4,499
Views
6
CrossRef citations to date
0
Altmetric
Infectious Diseases

Assessing the risk and costs of COVID-19 in immunocompromised populations in a large United States commercial insurance health plan: the EPOCH-US Study

, , , , , , , , & show all
Pages 1103-1118 | Received 24 May 2023, Accepted 04 Jul 2023, Published online: 17 Jul 2023

Abstract

Objective

To estimate the prevalence of patients with an immunocompromising condition at risk for COVID-19, estimate COVID-19 prevalence rate (PR) and incidence rate (IR) by immunocompromising condition, and describe COVID-19-related healthcare resource utilization (HCRU) and costs.

Methods

Using the Healthcare Integrated Research Database (HIRD), patients with ≥1 claim for an immunocompromising condition of interest or ≥2 claims for an immunosuppressive (IS) treatment and COVID-19 diagnosis during the infection period (1 April 2020–31 March 2022) and had ≥12 months baseline data were included. Cohorts (other than the composite cohort) were not mutually exclusive and were defined by each immunocompromising condition. Analyses were descriptive in nature.

Results

Of the 16,873,161 patients in the source population, 2.7% (n = 458,049) were immunocompromised (IC). The COVID-19 IR for the composite IC cohort during the study period was 101.3 per 1000 person-years and the PR was 13.5%. The highest IR (195.0 per 1000 person-years) and PR (20.1%) were seen in the end-stage renal disease (ESRD) cohort; the lowest IR (68.3 per 1000 person-years) and PR (9.4%) were seen in the hematologic or solid tumor malignancy cohort. Mean costs for hospitalizations associated with the first COVID-19 diagnosis were estimated at nearly $1 billion (2021 United States dollars [USD]) for 14,516 IC patients, with a mean cost of $64,029 per patient.

Conclusions

Immunocompromised populations appear to be at substantial risk of severe COVID-19 outcomes, leading to increased costs and HCRU. Effective prophylactic options are still needed for these high-risk populations as the COVID-19 landscape evolves.

PLAIN LANGUAGE SUMMARY

People who have a medical condition or take a medicine that can suppress their immune system (immunocompromised) have a high risk of getting COVID-19. Our study looked at how many immunocompromised people got COVID-19. We also looked at the costs and lengths of hospital stays for people with COVID-19. We found that 2.7% of the people in this large US population with health insurance were immunocompromised. People who were immunocompromised were more likely to get COVID-19 than people who were not immunocompromised. About 14% of the immunocompromised people in this study got COVID-19 and, of those, 24% were hospitalized. Immunocompromised patients in this study had long hospital stays and high costs associated with COVID-19. The risk of getting COVID-19 and having a severe case seemed to be highest for people with advanced kidney disease. The study results showed that COVID-19 can cause severe health issues in immunocompromised people and the use of vaccinations, medications, and other measures to prevent COVID-19 are especially important for immunocompromised people.

Introduction

The global coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused more than 757 million confirmed infections and over 6.8 million deaths globally since the start of the pandemic in 2019Citation1. In the United States (US), more than 103 million cases and 1.1 million deaths have been attributed to COVID-19Citation2. Certain populations, such as those with hematologic malignancies and recipients of hematopoietic stem cell or solid organ transplants, have significantly impaired immune systems; these immunocompromised (IC) patients have an increased vulnerability to SARS-CoV-2 and are at higher risk for severe COVID-19 outcomes, including prolonged hospitalization, greater healthcare resource utilization (HCRU), and deathCitation3–9. A representative sample of 22,345 hospitalized adults from 10 states in the COVID-19 Associated Hospital Surveillance Network (COVID-NET; 1 March 2020–28 February 2022) showed that 12.2% of all hospitalized patients were immunocompromisedCitation10. Further, the IC patients had a higher risk of intensive care unit (ICU) admission and death compared to immunocompetent patients.

In December 2020, approval to begin vaccination against COVID-19 caused by SARS-CoV-2 was granted by the US Food and Drug Administration (FDA)Citation11. Despite appropriate COVID-19 vaccination uptake, some populations may not mount an adequate immune response to the COVID-19 vaccines due to underlying immunocompromising conditions and/or use of immunosuppressive (IS) treatment, which may place them at increased risk for more severe COVID-related outcomesCitation12–20. Underlying conditions that have been shown to decrease COVID-19 vaccine effectiveness and place individuals at risk for more severe outcomes related to COVID-19 include hematologic malignanciesCitation12,Citation13,Citation21,Citation22 and a history of solid organ transplantCitation12–15,Citation23–26. Immunosuppressive medications such as those used to prevent allograft rejection among solid organ transplant recipients can impact immune response to vaccination in generalCitation27,Citation28, with documented poor antibody response to COVID-19 vaccinationCitation12–15,Citation23–26. Chronic kidney disease (CKD; particularly end-stage renal disease or dialysis)Citation29,Citation30, primary immunodeficienciesCitation12,Citation31–33, and use of IS or immunomodulatory medications (e.g. chemotherapy in patients with cancer; sphingosine-1-phosphate receptor modulators in patients with multiple sclerosis) may also cause immune system impairment leading to a higher risk of severe COVID-19-related outcomesCitation8,Citation9. Age-related decline in immune function has also been observed, which may lead to a greater risk of SARS-CoV-2 infection and severe outcomesCitation15,Citation34. However, post-immunization measurement to assess levels of immunity to SARS-CoV-2 is not recommended based on current guidelinesCitation35.

The prevalence of immune suppression/compromise due to conditions or medications has been estimated to be between 2% and 3% in the US populationCitation36,Citation37. Although several studies have examined immunocompromising risk factors associated with breakthrough COVID-19Citation12,Citation15,Citation34, the incidence and severity of COVID-19 in these IC populations are not well understood. This is in part because patients with immunocompromising conditions were largely excluded from SARS-CoV-2 vaccine clinical trialsCitation38–40. Furthermore, there is limited information on the impact of COVID-19 on HCRU and healthcare costs in IC populations in the US. This study aimed to estimate the prevalence of IC populations who are potentially at risk for a suboptimal COVID-19 vaccine immune response and subsequent SARS-CoV-2 infection; estimate the incidence of COVID-19 stratified by immunocompromising condition; and describe the HCRU and healthcare costs associated with COVID-19 in these populations. Results from this study will further the general understanding of the impact of COVID-19 on IC populations and inform the management of these patients and their prevention and treatment options, particularly among a commercially insured population.

Methods

Study design

This retrospective, observational cohort study was conducted among US patients identified in the Healthcare Integrated Research Database (HIRD)® (formerly the HealthCore Integrated Research Database) between 1 April 2018, and 31 March 2022 (study period). The HIRD is a large healthcare database curated by Carelon Research (formerly HealthCore) that is built on a broad, clinically rich, and geographically diverse spectrum of longitudinal medical and pharmacy claims from health plan members across the US. Member enrollment, medical care (professional and facility claims), outpatient prescription drug use (pharmacy claims), and healthcare utilization is tracked for health plan members in the database dating back to January 2006. The HIRD contains neighborhood-level characteristics at the census block group level relevant to social determinants of health (SDOH) obtained from the 2017 American Community SurveyCitation41.

