D4)NAFevers, weakness, headacheLNMMF, hydroxychloroquine, prednisoneCR Scr was 0.65 mg/dl, UPCR was 1.0 g/g at D56
10.Schaubschlager et al.5839/FUSASLE, hashimoto thyroiditis, LN class V (2015)mRNAModernaSecondD230.9–1.15.6 (peak)NALower extremity cellulitis, deep vein thrombosisATIN, ICGN with class II LNNANA
11.Martinez-Zayas et al.11353/MUSAANCA-associated vasculitis (2008), HIVmRNAPfizer-BioNTechSecondFew weeks0.92.49IFever, fatigue, shortness of breathICGNmPSL 1 g/d 1–3 days, RTX, prednisonePR

ACEi, angiotensin-converting enzyme inhibitor; AKI, acute kidney injury; AIH, autoimmune hepatitis; AIN, acute interstitial nephritis; ATN, acute tubular necrosis; ATIN, acute tubulointerstitial nephritis; ATI, acute tubular injury; APS, antiphospholipid syndrome; ANCA, antineutrophil cytoplasmic autoantibodies; ANCA GN, antineutrophil cytoplasmic autoantibody-associated glomerulonephritis; anti-PLA2R, antiphospholipase A2 receptor; aHUS, atypical hemolytic uremic syndrome; aTTP, acquired thrombotic thrombocytopenic purpura; BPH, benign prostatic hyperplasia; CKD, chronic kidney disease; CyC, cyclophosphamide; CR, complete remission; CRF, chronic renal failure; CrGN, crescentic glomerulonephritis; DM, diabetes mellitus; eGFR, estimated glomerular filtration rate; F, female; FGN, fibrillary glomerulonephritis; FSGS, focal segmental glomerulosclerosis; GBM, glomerular basement membrane; GDM, gestational diabetes; GERD, gastroesophageal reflux disease; HD, hemodialysis; HIV, human immunodeficiency virus; HT, hypertension; ICGN, immune-complex glomerulonephritis; IDA, iron deficiency anemia; IFTA, interstitial fibrosis and tubular atrophy; IgAN, IgA nephropathy; ITT, vaccine-immune thrombotic thrombocytopenia; IVIG, intravenous immunoglobulins; KT, kidney transplantation; LN, lupus nephritis; M, male; MCD, minimal change disease; mPSL, methylprednisolone; MMF, mycophenolate mofetil; MN, membranous nephropathy; MPA, mycophenolic acid; NA, not applicable; NR, no response; PR, partial remission; PLEX, plasma exchange; PSL, prednisolone; R, response; RAAS, renin–angiotensin system; RASB, renin–angiotensin system blockade; RBCs, red blood cells; RTX, rituximab; RRT, renal replacement therapy; Scr, serum creatinine; TAC, tacrolimus; T1DM, Type 1 diabetes mellitus; T2DM, Type 2 diabetes mellitus; UACR, urinary albumin-creatinine ratio; UTP, 24-h urine protein; UPCR, urine protein-to-creatinine ratio; VITT, vaccine-induced immune thrombotic thrombocytopenia; VTE, venous thromboembolism.

Table 3.

Clinical characteristics of acute kidney injury post-COVID-19 vaccination

CharacteristicsTotalNew-onset kidney disease with AKIRelapsed kidney disease with AKI
Age (years)55.4 (12–84)56.5 (12–84)47.8 (13–67)
Male sex, n (%)55 (56.7)50 (58.1)5 (45.5)
Medical history, n (%)
 Hypertension29 (35.4)27 (38.0)2 (18.2)
 Diabetes mellitus17 (20.7)15 (21.1)2 (18.2)
 Thyroid dysfunction11 (13.4)10 (14.1)1 (9.0)
 Dyslipidemia10 (12.2)9 (12.7)1 (9.0)
Vaccine type, n (%)
 mRNA70 (72.2)62 (72.1)8 (72.7)
 Vector20 (20.6)18 (20.9)2 (18.2)
 Inactivated7 (7.2)6 (7.0)1 (9.1)
Which dose, n (%)
 First dose44 (45.4)39 (45.3)5 (45.5)
 Second dose47 (48.4)41 (47.7)6 (54.5)
 Third dose5 (5.2)5 (5.0)0 (0)
 Fourth dose1 (1.0)1 (1.0)0 (0)
Onset time after vaccination, n (%)
 ≤7 days56 (57.7)51 (59.3)5 (45.5)
 8–14 days20 (20.6)18 (20.9)2 (18.2)
 15–28 days12 (12.4)9 (10.5)3 (27.3)
 ≥29 days9 (9.3)8 (9.3)1 (9.1)
Symptoms, n (%)
 Nephrotic syndrome29 (29.9)24 (27.9)5 (45.5)
 Gastrointestinal discomfort25 (25.8)23 (26.4)2 (18.2)
 Hematuria21 (21.6)18 (20.5)3 (27.3)
 Fever13 (13.4)11 (12.5)2 (18.2)
 Headache12 (12.4)11 (12.5)1 (9.1)
Pathological type, n (%)
 Crescentic glomerulonephritis29 (29.9)29 (32.6)0 (0)
 Acute tubular injury23 (23.7)22 (24.7)1 (9.1)
 IgA nephropathy18 (18.6)15 (16.9)3 (27.3)
 ANCA-associated vasculitis17 (17.5)17 (19.1)0 (0)
 Minimal change disease17 (17.5)15 (16.9)2 (18.2)
 Thrombotic microangiopathy10 (10.3)10 (11.2)0 (0)
 Anti-GBM glomerulonephritis5 (5.2)5 (5.6)0 (0)
AKI stage, n (%)
 AKI Stage 130 (30.9)26 (29.2)4 (36.4)
 AKI Stage 222 (22.7)18 (20.2)4 (36.4)
 AKI Stage 345 (46.4)42 (47.2)3(27.3)
Treatment, n (%)
 Conservative medication11 (12.8)9 (11.7)2 (22.2)
 Steroids77 (89.5)70 (90.9)7 (77.8)
 Cyclophosphamide24 (27.9)24 (31.2)0 (0)
 Rituximab17 (19.8)15 (19.5)2 (22.2)
 Mycophenolate mofetil4 (4.7)3 (3.9)1 (11.1)
 Intravenous immunoglobulins3 (3.5)3 (3.9)0 (0)
 Plasma exchange18 (20.9)18 (23.4)0 (0)
 Renal replacement therapy22 (25.6)21 (27.3)1 (11.1)
Outcome, n (%)
 CR in 3 months42 (49.4)36 (47.4)6 (66.7)
 PR in 3 months24 (28.2)22 (28.9)2 (22.2)
 NR in 3 months19 (22.4)18 (23.7)1 (11.1)
 Dependent on hemodialysis7 (8.2)7 (9.2)0 (0)
CharacteristicsTotalNew-onset kidney disease with AKIRelapsed kidney disease with AKI
Age (years)55.4 (12–84)56.5 (12–84)47.8 (13–67)
Male sex, n (%)55 (56.7)50 (58.1)5 (45.5)
Medical history, n (%)
 Hypertension29 (35.4)27 (38.0)2 (18.2)
 Diabetes mellitus17 (20.7)15 (21.1)2 (18.2)
 Thyroid dysfunction11 (13.4)10 (14.1)1 (9.0)
 Dyslipidemia10 (12.2)9 (12.7)1 (9.0)
Vaccine type, n (%)
 mRNA70 (72.2)62 (72.1)8 (72.7)
 Vector20 (20.6)18 (20.9)2 (18.2)
 Inactivated7 (7.2)6 (7.0)1 (9.1)
Which dose, n (%)
 First dose44 (45.4)39 (45.3)5 (45.5)
 Second dose47 (48.4)41 (47.7)6 (54.5)
 Third dose5 (5.2)5 (5.0)0 (0)
 Fourth dose1 (1.0)1 (1.0)0 (0)
Onset time after vaccination, n (%)
 ≤7 days56 (57.7)51 (59.3)5 (45.5)
 8–14 days20 (20.6)18 (20.9)2 (18.2)
 15–28 days12 (12.4)9 (10.5)3 (27.3)
 ≥29 days9 (9.3)8 (9.3)1 (9.1)
Symptoms, n (%)
 Nephrotic syndrome29 (29.9)24 (27.9)5 (45.5)
 Gastrointestinal discomfort25 (25.8)23 (26.4)2 (18.2)
 Hematuria21 (21.6)18 (20.5)3 (27.3)
 Fever13 (13.4)11 (12.5)2 (18.2)
 Headache12 (12.4)11 (12.5)1 (9.1)
Pathological type, n (%)
 Crescentic glomerulonephritis29 (29.9)29 (32.6)0 (0)
 Acute tubular injury23 (23.7)22 (24.7)1 (9.1)
 IgA nephropathy18 (18.6)15 (16.9)3 (27.3)
 ANCA-associated vasculitis17 (17.5)17 (19.1)0 (0)
 Minimal change disease17 (17.5)15 (16.9)2 (18.2)
 Thrombotic microangiopathy10 (10.3)10 (11.2)0 (0)
 Anti-GBM glomerulonephritis5 (5.2)5 (5.6)0 (0)
AKI stage, n (%)
 AKI Stage 130 (30.9)26 (29.2)4 (36.4)
 AKI Stage 222 (22.7)18 (20.2)4 (36.4)
 AKI Stage 345 (46.4)42 (47.2)3(27.3)
Treatment, n (%)
 Conservative medication11 (12.8)9 (11.7)2 (22.2)
 Steroids77 (89.5)70 (90.9)7 (77.8)
 Cyclophosphamide24 (27.9)24 (31.2)0 (0)
 Rituximab17 (19.8)15 (19.5)2 (22.2)
 Mycophenolate mofetil4 (4.7)3 (3.9)1 (11.1)
 Intravenous immunoglobulins3 (3.5)3 (3.9)0 (0)
 Plasma exchange18 (20.9)18 (23.4)0 (0)
 Renal replacement therapy22 (25.6)21 (27.3)1 (11.1)
Outcome, n (%)
 CR in 3 months42 (49.4)36 (47.4)6 (66.7)
 PR in 3 months24 (28.2)22 (28.9)2 (22.2)
 NR in 3 months19 (22.4)18 (23.7)1 (11.1)
 Dependent on hemodialysis7 (8.2)7 (9.2)0 (0)

AKI, acute kidney injury; CKD, chronic kidney disease; CR, complete remission; NR, no response; PR, partial remission.

Table 3.

