Next Article in Journal
Genotypic Findings in Noonan and Non-Noonan RASopathies and Patient Eligibility for Growth Hormone Treatment
Next Article in Special Issue
Effects of Aerobic Exercise Therapy through Nordic Walking Program in Lactate Concentrations, Fatigue and Quality-of-Life in Patients with Long-COVID Syndrome: A Non-Randomized Parallel Controlled Trial
Previous Article in Journal
Stroke in Young Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Extracorporeal Photopheresis as a Possible Therapeutic Approach for Adults with Severe and Critical COVID-19 Non-Responsive to Standard Treatment: A Pilot Investigational Study

1
Departmental Group of Infectious Diseases, Department of Haematology and Internal Medicine, Semmelweis University, H-1097 Budapest, Hungary
2
South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Florian út 5–7., H-1097 Budapest, Hungary
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work (in aequo loco).
These authors contributed equally to the work (in aequo loco).
J. Clin. Med. 2023, 12(15), 5000; https://doi.org/10.3390/jcm12155000
Submission received: 17 June 2023 / Revised: 8 July 2023 / Accepted: 26 July 2023 / Published: 29 July 2023
(This article belongs to the Special Issue COVID-19 Treatments and Therapeutics)

Abstract

:
Background: The optimal approach for adult patients hospitalized with severe and critical coronavirus disease 2019 (COVID-19), non-responsive to antiviral and immunomodulatory drugs, is not well established. Our aim was to evaluate feasibility and safety of extracorporeal photopheresis (ECP) in this setting. Methods: A prospective, single-center investigational study was performed between 2021 and 2022 at a tertiary referral center for COVID-19. Patients diagnosed with COVID-19 were screened, and cases with severe or critical disease fulfilling pre-defined clinical and biochemical criteria of non-response for >5 days, despite remdesivir, dexamethasone and immunomodulation (tocilizumab, baricitinib, ruxolitinib), were consecutively enrolled. After patient inclusion, two ECP sessions on two consecutive days per week for 2 weeks were applied. Patients were followed-up per protocol from study inclusion, and clinical, virological and radiological outcomes were assessed at the end of treatment (EOT) +28 days. Results: A total of seven patients were enrolled. At inclusion, four out of seven (57.1%) were admitted to the ICU, all patients had ongoing cytokine storm. Additionally, 3/7 (42.9%) had radiological progression on chest CT. At EOT+28 days, 2/7 (28.6%) patients died due to non-ECP-related causes. Among the survivors, no additional requirement for intensive care unit admission or radiological progression was observed, and invasive mechanical ventilation could be weaned off in 1/5 (20.0%). All patients achieved whole-blood SARS-CoV-2 RNAemia clearance, while 3/7 (42.9%) no longer showed detectable respiratory SARS-CoV-2 RNA. According to immune biomarker profiling, ECP mainly facilitated a decrease in plasma IL-6 and IL-17A levels, as well as the physiological regeneration of peripheral blood immunocyte subpopulations, notably CD8+/CD45RO+ memory T-cells. No safety signals were identified. Conclusions: ECP appears to be a safe and feasible option for adults hospitalized with severe or critical COVID-19 who do not respond to pharmacological interventions. Further trial data are warranted to assess its optimal use. Trial registration: ClinicalTrials.gov NCT05882331 (retrospectively registered).

Graphical Abstract

1. Introduction

In adult patients requiring hospitalization for coronavirus disease 2019 (COVID-19), an acute viral illness of zoonotic origin caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a systemic hyperinflammation may develop. This state is characterized by persistent fever, arterial hypoxaemia, and multi-organ dysfunction involving the respiratory and cardiovascular system, the kidneys and the immune system [1,2,3,4,5,6,7]. As of June 2023, more than 6 million people have lost their lives due to confirmed COVID-19 since the start of the pandemic. In the search for better treatment options, several blood purification strategies, such as hemoadsorption, hemoperfusion and artificial liver blood purification, have been attempted in patients with severe and critical COVID-19, with mixed clinical results [8,9,10].
Extracorporeal photopheresis (ECP) is a non-toxic anti-inflammatory /immunomodulatory treatment. During ECP, patient blood is harvested, and then white blood cells are ex vivo separated and treated with 8-methoxypsoralen, a photoactive drug activated by ultraviolet-A irradiation. Subsequently, the cells are retransfused to the patient. Since its first use, ECP has been considered as an effective and safe strategy in conditions with a dysregulated adaptive immune response, such as acute cellular rejection in solid organ transplantation, graft-versus-host-disease in allogeneic hematopoetic stem-cell transplantation, and some systemic autoimmune disorders. It has been postulated that hyperactivated peripheral T-cells, which are representative of these pathophysiological states, are forced to enter apoptosis during ECP and are consequentially destroyed by tissue macrophages upon reintroduction to the body. The clearance of apoptotic T-cells also leads to the abatement of systemic inflammation by homeostatic reprogramming of macrophages, normalization of regulatory T-cells counts and type-1/type-2 helper T-cell ratios, and an establishment of anti-inflammatory cytokine milieu in peripheral blood [11]. Based on the published data, COVID-19 with systemic hyperinflammation is also characterized by a pathological decrease of regulatory T-cells and type-2 helper T-cell inhibition, along with an over-activation of tissue macrophages [12,13,14]. Therefore, we hypothesized that ECP may be beneficial for adults hospitalized with severe and critical COVID-19 who do not respond to other pharmacological treatments.

2. Methods

2.1. Study Design

A prospective, single-center investigational study was performed between 2021 and 2022 at the South Pest Central Hospital, National Institute of Haematology and Infectious Diseases (Budapest, Hungary), a tertiary referral center with >250 beds for COVID-19 care. The study was conducted in accordance with the Declaration of Helsinki and national ethical standards. The study protocol was approved by the Institutional Review Board of South Pest Central Hospital, National Institute of Haematology and Infectious Diseases (IKEB-14/2020) and the Scientific and Research Ethics Committee of the Hungarian National Medical Scientific Council (ETT-TUKEB–IV/3937–1/2020/EKU). All patients provided written informed consent for anonymized data processing before study inclusion.

2.2. Patient Enrollment

Hospitalized adult patients (≥18 years at diagnosis) with diagnosed COVID-19 of any illness duration before hospitalization were eligible and screened for inclusion during daily on-site investigator visits. Patients were consecutively enrolled based on the following inclusion criteria: (1) severe or critical COVID-19, (2) clinical and biochemical non-response for >5 consecutive days, despite remdesivir, dexamethasone and immunomodulatory therapies (tocilizumab, baricitinib or ruxolitinib), with or without COVID-19 reconvalescent plasmatherapy, in absence of other plausible causes. Exclusion criteria were: (1) pregnancy or breastfeeding, (2) allergy or contraindications to 8-methoxypsoralen, (3) pre-COVID-19 ECP, (4) written informed consent was not obtainable. Clinical non-response was defined when ≥2 of the following were met compared to baseline: (1) persistent fever (non-contact tympanal measurement of >38.0 °C) for ≥48 h despite antipyretics, (2) progression of COVID-19 severity according to World Health Organization criteria by ≥1 stratum after ≥48 h, (3) persistent or worsening of partial arterial oxygen tension (PaO2)/inspired oxygen fraction (FiO2) index by ≥10% after ≥48 h despite respiratory support, (4) radiological progression by infiltrate extension on chest-computed tomography (CT) by ≥10% after ≥48 h, (5) new requirement of invasive mechanical ventilation, as determined necessary by an intensive care unit (ICU) team. Biochemical non-response was defined when ≥2 of the following laboratory analytes showed persistent or increasing levels by ≥20% after ≥48 h compared to baseline: (1) serum lactate dehydrogenase (LDH), (2) serum C-reactive protein (CRP), (3) serum ferritin, (4) plasma interleukin-6 (IL-6), (5) plasma D-dimer levels.

2.3. Data Collection

An electronic case report form was dedicated to the study purposes, and investigators manually uploaded anonymized patient data. The collected data during the study included: (1) age, sex, comorbidities and vaccination status, (2), symptom duration before diagnosis, (3) therapies administered for COVID-19, (4) outcomes and characteristics of follow-up (see below). Baseline data were recorded on the day of COVID-19 diagnosis. Inclusion data were registered on the day of the first ECP session.