All data were accessed as a limited data set for which a business associate and data use agreement was in place with the covered entities in compliance with the Health Insurance Portability and Accountability Act (HIPAA) Privacy Rule. Data was anonymized and thus exempt from Institutional Review Board review.

Study population

To be included in the study, patients with commercial or Medicare (Medicare Advantage, Supplemental, or Part D) health insurance coverage were required to have ≥1 claim for an immunocompromising condition of interest (hematologic or solid tumor malignancy [HSTM]; hematopoietic stem cell transplant [HSCT] or solid organ transplant [SOT]; primary immunodeficiency [PID]; or end-stage renal disease [ESRD; defined as patients with CKD stage 5, ESRD, or history of dialysis]) within the study period, or ≥2 fills or claims for an IS treatment from 1 October 2019 to the end of the study period (). The index date was defined as the date of diagnosis of an immunocompromising condition or use of an IS treatment in the study period, or 1 April 2020 (the first day of the infection period), whichever came last. Only actively treated patients with HSTM were included. Active treatment was identified as patients with ≥1 claim for an antineoplastic medication from 6 months before the index date to the date of the COVID-19 diagnosis, member’s health plan disenrollment, death, or end of the study period, whichever came first. Individual IC cohorts were not mutually exclusive and were defined by each immunocompromising condition; however, a mutually exclusive composite cohort of the overall IC population was also created. The HIRD source population was defined as patients with ≥12 months of continuous pharmacy and medical benefit enrollment between 1 April 2019 and 31 March 2022. The index date for the HIRD source population was set to day 365 after the first day of continuous enrollment. The infection period was defined as 1 April 2020 to 31 March 2022 (as 1 April 2020 was the date when the specific ICD-10 diagnosis code for COVID-19 [U07.1] was released in the US); the baseline period was defined as 1 April 2018 to the index date. All patients in the IC cohorts and the HIRD source population were required to have ≥12 months of continuous medical and pharmacy health plan enrollment prior to the index date.

Figure 1. Study timeline.

aUse of IS treatment was assessed starting on 1 October 2019. For all other cohorts, risk was assessed beginning on 1 April 2018.

Key: IS: immunosuppressive.

Figure 1. Study timeline.aUse of IS treatment was assessed starting on 1 October 2019. For all other cohorts, risk was assessed beginning on 1 April 2018.Key: IS: immunosuppressive.

Outcomes

Outcomes were assessed for each individual IC cohort and the overall IC population (composite IC cohort). Evaluated measures included the prevalence of immunocompromising conditions and use of IS treatments; baseline demographic and clinical characteristics; prevalence and incidence of COVID-19; and length of stay (LOS) and costs (health plan paid + patient paid) associated with COVID-19 hospitalization. All-cause HCRU and paid costs were also captured in the 30-day pre-COVID-19 period and the 30-day post-COVID-19 period among patients who had at least 30 days of post-infection continuous enrollment. Costs were adjusted to 2021 US dollars (USD) according to medical care price index information provided by the US Bureau of Labor StatisticsCitation42. The baseline period was defined as the study start date to the index date (inclusive). Baseline demographic characteristics included sex, age on index date, age group, and payor type. Baseline clinical characteristics included the Quan-Charlson Comorbidity Index (QCI) score (range: 0–24; higher scores are associated with a higher likelihood of death within 1 year of hospital admission)Citation43 and comorbidities of interest, as well as the categorical number of immunocompromising conditions and IS treatments of interest. Prevalence of COVID-19 and available baseline demographic and clinical characteristics were also reported for the overall HIRD cohort. Antiviral medication use was identified using medical and pharmacy claims for remdesivir, and pharmacy claims for nirmatrelvir/ritonavir, baricitinib, or molnupiravir in the 30-day pre- and post-COVID-19 periods.

A SARS-CoV-2 infection was identified based on a positive SARS-CoV-2 polymerase chain reaction (PCR) or antigen test analyzed by accredited laboratories, or a medical encounter (inpatient, emergency room [ER], office visit, urgent care, or telehealth visit) for treatment/evaluation and management/consultation with a COVID-19 diagnosis code at any position on the medical claim. Severity of COVID-19 was classified into the following mutually exclusive categories: severe/critical, moderate, or mild/asymptomatic. Severe/critical COVID-19 was defined as an inpatient admission for COVID-19 with an ICU admission and one of the following: evidence of extracorporeal membrane oxygenation (ECMO), mechanical ventilation use, administration of high-flow oxygen via nasal cannula, or discharge status of expired. Moderate COVID-19 was defined as an inpatient admission for COVID-19 that did not meet any of the criteria for severe/critical COVID-19. Mild COVID-19 was defined as an outpatient claim for COVID-19 (e.g. office, telehealth, or stand-alone ER visit) without any inpatient claims, while asymptomatic COVID-19 was defined as a positive SARS-CoV-2 lab result with no inpatient or outpatient COVID-19 claims. These two categories (mild and asymptomatic) were combined as it was not possible to fully ascertain whether the patient had symptoms but did not seek medical care.

Statistical analysis

This study was descriptive with no statistical comparisons between cohorts. Descriptive statistics were reported as mean and standard deviation (SD) or median and interquartile range (IQR) for continuous data, and absolute and relative frequencies for categorical data. The prevalence of immune compromise was calculated as the number of patients with an immunocompromising condition or using an IS treatment in the infection period divided by the total number of patients with ≥12 months of continuous pharmacy and medical benefit enrollment prior to the index date. The prevalence of COVID-19 was calculated as the number of patients who developed COVID-19 in the infection period divided by the total number of patients within each cohort of interest with ≥12 months of continuous pharmacy and medical benefit enrollment prior to the index date. Incidence rates (IRs) of COVID-19 during the infection period were reported with 95% confidence intervals (CIs). Incidence rates were calculated as the number of new cases of COVID-19 during the infection period divided by the total person-years in each individual IC cohort and the composite IC cohort during the infection period. Both overall and monthly IRs were calculated. In addition, overall IRs by severity of COVID-19 were assessed. The frequency, LOS, and total all-cause costs of hospitalizations associated with the first COVID-19 diagnosis were stratified by severity and reported for all IC cohorts. Total aggregate costs for COVID-19-related hospitalizations were calculated by multiplying the total number of ICU and non-ICU hospitalized patients by the mean total all-cause costs for ICU and non-ICU hospitalizations, respectively.

Sample selection, creation of analytic variables, and analyses were performed using the Instant Health Data (IHD) platform (Panalgo, Boston, MA, US) and Statistical Analysis System (SAS), version 9.3 (SAS Institute Inc, Cary, NC, US).