Clinical characteristics of acute kidney injury post-COVID-19 vaccination

CharacteristicsTotalNew-onset kidney disease with AKIRelapsed kidney disease with AKI
Age (years)55.4 (12–84)56.5 (12–84)47.8 (13–67)
Male sex, n (%)55 (56.7)50 (58.1)5 (45.5)
Medical history, n (%)
 Hypertension29 (35.4)27 (38.0)2 (18.2)
 Diabetes mellitus17 (20.7)15 (21.1)2 (18.2)
 Thyroid dysfunction11 (13.4)10 (14.1)1 (9.0)
 Dyslipidemia10 (12.2)9 (12.7)1 (9.0)
Vaccine type, n (%)
 mRNA70 (72.2)62 (72.1)8 (72.7)
 Vector20 (20.6)18 (20.9)2 (18.2)
 Inactivated7 (7.2)6 (7.0)1 (9.1)
Which dose, n (%)
 First dose44 (45.4)39 (45.3)5 (45.5)
 Second dose47 (48.4)41 (47.7)6 (54.5)
 Third dose5 (5.2)5 (5.0)0 (0)
 Fourth dose1 (1.0)1 (1.0)0 (0)
Onset time after vaccination, n (%)
 ≤7 days56 (57.7)51 (59.3)5 (45.5)
 8–14 days20 (20.6)18 (20.9)2 (18.2)
 15–28 days12 (12.4)9 (10.5)3 (27.3)
 ≥29 days9 (9.3)8 (9.3)1 (9.1)
Symptoms, n (%)
 Nephrotic syndrome29 (29.9)24 (27.9)5 (45.5)
 Gastrointestinal discomfort25 (25.8)23 (26.4)2 (18.2)
 Hematuria21 (21.6)18 (20.5)3 (27.3)
 Fever13 (13.4)11 (12.5)2 (18.2)
 Headache12 (12.4)11 (12.5)1 (9.1)
Pathological type, n (%)
 Crescentic glomerulonephritis29 (29.9)29 (32.6)0 (0)
 Acute tubular injury23 (23.7)22 (24.7)1 (9.1)
 IgA nephropathy18 (18.6)15 (16.9)3 (27.3)
 ANCA-associated vasculitis17 (17.5)17 (19.1)0 (0)
 Minimal change disease17 (17.5)15 (16.9)2 (18.2)
 Thrombotic microangiopathy10 (10.3)10 (11.2)0 (0)
 Anti-GBM glomerulonephritis5 (5.2)5 (5.6)0 (0)
AKI stage, n (%)
 AKI Stage 130 (30.9)26 (29.2)4 (36.4)
 AKI Stage 222 (22.7)18 (20.2)4 (36.4)
 AKI Stage 345 (46.4)42 (47.2)3(27.3)
Treatment, n (%)
 Conservative medication11 (12.8)9 (11.7)2 (22.2)
 Steroids77 (89.5)70 (90.9)7 (77.8)
 Cyclophosphamide24 (27.9)24 (31.2)0 (0)
 Rituximab17 (19.8)15 (19.5)2 (22.2)
 Mycophenolate mofetil4 (4.7)3 (3.9)1 (11.1)
 Intravenous immunoglobulins3 (3.5)3 (3.9)0 (0)
 Plasma exchange18 (20.9)18 (23.4)0 (0)
 Renal replacement therapy22 (25.6)21 (27.3)1 (11.1)
Outcome, n (%)
 CR in 3 months42 (49.4)36 (47.4)6 (66.7)
 PR in 3 months24 (28.2)22 (28.9)2 (22.2)
 NR in 3 months19 (22.4)18 (23.7)1 (11.1)
 Dependent on hemodialysis7 (8.2)7 (9.2)0 (0)
CharacteristicsTotalNew-onset kidney disease with AKIRelapsed kidney disease with AKI
Age (years)55.4 (12–84)56.5 (12–84)47.8 (13–67)
Male sex, n (%)55 (56.7)50 (58.1)5 (45.5)
Medical history, n (%)
 Hypertension29 (35.4)27 (38.0)2 (18.2)
 Diabetes mellitus17 (20.7)15 (21.1)2 (18.2)
 Thyroid dysfunction11 (13.4)10 (14.1)1 (9.0)
 Dyslipidemia10 (12.2)9 (12.7)1 (9.0)
Vaccine type, n (%)
 mRNA70 (72.2)62 (72.1)8 (72.7)
 Vector20 (20.6)18 (20.9)2 (18.2)
 Inactivated7 (7.2)6 (7.0)1 (9.1)
Which dose, n (%)
 First dose44 (45.4)39 (45.3)5 (45.5)
 Second dose47 (48.4)41 (47.7)6 (54.5)
 Third dose5 (5.2)5 (5.0)0 (0)
 Fourth dose1 (1.0)1 (1.0)0 (0)
Onset time after vaccination, n (%)
 ≤7 days56 (57.7)51 (59.3)5 (45.5)
 8–14 days20 (20.6)18 (20.9)2 (18.2)
 15–28 days12 (12.4)9 (10.5)3 (27.3)
 ≥29 days9 (9.3)8 (9.3)1 (9.1)
Symptoms, n (%)
 Nephrotic syndrome29 (29.9)24 (27.9)5 (45.5)
 Gastrointestinal discomfort25 (25.8)23 (26.4)2 (18.2)
 Hematuria21 (21.6)18 (20.5)3 (27.3)
 Fever13 (13.4)11 (12.5)2 (18.2)
 Headache12 (12.4)11 (12.5)1 (9.1)
Pathological type, n (%)
 Crescentic glomerulonephritis29 (29.9)29 (32.6)0 (0)
 Acute tubular injury23 (23.7)22 (24.7)1 (9.1)
 IgA nephropathy18 (18.6)15 (16.9)3 (27.3)
 ANCA-associated vasculitis17 (17.5)17 (19.1)0 (0)
 Minimal change disease17 (17.5)15 (16.9)2 (18.2)
 Thrombotic microangiopathy10 (10.3)10 (11.2)0 (0)
 Anti-GBM glomerulonephritis5 (5.2)5 (5.6)0 (0)
AKI stage, n (%)
 AKI Stage 130 (30.9)26 (29.2)4 (36.4)
 AKI Stage 222 (22.7)18 (20.2)4 (36.4)
 AKI Stage 345 (46.4)42 (47.2)3(27.3)
Treatment, n (%)
 Conservative medication11 (12.8)9 (11.7)2 (22.2)
 Steroids77 (89.5)70 (90.9)7 (77.8)
 Cyclophosphamide24 (27.9)24 (31.2)0 (0)
 Rituximab17 (19.8)15 (19.5)2 (22.2)
 Mycophenolate mofetil4 (4.7)3 (3.9)1 (11.1)
 Intravenous immunoglobulins3 (3.5)3 (3.9)0 (0)
 Plasma exchange18 (20.9)18 (23.4)0 (0)
 Renal replacement therapy22 (25.6)21 (27.3)1 (11.1)
Outcome, n (%)
 CR in 3 months42 (49.4)36 (47.4)6 (66.7)
 PR in 3 months24 (28.2)22 (28.9)2 (22.2)
 NR in 3 months19 (22.4)18 (23.7)1 (11.1)
 Dependent on hemodialysis7 (8.2)7 (9.2)0 (0)

AKI, acute kidney injury; CKD, chronic kidney disease; CR, complete remission; NR, no response; PR, partial remission.

Edema, hematuria, headache and proteinuria were the four main initial signs in patients with AKI after vaccination. AKI developed after vaccination with any of the commercially available COVID-19 vaccines, but 72.2% (70/97) of patients received an mRNA vaccine, and 20.6% (20/97) received a vector vaccine. In addition, 7.2% (7/97) received an inactivated vaccine. Of these, 45.4% (44/97) of patients developed AKI after the first dose, while 48.4% (47/97) of patients developed AKI after the second dose. In addition, 5.2% (5/97) of patients developed AKI after the third dose, and 1.0% (1/97) of patients developed AKI after the fourth dose. The proportions of patients with AKI Stages 1, 2 and 3 after COVID-19 vaccination in case reports were 30.9%, 22.7% and 46.4%, respectively. Twenty-two (25.6%) patients were treated with hemodialysis (HD).

Common clinical symptoms included edema, hematuria, fever, body aches, fever, dizziness, nausea, vomiting, asthenia, headache and foamy urine. According to research, the medical conditions most commonly associated with the development of AKI after receiving COVID-19 vaccines were HT (35.4%), DM (20.7%) and thyroid dysfunction (including hypothyroidism, hyperthyroidism, total thyroidectomy, autoimmune thyroiditis and Hashimoto's thyroiditis, 13.4%) and dyslipidemia (12.2%).

Treatment and prognosis

Seventy-seven (89.5%) patients received steroid therapy, 24 (27.9%) patients received cyclophosphamide (CyC) therapy, 17 (19.8%) patients were treated with rituximab (RTX), 18 (20.9%) patients received plasma exchange (PLEX) therapy and 11 (12.8) patients received conservative treatment. A small number of patients received mycophenolate mofetil (MMF)6,70,103 or intravenous immunoglobulins (IVIG).77,87,91 Regarding outcomes, 49.4% of patients achieved complete remission in 3 months, 28.2% of patients achieved partial remission in 3 months, 22.4% of patients achieved no response in 3 months and 8.2% of patients were still dependent on HD after discharge. Generally, clinical cases of new-onset and relapsed kidney disease with AKI after COVID-19 vaccination had a good prognosis. Some patients, especially those with new-onset kidney disease with AKI, achieved remission through conservative treatment.

Discussion

Potential mechanisms of AKI post-SARS-CoV-2 infection

The pathophysiology of AKI in patients with COVID-19 is complex and involves various factors. There is controversy about whether COVID-19 causes AKI primarily through direct or indirect mechanisms.10–12 What needs to be clarified is that in addition to the direct effects of the virus, indirect mechanisms also play an important role in COVID-19-related AKI.12 Many cases appear to be clearly attributable to indirect effects, such as ischemic injury, toxic injury, cytokine storm, activation and the dysregulation of the angiotensin (Ang) II pathway and complement system, endothelial dysfunction, abnormal platelet activation, hypercoagulation and microangiopathy (Figure 1).10,114–116

Proposed mechanisms of acute kidney injury caused by COVID-19. The virus may directly or indirectly damage the kidneys, possibly through organ crosstalk, cytokine storm and damage-associated molecular patterns (DAMPs) secretion, which can lead to cell death in specific renal cells like podocytes and proximal tubular cells. Additionally, complement activation and an abnormal immune system response may contribute to further damage of kidney tissue. Abnormal immune system and complement activation can cause additional harm to kidney tissue. Myositis and damage to endothelial cells can result in the formation of microthrombi. Immune system dysfunction or direct damage to blood vessels from a virus can cause abnormal clotting. AKI can also be caused by infection or drug-induced rhabdomyolysis. The figure refers to the pathogenesis of AKI in COVID-19 by Chavez-Valencia et al. (DOI: 10.3390/diseases10030053).
Figure 1.

Proposed mechanisms of acute kidney injury caused by COVID-19. The virus may directly or indirectly damage the kidneys, possibly through organ crosstalk, cytokine storm and damage-associated molecular patterns (DAMPs) secretion, which can lead to cell death in specific renal cells like podocytes and proximal tubular cells. Additionally, complement activation and an abnormal immune system response may contribute to further damage of kidney tissue. Abnormal immune system and complement activation can cause additional harm to kidney tissue. Myositis and damage to endothelial cells can result in the formation of microthrombi. Immune system dysfunction or direct damage to blood vessels from a virus can cause abnormal clotting. AKI can also be caused by infection or drug-induced rhabdomyolysis. The figure refers to the pathogenesis of AKI in COVID-19 by Chavez-Valencia et al. (DOI: 10.3390/diseases10030053).