2.4. Diagnostic and Therapeutic Strategies, Follow-Up

Symptoms of COVID-19 were defined according to the criteria of the European Centre for Disease Prevention and Control [15]. Diagnosis was based on the positivity of nasopharyngeal or endotracheal specimens for SARS-CoV-2 ribonucleic acid (RNA) using real-time polymerase chain reaction (RT-PCR). COVID-19 severity was determined based on the criteria of the World Health Organization [1]. Acute respiratory distress syndrome (ARDS) and acute respiratory failure were defined according to the Berlin criteria [16]. COVID-19-associated cytokine storm was previously defined by our group based on an interpretation published by Fajgenbaum et al. [17,18]. COVID-19 vaccination status was categorized as a primary series (two doses) or booster (≥1 additional dose), after ≥14 days of last vaccination with any of the vaccines authorized in Hungary (Janssen, Moderna, Oxford-AstraZeneca, Pfizer-BioNTech, Sinopharm-BBIBP-CorV, Sputnik-V).
Therapies against COVID-19 were allocated based on the disease severity following national and international guidelines [19,20]. The standard of care consisted of oxygen/respiratory support, intravenous fluids, antipyretics, antitussives, and bronchodilators as determined necessary by attending physicians. Patients requiring oxygen support received remdesivir (1 × 200 mg loading dose, 1 × 100 mg maintenance intravenously, 5 days minimum) and dexamethasone (1 × 6 mg orally, 5 days minimum). Either tocilizumab (8 mg/kg intravenously, single dose), baricitinib (1 × 4 mg orally, 7 days minimum) or ruxolitinib (2 × 5 mg orally, 7 days minimum) were administered to patients with a COVID-19-associated cytokine storm. The choice of therapy was based on drug accessibility, available route of administration and patient-specific contraindications. Patients also received COVID-19 reconvalescent plasmatherapy when compatible products were available.
Details of patient follow-up are demonstrated in Supplementary Table S1. Briefly, in-hospital follow-up of patients was conducted according to a standardized protocol from study inclusion until ECP end of treatment (EOT) plus 28 days or patient death. The EOT was declared after the last ECP session. During the study period, patients who were planned for discharge from the hospital before EOT+28 days were scheduled for follow-up by study investigators at our outpatient clinic at regular time intervals of 3 days. Physical examination, routine laboratory studies, arterial blood gas analyses were performed daily. Immune biomarker profiling, virological markers and chest CT scans were performed at baseline, inclusion, and on days EOT+3, EOT+7, EOT+14 and EOT+28. Immune biomarker profiling included a multi-cytokine analysis to measure concentrations of 20 cytokines/chemokines, as well as serum electrophoresis with immunofixation, immunoglobulins, complement-3/complement-4 and autoantibody levels, and fluorescence-activated cell sorting (FACS) of different subpopulations of T and B lymphocytes and natural killer (NK) cells from whole blood. Virological markers involved testing respiratory and whole-blood samples for the presence of SARS-CoV-2 nucleic acid by RT-PCR. All tests were performed according to the descriptions provided by the manufacturers.

2.5. Protocol of Extracorporeal Photopheresis

ECP was initiated on the day of study inclusion using the TherakosTM CellexTM system, following the instructions of the manufacturer. Each patient underwent two ECP cycles for 2 weeks, with each cycle consisting of two sessions on 2 consecutive days per week (a total of four sessions), conducted through a peripheral or central venous access device. Cycles 1 and 2 were separated by 5 consecutive days. One ECP session took approximately 3 h to complete, and was divided into 4 phases: (1) during priming, the system performed a series of calibrations to ensure proper operation, (2) during collection, 1500 mL of whole blood was processed to collect a concentrated buffy coat containing white blood cells, while other cells and plasma were reinfused, (3) during the photoactive phase, a prescribed dose of 8-methoxypsoralen was added to the buffy coat, which was then circulated through ultraviolet-A photoactivation, (4) during reinfusion phase, treated cells were automatically reinfused to the patient. Multi-cytokine analysis and FACS for early and late apoptosis markers were performed from each ECP mononuclear cell collection (MNC), a sample type representative of the buffy coat.

2.6. Outcomes and Statistical Analysis

The clinical outcomes assessed were all-cause death, requirement for invasive mechanical ventilation and admission to the ICU. Virological outcomes included respiratory and whole-blood SARS-CoV-2 RT-PCR positivity. Radiological outcomes were evaluated based on radiological progression/regression or fixed infiltration on chest CT scan, measured by the relative extension of COVID-19 infiltration compared to the proportion of normal lung tissue. All outcomes were assessed at EOT+28 days and compared to data at inclusion. Continuous variables are expressed as median ± interquartile ranges and minimum–maximum values; categorical values are reported in absolute numbers and relative percentages. Statistical comparisons were performed using Fisher’s exact-test or the Mann–Whitney U-test. A two-tailed p value of <0.05 determined statistical significance. For reporting, we adhered to the “Better reporting of interventions: template for intervention description and replication” (TIDieR) checklist (http://www.tidierguide.org, accessed on 1 March 2023).

3. Results

3.1. Demographic Characteristics of Patients

The study flowchart is presented in Supplementary Figure S1. During the study period, 1035 eligible patients were screened. Among them, 296 patients had severe or critical COVID-19, and 7 met study inclusion criteria. Demographic data and therapeutic strategies are reported in Table 1.
Median age was 54 ± 14 (32–72) years, with a male dominance in the cohort (5/7, 71.4%). Median duration of symptom before diagnosis was 2 ± 4 (1–7) days. Prevalent comorbidities included essential hypertension (5/7, 71.4%), chronic cardiac and pulmonary diseases (3/7, 42.9% each). Four patients received the primary series of COVID-19 vaccinations, and two also received boosters.

3.2. Clinical Characteristics of Patients

Clinical characteristics are reported in Table 2.
At baseline, 4/7 (57.1%) patients had severe COVID-19, with a median infiltrate extension of 50 ± 60% (20–80) on chest CT. Cytokine storm was documented in six cases (85.7%), 3/7 (42.9%) patients had ARDS. Two patients were admitted to the ICU, requiring invasive mechanical ventilation (28.6%). At inclusion, all patients had ongoing cytokine storm, and an additional patient progressed to critical severity (5/7, 71.4% in total). Three (42.9%) more patients showed radiological progression on chest CT. An additional two patients had to be admitted to the ICU, without any further ARDS development (4/7, 57.1% in total).

3.3. Laboratory Characteristics of Patients

Laboratory characteristics are reported in Table 3.
From baseline to inclusion, an increase in the absolute neutrophil count (ANC) with a median of 1.5 ± 5.9 × 109/L and an absolute lymphocyte count (ALC) with a median of 0.7 ± 0.4 × 109/L, along with serum ferritin (381 ± 230 μg/L) and LDH (153 ± 96 UI/L) was documented, while serum CRP decreased with a median of 75 ± 14 mg/L (p > 0.05 for all). At follow-up, ANC and ALC, serum ferritin, CRP and plasma D-dimer values showed a statistical trend towards their reference ranges (p > 0.05 for all), while serum LDH declined significantly (1096 ± 379 vs. 522 ± 214 UI/L, p = 0.02).

3.4. Immune Biomarker Profiling of Patients

Results of immune biomarker profiling are reported in Table 4.
At inclusion, multi-cytokine analysis revealed an increasing trend of serum RANTES, plasma interleukin- (IL-) 4 and IL-6 levels compared to baseline (p > 0.05 for all differences). During follow-up, both plasma IL-6 (203.5 ± 1688.0 vs. 6.5 ± 24.0 pg/mL, p = 0.04) and interleukin-17A (4.1 ± 3.8 vs. 1.3 ± 0.1 pg/mL, p < 0.01) levels decreased significantly, while the concentrations of serum interferon- (IFN-) γ and IP-10, and plasma IL-5, IL-8 and IL-10 showed trends of increase. Complement consumption or paraprotein/autoantibody generation was not documented among patients. Peripheral whole-blood FACS parameters are reported in Table 5.
From baseline to inclusion, a statistically non-significant decrease among CD3+, CD3+/CD8+ and CD3+/CD4+ T, CD3-/CD16+/CD56+ NK and CD19+ B-cell subpopulations, with an increase in double-negative T-cell ratios, CD3+/TCRγδ+, CD8+/CD45RO+ and CD4+/CD45RO+ memory T-cell counts was documented (p > 0.05 for all differences). During follow-up, double-negative T-cell ratios, CD3+/TCRγδ+ and CD8+/CD45RA+ naive T-cell numbers decreased. Other lymphoid subpopulations showed a trend for physiological regeneratior, with a statistically significant increase in CD8+/CD45RO+ memory T-cell counts (76.2 ± 94.1 vs. 144.4 ± 127.0 cells/μL, p = 0.03).

3.5. Outcome Characteristics of Patients

Outcomes and therapeutic strategies against COVID-19 are reported in Table 1. Two (28.6%) patients died due to septic shock unrelated to ECP (Staphylococcus aureus and Klebsiella pneumoniae bloodstream-infections) at EOT+22 and EOT+24 days, respectively. Invasive mechanical ventilation could be weaned in one, and had to be continued in two patients. The rate of ICU admission requirement (57.1%) among patients did not increase at EOT+28 days compared to inclusion. During follow-up, all patients reached peripheral blood clearance of SARS-CoV-2 RNAemia, while respiratory SARS-CoV-2 PCR positivity remained detectable in four patients (57.1%). Radiological progression was only found in patients with a fatal outcome. The median length of stay was 67 ± 75 (22–155) days. Both tocilizumab and baricitinib were administered to four patients each (57.1% each). Reconvalescent plasmatherapy could be administered to six patients (85.7%).

3.6. ECP MNC Apoptosis Assessment

FACS with early/late apoptosis markers and multi-cytokine analysis from ECP MNC are reported in Supplementary Table S2 and Supplementary Material S1. During ECP, 35–51% of lymphocytes, 38–47% of T-cells, and 95–97% of monocytes entered the early apoptotic stage. Additionally, from the first to last MNC collections, there was a statistically nonsignificant trend towards rising the rates of cells entering early apoptosis, a phenomenon more pronounced among lymphocytes and T-cells. Necrotic cells were not detected, while rates of lymphocytes, T-cells and monocytes entering late apoptotic stage remained low. Cytokines were detected in both increasing (IL-1α, IL-10 and IL-15) and decreasing (IFN-α, IL-5, IL-6, IL-8, IL-13, IP-10, MIP-1α, TNF-β) concentrations, while other cytokines did not show relevant fluctuations (IFN-γ, IL-1β, IL-2, IL-4, IL-12, IL-17A, MCP-1, RANTES, TNF-α) in serial samples.