Results

Study population

A total of 22,858,635 patients with commercial or Medicare Advantage/Supplemental/Part D health insurance were identified within the HIRD during the infection period. Of these, there were 1,518,358 (6.6%) unique members with ≥1 claim for an immunocompromising condition or ≥2 claims for an IS treatment during the study period (). After applying the study's continuous enrollment criteria, a total of 16,873,161 patients remained in the overall HIRD cohort. Once the active treatment criteria for patients with HSTM were applied, there were 458,049 (2.7%) unique members identified as IC patients in the composite IC cohort. The individual IC cohorts were not mutually exclusive, and they included 259,914 (1.5%) patients in the IS treatment cohort; 165,081 (1.0%) patients in the HSTM cohort; 84,610 (0.5%) patients in the PID cohort; 48,131 (0.3%) patients in the ESRD cohort; and 28,698 (0.2%) patients in the HSCT/SOT cohort.

Figure 2. Patient selection and attrition.

Study period: 1 April 2018 through 31 March 2022.

Use of IS treatment was assessed starting on 1 October 2019. For all other cohorts, risk was assessed beginning on 1April 2018.

Key: ESRD: end-stage renal disease; HIRD: healthcare integrated research database; HSCT: hematopoietic stem cell transplant; HSTM: hematologic or solid tumor malignancy; IS: immunosuppressive; PID: primary immunodeficiency; SOT: solid organ transplant.

Figure 2. Patient selection and attrition.Study period: 1 April 2018 through 31 March 2022.Use of IS treatment was assessed starting on 1 October 2019. For all other cohorts, risk was assessed beginning on 1April 2018.Key: ESRD: end-stage renal disease; HIRD: healthcare integrated research database; HSCT: hematopoietic stem cell transplant; HSTM: hematologic or solid tumor malignancy; IS: immunosuppressive; PID: primary immunodeficiency; SOT: solid organ transplant.

Baseline patient characteristics

Baseline and clinical characteristics varied among the cohorts. The median age for the composite IC cohort was 58 years (). Depending on the cohort, 42.1% to 64.8% of IC patients were females, and 64.3% to 85.8% of IC patients had commercial insurance coverage. Among all IC cohorts, the mean proportions of distribution of ethno-racial groups at the census level were: 4.6% to 5.4% Asian; 9.5% to 14.2% non-Hispanic Black; 63.9% to 70.7% non-Hispanic White; and 11.6% to 14.2% Hispanic/Latino of any race. Approximately twice as many patients across all IC cohorts were in the highest socioeconomic status (SES) quartile compared to the lowest SES quartile, with the exception of the ESRD cohort (which had a similar number of patients in the highest and lowest SES quartiles). The overall HIRD population was substantially younger than the IC population (median age: 38 years), 50.5% were females, and 93.4% were commercially insured.

Table 1. Demographics on index date.

Patients in the overall HIRD population appeared healthier, with fewer comorbidities than patients in the IC population; the HIRD population had a median QCI score of 0, while the median QCI scores ranged from 1 to 4 across the IC cohorts (). The ESRD cohort had the highest QCI median score (4), and the IS treatment cohort had the lowest median score (1). The five comorbidities with the highest prevalence in the composite IC cohort included hypertension (55.1%), cancer (44.2%), obesity (31.0%), atherosclerotic cardiovascular disease (ASCVD; 27.8%), and chronic pulmonary disease (27.2%; including asthma and chronic obstructive pulmonary disease [COPD]). In contrast, the five comorbidities with the highest prevalence in the HIRD population were hypertension (21.7%), obesity (15.2%), anxiety (14.5%), depression (11.6%), and chronic pulmonary disease (10.7%). Comorbidities for the individual cohorts are shown in .

Table 2. Baseline (variable) clinical characteristics assessed from study start to index date.

While the median number of immunocompromising conditions for patients in most of the individual IC cohorts was 1 (except for PID and HSCT/SOT, where the median was 2), 19.8% of the composite IC cohort had ≥2 immunocompromising conditions (). The majority of patients in the HSTM cohort (90.8%) had solid tumor malignancies (). Among the HSCT/SOT cohort, the majority (73.3%) received a solid organ transplant (with kidney transplants being the most common [42.8%]). Among the ESRD cohort, the majority of patients (84.1%) were on dialysis. Immunocompromising conditions at baseline for the individual cohorts are shown in .

Figure 3. Categorical number of immunocompromising conditions per IC cohort.

Key: ESRD: end-stage renal disease; HSCT: hematopoietic stem cell transplant; HSTM: hematologic or solid tumor malignancy; IC: immunocompromised; IS: immunosuppressive; PID: primary immunodeficiency; SOT: solid organ transplant.

Note: Due to rounding, percentages may not add up to 100%.

Figure 3. Categorical number of immunocompromising conditions per IC cohort.Key: ESRD: end-stage renal disease; HSCT: hematopoietic stem cell transplant; HSTM: hematologic or solid tumor malignancy; IC: immunocompromised; IS: immunosuppressive; PID: primary immunodeficiency; SOT: solid organ transplant.Note: Due to rounding, percentages may not add up to 100%.

Table 3. Baseline immunocompromising conditions and is therapies.

Within the IS treatment cohort, the most commonly used treatments were high-dose corticosteroids (33.1%; defined as ≥20 mg of prednisone or equivalent daily for ≥2 weeks), antimetabolites (30.0%), tumor necrosis factor (TNF) inhibitors (22.6%), and alkylating agents (14.9%). A majority (57.8%) of patients in the HSCT/SOT cohort used IS treatments at baseline; of the remaining IC cohorts, 53.1% in the composite IC cohort, 46.8% in the PID cohort, 23.0% in the HSTM cohort, and 22.0% in the ESRD cohort used IS treatments at baseline. Specific IS treatments used by patients across all IC cohorts are shown in .

COVID-19 prevalence and incidence

Overall, 13.5% (n = 61,865) of IC patients developed COVID-19 during the follow-up period. Within the individual IC cohorts, ESRD had the highest proportion of patients who developed COVID-19 (20.1%), followed by PID (15.8%), HSCT/SOT (15.7%), IS treatment (13.8%), and HSTM (9.4%). In the HIRD source population, 9.7% of members developed COVID-19 during the follow-up period.

Monthly IRs of COVID-19 per 1000 person-years for all IC cohorts are shown in . displays the weekly COVID-19 case rates per 100,000 population published by the Centers for Disease Control and Prevention (CDC)Citation44, which shows a similar pattern of COVID-19 rate surges and troughs. The overall IR of COVID-19 was 101.3 per 1000 person-years in the composite IC cohort. Within the individual IC cohorts, ESRD had the highest overall IR of COVID-19 (195.0 per 1000 person-years) and the highest IR of severe/critical COVID-19 (79.4 per 1000 person-years), while HSTM had the lowest overall IR of COVID-19 (68.3 per 1000 person-years) and the lowest IR of severe/critical COVID-19 (8.4 per 1000 person-years). Incidence rates of COVID-19 by severity are shown for each IC cohort in . Overall and monthly IRs of COVID-19 in the HIRD source population were not available at the time of submission.