SARS-CoV-2 enters host cells mainly by binding to the ACE2 receptor. This receptor aids virus entry through two pathways: clathrin-dependent endocytosis and ACE2 receptor-mediated transmembrane protease serine 2 (TMPRSS2)-dependent membrane fusion.117–119 Recent research findings indicate that ACE2 and TMPRSS2 are present in proximal tubular epithelial cells and renal podocytes.120–122 These receptors are essential for the virus to enter the cells and replicate.123 Once SARS-CoV-2 interacts with cells, ACE2 binds to the spike (S) protein of SARS-CoV-2,117 while TMPRSS2 interacts with the spike protein and promotes internalization. Additionally, a disintegrin and a metalloproteinase 17 (ADAM17) leads to ACE2 shedding by converting ACE2 to a soluble form. After binding to ACE2, SARS-CoV-2 may lead to a downregulation of ACE2, resulting in increased levels of Ang II and decreased levels of Ang (1–7) and thus causing RAS dysregulation.124 Ang II can induce the release of cytokines, activate nuclear factor kappa B (NF-κB), trigger inflammatory reactions and lead to necrotic sequelae with the release of neutrophil extracellular traps (NETs). However, there are no human data at all that the role of Ang II matters in COVID-19. The findings of several large randomized trials examining RAS modulators of various types and in various scenarios (starting vs. stopping RAS blockade, continuing vs. discontinuing chronic RAS blockade, RCTs of novel RAS modulators) have essentially all been negative.125–128 One possibility is that COVID-19 infection may not result in unopposed Ang II activity in the organs, as hypothesized.

In addition, an analysis of renal biopsy samples from 19 SARS-CoV-2-related AKI patients revealed evidence of complement system activation in the patients’ renal tissue after viral infection.129 Complement components may collaborate with other factors to trigger inflammation, coagulation and endothelial damage. There is a hypothesis that Ang II may activate the terminal components of the complement system (C5a, C5b-9) in a destructive manner through the angiotensin-1 (AT1) receptor.130 C5a can also stimulate the production of NETs in vitro,131 triggering an increase in mitochondrial oxygen species (ROS) production.

Studies have also found that viral particles exist in renal endothelial cells, and these particles mediate endothelial injury and promote vasoconstriction and a hypercoagulable state, leading to microthrombosis and renal microvasculature damage.34,120,132 According to Wang et al.,133 CD147 (extracellular matrix metalloproteinase inducer/basigin) has a high affinity for SARS-CoV-2 and is expressed within the transmembrane, which helps the virus enter renal epithelial cells. Normally, CD147 is highly expressed only on the basolateral side of renal tubular epithelial cells in healthy kidneys.134 However, in patients with viremia, SARS-CoV-2 may invade renal proximal tubular epithelial cells through both the luminal surface and the basolateral side, leading to the production of inflammatory cytokines.135 The innate immune system directly responds to SARS-CoV-2 by recognizing pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs) and indirectly responds to the virus through damage-associated molecular patterns (DAMPs), releasing cytokines locally, recruiting inflammatory cells and stimulating adaptive immune responses.10

Although the theory of a cytokine storm being a prominent part of severe COVID-19 was popular early in the pandemic, how much of a role a true cytokine storm plays in COVID-19 or COVID-19-associated AKI remains controversial.10–12 A meta-analysis reported that serum IL-6 levels were much lower in patients with severe COVID-19 than in patients with sepsis and non-COVID-19 acute respiratory distress syndrome (ARDS).136 Importantly, a role for local inflammation in the pathogenicity of COVID-19 has not been ruled out. Inflammatory mediators can directly bind to their specific receptors expressed by renal endothelial cells and tubular epithelial cells to cause cellular damage, but their role in COVID-19-related AKI has not been clearly demonstrated.136–138

Organ crosstalk among the lung, heart and kidneys has also been found in critically ill patients with COVID-19, and this process is complex.40 Acute hypoxemia can affect renal function and increase renal vascular resistance.139,140 Pneumonia may cause right ventricular failure, leading to renal congestion, while left ventricular dysfunction can cause reduced cardiac output and renal hypoperfusion.141 In patients with severe COVID-19, excessive positive pressure ventilation can have negative effects on cardiac output and renal hypoperfusion.142 Additionally, extracorporeal membrane oxygenation (ECMO) may cause AKI via venous congestion, hemolysis, bleeding, secondary infections and inflammation.10,143 Severe COVID-19 can also cause rhabdomyolysis and myositis, resulting in tubular obstruction and toxicity.144 Furthermore, exposure to certain drugs such as antibiotics, antivirals and other nephrotoxic drugs can also lead to AKI in patients.40 In addition, patients' underlying diseases such as CKD, DM, cardiovascular disease17,32,37,38 and genetic risk factors such as high-risk APOL1 alleles may further induce the occurrence of AKI.145,146

Potential mechanisms of AKI post-COVID-19 vaccination

According to recent studies, the SARS-CoV-2 spike protein found in vaccines may lead to kidney damage, either directly or indirectly (Figure 2). These studies have shown that certain tissue antigens, including transglutaminase 3, antiextraction nuclear antigen and thyroid peroxidase, can strongly react with SARS-CoV-2 antibodies.147 COVID-19 vaccines activate antigen-presenting cells (APCs), which, upon receiving a second vaccination, trigger robust CD4+ T-cell and CD8+ T-cell responses. This process results in the release of significant amounts of inflammatory cytokines (IFN-c, TNF-α, IL-2, IFN-γ and TNF), which can promote a cytokine storm and lead to damage to renal tissue.148,149 The receptor-binding domain (RBD) of the SARS-CoV-2 spike protein (S) has the ability to bind to natural antibodies in the body, which can lead to the formation of circulating immune complexes and their deposition in the glomeruli.150

Hypotheses for the pathogenesis of acute kidney injury after COVID-19 vaccination. First, vaccination triggers the activation of antigen-presenting cells (APCs) and B cells. These cells then stimulate T cells through antigen presentation and cytokine production, which has an impact on the immune and complement systems. This process may result in the formation of a cellular inflammatory cytokine storm, dysregulation of neutrophil extracellular traps (NETs) and activation of the membrane attack complex (MAC). Besides, SARS-CoV-2 is capable of infecting renal tissue by utilizing angiotensin-converting enzyme 2 (ACE2), CD147 and glucose regulatory protein 78 (GRP78) in conjunction with furin-like cleavage on transmembrane protease serine 2 (TMPRESS2) and spike protein (S), damaging proximal tubular cells and podocytes. What’s more, the activation of platelets, endothelial cell damage and abnormal coagulation can be caused by an abnormal immune system or direct viral damage. AKI may also be attributed to infection or drug-induced rhabdomyolysis, underlying chronic or autoimmune disease, genetic predisposition and elevated levels of ANCA antibodies.
Figure 2.

Hypotheses for the pathogenesis of acute kidney injury after COVID-19 vaccination. First, vaccination triggers the activation of antigen-presenting cells (APCs) and B cells. These cells then stimulate T cells through antigen presentation and cytokine production, which has an impact on the immune and complement systems. This process may result in the formation of a cellular inflammatory cytokine storm, dysregulation of neutrophil extracellular traps (NETs) and activation of the membrane attack complex (MAC). Besides, SARS-CoV-2 is capable of infecting renal tissue by utilizing angiotensin-converting enzyme 2 (ACE2), CD147 and glucose regulatory protein 78 (GRP78) in conjunction with furin-like cleavage on transmembrane protease serine 2 (TMPRESS2) and spike protein (S), damaging proximal tubular cells and podocytes. What’s more, the activation of platelets, endothelial cell damage and abnormal coagulation can be caused by an abnormal immune system or direct viral damage. AKI may also be attributed to infection or drug-induced rhabdomyolysis, underlying chronic or autoimmune disease, genetic predisposition and elevated levels of ANCA antibodies.

CoronaVac is a vaccine that was developed using whole virion inactivated Vero cells that retain the antigenic components of SARS-CoV-2 to induce antiviral neutralizing immunoglobulins.151 Additionally, CoronaVac contains aluminum, a complex adjuvant that enhances the antigen-specific immune response, making the recognition of ‘nonself’ factors easier and more effective.152 Autoinflammatory syndrome induced by adjuvants (ASIA) can be triggered by the injection of adjuvant, which can induce the release of risk-related molecular patterns in vivo.153 This, in turn, can activate the intracellular Nalp3 inflammasome system and caspase-1, leading to autoimmune or autoinflammatory disease (AI/AIFD). Additionally, aluminum may stimulate the production and secretion of cytokines, such as IL-1β, IL-18 and IL-33.154

Elevated levels of complement markers (sC5b-9 and C5a) have been observed in COVID-19 patients, and these markers were found to be correlated with the severity of the disease.155,156 Furthermore, in vitro studies have shown that the spike protein of SARS-CoV-2 can directly activate the alternative pathway of complement (APC) by impeding the inactivation of the cell surface APC convertase.157 It is currently speculated that COVID-19 vaccines, which target the spike protein as an immunogenic target, may also activate the complement system and lead to endothelial damage.

NETs play a crucial role in the pathogenesis of complement activation and cytokine storms in COVID-19 vaccine-induced AKI.158 In individuals with compromised immune systems, NET nucleic acid clearance may be reduced.159 Neutrophils are activated by proinflammatory cytokines, which in turn release NETs. These NETs are crucial in local inflammatory responses, pathogen clearance and thrombus formation and play a significant role in host defense. However, NET dysregulation can lead to vasculopathy and ANCA-associated vasculitis.160 NETs are often accompanied by numerous harmful proteins, such as myeloperoxidase (MPO) and antiproteinase 3 antibody. These proteins can inflict direct harm on the vascular endothelium, activate the alternative complement pathway and contribute to the onset and progression of vasculitis.