4. Discussion

4.1. Main Study Findings

To the best of our knowledge, this is the first study assessing feasibility and safety of extracorporeal photopheresis among adults hospitalized for severe and critical COVID-19 who showed non-responsiveness to pharmacological treatment modalities. Our hypothesis was that ECP might attenuate the systemic hyperinflammation associated with COVID-19 in this patient population.
Seven mostly middle-aged male patients with a significant comorbidity burden requiring oxygen supportation and showing prolonged clinical and biochemical non-responsiveness, despite the administration of remdesivir, dexamethasone and immunomodulation (tocilizumab, baricitinib, ruxolitinib), were enrolled in our pilot investigational study. At inclusion, the majority of patients had to be admitted to the ICU in critical condition, with an ongoing COVID-19-associated cytokine storm and progression of infiltrates on chest CT scans. At EOT+28 days, two patients died due to non-ECP-related sepsis. Among those who survived, invasive mechanical ventilation could be discontinued in one patient; additional patients did not require ICU admission, and further radiological progression was not observed on chest CT scans. All the included patients cleared SARS-CoV-2 RNAemia, while a minority also lost respiratory SARS-CoV-2 PCR detectability.
During ECP cycles, it was proven that 35–50% of all lymphocytes and T-cells, as well as almost all monocytes entered the early apoptotic phase. Specific cytokine fluctuations pointed towards an evolving anti-inflammatory milieu, further supporting our hypothesis. After ECP, blood absolute lymphocyte counts, serum ferritin, CRP, LDH and plasma D-dimer levels showed a trend towards their physiological reference ranges. ECP also decreased plasma concentrations of IL-6 and IL-17A, while the regeneration of peripheral blood immunocyte subpopulations, including CD3+/CD8+ and CD3+/CD4+ T-cells, CD3-/CD16+/CD56+ NK-cells and CD19+ B-cells, was documented with a significant increment in CD8+/CD45RO+ memory T-cell counts. No biochemical or clinical safety signals for ECP were identified during the study period.

4.2. Previous Literature Findings

Since the start of the pandemic, the literature evidence has been suggesting that a dysregulated cellular and humoral host immune response could aggravate the severity of COVID-19, leading to critical organ dysfunction with poor disease prognosis [12].
In one of the earliest studies, 435 patients with confirmed SARS-CoV-2 infection were followed for 6 weeks. Counts of all measured lymphocyte subpopulations, including CD3+, CD4+, CD8+, CD19+ and CD16/56+ cells, remained below their respective reference ranges for ≥5 weeks. In addition, severe disease and fatal outcomes were associated with pronounced lymphopenia in peripheral blood compared to non-severe cases and survivors [13]. These findings were mirrored in other studies, mostly investigating patients with severe COVID-19, and suggested that peripheral neutrophil-to-lymphocyte ratios, and especially neutrophil-to-CD8+ T-cell ratios, as well as absolute CD8+ T-cell counts correlated well with poor clinical outcomes, including mortality [21,22]. A later study proved that the acute immunodeficiency character of COVID-19 stems from the immunoparesis of the CD4+ T-cell pool, and can also be demonstrated by peripheral T-cell exhaustion [23]. Other authors have also highlighted CD4+ T-cell peripheral depletion and altered functionality as negative prognostic markers for severity and survival in COVID-19 [24,25]. In the context of COVID-19 pulmonary infiltrations, histopathological evidence gathered from 60 autopsies demonstrated that CD8+ T-cells were abundant, whereas the CD4+ T cell presence was low in the pulmonary interstitium, in contrast to the cases of control patients without infection [26]. These findings were later mirrored by an in vivo clinical study which proved a correlation between CD4+ T-cell lymphopenia and pulmonary inflammation on chest CT [27,28]. We note that other cellular subpopulations also play a role in disease progression. For example, it has been shown that phenotypically distinct CD3+/TCRγδ+, double-negative, and double-positive T-cells proliferate during the acute stage of SARS-CoV-2 infection, parallel to prominent expansions of CD14+ classical and CD16+ non-classical monocyte subpopulations. While CD4+/CD45RA+ and CD8+/CD45RA+ naive T-cells, as well as CD4+/CD45RO- central memory T-cells decreased in number, the number of CD8+/CD45RO+ and CD4+/CD45RO+ effector memory T-cells increased, compared to healthy controls and reconvalescent patients alike, strengthening the idea that abnormal peripheral T-cell activation and differentiation are key steps in SARS-CoV-2 pathophysiology [29].
On the other hand, dysregulated cytokine responses, most notably fluctuations of serum IL-6 during COVID-19, have also been established in the literature [17]. Among critically ill patients with COVID-19, serum IL-6 and IL-10 levels are usually higher, while serum GM-CSF, TNF- α, IFN-γ, IL-2 and IL-8 serve as ancillary cytokines with clinical prognostic value in contrast to patients without critical disease [14,30,31]. Since tocilizumab is recommended for patients with COVID-19 of critical severity, its effect on cytokine levels should also be mentioned. For example, in a study carried out by Azmy et al., post-tocilizumab serum IL-6 levels spiked and then showed a steady decrease, while the levels of the anti-inflammatory IL-10 were not affected [31]. Our group previously documented similar trends of serum IL-6 among critically ill patients receiving immunomodulatory therapies [18,32]. Other studies have demonstrated that persistently elevated serum IL-6 levels during COVID-19 are independently associated with in-hospital mortality, and rises of serum IL-6, IL-8 and TNF-α are probably age-dependent, translating to poorer outcomes among the elderly [22,24,33]. Pro-inflammatory cytokine responses in blood were detectable for ≥3 weeks after SARS-CoV-2 infection [34]. In addition, serum levels of both IL-6 and IL-10 correlated positively with pulmonary lesion volumes on chest CT scans caused by COVID-19, while others argued that the TNF-α/IL-10 ratio could readily estimate progression to acute respiratory failure during COVID-19 [27,35]. Lastly, hyperinflammation during COVID-19 is probably accompanied by a simultaneous anti-inflammatory immune response driven by serum IL-10, which therefore also bears a deterministic effect on disease severity [36].
During cytokine storm, the complex interplay between immunocytes and secreted cytokines is obvious. For example, serum IL-6 and IL-8 levels correlate well with CD4+ and CD8+ T-cell counts and ongoing pulmonary inflammation [28]. Furthermore, blood levels of T-cell-associated cytokines, mainly IFN-γ, TNF-α, IL-2, IL-6 and IL-17, increase during severe COVID-19, as opposed to measurements in controls [23]. Lung samples from COVID-19 autopsies revealed that the inflamed pulmonary endothelium expressed IL-1β, IL-6, IL-15 and TNF-α, in contrast to non-infected tissue samples, recruiting inflammocytes along the alveolar epithelium [26]. Overall, while much has been discovered about COVID-19-associated inflammation, a therapeutic strategy for COVID-19 non-responsive to antiviral and immunomodulatory drugs has not yet been established in the literature. Therefore, given the high level of cellular and humoral immune dysregulation accompanying severe and critical COVID-19, non-toxic modalities aiming to reduce inflammation-related multi-organ damage, such as extracorporeal photopheresis, are probably crucial for clinical practice.

4.3. Limitations

A comparator arm was not feasible for this study. Since this was a pilot study, the number of enrolled patients is low. Despite limitations, we feel that results might serve as a basis for future research and possibly introduce a novel therapeutic alternative for a vulnerable population with limited treatment options and an overall negative prognosis.

5. Conclusions

Extracorporeal photopheresis might be considered a feasible, safe novel treatment for adults hospitalized with severe and critical COVID-19 who do not respond to other pharmacological therapies. Further research is warranted to confirm the findings of this pilot study.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm12155000/s1, Figure S1: Study flow chart; Materials S1: FACS plots from MNCs of patients enrolled in the study; Table S1: Schematic diagram for data acquisition at baseline, inclusion and follow-up; Table S2: FACS with early and late apoptosis markers and multi-cytokine analysis from ECP mononuclear cell collections (MNC) after ECP sessions 1 (first) to 4 (last) of patients enrolled in the study.

Author Contributions

B.G.S. and P.R. contributed equally to the manuscript (in aequo loco). IB and IVN share last authorship of the manuscript (in aequo loco). B.G.S.: management of patients, data collection, data analysis, preparation of study protocol, preparation of the manuscript, conception and design of the article, literature search and interpretation; P.R.: management of patients, data collection, data analysis, preparation of study protocol, preparation of the manuscript, conception and design of the article, literature search and interpretation; S.T.: laboratory data analysis, review of the manuscript; D.K.: data collection, literature search and interpretation; L.G.: preparation of study protocol, literature search and interpretation; M.R.: preparation of study protocol, preparation and review of the manuscript; A.V.: preparation of study protocol, preparation and review of the manuscript; J.S. (János Sinkó): preparation of study protocol, preparation and review of the manuscript; B.L.: preparation of study protocol, preparation and review of the manuscript, J.S. (János Szlávik): management of patients, review of the manuscript, G.B.: preparation of study protocol, preparation and review of the manuscript; I.B.: management of patients, review of the manuscript, I.V.-N.: preparation of study protocol, preparation and review of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Projects no. 2020–1.1.6-JÖVŐ-2021–00011 and no. TKP2021-EGA-08 have been implemented with the support provided from the National Research, Development and Innovation Fund of Hungary, financed under the “Investment in the Future Fund” and “Thematic Excellence Program 2021” funding scheme. The study was also supported by the Ministry of Human Capacities (EMMI IV/365–2/2022/EKF). BGSz received the EFOP-3.6.3-VEKOP-16–2017-00009 Doctorate Grant, and was supported by the NTP-NFTÖ-21-B “Grant for Young Talents of the Nation” and the ÚNKP-19–3-I-SE-74 New National Excellence Program of the Ministry of Innovation and Technology of Hungary. The funding sources were not involved in the preparation, writing, interpretation, or submission of this article.