Figure 4. A. Post-index incidence rate of COVID-19 per 1000 person-years. B. US weekly case rate of COVID-19 per 100,000 population as reported by the CDC.

Key: CDC: Centers for Disease Control and Prevention; US: United States.

The dotted lines in represent the transition from different variants throughout the COVID-19 pandemic. Data on variants were obtained from GISAID and CDC Nowcast. [Citation62,Citation63]

The shaded portion in represents the weekly case rate per 100,000 population as reported by the CDC from 1 April 2020 to 31 March 2022Citation44.

Figure 4. A. Post-index incidence rate of COVID-19 per 1000 person-years. B. US weekly case rate of COVID-19 per 100,000 population as reported by the CDC.Key: CDC: Centers for Disease Control and Prevention; US: United States.The dotted lines in Figures 4(A,B) represent the transition from different variants throughout the COVID-19 pandemic. Data on variants were obtained from GISAID and CDC Nowcast. [Citation62,Citation63]The shaded portion in Figure 4(B) represents the weekly case rate per 100,000 population as reported by the CDC from 1 April 2020 to 31 March 2022Citation44.

Figure 5. Post-index incidence rate of COVID-19 by severity per 1000 person-years.

Key: ESRD: end-stage renal disease; HSCT: hematopoietic stem cell transplant; HSTM: hematologic or solid tumor malignancy; IC: immunocompromised; IS: immunosuppressive; PID: primary immunodeficiency; SOT: solid organ transplant.

Figure 5. Post-index incidence rate of COVID-19 by severity per 1000 person-years.Key: ESRD: end-stage renal disease; HSCT: hematopoietic stem cell transplant; HSTM: hematologic or solid tumor malignancy; IC: immunocompromised; IS: immunosuppressive; PID: primary immunodeficiency; SOT: solid organ transplant.

HCRU and healthcare costs associated with COVID-19 hospitalization

Within the composite IC cohort, 23.5% of patients (n = 14,516) had hospitalizations associated with their first COVID-19 diagnosis, with a fairly even split between moderate/non-ICU (n = 7446) and severe/critical/ICU (n = 7070) hospitalizations. Rates of COVID-19-related hospitalizations varied across the individual IC cohorts, from 15.8% (n = 5646) in the IS treatment cohort to 65.1% (n = 6294) in the ESRD cohort (). The mean LOS associated with the first COVID-19 hospitalization was 15 days in the composite IC cohort, with a mean LOS of 11 days for moderate disease (non-ICU hospitalization) and 20 days for severe/critical disease (ICU hospitalization). Within the individual IC cohorts, LOS ranged from 10 days in the HSTM cohort to 23 days in the ESRD cohort.

Table 4. HCRU and costs associated with hospitalization for first COVID-19 diagnosis.

Mean total costs (health plan paid + patient paid) associated with the first COVID-19 hospitalization were $64,029 in the composite IC cohort, with costs for severe/critical disease ($96,829) noted to be roughly 3 times higher than those for moderate disease ($32,885). Within the individual IC cohorts, overall hospitalization costs ranged from $30,671 in the HSTM cohort to $102,224 in the ESRD cohort (). Median costs were generally lower, regardless of COVID-19 severity or IC cohort. Mean aggregate costs totaled $929,442,740 for the 14,516 IC patients with hospitalizations associated with their first COVID-19 diagnosis.

All-cause HCRU and costs paid by health plans and patients were captured in the 30-day pre-COVID-19 period (as a pre-COVID-19 “baseline”) and the 30-day post-COVID-19 period for IC patients who had at least 30 days of post-infection continuous enrollment (N = 30,437). The number and percentage of patients who had at least one hospitalization was almost 4 times higher (8173 [26.9%] vs 2143 [7.0%]) post-COVID-19 compared to pre-COVID-19 in the composite IC cohort. For ER visits, the number of patients who had at least one visit was more than 3 times greater (5997 [19.7%] vs 1806 [5.9%]) between the two periods, respectively. Total all-cause paid costs were approximately 2.5 times higher ($18,673 vs $7613) in the post-COVID-19 period than the pre-COVID-19 period, primarily driven by inpatient costs. Healthcare resource utilization and costs in the 30-day pre-COVID-19 and 30-day post-COVID-19 periods for all IC cohorts may be found in . Antiviral medications were used in the 30-day pre- and post-COVID-19 periods for less than 1% population among all cohorts.

Table 5. All-cause HCRU and costs in the 30-day pre-COVID-19 (“baseline”) period and the 30-day post-COVID-19 periodTable Footnotea.

Discussion

This analysis provides robust real-world data on COVID-19 risks and outcomes in IC populations in the US. Within the overall population, 2.7% of patients in this study were identified as IC (either due to an immunocompromising condition or the use of an IS treatment). This result aligns with the most recently available evidence showing an estimated prevalence of immune suppression/compromise of 2% to 3% in US adultsCitation36,Citation37.

The prevalence of COVID-19 among the IC cohorts in this study varied, ranging from 9.4% in the HSTM cohort to 20.1% in the ESRD cohort. In a US healthcare claims analysis of Medicare patients (median age: 66 years) undergoing chronic dialysis in 2020, the prevalence rate of COVID-19 was 12.1%Citation45. We surmise that the higher prevalence of COVID-19 seen in our ESRD cohort is likely due to the longer study period (1 April 2020–31 March 2022) and the different predominant variants, as well as evolving transmissibility and virulence seen with each variant over time. Patients receiving hemodialysis may also have a greater susceptibility to COVID-19 due to the procedure typically being performed in an outpatient setting three times per weekCitation46,Citation47. Interestingly, the prevalence of COVID-19 in the HIRD source population (9.7%) was slightly higher than that of the HSTM cohort (9.4%); this could be due to enhanced and consistent education from healthcare professionals, self-recognition of immune compromise, and continued lifestyle modifications to prevent SARS-CoV-2 infection (e.g. staying current with COVID-19 vaccinations/boosters, wearing a mask, washing hands frequently, physical distancing, and even self-isolation) in patients being actively treated for hematologic or solid tumor malignancies. While we did not examine patients with hematologic malignancies and solid tumor malignancies separately in this analysis, it is an area of interest for additional research.