According to research, the expression of ACE2 in kidney tissue is 100 times higher than that in lung tissue.161 The binding of SARS-CoV-2 to ACE2 is believed to downregulate the production of ACE2, which results in an increase in Ang II levels and a decrease in Ang 1–7 levels. This increase in Ang II levels can lead to renal injury by triggering inflammation and apoptosis.162 Additionally, increased Ang II levels can also lead to endothelial and platelet activation, vasoconstriction and microthrombosis, which are all unfavorable outcomes.132 The prothrombotic state can increase the likelihood of microthrombosis formation in renal tissue. These factors collectively contribute to the development of ATI or ATN, as well as glomerulonephritis. Overall, all of the proposed pathogenic processes described are simply hypotheses and may not be consistently confirmed in real-world clinical studies. For example, drugs aimed at reversing elevated Ang II to Ang (1–7) ratios did not confer benefit in COVID-19 patients in large RCTs.163,164

The risk of kidney disease after COVID-19 vaccination

Although there have been numerous case reports of glomerulonephritis and kidney injury following SARS-CoV-2 vaccination, a causal relationship between them remains to be proven. In a study that included data on the incidence of glomerulonephritis in 7.1 million people, 69% of whom received at least one dose of the vaccine during the study period, it was found that patients who developed glomerulonephritis within four weeks after receiving the mRNA vaccine showed no differences from patients with vaccination-unrelated glomerulonephritis.165 Another study, a self-controlled case series of Hong Kong-wide SARS-CoV-2 vaccination records, found no evidence of an increased risk of new or recurrent glomerular diseases with vaccination with the BNT162b2 or CoronaVac vaccines.166 A retrospective cohort study in Canada included 1105 adult patients with clinically stable glomerular disease (including MCD, FSGS, MN, IgAN, LN, ANCA-associated vasculitis and C3 glomerulonephritis) as confirmed by renal biopsy sample analysis. Exposure to a second or third dose of the COVID-19 vaccine in this population was associated with a 2.23-fold risk of relapse, but the absolute increase in risk of relapse remained low (1–5%).167 Overall, further research is needed on the risk of kidney disease following SARS-CoV-2 vaccination. The emergence of new cases of glomerulonephritis shortly after SARS-CoV-2 vaccination may be attributed to timing coincidence. Patients with preexisting glomerular disease should be monitored more closely after receiving a second or third COVID-19 vaccine.

Limitations

This study has limitations. The literature on the relationship between vaccines and AKI is primarily based on single-case reported studies, making it difficult to establish a definitive causal relationship. Additionally, there may be potential unmeasured confounders that could impact the results. Furthermore, relying solely on electronic health information systems as data sources may lead to an underreporting of AKI cases in patients with COVID-19. On the other hand, as the data presented on AKI after COVID-19 vaccination are based primarily on case reports, our analysis provides no information on the true incidence of AKI after COVID-19 vaccination. Though these observational data suggest that COVID-19 vaccination may predispose to kidney disease, given that billions of people have received COVID-19 vaccinations worldwide, whether COVID-19 vaccination truly increases the risk of kidney disease or if these case reports represent association rather than causation remains unclear. Finally, while there are hypotheses regarding the mechanism of action, a mechanism of action has not been definitively proven and hypotheses are based on a combination of case reports and literature.

Conclusion

The associated between COVID-19 and AKI is significant, leading to increased morbidity and mortality. However, with the widespread availability of vaccines, the death toll has decreased significantly, highlighting the importance of vaccination in the battle against the outbreak. This narrative review offers fresh perspectives on AKI in relation to COVID-19 infection and vaccination. We reviewed the pathomorphological, and clinical features and the prognosis of AKI associated with COVID-19 infection and vaccination. Additionally, the study explored the underlying pathophysiological mechanisms of AKI in these cases.

Supplementary material

Supplementary material is available at QJMED online.

Author contributions

Yebei Li and Yan Gong conducted data collection and wrote the manuscript. Gaosi Xu was responsible for the idea, funds and paper revision. All authors contributed to the article and approved the submitted version.

Yebei Li (Data curation, Investigation, Methodology, Writing—original draft, Writing—review & editing [equal]), Yan Gong (Data curation, Formal analysis, Investigation, Methodology, Writing—original draft, Writing—review & editing [equal]) and Gaosi Xu (Conceptualization, Funding acquisition, Supervision, Writing—review & editing [lead])

Funding

This work was supported by the National Natural Science Foundation of China (Nos. 81970583 and 82060138), the Key Project of Jiangxi Provincial Nature Science Foundation (No. 20224ACB206008), the Kidney Disease Engineering Technology Research Centre Foundation of Jiangxi Province (No. 20164BCD40095), Jiangxi Province introduces and trains innovative and entrepreneurial high-level talents “Thousand Talents Plan” project (JXSQ2023201030) and the Key Project of Clinical Research of the Second Affiliated Hospital of Nanchang University (2022efyB01).

Conflict of interest

None declared.

References

1

Thomas
SJ
,
Moreira
ED
Jr
,
Kitchin
N
,
Absalon
J
,
Gurtman
A
,
Lockhart
S
, et al. ;
C4591001 Clinical Trial Group
.
Safety and efficacy of the BNT162b2 mRNA COVID-19 vaccine through 6 months
.
N Engl J Med
2021
;
385
:
1761
73
.

2

Voysey
M
,
Clemens
SAC
,
Madhi
SA
,
Weckx
LY
,
Folegatti
PM
,
Aley
PK
, et al. ;
Oxford COVID Vaccine Trial Group
.
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
:
99
111
.

3

Emary
KRW
,
Golubchik
T
,
Aley
PK
,
Ariani
CV
,
Angus
B
,
Bibi
S
, et al. ;
Oxford COVID-19 Vaccine Trial Group
.
Efficacy of ChAdOx1 nCoV-19 (AZD1222) vaccine against SARS-CoV-2 variant of concern 202012/01 (B.1.1.7): an exploratory analysis of a randomised controlled trial
.
Lancet
2021
;
397
:
1351
62
.

4

Lebedev
L
,
Sapojnikov
M
,
Wechsler
A
,
Varadi-Levi
R
,
Zamir
D
,
Tobar
A
, et al.
Minimal change disease following the Pfizer-BioNTech COVID-19 vaccine
.
Am J Kidney Dis
2021
;
78
:
142
5
.

5

D'Agati
VD
,
Kudose
S
,
Bomback
AS
,
Adamidis
A
,
Tartini
A.
Minimal change disease and acute kidney injury following the Pfizer-BioNTech COVID-19 vaccine
.
Kidney Int
2021
;
100
:
461
3
.

6

Klomjit
N
,
Alexander
MP
,
Fervenza
FC
,
Zoghby
Z
,
Garg
A
,
Hogan
MC
, et al.
COVID-19 vaccination and glomerulonephritis
.
Kidney Int Rep
2021
;
6
:
2969
78
.

7

Levey
AS
,
Eckardt
KU
,
Tsukamoto
Y
,
Levin
A
,
Coresh
J
,
Rossert
J
, et al.
Definition and classification of chronic kidney disease: a position statement from kidney disease: improving global outcomes (KDIGO)
.
Kidney Int
2005
;
67
:
2089
100
.

8

Kellum
JA
,
Lameire
N
;
KDIGO AKI Guideline Work Group
.
Diagnosis, evaluation, and management of acute kidney injury: a KDIGO summary (part 1)
.
Crit Care
2013
;
17
:
204
.

9

Pacheco
ICR
,
Costa
D
,
Sousa
DS
,
Salgado Filho
N
,
Silva
GEB
,
Neves
P.
Kidney injury associated with COVID-19 infection and vaccine: a narrative review
.
Front Med (Lausanne)
2022
;
9
:
956158
.

10

Legrand
M
,
Bell
S
,
Forni
L
,
Joannidis
M
,
Koyner
JL
,
Liu
K
, et al.
Pathophysiology of COVID-19-associated acute kidney injury
.
Nat Rev Nephrol
2021
;
17
:
751
64
.

11

Nadim
MK
,
Forni
LG
,
Mehta
RL
,
Connor
MJ
Jr
,
Liu
KD
,
Ostermann
M
, et al.
COVID-19-associated acute kidney injury: consensus report of the 25th acute disease quality initiative (ADQI) workgroup
.
Nat Rev Nephrol
2020
;
16
:
747
64
.

12

Teixeira
JP
,
Barone
S
,
Zahedi
K
,
Soleimani
M.
Kidney injury in COVID-19: epidemiology, molecular mechanisms and potential therapeutic targets
.
Int J Mol Sci
2022
;
23
:
2242
.

13

Sindhu
C
,
Prasad
P
,
Elumalai
R
,
Matcha
J.
Clinical profile and outcomes of COVID-19 patients with acute kidney injury: a tertiary centre experience from South India
.
Clin Exp Nephrol
2022
;
26
:
36
44
.

14

Bell
JS
,
James
BD
,
Al-Chalabi
S
,
Sykes
L
,
Kalra
PA
,
Green
D.
Community- versus hospital-acquired acute kidney injury in hospitalised COVID-19 patients
.
BMC Nephrol
2021
;
22
:
269
.

15

Scarpioni
R
,
Valsania
T
,
Albertazzi
V
,
Blanco
V
,
DeAmicis
S
,
Manini
A
, et al.
Acute kidney injury, a common and severe complication in hospitalized patients during the COVID-19 pandemic
.
J Nephrol
2021
;
34
:
1019
24
.

16

Chen
K
,
Lei
Y
,
He
Y
,
Xiao
F
,
Yu
Y
,
Lai
X
, et al.
Clinical outcomes of hospitalized COVID-19 patients with renal injury: a multi-hospital observational study from Wuhan
.
Sci Rep
2021
;
11
:
15205
.

17

Sullivan
MK
,
Lees
JS
,
Drake
TM
,
Docherty
AB
,
Oates
G
,
Hardwick
HE
, et al. ;
ISARIC4C Investigators
.
Acute kidney injury in patients hospitalized with COVID-19 from the ISARIC WHO CCP-UK study: a prospective, multicentre cohort study
.
Nephrol Dial Transplant
2022
;
37
:
271
84
.

18

Rahimzadeh
H
,
Kazemian
S
,
Rahbar
M
,
Farrokhpour
H
,
Montazeri
M
,
Kafan
S
, et al.
The risk factors and clinical outcomes associated with acute kidney injury in patients with COVID-19: data from a large cohort in Iran
.
Kidney Blood Press Res
2021
;
46
:
620
8
.

19

Hung
AM
,
Shah
SC
,
Bick
AG
,
Yu
Z
,
Chen
HC
,
Hunt
CM
, et al. ;
VA Million Veteran Program COVID-19 Science Initiative
.
APOL1 risk variants, acute kidney injury, and death in participants with African ancestry hospitalized with COVID-19 from the million veteran program
.
JAMA Intern Med
2022
;
182
:
386
95
.

20

Marques
F
,
Gameiro
J
,
Oliveira
J
,
Fonseca
JA
,
Duarte
I
,
Bernardo
J
, et al.
Acute kidney disease and mortality in acute kidney injury patients with COVID-19
.
J Clin Med
2021
;
10
:
4599
.

21

Strohbehn
IA
,
Zhao
S
,
Seethapathy
H
,
Lee
M
,
Rusibamayila
N
,
Allegretti
AS
, et al.
Acute kidney injury incidence, recovery, and long-term kidney outcomes among hospitalized patients with COVID-19 and influenza
.
Kidney Int Rep
2021
;
6
:
2565
74
.

22

Hsu
CM
,
Gupta
S
,
Tighiouart
H
,
Goyal
N
,
Faugno
AJ
,
Tariq
A
, et al. ;
STOP-COVID Investigators
.
Kidney recovery and death in critically ill patients with COVID-19-associated acute kidney injury treated with dialysis: the STOP-COVID cohort study
.
Am J Kidney Dis
2022
;
79
:
404
16 e1
.