Institutional Review Board Statement

The study was in accordance with the Declaration of Helsinki and national ethical standards. The study protocol has been approved by Institutional Review Board of South Pest Central Hospital, National Institute of Haematology and Infectious Diseases (IKEB-14/2020), and the Scientific and Research Ethics Committee of the Hungarian National Medical Scientific Council (ETT-TUKEB–IV/3937–1/2020/EKU). All patients provided written informed consent for anonymized data collection and processing before study inclusion. Approval of use of off-label drugs for COVID-19 was granted by the National Institute of Pharmacy and Nutrition (www.ogyei.gov.hu/tajekoztato_a_veszelyhelyzet_megszunesevel_kapcsolatos_a_covid_19_jarvany_idejen_kulonos_meltanylast_erdemlo_betegellatasi_erdekhez_kotheto_gyogyszeralkalmazasok_bejelenteserol, accessed 1 March 2023).

Informed Consent Statement

All patients provided written informed consent for anonymized data collection and processing before study inclusion. The authors affirm that patients provided written informed consent for publication of their anonymized and statistically cumulated data.

Data Availability Statement

Anonymized data of patients are available from the corresponding author on reasonable request.

Acknowledgments

The image in the Graphical Abstract was kindly provided and is courtesy of Mallinckrodt Pharmaceuticals. The following physicians from South Pest Central Hospital, National Institute of Hematology and Infectious Diseases collaborated in patient care: Zsofia BALOGH, Zsuzsanna BANYAI, Emese BANYASZ, Ilona BOBEK, Tibor BOKROS, Jozsef BUDAI, Eszter CZEL, Katalin FRIED, Adrienn HANUSKA, Andras HORNYAK, Csaba LORINCZI, Krisztina NEMESI, Janos KADAR, Igor KAPRAN, Erzsebet KADLECSIK, Rebeka KISS, Bernadett KONDOR, Viola MALIK, Mariann MAYER, Eszter MOLNAR, Eva Livia NAGY, Marton NYULI, Akos OSVALD, Anita PETO, Edina PETROVICZ, Alexandra RICZU, Vlagyiszlav RUDENKO, Gabriella SEBESTYEN, Judit SZANKA, Andrea SZOMBATI, Balazs SZIVOS, Szilvia TOTH, Márta SZTRIKO KOVACS, Zsuzsanna VARNAI, Orsolya WOLLER. We would like to thank the institutional pharmacists for their contribution in the study through accelerated procurement and precise allocation of the medicines: Matyas HORVATH, Katalin KLENJANSZKI, Kitti RADACSI-RACZ, Zsuzsanna STELBACZKY, Eva TEREK, Anita ZEMPLENI-TOTH. The authors would like to thank all the healthcare workers of our centre for their sacrifice during the COVID-19 pandemic.

Conflicts of Interest

The authors declare no competing interests. The Copyright Transfer Statement was completed by the corresponding author on behalf of all co-authors.

Abbreviations

ANAanti-nuclear antibody
ANCAanti-neutrophil cytoplasmic antibody
ARDSacute respiratory distress syndrome
Ccomplement
CDcluster of differentiation
COVID-19coronavirus disease 2019
CTcomputed tomography
ECPextracorporeal photopheresis
ENAextractable nuclear antigen
EOTend of treatment
FACSfluorescence-activated cell sorting
FiO2fraction of inspired oxygen
ICUintensive care unit
IFNinterferon
Igimmunoglobulin
ILinterleukin
LDHlactate dehydrogenase
LOSlength of stay
NKnatural killer
MNCmononuclear cell collection
PaO2partial arterial oxygen tension
RNAribonucleic acid
RT-PCRreal-time polymerase chain reaction
SARS-CoV-2severe acute respiratory syndrome coronavirus 2