Direct HCRU and costs paid by health plans and patients associated with COVID-19 hospitalizations were high across the IC cohorts, with the highest HCRU and costs seen in the ESRD cohort. Healthcare utilization and paid costs for IC patients in the 30 days prior to COVID-19 diagnosis provided a “baseline” and allowed us to observe the incremental impact of HCRU and costs due to COVID-19. In our study, the mean all-cause paid costs for the composite IC cohort in the 30-day pre-COVID-19 period were $7613 and increased to $18,673 in the 30-day post-COVID-19 period. In DeMartino 2022Citation48, a separate analysis of a US commercial healthcare plan, mean total incurred costs in patients without a COVID-19 diagnosis were $660 (2020 USD), and incurred costs in patients with a COVID-19 diagnosis were $4366 in the first month after the diagnosis. While the results from these studies are not directly comparable, we observed that the mean “baseline” paid costs for our composite IC population were 11.5 times higher than the mean total costs for the general population without COVID-19 in DeMartino 2022. Further, the mean paid costs for our composite IC population in the 30-day post-COVID-19 period were more than 4 times higher than the mean total costs for the general population with COVID-19 diagnosis in DeMartino 2022. Although our study and the DeMartino 2022 study differed in design and patient characteristics, both utilized administrative claims data sourced from large, nationally representative and commercially insured populations. Our results would indicate that total all-cause costs related to COVID-19 for IC populations appear to be substantially greater than those seen for the general population, even when taking the pre-COVID-19 “baseline” costs into account. This difference is likely due to the increased complexity of the IC population due to their underlying health condition(s), which may also complicate the SARS-CoV-2 infection.

In the composite IC cohort in the current study, the total aggregate paid costs associated with the first COVID-19 hospitalization for 14,516 IC patients were nearly $1 billion based on the mean cost per hospitalization; this suggests that these relatively few IC patients not only incur significant cost burden to the healthcare system but also that there are outliers with risks for much more severe COVID-19 outcomes within both the composite IC cohort and the individual IC cohorts who require additional protection. Another potential cost driver that was not analyzed in the current study was the impact of delayed or missed treatment for existing conditions due to COVID-19. In a 2020 survey of 11,114 Medicare beneficiaries, 36% reported missing treatment for an ongoing conditionCitation49. Missing or delaying treatment for IC conditions could potentially lead to worse outcomes and greater overall costs.

Based on the clinical characteristics observed within the IC population in this study, it is possible that an increased risk of severe COVID-19 outcomes is not solely due to immune compromise; it could also be related to the older median age and the increased number of comorbidities and other concomitant diseases seen in the IC cohorts compared to the overall HIRD source population. While we chose to investigate these specific IC populations for COVID-19-related risks and outcomes, it is important to note that these are not the only patients who can be classified as IC or may be at greater risk for severe COVID-19 and hospitalization. In addition, there are likely specific subgroups of patients within these individual IC cohorts, such as those with specific tumor types, longer duration of immunocompromising conditions, and longer duration/higher intensity of IS treatment, who are at even greater risk of severe COVID-19 outcomes than what was observed in the composite IC cohort. As the total aggregated costs in this population were quite significant, the identification of these patients warrants further study to ensure appropriate protection from COVID-19 (particularly with the emergence of new variants) and could result in the avoidance of some of these costs.

As the COVID-19 landscape has evolved, vaccinations and treatments have become available to help prevent and treat COVID-19. While patients with immunocompromising conditions tend to be highly vaccinated, they may not be able to mount a complete immune response, even when considered fully vaccinated and boostedCitation50–53. Immunocompromised patients have been shown to have lower concentrations of antibodies, and the antibodies that are present have less neutralization activity against SARS-CoV-2Citation54. In addition, the protection that vaccinations provide declines more quickly in comparison with immunocompetent patientsCitation55. Monoclonal antibodies that were previously available under an emergency use authorization are no longer authorized for use as they are not effective against the currently circulating predominant variants of SARS-CoV-2Citation56. Antiviral medications such as nirmatrelvir/ritonavir and molnupiravir that can decrease the risk of progression to severe COVID-19 remain available and can decrease the risk of progression to severe COVID-19 although each drug comes with related challengesCitation57,Citation58. Additionally, as much of the US population now has some form of immunity to SARS-CoV-2 (either through vaccination or exposure)Citation59, the use of other preventive measures such as masking and physical distancing has decreased in the general population. Staying current with COVID-19 vaccinations, masking, hand washing, and avoiding poorly ventilated areas are still recommended for IC patients, but an existing unmet need for additional pre-exposure prophylaxis for COVID-19 in patients with immunocompromising conditions remains. Several monoclonal antibodies are being studied in response to the emerging variants; one such long-acting monoclonal antibody is AZD3152, which is being evaluated for the pre-exposure prophylaxis of COVID-19 in patients aged ≥12 years with immunocompromising conditionsCitation60.

Additional interventions to prevent COVID-19-related hospitalizations in the IC population are not only important from an individual perspective but also from a broader healthcare system perspective. The US healthcare system was under immense strain at the onset of the pandemic, with limited resources and increased healthcare worker shortages. Hospitalizations in the IC population likely caused additional strain on the system. Though the COVID-19 public health emergency has ended, implementation of effective pre-exposure prophylactic options that can decrease the risk of COVID-19-related hospitalizations in the IC population is still needed and may have a positive impact on the healthcare system as the COVID-19 landscape continues to evolve.

While claims data are extremely valuable for the efficient and effective examination of healthcare outcomes, treatment patterns, HCRU, and healthcare costs, they are ultimately collected for the purpose of payment (and not for research). Therefore, limitations associated with the use of claims data exist. Any COVID-19 vaccination or testing data that did not initiate a health insurance claim (e.g. federal COVID-19 testing sites, at-home antigen tests, federal vaccination sites, etc.) would not be captured in the claims data. Some patients who are asymptomatic or have mild cases of COVID-19 may never seek testing or treatment and would not be captured in the claims data. In addition, individuals with immunocompromising conditions may be more likely to be tested for COVID-19 due to the risks associated with their underlying condition or overall increased interactions within the healthcare system. The sensitivity and specificity of PCR and antigen tests for diagnosis of COVID-19 are different and individuals may be incorrectly identified as COVID-19 cases due to the inclusion of results from these types of tests, although no home testing results were utilized. The presence of a diagnosis code on a medical claim does not guarantee the positive presence of a disease, as the diagnosis code may be incorrectly coded or included as a rule-out criterion. Certain important patient clinical characteristics (e.g. individual-level SDOH, family history, etc.) are not available in claims data, and their effect cannot be measured in the analyses. Further, it is not possible to fully assess the impact of COVID-19 on the severity of underlying conditions from claims data alone; however, the worsening of underlying conditions by COVID-19 has been observed in patients with some neurological disorders by other researchersCitation61. While this concept was not directly explored in this analysis, it could have an impact on multiple conditions that were studied. The observed utilization of antiviral medication may vary throughout the study period due to the evolving regulatory and clinical guidance associated with their use. As with all claims-based analyses, the study results may not be fully generalizable to the overall population because patients who have commercial or Medicare health insurance may have different characteristics from those with public or no health insurance. Patients who are uninsured, underinsured, or have limited access to general healthcare may have higher rates and related costs of COVID-19 than what was observed in the IC population in this study. Finally, this initial analysis only captures data through 31 March 2022 (through the early Omicron [BA.1/BA.2] wave) and does not include new variants. Additional data and ongoing analyses will be needed to understand the impact of new variants on the IC population.