23

Jewell
PD
,
Bramham
K
,
Galloway
J
,
Post
F
,
Norton
S
,
Teo
J
, et al.
COVID-19-related acute kidney injury; incidence, risk factors and outcomes in a large UK cohort
.
BMC Nephrol
2021
;
22
:
359
.

24

Procaccini
FL
,
Alcazar Arroyo
R
,
Albalate Ramon
M
,
Torres Aguilera
E
,
Martin Navarro
J
,
Ryan Murua
P
, et al.
Acute kidney injury in 3182 patients admitted with COVID-19: a single-center, retrospective, case-control study
.
Clin Kidney J
2021
;
14
:
1557
69
.

25

Wan
YI
,
Bien
Z
,
Apea
VJ
,
Orkin
CM
,
Dhairyawan
R
,
Kirwan
CJ
, et al.
Acute kidney injury in COVID-19: multicentre prospective analysis of registry data
.
Clin Kidney J
2021
;
14
:
2356
64
.

26

Hirsch
JS
,
Ng
JH
,
Ross
DW
,
Sharma
P
,
Shah
HH
,
Barnett
RL
, et al. ;
Northwell Nephrology COVID-19 Research Consortium
.
Acute kidney injury in patients hospitalized with COVID-19
.
Kidney Int
2020
;
98
:
209
18
.

27

Kilis-Pstrusinska
K
,
Akutko
K
,
Braksator
J
,
Dancewicz
A
,
Grosman-Dziewiszek
P
,
Jamer
T
, et al.
Kidney dysfunction and its progression in patients hospitalized duo to COVID-19: contribution to the clinical course and outcomes
.
J Clin Med
2021
;
10
:
5522
.

28

Moledina
DG
,
Simonov
M
,
Yamamoto
Y
,
Alausa
J
,
Arora
T
,
Biswas
A
, et al.
The association of COVID-19 with acute kidney injury independent of severity of illness: a multicenter cohort study
.
Am J Kidney Dis
2021
;
77
:
490
9 e1
.

29

Magalhaes
LE
,
de Oliveira
PGS
,
Favarin
AJ
,
Yuasa
BK
,
Cardoso
PA
,
Zamoner
W
, et al.
Acute kidney injury in coronavirus infectious disease: a study of incidence, risk factors, and prognosis during the first wave of the disease in Brazil
.
Int Urol Nephrol
2022
;
55
:
1501
8
.

30

Tan
BWL
,
Tan
BWQ
,
Tan
ALM
,
Schriver
ER
,
Gutierrez-Sacristan
A
,
Das
P
, et al. ;
Consortium for Clinical Characterization of COVID-19 by EHR (4CE)
.
Long-term kidney function recovery and mortality after COVID-19-associated acute kidney injury: an international multi-centre observational cohort study
.
EClinicalMedicine
2023
;
55
:
101724
.

31

Fabrizi
F
,
Alfieri
CM
,
Molinari
P
,
Tamborini
F
,
Tangredi
M
,
Sikharulidze
A
, et al.
Acute kidney injury in non-intensive care unit (ICU) hospitalizations for coronavirus disease (COVID-19)
.
Pathogens
2022
;
11
:
1272
.

32

Chan
L
,
Chaudhary
K
,
Saha
A
,
Chauhan
K
,
Vaid
A
,
Zhao
S
, et al. ; on behalf of the
Mount Sinai COVID Informatics Center (MSCIC)
.
AKI in hospitalized patients with COVID-19
.
J Am Soc Nephrol
2021
;
32
:
151
60
.

33

Tarragon
B
,
Valdenebro
M
,
Serrano
ML
,
Maroto
A
,
Llopez-Carratala
MR
,
Ramos
A
, et al.
Acute kidney failure in patients admitted due to COVID-19
.
Nefrologia (Engl Ed)
2021
;
41
:
34
40
.

34

Su
H
,
Yang
M
,
Wan
C
,
Yi
LX
,
Tang
F
,
Zhu
HY
, et al.
Renal histopathological analysis of 26 postmortem findings of patients with COVID-19 in China
.
Kidney Int
2020
;
98
:
219
27
.

35

Huang
C
,
Wang
Y
,
Li
X
,
Ren
L
,
Zhao
J
,
Hu
Y
, et al.
Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China
.
Lancet
2020
;
395
:
497
506
.

36

Algahtani
FD
,
Elabbasy
MT
,
Alshammari
F
,
Atta
A
,
El-Fateh
AM
,
Ghoniem
ME.
Evolving risk of acute kidney injury in COVID-19 hospitalized patients: a single center retrospective study
.
Medicina (Kaunas)
2022
;
58
:
443
.

37

Chen
N
,
Zhou
M
,
Dong
X
,
Qu
J
,
Gong
F
,
Han
Y
, et al.
Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study
.
Lancet
2020
;
395
:
507
13
.

38

Gharge
A
,
Nayak-Rao
S.
Acute kidney injury associated with coronavirus disease 2019 - one year later, what do We know so far?
Saudi J Kidney Dis Transpl
2021
;
32
:
1543
51
.

39

Chaudhri
I
,
Moffitt
R
,
Taub
E
,
Annadi
RR
,
Hoai
M
,
Bolotova
O
, et al.
Association of proteinuria and hematuria with acute kidney injury and mortality in hospitalized patients with COVID-19
.
Kidney Blood Press Res
2020
;
45
:
1018
32
.

40

Joannidis
M
,
Forni
LG
,
Klein
SJ
,
Honore
PM
,
Kashani
K
,
Ostermann
M
, et al.
Lung-kidney interactions in critically ill patients: consensus report of the acute disease quality initiative (ADQI) 21 workgroup
.
Intensive Care Med
2020
;
46
:
654
72
.

41

Li
Z
,
Wu
M
,
Yao
J
,
Guo
J
,
Liao
X
,
Song
S
, et al. Caution on kidney dysfunctions of COVID-19 patients. medRxiv.
2020
; doi: 10.1101/2020.02.08.20021212.

42

Leclerc
S
,
Royal
V
,
Lamarche
C
,
Laurin
LP.
Minimal change disease with severe acute kidney injury following the Oxford-AstraZeneca COVID-19 vaccine: a case report
.
Am J Kidney Dis
2021
;
78
:
607
10
.

43

Lim
JH
,
Han
MH
,
Kim
YJ
,
Kim
MS
,
Jung
HY
,
Choi
JY
, et al.
New-onset nephrotic syndrome after Janssen COVID-19 vaccination: a case report and literature review
.
J Korean Med Sci
2021
;
36
:
e218
.

44

Maas
RJ
,
Gianotten
S
,
van der Meijden
WAG.
An additional case of minimal change disease following the Pfizer-BioNTech COVID-19 vaccine
.
Am J Kidney Dis
2021
;
78
:
312
.

45

Holzworth
A
,
Couchot
P
,
Cruz-Knight
W
,
Brucculeri
M.
Minimal change disease following the Moderna mRNA-1273 SARS-CoV-2 vaccine
.
Kidney Int
2021
;
100
:
463
4
.

46

Weijers
J
,
Alvarez
C
,
Hermans
MMH.
Post-vaccinal minimal change disease
.
Kidney Int
2021
;
100
:
459
61
.

47

Lim
JH
,
Kim
MS
,
Kim
YJ
,
Han
MH
,
Jung
HY
,
Choi
JY
, et al.
New-onset kidney diseases after COVID-19 vaccination: a case series
.
Vaccines (Basel)
2022
;
10
:
302
.

48

Park
HJ
,
An
WS
,
Rha
SH
,
Kim
SE
,
Lee
SM.
Minimal change glomerulonephritis following the second dose of the Moderna COVID-19 vaccine
.
QJM
2022
;
115
:
490
1
.

49

Annicchiarico Petruzzelli
L
,
Minale
B
,
Serio
V
,
De Luca
A
,
Marino Marsilia
G
,
Campione
S
, et al.
Pediatric minimal change disease and AKI following the Pfizer-BioNTech COVID-19 vaccine: causal or incidental correlation?
G Ital Nefrol
2022
;
39
.

50

Baskaran
K
,
Cohen
AWS
,
Weerasinghe
N
,
Vilayur
E.
Report of two cases of minimal change disease following vaccination for COVID-19
.
Nephrology (Carlton)
2022
;
27
:
111
2
.

51

Hanna
J
,
Ingram
A
,
Shao
T.
Minimal change disease after first dose of Pfizer-BioNTech COVID-19 vaccine: a case report and review of minimal change disease related to COVID-19 vaccine
.
Can J Kidney Health Dis
2021
;
8
:
1
5
.

52

Psyllaki
A
,
Stavrakaki
I
,
Androvitsanea
A
,
Gakiopoulou
H
,
Petrakis
I
,
Stylianou
K.
Two cases of glomerular involvement after vaccination against COVID-19: epiphenomenon or causality?
Clin Kidney J
2022
;
15
:
574
5
.

53

Mancianti
N
,
Guarnieri
A
,
Tripodi
S
,
Salvo
DP
,
Garosi
G.
Minimal change disease following vaccination for SARS-CoV-2
.
J Nephrol
2021
;
34
:
1039
40
.

54

Da
Y
,
Goh
GH
,
Khatri
P.
A case of membranous nephropathy following Pfizer-BioNTech mRNA vaccination against COVID-19
.
Kidney Int
2021
;
100
:
938
9
.

55

Thammathiwat
T
,
Chompuk
L
,
Worawichawong
S
,
Boonpucknavig
V
,
Sirilak
S
,
Pongcharoen
S
, et al.
Membranous nephropathy following full-dose of inactivated SARS-CoV-2 virus vaccination: a case report and literature review
.
Vaccines (Basel)
2022
;
11
:
80
.

56

Huang
YT
,
Chang
FP
,
Hung
SC
,
Wang
YC.
Relapsed focal segmental glomerulosclerosis and severe acute kidney injury after Oxford-AstraZeneca (AZ) COVID-19 vaccine
.
Arch Med Sci
2022
;
18
:
1693
5
.

57

Fernandez
P
,
Alaye
ML
,
Chiple
MEG
,
Arteaga
J
,
Douthat
W
,
Fuente
J
et al. Glomerulopathies after vaccination against COVID-19. Four cases with three different vaccines in Argentina. Nefrologia.
2021
; doi: 10.1016/j.nefro.2021.09.003.

58

Schaubschlager
T
,
Rajora
N
,
Diep
S
,
Kirtek
T
,
Cai
Q
,
Hendricks
AR
, et al.
De novo or recurrent glomerulonephritis and acute tubulointerstitial nephritis after COVID-19 vaccination: a report of six cases from a single center
.
Clin Nephrol
2022
;
97
:
289
97
.

59

Kudose
S
,
Friedmann
P
,
Albajrami
O
,
D'Agati
VD.
Histologic correlates of gross hematuria following Moderna COVID-19 vaccine in patients with IgA nephropathy
.
Kidney Int
2021
;
100
:
468
9
.