References

  1. World Health Organization. Living Guidance for Clinical Management of COVID-19; World Health Organization: Geneva, Switzerland, 2021.
  2. Huang, S.; Vignon, P.; Mekontso-Dessap, A.; Tran, S.; Prat, G.; Chew, M.; Balik, M.; Sanfilippo, F.; Banauch, G.; Clau-Terre, F.; et al. Echocardiography findings in COVID-19 patients admitted to intensive care units: A multi-national observational study (the ECHO-COVID study). Intensive Care Med. 2022, 48, 667–678. [Google Scholar] [CrossRef] [PubMed]
  3. La Via, L.; Dezio, V.; Santonocito, C.; Astuto, M.; Morelli, A.; Huang, S.; Vieillard-Baron, A.; Sanfilippo, F. Full and simplified assessment of left ventricular diastolic function in COVID-19 patients admitted to ICU: Feasibility, incidence, and association with mortality. Echocardiography 2022, 39, 1391–1400. [Google Scholar] [CrossRef] [PubMed]
  4. Gibson, P.G.; Qin, L.; Puah, S.H. COVID-19 acute respiratory distress syndrome (ARDS): Clinical features and differences from typical pre-COVID-19 ARDS. Med. J. Aust. 2020, 213, 54–56.e1. [Google Scholar] [CrossRef] [PubMed]
  5. Ahmadian, E.; Khatibi, S.M.H.; Soofiyani, S.R.; Abediazar, S.; Shoja, M.M.; Ardalan, M.; Vahed, S.Z. COVID-19 and kidney injury: Pathophysiology and molecular mechanisms. Rev. Med. Virol. 2021, 31, e2176. [Google Scholar] [CrossRef] [PubMed]
  6. Qin, R.; He, L.; Yang, Z.; Jia, N.; Chen, R.; Xie, J.; Fu, W.; Chen, H.; Lin, X.; Huang, R.; et al. Identification of Parameters Representative of Immune Dysfunction in Patients with Severe and Fatal COVID-19 Infection: A Systematic Review and Meta-analysis. Clin. Rev. Allergy Immunol. 2023, 64, 33–65. [Google Scholar] [CrossRef]
  7. Swenson, K.E.; Swenson, E.R. Pathophysiology of Acute Respiratory Distress Syndrome and COVID-19 Lung Injury. Crit. Care Clin. 2021, 37, 749–776. [Google Scholar] [CrossRef]
  8. Asgharpour, M.; Mehdinezhad, H.; Bayani, M.; Zavareh, M.S.H.; Hamidi, S.H.; Akbari, R.; Ghadimi, R.; Bijani, A.; Mouodi, S. Effectiveness of extracorporeal blood purification (hemoadsorption) in patients with severe coronavirus disease 2019 (COVID-19). BMC Nephrol. 2020, 21, 356. [Google Scholar] [CrossRef]
  9. Sanfilippo, F.; Martucci, G.; La Via, L.; Cuttone, G.; Dimarco, G.; Pulizzi, C.; Arcadipane, A.; Astuto, M. Hemoperfusion and blood purification strategies in patients with COVID-19: A systematic review. Artif. Organs 2021, 45, 1466–1476. [Google Scholar] [CrossRef]
  10. Dai, X.; Zhang, Y.; Yu, L.; Jiang, Y.; Chen, L.; Chen, Y.; Li, M.; Gao, C.; Shang, J.; Xiang, S.; et al. Effect of artificial liver blood purification treatment on the survival of critical ill COVID-19 patients. Artif. Organs 2021, 45, 762–769. [Google Scholar] [CrossRef]
  11. Cho, A.; Jantschitsch, C.; Knobler, R. Extracorporeal Photopheresis-An Overview. Front. Med. 2018, 5, 236. [Google Scholar] [CrossRef]
  12. Yang, A.P.; Li, H.M.; Tao, W.Q.; Yang, X.J.; Wang, M.; Yang, W.J.; Liu, J.P. Infection with SARS-CoV-2 causes abnormal laboratory results of multiple organs in patients. Aging 2020, 12, 10059–10069. [Google Scholar] [CrossRef] [PubMed]
  13. Deng, Z.; Zhang, M.; Zhu, T.; Zhili, N.; Liu, Z.; Xiang, R.; Zhang, W.; Xu, Y. Dynamic changes in peripheral blood lymphocyte subsets in adult patients with COVID-19. Int. J. Infect. Dis. 2020, 98, 353–358. [Google Scholar] [CrossRef] [PubMed]
  14. Essa, S.; Shamsah, M.; Alsarraf, A.H.; Esmaeil, A.; Al-Shammasi, A.; Raghupathy, R. Influence of SARS-CoV-2 Infection on Cytokine Production by Mitogen-Stimulated Peripheral Blood Mononuclear Cells and Neutrophils in COVID-19 Intensive Care Unit Patients. Microorganisms 2022, 10, 2194. [Google Scholar] [CrossRef]
  15. European Centre for Disease Prevention and Control. Case Definition for Coronavirus Disease 2019 (COVID-19), as of 3 December 2020. 2020. Available online: https://www.ecdc.europa.eu/en/covid-19/surveillance/case-definition (accessed on 30 January 2023).
  16. ARDS Definition of Task Force; Ranieri, V.M.; Rubenfeld, G.D.; Thompson, B.T.; Ferguson, N.D.; Caldwell, E.; Fan, E.; Camporota, L.; Slutsky, A.S. Acute respiratory distress syndrome: The Berlin Definition. JAMA 2012, 307, 2526–2533. [Google Scholar] [CrossRef]
  17. Fajgenbaum, D.C.; June, C.H. Cytokine Storm. N. Engl. J. Med. 2020, 383, 2255–2273. [Google Scholar] [CrossRef]
  18. Lakatos, B.; Szabó, B.G.; Bobek, I.; Kiss-Dala, N.; Gáspár, Z.; Riczu, A.; Petrik, B.; Farkas, B.F.; Sebestyén, G.; Gopcsa, L.; et al. Baricitinib vs tocilizumab treatment for hospitalized adult patients with severe COVID-19 and associated cytokine storm: A prospective, investigational, real-world study. Int. J. Infect. Dis. 2022, 125, 233–240. [Google Scholar] [CrossRef]
  19. Babarczy, B.; Bertókné, T.; Biró, K.; Bobek, I.; Bognár, Z.; Bogos, K. Hungarian Coronavirus Handbook; Hungarian Ministry of Human Resources: Budapest, Hungary, 2020. [Google Scholar]
  20. Bartoletti, M.; Azap, O.; Barac, A.; Bussini, L.; Ergonul, O.; Krause, R.; Paño-Pardo, J.R.; Power, N.R.; Sibani, M.; Szabo, B.G.; et al. ESCMID COVID-19 living guidelines: Drug treatment and clinical management. Clin. Microbiol. Infect. 2022, 28, 222–238. [Google Scholar] [CrossRef]
  21. Liu, J.; Li, S.; Liu, J.; Liang, B.; Wang, X.; Wang, H.; Li, W.; Tong, Q.; Yi, J.; Zhao, L.; et al. Longitudinal characteristics of lymphocyte responses and cytokine profiles in the peripheral blood of SARS-CoV-2 infected patients. EBioMedicine 2020, 55, 102763. [Google Scholar] [CrossRef]
  22. Luo, M.; Liu, J.; Jiang, W.; Yue, S.; Liu, H.; Wei, S. IL-6 and CD8+ T cell counts combined are an early predictor of in-hospital mortality of patients with COVID-19. JCI Insight 2020, 5, e139024. [Google Scholar] [CrossRef]
  23. Arcanjo, A.; Pinto, K.G.; Logullo, J.; Leite, P.E.C.; Menezes, C.C.B.; Freire-de-Lima, L.; Diniz-Lima, I.; Decoté-Ricardo, D.; Rodrigues-DA-Silva, R.N.; Freire-De-Lima, C.G.; et al. Critically Ill Coronavirus Disease 2019 Patients Exhibit Hyperactive Cytokine Responses Associated with Effector Exhausted Senescent T Cells in Acute Infection. J. Infect. Dis. 2021, 224, 1672–1683. [Google Scholar] [CrossRef]
  24. Luo, Y.; Mao, L.; Yuan, X.; Xue, Y.; Lin, Q.; Tang, G.; Song, H.; Wang, F.; Sun, Z. Prediction Model Based on the Combination of Cytokines and Lymphocyte Subsets for Prognosis of SARS-CoV-2 Infection. J. Clin. Immunol. 2020, 40, 960–969. [Google Scholar] [CrossRef]
  25. Schub, D.; Klemis, V.; Schneitler, S.; Mihm, J.; Lepper, P.M.; Wilkens, H.; Bals, R.; Eichler, H.; Gärtner, B.C.; Becker, S.L.; et al. High levels of SARS-CoV-2-specific T cells with restricted functionality in severe courses of COVID-19. JCI Insight 2020, 5, e142167. [Google Scholar] [CrossRef]