Conclusion

The IC population tends to be highly vaccinated against COVID-19, yet they may remain inadequately protected due to an inability to produce a robust protective response to the vaccine. Results from this large US healthcare claims analysis show that patients with an immunocompromising condition or using IS treatment are at greater risk for COVID-19 and have a high prevalence of severe outcomes such as hospitalizations, as well as high HCRU and healthcare costs. While only 2.7% of the overall study population was IC, 13.5% of all IC patients in our study developed COVID-19 and 23.5% of those patients required hospitalization related to their initial COVID-19 diagnosis. Mean paid costs associated with these 14,516 COVID-19-related hospitalizations totaled nearly $1 billion, but median hospitalization costs suggest that there are outliers who are at even greater risk of very severe COVID-19 outcomes within both the overall IC population and individual IC cohorts. Given the general risks observed within the IC population and the known incomplete protection from vaccination, these patients require additional protective measures in order to resume social interactions and activities. Identification of these patients is an area of interest for further research, particularly as emerging variants may present new, unforeseen risks within the IC population. As the COVID-19 landscape continues to evolve, effective prophylactic options against COVID-19 are still needed to provide additional protection to IC patients.

Transparency

Declaration of financial/other relationships

C. Dube, L. Glasser, and M. Pollack are employees of AstraZeneca. M. Verduzco-Gutierrez has received honoraria and travel to give lectures related to long COVID-19. Unrelated to this work, she has been a consultant with AbbVie (consultant/advisor, speakers bureau), Merz (consultant/advisor, speakers bureau), Ipsen (grant/research, consultant/advisor, speakers bureau), Medtronic (consultant/advisor), and Piramal (speakers bureau). D. Cunningham has been a consultant/advisor for AstraZeneca. Carelon Research received funding from AstraZeneca to conduct this study. A. Ketkar, V. Willey, C. Wenziger, and C-C Teng are employees of Carelon Research (formerly HealthCore). A. Ketkar and V. Willey are shareholders of Elevance Health (formerly Anthem), which is a parent company of Carelon Research (formerly HealthCore). C. Dobie is an employee of Xcenda LLC and a consultant for AstraZeneca.

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Author contributions

Conception/design of the study: AK, VW, MP, LG, MVG, DC; Acquisition/analysis of the data: AK, VW, CW, CCT; Interpretation of the data: All authors; Drafting of the paper or revising it critically for intellectual content: All authors; Final approval of the version to be published: All authors. All authors agree to be accountable for all aspects of the work.

Acknowledgements

Authors would like to acknowledge Andi Gundlach, PharmD, CMPP of Xcenda LLC (Carrollton, TX) for her writing and editorial assistance, which was funded by AstraZeneca.

Data availability statement

Due to contractual obligations to the data sources, these data are not permitted to be available for use by outside parties.

Additional information

Funding

This work was supported by AstraZeneca.