60

Tan
HZ
,
Tan
RY
,
Choo
JCJ
,
Lim
CC
,
Tan
CS
,
Loh
AHL
, et al.
Is COVID-19 vaccination unmasking glomerulonephritis?
Kidney Int
2021
;
100
:
469
71
.

61

Hanna
C
,
Herrera Hernandez
LP
,
Bu
L
,
Kizilbash
S
,
Najera
L
,
Rheault
MN
, et al.
IgA nephropathy presenting as macroscopic hematuria in 2 pediatric patients after receiving the Pfizer COVID-19 vaccine
.
Kidney Int
2021
;
100
:
705
6
.

62

Anderegg
MA
,
Liu
M
,
Saganas
C
,
Montani
M
,
Vogt
B
,
Huynh-Do
U
, et al.
De novo vasculitis after mRNA-1273 (Moderna) vaccination
.
Kidney Int
2021
;
100
:
474
6
.

63

Ritter
A
,
Helmchen
B
,
Gaspert
A
,
Bleisch
J
,
Fritschi
B
,
Buchkremer
F
, et al.
Clinical spectrum of gross haematuria following SARS-CoV-2 vaccination with mRNA vaccines
.
Clin Kidney J
2022
;
15
:
961
73
.

64

Chen
YS
,
Yang
CW
,
Tsai
CC
,
Ang
MD
,
Chou
SF
,
Chiang
WC
, et al.
Newly-diagnosed immunoglobulin a nephropathy with increased plasma galactose-deficient-IgA(1) antibody associated with mRNA COVID-19 vaccination: a case report
.
J Int Med Res
2022
;
50
:
3000605221129674
.

65

Park
K
,
Miyake
S
,
Tai
C
,
Tseng
M
,
Andeen
NK
,
Kung
VL.
Letter regarding: "a case of gross hematuria and IgA nephropathy Flare-Up following SARS-CoV-2 vaccination"
.
Kidney Int Rep
2021
;
6
:
2246
7
.

66

Sugita
K
,
Kaneko
S
,
Hisada
R
,
Harano
M
,
Anno
E
,
Hagiwara
S
, et al.
Development of IgA vasculitis with severe glomerulonephritis after COVID-19 vaccination: a case report and literature review
.
CEN Case Rep
2022
;
11
:
436
41
.

67

Perrin
P
,
Bassand
X
,
Benotmane
I
,
Bouvier
N.
Gross hematuria following SARS-CoV-2 vaccination in patients with IgA nephropathy
.
Kidney Int
2021
;
100
:
466
8
.

68

Watanabe
S
,
Zheng
S
,
Rashidi
A.
IgA nephropathy relapse following COVID-19 vaccination treated with corticosteroid therapy: case report
.
BMC Nephrol
2022
;
23
:
135
.

69

Gillion
V
,
Jadoul
M
,
Demoulin
N
,
Aydin
S
,
Devresse
A.
Granulomatous vasculitis after the AstraZeneca anti-SARS-CoV-2 vaccine
.
Kidney Int
2021
;
100
:
706
7
.

70

Kim
S
,
Jung
J
,
Cho
H
,
Lee
J
,
Go
H
,
Lee
JH.
A child with crescentic glomerulonephritis following SARS-CoV-2 mRNA (Pfizer-BioNTech) vaccination
.
Pediatr Nephrol
2023
;
38
:
299
302
.

71

Sacker
A
,
Kung
V
,
Andeen
N.
Anti-GBM nephritis with mesangial IgA deposits after SARS-CoV-2 mRNA vaccination
.
Kidney Int
2021
;
100
:
471
2
.

72

Coorey
CP
,
Phua
E
,
Chou
A
,
Shen
Y
,
Mather
A.
Anti-GBM disease after Oxford-AstraZeneca ChAdOx1 nCoV-19 vaccination: a report of two cases
.
Case Rep Nephrol Dial
2022
;
12
:
234
7
.

73

Tan
FS
,
Kabir
ME
,
Bhandari
S.
Acute interstitial nephritis after COVID-19 vaccination
.
BMJ Case Rep
2022
;
15
:
e246841
.

74

Mira
FS
,
Costa Carvalho
J
,
de Almeida
PA
,
Pimenta
AC
,
Alen Coutinho
I
,
Figueiredo
C
, et al.
A case of acute interstitial nephritis after two doses of the BNT162b2 SARS-CoV-2 vaccine
.
Int J Nephrol Renovasc Dis
2021
;
14
:
421
6
.

75

Unver
S
,
Haholu
A
,
Yildirim
S.
Nephrotic syndrome and acute kidney injury following CoronaVac anti-SARS-CoV-2 vaccine
.
Clin Kidney J
2021
;
14
:
2608
11
.

76

Choi
JH
,
Kang
KS
,
Han
KH.
Two adolescent cases of acute tubulointerstitial nephritis after second dose of COVID-19 mRNA vaccine
.
Hum Vaccin Immunother
2022
;
18
:
2059308
.

77

Al Rawahi
B
,
BaTaher
H
,
Jaffer
Z
,
Al Balushi
A
,
Al-Mazrouqi
A
,
Al-Balushi
N.
Vaccine-induced immune thrombotic thrombocytopenia following AstraZeneca (ChAdOx1 nCOV19) vaccine-A case report
.
Res Pract Thromb Haemost
2021
;
5
:
e12578
.

78

Yocum
A
,
Simon
EL.
Thrombotic thrombocytopenic purpura after Ad26.COV2-S vaccination
.
Am J Emerg Med
2021
;
49
:
441
e3–
e4
.

79

Osmanodja
B
,
Schreiber
A
,
Schrezenmeier
E
,
Seelow
E.
First diagnosis of thrombotic thrombocytopenic purpura after SARS-CoV-2 vaccine - case report
.
BMC Nephrol
2021
;
22
:
411
.

80

Alislambouli
M
,
Veras Victoria
A
,
Matta
J
,
Yin
F.
Acquired thrombotic thrombocytopenic purpura following Pfizer COVID-19 vaccination
.
EJHaem
2022
;
3
:
207
10
.

81

Yoshida
K
,
Sakaki
A
,
Matsuyama
Y
,
Mushino
T
,
Matsumoto
M
,
Sonoki
T
, et al.
Acquired thrombotic thrombocytopenic purpura following BNT162b2 mRNA coronavirus disease vaccination in a Japanese patient
.
Intern Med
2022
;
61
:
407
12
.

82

Ruhe
J
,
Schnetzke
U
,
Kentouche
K
,
Prims
F
,
Baier
M
,
Herfurth
K
, et al.
Acquired thrombotic thrombocytopenic purpura after first vaccination dose of BNT162b2 mRNA COVID-19 vaccine
.
Ann Hematol
2022
;
101
:
717
9
.

83

Chamarti
K
,
Dar
K
,
Reddy
A
,
Gundlapalli
A
,
Mourning
D
,
Bajaj
K.
Thrombotic thrombocytopenic purpura presentation in an elderly gentleman following COVID vaccine circumstances
.
Cureus
2021
;
13
:
e16619
.

84

Claes
KJ
,
Geerts
I
,
Lemahieu
W
,
Wilmer
A
,
Kuypers
DRJ
,
Koshy
P
, et al.
Atypical hemolytic uremic syndrome occurring after receipt of mRNA-1273 COVID-19 vaccine booster: a case report
.
Am J Kidney Dis
2022
; 81:364–7.

85

Sekar
A.
Lupus nephritis flare post Moderna mRNA-1273 coronavirus vaccine
.
QJM
2022
;
114
:
882
3
.

86

Kim
HJ
,
Jung
M
,
Lim
BJ
,
Han
SH.
New-onset class III lupus nephritis with multi-organ involvement after COVID-19 vaccination
.
Kidney Int
2022
;
101
:
826
8
.

87

Al-Rasbi
S
,
Al-Maqbali
JS
,
Al-Farsi
R
,
Al Shukaili
MA
,
Al-Riyami
MH
,
Al Falahi
Z
, et al.
Myocarditis, pulmonary hemorrhage, and extensive myositis with rhabdomyolysis 12 days after first dose of Pfizer-BioNTech BNT162b2 mRNA COVID-19 vaccine: a case report
.
Am J Case Rep
2022
;
23
:
e934399
.

88

Unger
K
,
Ponte
CD
,
Anderson
D.
A possible case of COVID-19 booster vaccine-associated rhabdomyolysis and acute kidney injury
.
J Pharm Technol
2022
;
38
:
247
50
.

89

Banamah
TA
,
Bogari
AA
,
Neyazi
A
,
Kotbi
E
,
Almaghraby
H
,
Atwah
F.
Severe rhabdomyolysis complicated with acute kidney injury required renal replacement therapy after Pfizer COVID-19 vaccine  
.
Cureus
2022
;
14
:
e25199
.

90

Missoum
S
,
Lahmar
M
,
Khellaf
G.
Leukocytoclastic vasculitis and acute renal failure following inactivated SARS-CoV-2 vaccine
.
Nephrol Ther
2021
; 18:287–90.

91

Vnučák
M
,
Graňák
K
,
Beliančinová
M
,
Jeseňák
M
,
Macháleková
KK
,
Benko
J
, et al.
Acute kidney rejection after anti-SARS-CoV-2 virus-vectored vaccine-case report
.
NPJ Vaccines
2022
;
7
:
30
.

92

Gulumsek
E
,
Ozturk
DD
,
Ozturk
HA
,
Saler
T
,
Erdogan
KE
,
Bashir
AM
, et al.
Minimal change nephrotic syndrome with acute kidney injury after the administration of Pfizer-BioNTech COVID-19 vaccine
.
Case Rep Infect Dis
2023
;
2023
:
5122228
.

93

Morimoto
N
,
Mori
T
,
Shioji
S
,
Taguchi
T
,
Watanabe
H
,
Sakai
K
, et al.
Rapidly progressive IgA nephropathy with membranoproliferative glomerulonephritis-like lesions in an elderly man following the third dose of an mRNA COVID-19 vaccine: a case report
.
BMC Nephrol
2023
;
24
:
108
.

94

Avalos
C
,
Ahmadzadeh
Y
,
Gatsak
D
,
Moosa
SA
,
Mozaffari
MA
,
Imas
AS
, et al.
Cardiac tamponade as a complication of microscopic polyangiitis: a case associated with a COVID-19 mRNA vaccine
.
Cureus
2023
;
15
:
e37569
.

95

Wisnik-Rainville
JM
,
Czyzewski
R
,
Demola
C
,
Czyzewski
K.
Focal crescentic glomerulonephritis following COVID-19 viral vector vaccination
.
Cureus
2023
;
15
:
e35305
.

96

Soma
Y
,
Kitaji
D
,
Hoshino
K
,
Sunohara
S
,
Iwano
T
,
Kawano
N.
Renal biopsy diagnosis of acute tubular injury after Pfizer-BioNTech COVID-19 vaccination: a case report
.
Vaccines (Basel)
2023
;
11
:
464
.