  26. Frisoni, P.; Neri, M.; D’errico, S.; Alfieri, L.; Bonuccelli, D.; Cingolani, M.; Di Paolo, M.; Gaudio, R.M.; Lestani, M.; Marti, M.; et al. Cytokine storm and histopathological findings in 60 cases of COVID-19-related death: From viral load research to immunohistochemical quantification of major players IL-1beta, IL-6, IL-15 and TNF-alpha. Forensic Sci. Med. Pathol. 2022, 18, 4–19. [Google Scholar] [CrossRef]
  27. He, S.; Zhou, C.; Lu, D.; Yang, H.; Xu, H.; Wu, G.; Pan, W.; Zhu, R.; Jia, H.; Tang, X.; et al. Relationship between chest CT manifestations and immune response in COVID-19 patients. Int. J. Infect. Dis. 2020, 98, 125–129. [Google Scholar] [CrossRef]
  28. Wan, S.; Yi, Q.; Fan, S.; Lv, J.; Zhang, X.; Guo, L.; Lang, C.; Xiao, Q.; Xiao, K.; Yi, Z.; et al. Relationships among lymphocyte subsets, cytokines, and the pulmonary inflammation index in coronavirus (COVID-19) infected patients. Br. J. Haematol. 2020, 189, 428–437. [Google Scholar] [CrossRef]
  29. Shi, W.; Liu, X.; Cao, Q.; Ma, P.; Le, W.; Xie, L.; Ye, J.; Wen, W.; Tang, H.; Su, W.; et al. High-dimensional single-cell analysis reveals the immune characteristics of COVID-19. Am. J. Physiol. Lung Cell Mol. Physiol. 2021, 320, L84–L98. [Google Scholar] [CrossRef]
  30. Han, H.; Ma, Q.; Li, C.; Liu, R.; Zhao, L.; Wang, W.; Zhang, P.; Liu, X.; Gao, G.; Liu, F.; et al. Profiling serum cytokines in COVID-19 patients reveals IL-6 and IL-10 are disease severity predictors. Emerg. Microbes Infect. 2020, 9, 1123–1130. [Google Scholar] [CrossRef]
  31. Azmy, V.; Kaman, K.; Tang, D.; Zhao, H.; Cruz, C.D.; Topal, J.E.; Malinis, M.; Price, C.C. Cytokine Profiles Before and After Immune Modulation in Hospitalized Patients with COVID-19. J. Clin. Immunol. 2021, 41, 738–747. [Google Scholar] [CrossRef]
  32. Lakatos, B.; Szabo, B.G.; Bobek, I.; Gopcsa, L.; Beko, G.; Kiss-Dala, N.; Petrik, B.; Gaspar, Z.; Farkas, B.F.; Sinko, J.; et al. Laboratory parameters predicting mortality of adult in-patients with COVID-19 associated cytokine release syndrome treated with high-dose tocilizumab. Acta Microbiol. Immunol. Hung. 2021, 68, 145–152. [Google Scholar] [CrossRef]
  33. Pirabe, A.; Heber, S.; Schrottmaier, W.C.; Schmuckenschlager, A.; Treiber, S.; Pereyra, D.; Santol, J.; Pawelka, E.; Traugott, M.; Schörgenhofer, C.; et al. Age Related Differences in Monocyte Subsets and Cytokine Pattern during Acute COVID-19-A Prospective Observational Longitudinal Study. Cells 2021, 10, 3373. [Google Scholar] [CrossRef]
  34. Vu, D.-L.; Martinez-Murillo, P.; Pigny, F.; Vono, M.; Meyer, B.; Eberhardt, C.S.; Lemeille, S.; Von Dach, E.; Blanchard-Rohner, G.; Eckerle, I.; et al. Longitudinal Analysis of Inflammatory Response to SARS-CoV-2 in the Upper Respiratory Tract Reveals an Association with Viral Load, Independent of Symptoms. J. Clin. Immunol. 2021, 41, 1723–1732. [Google Scholar] [CrossRef]
  35. Anderberg, S.B.; Luther, T.; Berglund, M.; Larsson, R.; Rubertsson, S.; Lipcsey, M.; Larsson, A.; Frithiof, R.; Hultström, M. Increased levels of plasma cytokines and correlations to organ failure and 30-day mortality in critically ill COVID-19 patients. Cytokine 2021, 138, 155389. [Google Scholar] [CrossRef]
  36. Henry, B.M.; Benoit, S.W.; Vikse, J.; Berger, B.A.; Pulvino, C.; Hoehn, J.; Rose, J.; de Oliveira, M.H.S.; Lippi, G.; Benoit, J.L. The anti-inflammatory cytokine response characterized by elevated interleukin-10 is a stronger predictor of severe disease and poor outcomes than the pro-inflammatory cytokine response in coronavirus disease 2019 (COVID-19). Clin. Chem. Lab. Med. 2021, 59, 599–607. [Google Scholar] [CrossRef] [PubMed]
Table 1. Demographic data, therapeutic strategies and outcomes of patients enrolled in the study.
Table 1. Demographic data, therapeutic strategies and outcomes of patients enrolled in the study.
Patient IdentificationPt. 1.Pt. 2.Pt. 3.Pt. 4.Pt. 5.Pt. 6.Pt. 7.Total
Age (years)5565325451417254 ± 14 (32–72)
Gender (n, %)MMMMMFFM: 5/7 (71.4)
F: 2/7 (28.6)
Comorbidities (n, %)ACHT
OB
CCD
CHD
CRD
CVD
HT
CPD
HEM
TOB
CPD
HT
OB
AID
CHD
CPD
CVD
HT
TOB
AID
CHD
CRD
CVD
HEM
HT
AC: 1/7 (14.3)
AID: 2/7 (28.6)
CCD: 1/7 (14.3)
CHD: 3/7 (42.9)
CPD: 3/7 (42.9)
CRD: 2/7 (28.6)
CVD: 3/7 (42.9)
HEM: 2/7 (28.6)
HT: 5/7 (71.4)
OB: 2/7 (28.6)
TOB: 2/7 (28.6)
COVID-19 vaccination status:
- Primary series (n, %)N2 × AZ2 × Sp2 × Pf2 × Pf2 × Pf2 × PfY: 6/7 (85.7)
- Booster vaccination (n, %)n.a.1 × PfNNNN1 × PfY: 2/6 (33.3)
- Time since last vaccine (months)n.a.1876626 ± 5 (1–8)
Symptom duration before
COVID-19 diagnosis (days)
76112242 ± 4 (1–7)
Therapies against COVID-19 (n, %):
- RemdesivirYYYYYYYY: 7/7 (100)
- DexamethasoneYYYYYYYY: 7/7 (100)
- TocilizumabYYYNNYNY: 4/7 (57.1)
- BaricitinibYYNNYNYY: 4/7 (57.1)
- RuxolitinibNNNYNNNY: 1/7 (14.3)
- Reconvalescent plasmatherapyYNYYYYYY: 6/7 (85.7)
Clinical outcomes (n, %):
- All-cause deathNNNNNYYY: 2/7 (28.6)
- Invasive mechanical ventilationYNYNNN *N *Y: 2/7 (28.6)
- ICU admissionYNYNYN *Y *Y: 4/7 (57.1)
Virological outcomes (n, %):
- Respiratory SARS-CoV-2 RT-PCR positivityNYNNYY *Y *Y: 4/7 (57.1)
- Blood SARS-CoV-2 RT-PCR positivityNNNNNN *N *Y: 0 (0)
Radiological outcomes (n, %)
- Radiological progression on chest CT scanNNNNNY *Y *Y: 2/7 (28.6)
- Radiological regression on chest CT scanNYNYYNNY: 3/7 (42.9)
- Fixed infiltration on chest CT scanYNYNNNNY: 2/7 (28.6)
Hospital ward length of stay (days) **86450305124045 ± 42 (24–86)
Intensive care unitlength of stay (days)6909104802259 ± 45 (22–91)
Time to death from hospital admission (days)n.a.n.a.n.a.n.a.n.a.242223 ± n.a. (22–24)
* Outcome assessed on day of patient death by last available data. ** Excluding length of stay at the intensive care unit. AC: chronic alcohol consumption, AID: systemic autoimmune disease, ARDS: acute respiratory distress syndrome, AZ: Oxford–AstraZeneca COVID-19 vaccine, CCD: chronic cerebral disease, CHD: chronic heart disease, CPD: chronic pulmonary disease, CRD: chronic renal disease, CT: computed tomography, CVD: chronic vascular disease, F: female, HEM: active hematological malignancy, HT: essential hypertension, ICU: intensive care unit, M: male, N: no, n.a.: not applicable, OB: obesity, Pf: Pfizer–BioNTech COVID-19 vaccine, RNA: ribonucleic acid, SARS-CoV-2: severe acute respiratory syndrome coronavirus 2, Sp: Sputnik-V COVID-19 vaccine, TOB: chronic tobacco use, Y: yes.
Table 2. Clinical characteristics at baseline and inclusion of patients enrolled in the study.
Table 2. Clinical characteristics at baseline and inclusion of patients enrolled in the study.
Patient IdentificationPt. 1.Pt. 2.Pt. 3.Pt. 4.Pt. 5.Pt. 6.Pt. 7.Totalp Value **
Time of AssessmentBaseline InclusionBaseline InclusionBaseline InclusionBaseline InclusionBaseline InclusionBaseline InclusionBaseline InclusionBaseline Inclusion