References

  • World Health Organization Emergency Response Team. COVID-19 Weekly Epidemiological Update: World Health Organization; 2023 [updated 22 February 2023; cited 2023 1 March]; 131:[Available from: https://www.who.int/publications/m/item/weekly-epidemiological-update-on-covid-19–-22-february-2023.
  • Centers for Disease Control and Prevention. COVID Data Tracker. Daily Update for the United States Atlanta, Georgia: Centers for Disease Control and Prevention; 2023 updated 2 March 2023; cited 2023 3 March]. Available from: https://covid.cdc.gov/covid-data-tracker/#datatracker-home.
  • Clark A, Jit M, Warren-Gash C, et al. Global, regional, and national estimates of the population at increased risk of severe COVID-19 due to underlying health conditions in 2020: a modelling study. Lancet Glob Health. 2020; 8(8):e1003–e1017. doi: 10.1016/S2214-109X(20)30264-3.
  • Ryan C, Minc A, Caceres J, et al. Predicting severe outcomes in Covid-19 related illness using only patient demographics, comorbidities and symptoms. Am J Emerg Med. 2021; 45:378–384. doi: 10.1016/j.ajem.2020.09.017.
  • Velayos FS, Dusendang JR, Schmittdiel JA. Prior immunosuppressive therapy and severe illness among patients diagnosed with SARS-CoV-2: a community-based study. J Gen Intern Med. 2021; 36(12):3794–3801. doi: 10.1007/s11606-021-07152-2.
  • SeyedAlinaghi S, Karimi A, Barzegary A, et al. COVID-19 mortality in patients with immunodeficiency and its predictors: a systematic review. Eur J Med Res. 2022; 27(1):195. doi: 10.1186/s40001-022-00824-7.
  • Choi JH, Choi SH, Yun KW. Risk factors for severe COVID-19 in children: a systematic review and meta-analysis. J Korean Med Sci. 2022; 37(5):e35. doi: 10.3346/jkms.2022.37.e35.
  • Abbasi J. Researchers tie severe immunosuppression to chronic COVID-19 and virus variants. JAMA. 2021; 325(20):2033–2035. doi: 10.1001/jama.2021.7212.
  • Centers for Disease Control and Prevention. People with certain medical condtions [updated February 10. 2023; April 25, 2023]. Available from: https://www.cdc.gov/coronavirus/2019-ncov/need-extra-precautions/people-with-medical-conditions.html.
  • Singson JRC, Kirley PD, Pham H, et al. Factors associated with severe outcomes among immunocompromised adults hospitalized for COVID-19 - COVID-NET, 10 states, march 2020-February 2022. MMWR Morb Mortal Wkly Rep. 2022; 71(27):878–884. doi: 10.15585/mmwr.mm7127a3.
  • Pfizer and BioNTech Celebrate Historic First Authorization in the U.S. of Vaccine to Prevent COVID-19. [Internet]. New York; 2020. Available from: businesswire.com/news/home/20201211005640/en/.
  • Di Fusco M, Moran MM, Cane A, et al. Evaluation of COVID-19 vaccine breakthrough infections among immunocompromised patients fully vaccinated with BNT162b2. J Med Econ. 2021; 24(1):1248–1260. doi: 10.1080/13696998.2021.2002063.
  • Galmiche S, Luong Nguyen LB, Tartour E, et al. Immunological and clinical efficacy of COVID-19 vaccines in immunocompromised populations: a systematic review. Clin Microbiol Infect. 2022; Feb28(2):163–177. doi: 10.1016/j.cmi.2021.09.036.
  • Speich B, Chammartin F, Abela IA, et al. Antibody response in immunocompromised patients after the administration of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine BNT162b2 or mRNA-1273: a randomized controlled trial. Clin Infect Dis. 2022; 75(1):e585–e593. doi: 10.1093/cid/ciac169.
  • Sun J, Zheng Q, Madhira V, et al. Association between immune dysfunction and COVID-19 breakthrough infection after SARS-CoV-2 vaccination in the US. JAMA Intern Med. 2022;182(2):153. doi: 10.1001/jamainternmed.2021.7024.
  • Bergman P, Blennow O, Hansson L, et al. Safety and efficacy of the mRNA BNT162b2 vaccine against SARS-CoV-2 in five groups of immunocompromised patients and healthy controls in a prospective open-label clinical trial. EBioMedicine. 2021; 74:103705. doi: 10.1016/j.ebiom.2021.103705.
  • Abhishek A, Boyton RJ, Peckham N, et al. Effect of a 2-week interruption in methotrexate treatment versus continued treatment on COVID-19 booster vaccine immunity in adults with inflammatory conditions (VROOM study): a randomised, open label, superiority trial. Lancet Respir Med. 2022;10(9):840–850. doi: 10.1016/S2213-2600(22)00186-2.
  • Doherty J, Morain NO, Stack R, et al. Reduced serological response to COVID-19 vaccines in patients with IBD is further diminished by TNF inhibitor therapy; early results of the VARIATION study (VAriability in response in IBD against SARS-COV-2 immunisation). J Crohns Colitis. 2022;16(9):1354–1362. doi: 10.1093/ecco-jcc/jjac029.
  • Krasselt M, Wagner U, Nguyen P, et al. Humoral and cellular response to COVID-19 vaccination in patients with autoimmune inflammatory rheumatic diseases under real-life conditions. Rheumatology. 2022;61(SI2):SI180–SI188. doi: 10.1093/rheumatology/keac089.
  • Mandl P, Tobudic S, Haslacher H, et al. Response to SARS-CoV-2 vaccination in systemic autoimmune rheumatic disease depends on immunosuppressive regimen: a matched, prospective cohort study. Ann Rheum Dis. 2022;81(7):1017–1022. doi: 10.1136/annrheumdis-2021-221788.
  • Dhodapkar MV, Dhodapkar KM, Ahmed R. Viral immunity and vaccines in hematologic malignancies: implications for COVID-19. Blood Cancer Discov. 2021; 2(1):9–12. doi: 10.1158/2643-3230.bcd-20-0177.
  • Haggenburg S, Lissenberg-Witte BI, van Binnendijk RS, et al. Quantitative analysis of mRNA-1273 COVID-19 vaccination response in immunocompromised adult hematology patients. Blood Adv. 2022; 6(5):1537–1546. doi: 10.1182/bloodadvances.2021006917.
  • Boyarsky BJ, Werbel WA, Avery RK, et al. Antibody response to 2-dose SARS-CoV-2 mRNA vaccine series in solid organ transplant recipients. JAMA. 2021;325(21):2204–2206. doi: 10.1001/jama.2021.7489.
  • Boyarsky BJ, Werbel WA, Avery RK, et al. Immunogenicity of a single dose of SARS-CoV-2 messenger RNA vaccine in solid organ transplant recipients. JAMA. 2021;325(17):1784–1786. doi: 10.1001/jama.2021.4385.
  • Kwon JH, Tenforde MW, Gaglani M, et al. mRNA vaccine effectiveness against COVID-19 hospitalization among solid organ transplant recipients. J Infect Dis. 2022;226(5):797–807. doi: 10.1093/infdis/jiac118.
  • Chukwu CA, Mahmood K, Elmakki S, et al. Evaluating the antibody response to SARS-COV-2 vaccination amongst kidney transplant recipients at a single nephrology centre. PLOS One. 2022;17(3):e0265130. doi: 10.1371/journal.pone.0265130.
  • Boey L, Curinckx A, Roelants M, et al. Immunogenicity and safety of the 9-valent human papillomavirus vaccine in solid organ transplant recipients and adults infected with human immunodeficiency virus (HIV). Clin Infect Dis. 2021;73(3):e661–e671. doi: 10.1093/cid/ciaa1897.
  • Eckerle I, Rosenberger KD, Zwahlen M, et al. Serologic vaccination response after solid organ transplantation: a systematic review. PLOS One. 2013;8(2):e56974. doi: 10.1371/journal.pone.0056974.
  • Trakarnvanich T, Ngamvichchukorn T, Phumisantiphong U, et al. Immune response after COVID-19 vaccination among patients with chronic kidney disease and kidney transplant. Vaccine. 2022; 40(45):6499–6511. doi: 10.1016/j.vaccine.2022.09.067.
  • Park JS, Minn D, Hong S, et al. Immunogenicity of COVID-19 vaccination in patients with End-Stage renal disease undergoing maintenance hemodialysis: the efficacy of a mix-and-match strategy. J Korean Med Sci. 2022; 37(23):e180. doi: 10.3346/jkms.2022.37.e180.