97

Tawhari
M
,
Alhamadh
MS
,
Alhabeeb
AY
,
Almudayfir
Z
,
Radwi
M.
End-stage kidney disease resulting from atypical hemolytic uremic syndrome after receiving AstraZeneca SARS-CoV-2 vaccine: a case report
.
Vaccines (Basel)
2023
;
11
:
679
.

98

Zamoner
W
,
Scardini
JB
,
De Dio
BJ
,
Marques
AM
,
Silva
VDS
,
Garcia
AL
, et al.
ANCA-associated vasculitis following Oxford-AstraZeneca COVID-19 vaccine in Brazil: is there a causal relationship? A case report
.
Front Med (Lausanne)
2022
;
9
:
1003332
.

99

Bansal
SB
,
Rana
AS
,
Manhas
N
,
Rana
A.
Post COVID vaccination (COVAXIN-BB152 V) pauci-immune crescentic glomerulonephritis
.
Indian J Nephrol
2022
;
32
:
495
7
.

100

Sekar
A
,
Campbell
R
,
Tabbara
J
,
Rastogi
P.
ANCA glomerulonephritis after the Moderna COVID-19 vaccination
.
Kidney Int
2021
;
100
:
473
4
.

101

Feghali
EJ
,
Zafar
M
,
Abid
S
,
Santoriello
D
,
Mehta
S.
De-novo antineutrophil cytoplasmic antibody-associated vasculitis following the mRNA-1273 (Moderna) vaccine for COVID-19
.
Cureus
2021
;
13
:
e19616
.

102

Villa
M
,
Diaz-Crespo
F
,
Perez de Jose
A
,
Verdalles
U
,
Verde
E
,
Almeida Ruiz
F
, et al.
A case of ANCA-associated vasculitis after AZD1222 (Oxford-AstraZeneca) SARS-CoV-2 vaccination: casualty or causality?
Kidney Int
2021
;
100
:
937
8
.

103

Noel
E
,
Rashid
U
,
Rabbani
R
,
Khan
WA
,
Benjamin
YS
,
Lee
I.
Antineutrophil cytoplasmic autoantibody-associated glomerulonephritis as a possible side effect of COVID-19 vaccination
.
Cureus
2022
;
14
:
e30565
.

104

Hakroush
S
,
Tampe
B.
Case report: ANCA-Associated vasculitis presenting with rhabdomyolysis and Pauci-Immune crescentic glomerulonephritis after Pfizer-BioNTech COVID-19 mRNA vaccination
.
Front Immunol
2021
;
12
:
762006
.

105

Shakoor
MT
,
Birkenbach
MP
,
Lynch
M.
ANCA-associated vasculitis following Pfizer-BioNTech COVID-19 vaccine
.
Am J Kidney Dis
2021
;
78
:
611
3
.

106

So
D
,
Min
KW
,
Jung
WY
,
Han
SW
,
Yu
MY.
Microscopic polyangiitis following mRNA COVID-19 vaccination: a case report
.
J Korean Med Sci
2022
;
37
:
e154
.

107

Dube
GK
,
Benvenuto
LJ
,
Batal
I.
Antineutrophil cytoplasmic autoantibody-associated glomerulonephritis following the Pfizer-BioNTech COVID-19 vaccine
.
Kidney Int Rep
2021
;
6
:
3087
9
.

108

Ma
Y
,
Huang
T
,
Xu
G.
ANCA-associated vasculitis following the CoronaVac vaccination
.
Ther Adv Chronic Dis
2022
;
13
:
20406223221125708
.

109

Kim
BC
,
Kim
HS
,
Han
KH
,
Han
SY
,
Jo
HA.
A case report of MPO-ANCA-associated vasculitis following heterologous mRNA1273 COVID-19 booster vaccination
.
J Korean Med Sci
2022
;
37
:
e204
.

110

Suzuki
M
,
Sekiguchi
Y
,
Sasaki
M
,
Inaba
S
,
Oyama
S
,
Inoue
Y
, et al.
Antineutrophil cytoplasmic antibody-associated vasculitis after COVID-19 vaccination with Pfizer-BioNTech
.
Intern Med
2022
;
61
:
2925
9
.

111

Nune
A
,
Iyengar
KP
,
Goddard
C
,
Ahmed
AE.
Multisystem inflammatory syndrome in an adult following the SARS-CoV-2 vaccine (MIS-V)
.
BMJ Case Rep
2021
;
14
:
e243888
.

112

Aydin
MF
,
Yildiz
A
,
Oruc
A
,
Sezen
M
,
Dilek
K
,
Gullulu
M
, et al.
Relapse of primary membranous nephropathy after inactivated SARS-CoV-2 virus vaccination
.
Kidney Int
2021
;
100
:
464
5
.

113

Martinez-Zayas
G
,
Savino
D
,
Kumar
S
,
Dao
KH.
An unusually "complex" glomerulonephritis
.
Proc (Bayl Univ Med Cent)
2022
;
35
:
531
3
.

114

Qian
JY
,
Wang
B
,
Lv
LL
,
Liu
BC.
Pathogenesis of acute kidney injury in coronavirus disease 2019
.
Front Physiol
2021
;
12
:
586589
.

115

Omer
D
,
Pleniceanu
O
,
Gnatek
Y
,
Namestnikov
M
,
Cohen-Zontag
O
,
Goldberg
S
, et al.
Human kidney spheroids and monolayers provide insights into SARS-CoV-2 renal interactions
.
J Am Soc Nephrol
2021
;
32
:
2242
54
.

116

Sang
L
,
Chen
S
,
Zheng
X
,
Guan
W
,
Zhang
Z
,
Liang
W
, et al.
The incidence, risk factors and prognosis of acute kidney injury in severe and critically ill patients with COVID-19 in mainland China: a retrospective study
.
BMC Pulm Med
2020
;
20
:
290
.

117

Kai
H
,
Kai
M.
Interactions of coronaviruses with ACE2, angiotensin II, and RAS inhibitors-lessons from available evidence and insights into COVID-19
.
Hypertens Res
2020
;
43
:
648
54
.

118

Li
W
,
Moore
MJ
,
Vasilieva
N
,
Sui
J
,
Wong
SK
,
Berne
MA
, et al.
Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus
.
Nature
2003
;
426
:
450
4
.

119

Kuba
K
,
Imai
Y
,
Ohto-Nakanishi
T
,
Penninger
JM.
Trilogy of ACE2: a peptidase in the renin-angiotensin system, a SARS receptor, and a partner for amino acid transporters
.
Pharmacol Ther
2010
;
128
:
119
28
.

120

Pan
XW
,
Xu
D
,
Zhang
H
,
Zhou
W
,
Wang
LH
,
Cui
XG.
Identification of a potential mechanism of acute kidney injury during the COVID-19 outbreak: a study based on single-cell transcriptome analysis
.
Intensive Care Med
2020
;
46
:
1114
6
.

121

Dong
M
,
Zhang
J
,
Ma
X
,
Tan
J
,
Chen
L
,
Liu
S
, et al.
ACE2, TMPRSS2 distribution and extrapulmonary organ injury in patients with COVID-19
.
Biomed Pharmacother
2020
;
131
:
110678
.

122

Qi
J
,
Zhou
Y
,
Hua
J
,
Zhang
L
,
Bian
J
,
Liu
B
, et al.
The scRNA-seq expression profiling of the receptor ACE2 and the cellular protease TMPRSS2 reveals human organs susceptible to SARS-CoV-2 infection
.
Int J Environ Res Public Health
2021
;
18
:
284
.

123

Pacciarini
F
,
Ghezzi
S
,
Canducci
F
,
Sims
A
,
Sampaolo
M
,
Ferioli
E
, et al.
Persistent replication of severe acute respiratory syndrome coronavirus in human tubular kidney cells selects for adaptive mutations in the membrane protein
.
J Virol
2008
;
82
:
5137
44
.

124

Cheng
Y
,
Wang
W
,
Wu
L
,
Cai
G.
SARS-CoV-2-related kidney injury: current concern and challenges
.
SN Compr Clin Med
2020
;
2
:
2015
24
.

125

Self
WH
,
Shotwell
MS
,
Gibbs
KW
,
de Wit
M
,
Files
DC
,
Harkins
M
, et al. ;
ACTIV-4 Host Tissue Investigators
.
Renin-angiotensin system modulation with synthetic angiotensin (1-7) and angiotensin II type 1 receptor-Biased ligand in adults with COVID-19: two randomized clinical trials
.
JAMA
2023
;
329
:
1170
82
.

126

Puskarich
MA
,
Ingraham
NE
,
Merck
LH
,
Driver
BE
,
Wacker
DA
,
Black
LP
, et al. ;
Angiotensin Receptor Blocker Based Lung Protective Strategies for Inpatients With COVID-19 (ALPS-IP) Investigators
.
Efficacy of losartan in hospitalized patients with COVID-19-induced lung injury: a randomized clinical trial
.
JAMA Netw Open
2022
;
5
:
e222735
.

127

Lopes
RD
,
Macedo
AVS
,
de Barros
E
,
Moll-Bernardes
RJ
,
Dos Santos
TM
,
Mazza
L
, et al. ;
BRACE CORONA Investigators
.
Effect of discontinuing vs continuing angiotensin-converting enzyme inhibitors and angiotensin II receptor blockers on days alive and out of the hospital in patients admitted with COVID-19: a randomized clinical trial
.
JAMA
2021
;
325
:
254
64
.

128

Cohen
JB
,
Hanff
TC
,
William
P
,
Sweitzer
N
,
Rosado-Santander
NR
,
Medina
C
, et al.
Continuation versus discontinuation of renin-angiotensin system inhibitors in patients admitted to hospital with COVID-19: a prospective, randomised, open-label trial
.
Lancet Respir Med
2021
;
9
:
275
84
.

129

Ostermann
M
,
Zarbock
A
,
Goldstein
S
,
Kashani
K
,
Macedo
E
,
Murugan
R
, et al.
Recommendations on acute kidney injury biomarkers from the acute disease quality initiative consensus conference: a consensus statement
.
JAMA Netw Open
2020
;
3
:
e2019209
.

130

Musial
K.
Update on innate immunity in acute kidney Injury-Lessons taken from COVID-19
.
Int J Mol Sci
2022
;
23
:
12514
.

131

Chen
Y
,
Li
X
,
Lin
X
,
Liang
H
,
Liu
X
,
Zhang
X
, et al.
Complement C5a induces the generation of neutrophil extracellular traps by inhibiting mitochondrial STAT3 to promote the development of arterial thrombosis
.
Thromb J
2022
;
20
:
24
.

132

Dudoignon
E
,
Moreno
N
,
Deniau
B
,
Coutrot
M
,
Longer
R
,
Amiot
Q
, et al.
Activation of the renin-angiotensin-aldosterone system is associated with acute kidney injury in COVID-19
.
Anaesth Crit Care Pain Med
2020
;
39
:
453
5
.

133

Wang
K
,
Chen
W
,
Zhang
Z
,
Deng
Y
,
Lian
JQ
,
Du
P
, et al.
CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells
.
Signal Transduct Target Ther
2020
;
5
:
283
.