COVID-19 severityCCCCCCSSCCSCSSC: 4/7 (57.1)
S: 3/7 (42.9)
C: 5/7 (71.4)
S: 2/7 (28.6)
1.0
Infiltrate extension on chest CT (%) *808050508080203080802080205050 ± 60
(20–80)
80 ± 30
(30–80)
0.37
Partial arterial oxygen tension
per fraction of inspired oxygen (mmHg/%) *
95851758011590300200559020585300200175 ± 205
(55–300)
90 ± 115
(80–200)
0.16
ARDSYYNNYYNNYYNNNNY: 3/7 (42.9)Y: 3/7 (42.9)1.0
Cytokine stormYYYYYYYYYYNYYYY: 6/7 (85.7)Y: 7/7 (100%)1.0
Respiratory SARS-CoV-2
RT-PCR positivity
YYYYYYYYYYYYYYY: 7/7 (100%)Y: 7/7 (100%)1.0
Blood SARS-CoV-2
RT-PCR positivity
YYNYYYNYYYNNNYY: 3/7 (42.9)Y: 6/7 (85.7)0.31
ICU admissionNYNNYYNNYYNNNYY: 2/7 (28.6)Y: 4/7 (57.1)0.59
Invasive mechanical ventilationNYNNYYNNYYNNNNY: 2/7 (28.6)Y: 3/7 (42.9)1.0
* Parameters are reported in median ± interquartile ranges and minimum–maximum values. ** p values are calculated between cumulated data, measured at baseline and inclusion. ARDS: acute respiratory distress syndrome, C: critical COVID-19, CT: computed tomography, E: extracorporeal photopheresis, ICU: intensive care unit, N: no, RNA: ribonucleic acid, S: severe COVID-19, SARS-CoV-2: severe acute respiratory syndrome coronavirus 2, Y: yes.
Table 3. Routine laboratory parameters of patients enrolled in the study at baseline, inclusion and post-ECP follow-up.
Table 3. Routine laboratory parameters of patients enrolled in the study at baseline, inclusion and post-ECP follow-up.
ParameterBaselineInclusionPost-ECP Follow-Upp Value *
+3 Days+7 Days+14 Days+28 Days
Blood absolute white blood
cell count (×109/L)
8.1 ± 3.2
(2.1–10.5)
8.9 ± 7.4
(1.9–21.3)
14.3 ± 7.4
(3.7–23.5)
8.1 ± 2.4
(4.5–21.4)
6.4 ± 6.4
(2.3–19.7)
8.1 ± 2.5
(3.7–9.2)
0.97
Blood absolute neutrophil
granulocyte count (×109/L)
4.7 ± 2.0
(1.8–9.2)
6.2 ± 7.9
(1.4–16.9)
8.2 ± 6.9
(2.5–20.1)
6.1 ± 4.6
(3.0–17.7)
3.8 ± 5.1
(1.1–15.5)
4.4 ± 1.9
(2.1–6.1)
0.46
Blood absolute lymphocyte
count (×109/L)
0.8 ± 1.7
(0.1–4.1)
1.5 ± 1.3
(0.2–3.3)
1.3 ± 1.1
(0.4–5.4)
1.4 ± 1.7
(0.5–3.6)
1.4 ± 1.2
(0.3–2.6)
1.8 ± 1.6
(0.4–6.0)
0.52
Blood absolute monocyte
count (×109/L)
0.3 ± 0.2
(0.1–0.8)
0.3 ± 0.3
(0.2–1.5)
0.6 ± 0.5
(0.1–1.4)
0.4 ± 0.3
(0.1–1.7)
0.5 ± 0.7
(0.3–2.0)
0.7 ± 0.6
(0.3–0.9)
0.7
Blood hemoglobin (g/L)113 ± 18
(96–141)
136 ± 40
(86–140)
111 ± 31
(76–142)
98 ± 19
(82–131)
96 ± 13
(84–133)
85 ± 12
(75–139)
0.07
Blood platelet count (×109/L)177 ± 113
(49–368)
302 ± 208
(43–853)
230 ± 233
(26–432)
167 ± 220
(30–498)
111 ± 126
(43–705)
156 ± 368
(35–783)
0.87
Serum lactate dehydrogenase (IU/L)943 ± 283
(400–1168)
1096 ± 379
(530–1802)
807 ± 200
(586–2020)
693 ± 166
(505–847)
473 ± 88
(389–626)
522 ± 214
(274–626)
0.02
Serum glutamic pyruvate
transaminase (IU/L)
34 ± 16
(11–48)
31 ± 39
(18–76)
75 ± 41
(19–395)
85 ± 71
(16–253)
119 ± 192
(19–338)
38 ± 56
(16–117)
0.62
Serum creatinine (μmol/L)117 ± 134
(64–364)
127 ± 97
(61–315)
93 ± 38
(46–371)
113 ± 178
(33–317)
98 ± 80
(35–281)
68 ± 57
(29–120)
0.1
Serum ferritin (μg/L)1107 ± 1340
(35–2478)
1488 ± 1110
(249–6241)
1185 ± 2122
(284–4034)
1709 ± 1895
(470–3052)
822 ± 1247
(444–2439)
715 ± 505
(324–3753)
0.74
Serum C-reactive protein (mg/L)114 ± 142
(33–279)
39 ± 128
(1–245)
30 ± 50
(4–94)
44 ± 59
(2–192)
43 ± 160
(0–338)
35 ± 3
(0–46)
0.28
Serum procalcitonin (ng/mL)0.1 ± 0.1
(0–0.1)
0.1 ± 0.1
(0–1.2)
0 ± 0.1
(0–1.3)
0.1 ± 0.1
(0–2.1)
0.2 ± 0.6
(0–0.7)
0 ± 0
(0–2.4)
0.92
Plasma D-dimer (ng/mL)1184 ± 568
(642–2128)
1254 ± 1601
(659–3581)
1145 ± 327
(799–1473)
1411 ± 672
(496–2384)
1592 ± 1108
(261–13920)
763 ± 250
(597–965)
0.21
* p values are calculated between data measured at inclusion and +28 days post-ECP.
Table 4. Immune biomarker profiling of patients enrolled in the study at baseline, inclusion and post-ECP follow-up.
Table 4. Immune biomarker profiling of patients enrolled in the study at baseline, inclusion and post-ECP follow-up.
ParameterBaselineInclusionPost-ECP Follow-Upp Value *
+3 Days+7 Days+14 Days+28 Days
Serum interferon-α (pg/mL)33.6 ± 25.8 (8.0–60.4)14.8 ± 11.8 (1.5–34.7)9.0 ± 3.3 (1.5–47.3)9.0 ± 10.5 (8.0–60.4)8.0 ± 0.3 (8.0–9.0)8.0 ± 0.1 (8.0–14.8)0.14
Serum interferon-γ (pg/mL)7.7 ± 25.5 (1.3–48.6)1.3 ± 34.5 (1.3–37.7)10.0 ± 16.0 (1.3–44.7)1.3 ± 31.6 (1.3–148.3)7.4 ± 20.0 (1.3–44.9)6.0 ± 4.9 (1.3–22.1)0.33
Plasma interleukin-1α (pg/mL)40.7 ± 23.6 (4.8–67.4)23.3 ± 16.3 (4.8–91.9)55.7 ± 45.0 (4.8–68.2)34.9 ± 48.0 (4.8–68.2)20.4 ± 156.1 (4.8–535.2)8.2 ± 55.9 (4.8–60.7)0.18
Plasma interleukin-1β (pg/mL)7.2 ± 9.0 (1.6–11.9)1.6 ± 3.8 (1.6–9.2)4,2 ± 5,8 (1.6–10.6)1.6 ± 4.6 (1.6–40.1)1.9 ± 1.6 (1.6–6.2)1.6 ± 0.1 (1.6–1.7)0.12
Plasma interleukin-2 (pg/mL)1.0 ± 1.1 (0.6–10.6)0.6 ± 0.7 (0.6–2.2)0.6 ± 0.8 (0.6–6.9)0.6 ± 0.1 (0.6–16.8)0.8 ± 1.1 (0.6–4.5)0.6 ± 0.3 (0.6–1.7)1.0
Plasma interleukin-4 (pg/mL)2.2 ± 6.3 (0.9–15.2)3.2 ± 2.8 (0.6–10.1)1.7 ± 4.6 (0.6–6.2)1.0 ± 10.5 (0.6–45.2)2.6 ± 5.9 (0.6–12.6)1.3 ± 2.1 (0.6–8.4)0.44
Plasma interleukin-5 (pg/mL)4.7 ± 21.7 (0.6–44.9)2.7 ± 1.4 (0.6–3.0)1.2 ± 0.7 (1.0–5.0)1.6 ± 129.5 (0.6–494.3)3.7 ± 19.0 (1.1–63.9)6.3 ± 21.1 (1.0–23.6)0.44
Plasma interleukin-6 (pg/mL)38.0 ± 38.0
(11.5–168.0)
203.5 ± 1688.0
(8.5–2776.0)
134.5 ± 398.5
(7.5–904.0)
84.0 ± 461.5
(20.5–1952.0)
751.0 ± 1907.0
(12.5–4381.0)
6.5 ± 24.0
(3.0–634.5)
0.04
Plasma interleukin-8 (pg/mL)28.9 ± 12.6 (5.2–94.0)14.3 ± 31.9 (11.3–66.6)22.6 ± 8.1 (9.7–68.1)34.2 ± 25.1 (8.3 ± 107.3)30.0 ± 12.5 (7.0–36.3)26.5 ± 21.7 (11.9–44.0)0.41
Plasma interleukin-10 (pg/mL)20.9 ± 10.6 (2.6–26.2)8.5 ± 18.5 (2.6–39.6)35.7 ± 17.5 (2.6 ± 86.3)21.9 ± 90.1 (5.0–135.1)4.4 ± 160.7 (2.6–634.7)18.0 ± 36.3 (2.6–70.3)0.64
Plasma interleukin-12 (pg/mL)4.0 ± 3.5 (3.0–93.8)3.0 ± 4.3 (3.0–57.8)3.0 ± 0.1 (3.0–71.9)3.0 ± 0.9 (3.0–247.8)3.0 ± 15.2 (3.0–63.7)3.0 ± 0.3 (3.0–32.2)0.11
Plasma interleukin-13 (pg/mL)32.2 ± 62.2 (6.4–274.5)20.7 ± 100.2 (6.4–264.1)18.5 ± 76.5 (6.4–111.1)10.6 ± 109.7 (6.4–408.7)30.5 ± 68.8 (6.4 ± 137.2)19.7 ± 30.5 (6.4–79.9)0.59
Plasma interleukin-15 (pg/mL)13.8 ± 7.5 (6.9–21.6)9.7 ± 8.2 (7.9–26.3)13.5 ± 6.9 (6.4–25.6)7.6 ± 3.3 (5.3–32.9)4.3 ± 1.0 (3.0–5.9)7.2 ± 5.9 (4.0 ± 12.9)0.68
Plasma interleukin-17A (pg/mL)6.5 ± 5.9 (1.3–11.1)4.1 ± 3.8 (1.3–9.2)4.7 ± 2.8 (1.3–7.4)1.3 ± 1.4 (1.3–8.3)1.3 ± 0.1 (1.3–1.3)1.3 ± 0.1 (1.3–1.3)<0.01
Serum IP-10 (pg/mL)1732.8 ± 2939.5
(205.0–5024.9)
120.7 ± 587.7
(101.8–1071.0)
166.3 ± 357.7
(63.9–583.4)
216.3 ± 230.3
(79.9–374.1)
104.9 ± 135.3
(73.6–488.9)
135.5 ± 193.9
(95.0–310.1)
0.5
Serum MCP-1 (pg/mL)972.8 ± 2068.9
(362.9–4650.5)
761.2 ± 680.3
(255.9–1886.2)
600.5 ± 204.6
(372.0–1484.6)
682.2 ± 123.7
(296.5–4010.7)
526.6 ± 432.9
(337.5–1512.0)
468.3 ± 18.8
(335.4–1408.2)
0.5