  • Abo-Helo N, Muhammad E, Ghaben-Amara S, et al. Specific antibody response of patients with common variable immunodeficiency to BNT162b2 coronavirus disease 2019 vaccination. Ann Allergy Asthma Immunol. 2021;127(4):501–503. doi: 10.1016/j.anai.2021.07.021.
  • Barmettler S, DiGiacomo DV, Yang NJ, et al. Response to SARS-CoV-2 initial series and additional dose vaccine in patients with predominant antibody deficiency. J Allergy Clin Immunol Pract. 2022;10(6):1622–1634.e4. doi: 10.1016/j.jaip.2022.03.017.
  • van Leeuwen LPM, GeurtsvanKessel CH, Ellerbroek PM, et al. Immunogenicity of the mRNA-1273 COVID-19 vaccine in adult patients with inborn errors of immunity. J Allergy Clin Immunol. 2022; 149(6):1949–1957. doi: 10.1016/j.jaci.2022.04.002.
  • Yek C, Warner S, Wiltz JL, et al. Risk factors for severe COVID-19 outcomes among persons aged ≥18 years who completed a primary COVID-19 vaccination series - 465 health care facilities, United States, december 2020-October 2021. MMWR Morb Mortal Wkly Rep. 2022; 71(1):19–25. doi: 10.15585/mmwr.mm7101a4.
  • Centers for Disease Control and Prevention. Interim Clinical Considerations for Use of COVID-19 Vaccines in the United States [updated May 12, 2023; last reviewed June 14, 2023]. Available at: https://www.cdc.gov/vaccines/covid-19/clinical-considerations/interim-considerations-us.html#:∼:text=COVID%2D19%20vaccination%20is%20recommended,younger%20than%20age%206%20months.
  • Harpaz R, Dahl RM, Dooling KL. Prevalence of immunosuppression among US adults, 2013. JAMA. 2016;316(23):2547–2548. doi: 10.1001/jama.2016.16477.
  • Wallace BI, Kenney B, Malani PN, et al. Prevalence of immunosuppressive drug use among commercially insured US adults, 2018–2019. JAMA Netw Open. 2021; 4(5):e214920. doi: 10.1001/jamanetworkopen.2021.4920.
  • Walsh EE, Frenck RW, Jr., Falsey AR, et al. Safety and immunogenicity of two RNA-based covid-19 vaccine candidates. N Engl J Med. 2020;383(25):2439–2450. doi: 10.1056/NEJMoa2027906.
  • Sadoff J, Gray G, Vandebosch A, et al. Final analysis of efficacy and safety of single-dose Ad26.COV2.S. N Engl J Med. 2022;386(9):847–860. doi: 10.1056/NEJMoa2117608.
  • Jackson LA, Anderson EJ, Rouphael NG, et al. An mRNA vaccine against SARS-CoV-2 - preliminary report. N Engl J Med. 2020;383(20):1920–1931. doi: 10.1056/NEJMoa2022483.
  • US Census Bureau. American Community Survey 5-Year Data Profile (2013-2017) 2017 Available from: https://www.census.gov/programs-surveys/acs/data.html.
  • U.S. Bureau of Labor Statistics. Consumer Price Index [April 25, 2022]. Available from: https://www.bls.gov/cpi/home.htm.
  • Quan H, Li B, Couris CM, et al. Updating and validating the Charlson comorbidity index and score for risk adjustment in hospital discharge abstracts using data from 6 countries. Am J Epidemiol. 2011; Mar173(6):676–682. doi: 10.1093/aje/kwq433.
  • Centers for Disease Control and Prevention. COVID data tracker Atlanta, GA: US Department of Health and Human Services; [updated April 19, 2023;April 19, 2023]. Available from: https://covid.cdc.gov/covid-data-tracker/#trends_weeklycases_7daycasesper100k_00.
  • Salerno S, Messana JM, Gremel GW, et al. COVID-19 risk factors and mortality outcomes among medicare patients receiving long-term dialysis. JAMA Netw Open. 2021; 14(11):e2135379. doi: 10.1001/jamanetworkopen.2021.35379.
  • Kliger AS, Silberzweig J. Mitigating risk of COVID-19 in dialysis facilities. Clin J Am Soc Nephrol. 2020; May 715(5):707–709. doi: 10.2215/CJN.03340320.
  • Rabb H. Kidney diseases in the time of COVID-19: major challenges to patient care. J Clin Invest. 2020; 130(6):2749–2751. doi: 10.1172/JCI138871.
  • DeMartino JK, Swallow E, Goldschmidt D, et al. Direct health care costs associated with COVID-19 in the United States. J Manag Care Spec Pharm. 2022; 28(9):936–947. doi: 10.18553/jmcp.2022.22050.
  • Centers for Medicare and Medicaid Services. COVID-19 experiences among the Medicare population: summer 2020 [updated October 2020;March 28, 2023]. Available from: https://www.cms.gov/files/document/medicare-current-beneficiary-survey-summer-2020-covid-19-data-snapshot.pdf.
  • Tenforde MW, Patel MM, Ginde AA, et al. Effectiveness of severe acute respiratory syndrome coronavirus 2 messenger RNA vaccines for preventing coronavirus disease 2019 hospitalizations in the United States. Clin Infect Dis. 2022; 74(9):1515–1524. doi: 10.1093/cid/ciab687.
  • Kamar N, Abravanel F, Marion O, et al. Three doses of an mRNA covid-19 vaccine in solid-organ transplant recipients. N Engl J Med. 2021; 385(7):661–662. doi: 10.1056/NEJMc2108861.
  • Simon B, Rubey H, Treipl A, et al. Haemodialysis patients show a highly diminished antibody response after COVID-19 mRNA vaccination compared with healthy controls. Nephrol Dial Transplant. 2021;36(9):1709–1716. doi: 10.1093/ndt/gfab179.
  • Agha ME, Blake M, Chilleo C, et al. Suboptimal response to coronavirus disease 2019 messenger RNA vaccines in patients with hematologic malignancies: a need for vigilance in the postmasking era. Open Forum Infect Dis. 2021; 8(7):ofab353.
  • Cheung MW, Dayam RM, Shapiro JR, et al. Third and fourth vaccine doses broaden and prolong immunity to SARS-CoV-2 in immunocompromised adult patients. medRxiv. 2023:2023.03.01.23286513.
  • Tartof SY, Slezak JM, Puzniak L, et al. Effectiveness of a third dose of BNT162b2 mRNA COVID-19 vaccine in a large US health system: a retrospective cohort study. Lancet Reg Health Am. 2022; 9:100198. doi: 10.1016/j.lana.2022.100198.
  • Food and Drug Administration. Emergency use authorizations for drugs and non-vaccine biological products [updated March 17, 2023;March 29, 2023]. Available from: https://www.fda.gov/drugs/emergency-preparedness-drugs/emergency-use-authorizations-drugs-and-non-vaccine-biological-products.
  • De Vito A, Colpani A, Saderi L, et al. Impact of early SARS-CoV-2 antiviral therapy on disease progression. Viruses. 2022; 15(1):71. doi: 10.3390/v15010071.
  • Rahmah L, Abarikwu SO, Arero AG, et al. Oral antiviral treatments for COVID-19: opportunities and challenges. Pharmacol Rep. 2022; 74(6):1255–1278. doi: 10.1007/s43440-022-00388-7.
  • Klaassen F, Chitwood MH, Cohen T, et al. Changes in population immunity against infection and severe disease from SARS-CoV-2 Omicron variants in the United States between December 2021 and November 2022. Clin Infect Dis. 2023:ciad210. doi: 10.1093/cid/ciad210.
  • ClinicalTrials.gov. Study Understanding Pre-Exposure pRophylaxis of NOVel Antibodies (SUPERNOVA). NCT05648110 [updated March 17, 2023;March 29, 2023]. Available from: https://clinicaltrials.gov/ct2/show/NCT05648110.
  • Amruta N, Chastain WH, Paz M, et al. SARS-CoV-2 mediated neuroinflammation and the impact of COVID-19 in neurological disorders. Cytokine Growth Factor Rev. 2021; 58:1–15. doi: 10.1016/j.cytogfr.2021.02.002.
  • Khare S, Gurry C, Freitas L, et al. GISAID’s role in pandemic response. China CDC Wkly. 2021;3(49):1049–1051. doi: 10.46234/ccdcw2021.255.
  • Centers for Disease Control and Prevention. COVID data tracker. Nowcast estimates. Atlanta, GA: US Department of Health and Human Services; [updated May 6, 2023;January 20, 2023]. Available from: https://covid.cdc.gov/covid-data-tracker/#variant-proportions.