134

Kosugi
T
,
Maeda
K
,
Sato
W
,
Maruyama
S
,
Kadomatsu
K.
CD147 (EMMPRIN/Basigin) in kidney diseases: from an inflammation and immune system viewpoint
.
Nephrol Dial Transplant
2015
;
30
:
1097
103
.

135

Venkatesan
B
,
Valente
AJ
,
Prabhu
SD
,
Shanmugam
P
,
Delafontaine
P
,
Chandrasekar
B.
EMMPRIN activates multiple transcription factors in cardiomyocytes, and induces interleukin-18 expression via Rac1-dependent PI3K/Akt/IKK/NF-kappaB andMKK7/JNK/AP-1 signaling
.
J Mol Cell Cardiol
2010
;
49
:
655
63
.

136

Leisman
DE
,
Ronner
L
,
Pinotti
R
,
Taylor
MD
,
Sinha
P
,
Calfee
CS
, et al.
Cytokine elevation in severe and critical COVID-19: a rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes
.
Lancet Respir Med
2020
;
8
:
1233
44
.

137

Cantaluppi
V
,
Assenzio
B
,
Pasero
D
,
Romanazzi
GM
,
Pacitti
A
,
Lanfranco
G
, et al.
Polymyxin-B hemoperfusion inactivates circulating proapoptotic factors
.
Intensive Care Med
2008
;
34
:
1638
45
.

138

Zarbock
A
,
Gomez
H
,
Kellum
JA.
Sepsis-induced acute kidney injury revisited: pathophysiology, prevention and future therapies
.
Curr Opin Crit Care
2014
;
20
:
588
95
.

139

Darmon
M
,
Schortgen
F
,
Vargas
F
,
Liazydi
A
,
Schlemmer
B
,
Brun-Buisson
C
, et al.
Diagnostic accuracy of Doppler renal resistive index for reversibility of acute kidney injury in critically ill patients
.
Intensive Care Med
2011
;
37
:
68
76
.

140

Legrand
M
,
Mik
EG
,
Johannes
T
,
Payen
D
,
Ince
C.
Renal hypoxia and dysoxia after reperfusion of the ischemic kidney
.
Mol Med
2008
;
14
:
502
16
.

141

Ronco
C
,
Reis
T.
Kidney involvement in COVID-19 and rationale for extracorporeal therapies
.
Nat Rev Nephrol
2020
;
16
:
308
10
.

142

Koyner
JL
,
Murray
PT.
Mechanical ventilation and the kidney
.
Blood Purif
2010
;
29
:
52
68
.

143

Schmidt
M
,
Hajage
D
,
Lebreton
G
,
Monsel
A
,
Voiriot
G
,
Levy
D
, et al. ;
Paris-Sorbonne ECMO-COVID Investigators
.
Extracorporeal membrane oxygenation for severe acute respiratory distress syndrome associated with COVID-19: a retrospective cohort study
.
Lancet Respir Med
2020
;
8
:
1121
31
.

144

Wishahi
M
,
Kamal
NM.
Multidisciplinary basic and clinical research of acute kidney injury with COVID-19: pathophysiology, mechanisms, incidence, management and kidney transplantation
.
World J Nephrol
2022
;
11
:
105
14
.

145

Shetty
AA
,
Tawhari
I
,
Safar-Boueri
L
,
Seif
N
,
Alahmadi
A
,
Gargiulo
R
, et al.
COVID-19-associated glomerular disease
.
J Am Soc Nephrol
2021
;
32
:
33
40
.

146

Velez
JCQ
,
Caza
T
,
Larsen
CP.
COVAN is the new HIVAN: the re-emergence of collapsing glomerulopathy with COVID-19
.
Nat Rev Nephrol
2020
;
16
:
565
7
.

147

Vojdani
A
,
Kharrazian
D.
Potential antigenic cross-reactivity between SARS-CoV-2 and human tissue with a possible link to an increase in autoimmune diseases
.
Clin Immunol
2020
;
217
:
108480
.

148

Ejemel
M
,
Li
Q
,
Hou
S
,
Schiller
ZA
,
Tree
JA
,
Wallace
A
, et al.
A cross-reactive human IgA monoclonal antibody blocks SARS-CoV-2 spike-ACE2 interaction
.
Nat Commun
2020
;
11
:
4198
.

149

Ma
Y
,
Huang
Y
,
Xu
G.
New insights into the mucosal immune pathogenesis of IgA nephropathy from the perspective of COVID-19 vaccination
.
QJM
2023
;
116
:
181
95
.

150

Patel
C
,
Shah
HH.
Membranous nephropathy and severe acute kidney injury following influenza vaccination
.
Saudi J Kidney Dis Transpl
2015
;
26
:
1289
93
.

151

Wu
Z
,
Hu
Y
,
Xu
M
,
Chen
Z
,
Yang
W
,
Jiang
Z
, et al.
Safety, tolerability, and immunogenicity of an inactivated SARS-CoV-2 vaccine (CoronaVac) in healthy adults aged 60 years and older: a randomised, double-blind, placebo-controlled, phase 1/2 clinical trial
.
Lancet Infect Dis
2021
;
21
:
803
12
.

152

Perricone
C
,
Colafrancesco
S
,
Mazor
RD
,
Soriano
A
,
Agmon-Levin
N
,
Shoenfeld
Y.
Autoimmune/inflammatory syndrome induced by adjuvants (ASIA) 2013: unveiling the pathogenic, clinical and diagnostic aspects
.
J Autoimmun
2013
;
47
:
1
16
.

153

Shoenfeld
Y
,
Agmon-Levin
N.
‘ASIA’ - autoimmune/inflammatory syndrome induced by adjuvants
.
J Autoimmun
2011
;
36
:
4
8
.

154

Shoenfeld
Y
,
Gilburd
B
,
Abu-Shakra
M
,
Amital
H
,
Barzilai
O
,
Berkun
Y
, et al.
The mosaic of autoimmunity: genetic factors involved in autoimmune diseases–2008
.
Isr Med Assoc J
2008
;
10
:
3
7
.

155

Cugno
M
,
Meroni
PL
,
Gualtierotti
R
,
Griffini
S
,
Grovetti
E
,
Torri
A
, et al.
Complement activation in patients with COVID-19: a novel therapeutic target
.
J Allergy Clin Immunol
2020
;
146
:
215
7
.

156

Henry
BM
,
Szergyuk
I
,
de Oliveira
MHS
,
Lippi
G
,
Benoit
JL
,
Vikse
J
, et al.
Complement levels at admission as a reflection of coronavirus disease 2019 (COVID-19) severity state
.
J Med Virol
2021
;
93
:
5515
22
.

157

Yu
J
,
Yuan
X
,
Chen
H
,
Chaturvedi
S
,
Braunstein
EM
,
Brodsky
RA.
Direct activation of the alternative complement pathway by SARS-CoV-2 spike proteins is blocked by factor D inhibition
.
Blood
2020
;
136
:
2080
9
.

158

Izci Duran
T
,
Turkmen
E
,
Dilek
M
,
Sayarlioglu
H
,
Arik
N.
ANCA-associated vasculitis after COVID-19
.
Rheumatol Int
2021
;
41
:
1523
9
.

159

Pelka
K
,
Shibata
T
,
Miyake
K
,
Latz
E.
Nucleic acid-sensing TLRs and autoimmunity: novel insights from structural and cell biology
.
Immunol Rev
2016
;
269
:
60
75
.

160

Nakazawa
D
,
Masuda
S
,
Tomaru
U
,
Ishizu
A.
Pathogenesis and therapeutic interventions for ANCA-associated vasculitis
.
Nat Rev Rheumatol
2019
;
15
:
91
101
.

161

Xiao
G
,
Hu
H
,
Wu
F
,
Sha
T
,
Zeng
Z
,
Huang
Q
, et al.
Acute kidney injury in patients hospitalized with COVID-19 in Wuhan, China: a single-center retrospective observational study
.
Nan Fang Yi Ke Da Xue Xue Bao
2021
;
41
:
157
63
.

162

Werion
A
,
Belkhir
L
,
Perrot
M
,
Schmit
G
,
Aydin
S
,
Chen
Z
, et al. ;
Cliniques universitaires Saint-Luc (CUSL) COVID-19 Research Group
.
SARS-CoV-2 causes a specific dysfunction of the kidney proximal tubule
.
Kidney Int
2020
;
98
:
1296
307
.

163

Tornling
G
,
Batta
R
,
Porter
JC
,
Williams
B
,
Bengtsson
T
,
Parmar
K
, et al.
Seven days treatment with the angiotensin II type 2 receptor agonist C21 in hospitalized COVID-19 patients; a placebo-controlled randomised multi-Centre double-blind phase 2 trial
.
EClinicalMedicine
2021
;
41
:
101152
.

164

Bauer
A
,
Schreinlechner
M
,
Sappler
N
,
Dolejsi
T
,
Tilg
H
,
Aulinger
BA
, et al. ;
ACEI-COVID investigators
.
Discontinuation versus continuation of renin-angiotensin-system inhibitors in COVID-19 (ACEI-COVID): a prospective, parallel group, randomised, controlled, open-label trial
.
Lancet Respir Med
2021
;
9
:
863
72
.

165

Diebold
M
,
Locher
E
,
Boide
P
,
Enzler-Tschudy
A
,
Faivre
A
,
Fischer
I
, et al.
Incidence of new onset glomerulonephritis after SARS-CoV-2 mRNA vaccination is not increased
.
Kidney Int
2022
;
102
:
1409
19
.

166

Cheng
FWT
,
Wong
CKH
,
Qin
SX
,
Chui
CSL
,
Lai
FTT
,
Li
X
, et al.
Risk of glomerular diseases, proteinuria and hematuria following mRNA (BNT162b2) and inactivated (CoronaVac) SARS-CoV-2 vaccines
.
Nephrol Dial Transplant
2023
;
38
:
129
37
.

167

Canney
M
,
Atiquzzaman
M
,
Cunningham
AM
,
Zheng
Y
,
Er
L
,
Hawken
S
, et al.
A population-based analysis of the risk of glomerular disease relapse after COVID-19 vaccination
.
J Am Soc Nephrol
2022
;
33
:
2247
57
.

168

Chen
G
,
Ren
Q
,
Zhou
J
,
Zhou
Y
,
Luo
H
,
Wang
Y
, et al.
Association of current active illnesses and severe acute kidney injury after COVID-19 vaccines: a real-world study
.
Vaccines (Basel)
2022
;
10
:
706
.

169

Samaan
F
,
Carneiro de Paula
E
,
de Lima Souza
FBG
,
Mendes
LFC
,
Rossi
PRG
,
Freitas
RAP
, et al.
COVID-19-associated acute kidney injury patients treated with renal replacement therapy in the intensive care unit: a multicenter study in Sao Paulo, Brazil
.
PLoS One
2022
;
17
:
e0261958
.

Author notes

These authors contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data