Serum MIP-1α (pg/mL)22.7 ± 17.2 (3.0–43.5)15.1 ± 6.6 (3.0–47.9)15.1 ± 11.8 (3.0–47.9)13.1 ± 39.0 (3.0–87.5)9.8 ± 31.4 (3.0–87.9)11.3 ± 10.6 (3.0–33.8)0.4
Serum RANTES (pg/mL)2416.4 ± 2479.9
(831.1–6169.0)
4172.3 ± 1419.0
(1125.9–17271.7)
3523.7 ± 1976.1
(320.7–7093.2)
2576.8 ± 1186.9
(624.5–3021.3)
2040.4 ± 589.2
(1068.8–2959.3)
1834.2 ± 1262.6
(619.7–2887.7)
0.85
Serum TNF-α (pg/mL)59.9 ± 44.0
(24.7–155.6)
38.5 ± 33.3
(18.5–80.2)
39.6 ± 16.8
(28.8–60.2)
39.6 ± 152.4
(6.4–244.0)
32.0 ± 37.3
(13.4–100.3)
26.3 ± 18.7
(8.0–47.6)
0.86
Serum TNF-β (pg/mL)22.0 ± 54.2 (2.2–69.2)18.5 ± 35.4 (1.6–71.0)10.8 ± 13.0 (1.6–39.7)5.3 ± 56.7 (1.6–72.7)9.2 ± 15.8 (1.6–25.8)4.6 ± 11.6 (1.6–15.3)0.33
Serum IgG (g/L)7.8 ± 3.8 (1.3–15.4)8.0 ± 1.9 (4.8–12.0)7.7 ± 0.7 (6.7–8.0)6.1 ± 1.8 (4.9–11.9)7.6 ± 0.1 (7.0–9.5)7.6 ± 2.4 (7.2–9.8)0.05
Serum IgA (g/L)1.9 ± 0.9 (0.5–5.1)2.1 ± 0.8 (0.3–3.3)1.4 ± 1.0 (0.5–2.6)2.0 ± 1.8 (0.4–3.0)1.9 ± 0.1 (1.9–1.9)1.9 ± 0.7 (0.5–2.0)0.97
Serum IgM (g/L)1.3 ± 1.1 (0.1–2.4)0.8 ± 0.6 (0–1.9)0.8 ± 1.7 (0–3.5)1.0 ± 1.2 (0–2.6)0.9 ± 0.1 (0–0.9)0.9 ± 0.1 (0.1–3.1)0.9
Serum electrophoresis/immunofixationNormal/negativeNormal/negativeNormal/negativeNormal/negativeNormal/negativeNormal/negativen.a.
Serum C3 (g/L)1.3 ± 0.3 (0.6–1.7)1.1 ± 0.3 (1.0–1.6)1.3 ± 0.8 (0.9–2.0)1.7 ± 0.4 (0.9–1.7)0.9 ± 0.1 (0.8–1.0)1.0 ± 0.1 (0.8–1.8)1
Serum C4 (g/L)0.3 ± 0.1 (0.2–0.5)0.3 ± 0.1 (0.1–0.4)0.3 ± 0.2 (0–0.4)0.3 ± 0.3 (0–0.4)0.1 ± 0.1 (0.1–0.2)0.2 ± 0.1 (0.1–0.5)0.97
Serum ENA ELISABelow c/oBelow c/oBelow c/oBelow c/oBelow c/oBelow c/on.a.
Serum anti-dsDNA (IU/mL)4.0 ± 7.5 (1.0–16.0)3.0 ± 0.8 (1.0–4.0)2.5 ± 3.0 (1.0–10.0)4.0 ± 1.0 (3.0–5.0)6.5 ± 0.5 (6.0–7.0)3.0 ± 4.0 (1.0–7.0)0.46
Serum ANCA IFBelow c/oBelow c/oBelow c/oBelow c/oBelow c/oBelow c/on.a.
Serum ANA IFBelow c/oBelow c/oBelow c/oBelow c/oBelow c/oBelow c/on.a.
* p values are calculated between data measured at inclusion and +28 days post-ECP. ANA: antinuclear antibody, ANCA: anti-neutrophil cytoplasmic antibody, C3: complement component 3, C4: complement component 4, c/o: cutoff, dsDNA: double-stranded doxyribonucleic acid, ELISA: enzyme-linked immunosorbent assay, ENA: extractable nuclear antigens, IF: immunofluorescence, IgA: immunoglobulin A, IgG: immunoglobulin G, IgM: immunoglobulin M, n.a.: not applicable.
Table 5. Peripheral whole-blood FACS parameters of enrolled patients at baseline, inclusion and post-ECP follow-up.
Table 5. Peripheral whole-blood FACS parameters of enrolled patients at baseline, inclusion and post-ECP follow-up.
ParameterBaselineInclusionPost-ECP Follow-Upp Value *
+3 Days+7 Days+14 Days+28 Days
Regulatory T-cell ratio (%)0.1 ± 0.1 (0.1–0.3)0.1 ± 0.1 (0–0.3)0.4 ± 0.1 (0.4–0.5)0.1 ± 0.5 (0.1–1.0)0.3 ± 0.1 (0.3–0.3)0.2 ± 0.3 (0.1–0.3)0.74
CD3+ T-cell count (cells/μL)656 ± 654 (277–2377)367 ± 473 (321–1267)469 ± 113 (382–609)802 ± 560 (445–1990)1987 ± 447 (1536–2430)1888 ± 789 (1454–4324)0.28
CD3+/CD8+ T-cell count (cells/μL)215 ± 164 (158–630)117 ± 71 (97–318)193 ± 47 (119–213)194 ± 31 (150–246)189 ± 77 (89–244)287 ± 125 (112–361)0.87
CD3+/CD4+ T-cell count (cells/μL)406 ± 542 (213–1834)203 ± 677 (96–2187)481 ± 796 (342–1934)711 ± 741 (282–1766)1286 ± 889 (397–2175)1159 ± 1946 (61–3954)0.46
CD3-/CD16+/CD56+ NK-cell count (cells/μL)90 ± 33 (65–162)38 ± 77 (11–245)56 ± 48 (26–124)42 ± 24 (20–68)35 ± 5 (29–39)59 ± 25 (33–84)0.88
CD19+ B-cell count (cells/μL)126 ± 69 (59–198)44 ± 97 (6.0–191.7)136 ± 213 (96–523)188 ± 47 (107–202)235 ± 111 (43–267)212 ± 197 (59–453)0.2
Double-negative T-cell ratio (%)1.0 ± 0.7 (0–3.4)1.7 ± 2.1 (0.2–7.5)0.8 ± 2.5 (0.6–5.6)1.1 ± 0.8 (0.5–2.0)1.1 ± 0.7 (0.4–1.9)0.4 ± 2.0 (0.2–4.1)0.28
Double-positive T-cell ratio (%)0.1 ± 0.3 (0.01–0.8)0.1 ± 0.4 (0.04–1.2)0.1 ± 0.1 (0.03–0.2)0.1 ± 0.1 (0.01–0.2)0.1 ± 0.1 (0.05–0.12)0.2 ± 0.1 (0.1–0.2)1.0
Counts within the CD3+ gate:
CD3+/TCRαβ+ T-cell (cells/μL)949 ± 221 (687–1546)601 ± 631 (212–2368)489 ± 83 (435–601)906 ± 774 (434–1982)567 ± 1128 (164–2421)1440 ± 2067 (172–4301)1.0
CD3+/TCRγδ+ T-cell (cells/μL)23.5 ± 19.6 (14.3–53.7)43.1 ± 37.3 (7.1–77.3)27.6 ± 21.9 (6.7–45.6)10.2 ± 2.5 (6.0–11.0)14.1 ± 4.3 (9.7–18.4)13.1 ± 3.1 (11.0–17.3)0.23
CD3+/CD25+ T-cell (cells/μL)54.3 ± 26.5 (14.2–67.1)19.3 ± 9.9 (2.8–26.6)63.5 ± 23.4 (42.1–88.9)77.9 ± 148.0 (21.9–317.9)48.0 ± 33.5 (14.6–81.5)60.5 ± 108.6 (12.7–229.8)0.14
CD3+/HLA-DR+ T-cell (cells/μL)146.6 ± 211.5 (23.1–524.8)89.6 ± 203.2 (44.7–615.8)304.9 ± 297.1 (49.1–682.4)56.1 ± 333.8 (45.0–712.6)358.9 ± 307.4 (51.2–666.1)98.1 ± 375.0 (41.7–1370.8)0.56
Counts within the CD4+ gate:
CD4+/CD45RA+ naive T-cell (cells/μL)162.3 ± 487.3
(86.2–1548.2)
105.8 ± 599.3
(11.9–2045.4)
152.4 ± 110.1
(29.4–249.6)
335.6 ± 767.6
(102.4–1637.5)
1056.0 ± 869.6
(186.4–1925.6)
547.1 ± 1807.0
(16.2–3630.1)
0.69
CD4+/CD45RO+ memory T-cell (cells/μL)89.9 ± 53.9
(26.5–112.2)
113.4 ± 60.8
(52.0–140.0)
206.1 ± 21.9
(184.2–227.9)
178.2 ± 121.1
(129.0–371.2)
229.4 ± 16.4
(213.0–245.9)
286.7 ± 195.7
(45.3–602.7)
0.2
Counts within the CD8+ gate:
CD8+/CD45RA+ naive T-cell (cells/μL)167.2 ± 34.3 (147.8–216.4)175.0 ± 171.3 (46.6–506.6)173.5 ± 186.5 (65.3–438.1)102.2 ± 21.1 (65.2–107.6)74.0 ± 24.1 (49.9–98.1)90.5 ± 29.5 (78.5–136.9)0.93
CD8+/CD45RO+ memory T-cell (cells/μL)42.5 ± 24.8 (12.8–62.4)76.2 ± 94.1 (20.9–136.4)51.2 ± 2.2 (48.7–53.1)90.0 ± 53.3 (41.6–147.7)159.5 ± 33.1 (126.4–192.6)144.4 ± 127.0 (21.6–275.7)0.03
* p values are calculated between data measured at inclusion and +28 days post-ECP.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Szabó, B.G.; Reményi, P.; Tasnády, S.; Korózs, D.; Gopcsa, L.; Réti, M.; Várkonyi, A.; Sinkó, J.; Lakatos, B.; Szlávik, J.; et al. Extracorporeal Photopheresis as a Possible Therapeutic Approach for Adults with Severe and Critical COVID-19 Non-Responsive to Standard Treatment: A Pilot Investigational Study. J. Clin. Med. 2023, 12, 5000. https://doi.org/10.3390/jcm12155000

AMA Style

Szabó BG, Reményi P, Tasnády S, Korózs D, Gopcsa L, Réti M, Várkonyi A, Sinkó J, Lakatos B, Szlávik J, et al. Extracorporeal Photopheresis as a Possible Therapeutic Approach for Adults with Severe and Critical COVID-19 Non-Responsive to Standard Treatment: A Pilot Investigational Study. Journal of Clinical Medicine. 2023; 12(15):5000. https://doi.org/10.3390/jcm12155000

Chicago/Turabian Style

Szabó, Bálint Gergely, Péter Reményi, Szabolcs Tasnády, Dorina Korózs, László Gopcsa, Marienn Réti, Andrea Várkonyi, János Sinkó, Botond Lakatos, János Szlávik, and et al. 2023. "Extracorporeal Photopheresis as a Possible Therapeutic Approach for Adults with Severe and Critical COVID-19 Non-Responsive to Standard Treatment: A Pilot Investigational Study" Journal of Clinical Medicine 12, no. 15: 5000. https://doi.org/10.3390/jcm12155000

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop