Next Article in Journal
Nanog, in Cooperation with AP1, Increases the Expression of E6/E7 Oncogenes from HPV Types 16/18
Next Article in Special Issue
Protective Effects of Astodrimer Sodium 1% Nasal Spray Formulation against SARS-CoV-2 Nasal Challenge in K18-hACE2 Mice
Previous Article in Journal
In Vitro and In Vivo Evaluation of Human Adenovirus Type 49 as a Vector for Therapeutic Applications
Previous Article in Special Issue
Compelling Evidence for the Activity of Antiviral Peptides against SARS-CoV-2
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

What Is the Role of Therapeutic Plasma Exchange as an Adjunctive Treatment in Severe COVID-19: A Systematic Review

Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
*
Author to whom correspondence should be addressed.
Viruses 2021, 13(8), 1484; https://doi.org/10.3390/v13081484
Submission received: 1 July 2021 / Revised: 22 July 2021 / Accepted: 26 July 2021 / Published: 28 July 2021

Abstract

:
Introduction: Since the COVID-19 pandemic outbreak, multiple promising treatment modalities have been tested, however, only several of them were proven to be effective. Therapeutic plasma exchange (TPE) has been recently discussed as a possible supportive treatment for severe cases. Methods: To investigate a possible role of TPE in severe COVID-19 we used a structured systematic search strategy to retrieve all relevant publications in the field. We screened in PubMed, EMBASE, Web of Science, Cochrane Library and clinicaltrials.gov for data published until the 4 June 2021. Results: We identified 18 papers, enrolling 384 patients, 220 of whom received TPE. The number of TPE sessions ranged from 1 to 9 and the type of replacement fluid varied markedly between studies (fresh frozen plasma or 5% albumin solution, or convalescent plasma). Biochemical improvement was observed in majority of studies as far as C-reactive protein (CRP), interleukin-6 (IL-6), ferritin, lactate dehydrogenase (LDH), D-dimer concentrations and lymphocyte count are concerned. The improvement at a laboratory level was associated with enhancement of respiratory function. Adverse effects were limited to five episodes of transient hypotension and one femoral artery puncture and thrombophlebitis. Conclusions: Although the effect of therapeutic plasma exchange on mortality remains unclarified, the procedure seems to improve various secondary end-points such as PaO2/FiO2 ratio or biomarkers of inflammation. Therapeutic plasma exchange appears to be a safe treatment modality in COVID-19 patients in terms of side effects.

1. Introduction

Since the COVID-19 pandemic outbreak, multiple promising treatment modalities have been tested, however, only several of them were proven to be effective. Mortality of critically ill COVID-19 patients remains high, depending on population characteristics [1,2,3].
One of frequently discussed pathomechanisms for the severe course of COVID-19 is an excessive immune response leading to proinflammatory cytokine storm (often similar to the course of macrophage activation syndrome) that is associated with multiorgan dysfunction [4]. Moreover, a heavily studied process is hypercoagulability induced by numerous mechanisms: SARS-CoV2 tropism towards ACE II receptors, excessive complement activation, production of harmful antibodies (often similar to those found in antiphospholipid syndrome), formation of immunological complexes, release of procoagulant factors (e.g., von Willebrand factor) and diffused endothelialitis [5,6,7,8,9]. Usually, lactate dehydrogenase (LDH), ferritin, interleukin-6 (IL-6), C-reactive protein (CRP) and D-dimers are discussed as the biomarkers for predicting the severity of the disease [10,11,12,13,14].
Therapeutic plasma exchange (TPE) is a procedure in which plasma is separated from the morphotic elements of blood and is then replaced by either albumin solution or fresh frozen plasma (FFP). The aim of TPE is to eliminate morbific factors, often pathological antibodies [15]. Myasthenia gravis, inflammatory demyelinating polyneuropathies, thrombotic microangiopathy or macrophage activation syndrome are only narrow examples of the applications of TPE [16]. In the latter diseases, the elimination of pathological antibodies reduces the procoagulable state and, therefore, improves survival of patients.
As organ injury is triggered by cytokine storm-mediated immune reaction, theoretically, the elimination of cytokines and harmful antibodies could attenuate the severity of the disease. Additional removal of fibrin degradation products (e.g., D-dimers) could also improve the hemostatic balance [17]. For those reasons, TPE has been recently discussed as a possible supportive treatment for severe COVID-19 cases [18]. The purpose of this review was to investigate efficacy and safety of TPE in severe COVID-19 in a systematic manner. Participants, interventions, controls and outcomes (PICO) criteria are presented in Table 1.

2. Methods

By following the PRISMA guidelines, we used a structured systematic search strategy to retrieve all relevant publications regarding TPE use in severe COVID-19 [19]. We screened for data that were published until 4 June 2021 in PubMed, EMBASE, Web of Science, Cochrane Library and clinicaltrials.gov. The search string was as follows: (plasmapheresis) OR (therapeutic plasma exchange) OR (total plasma exchange) OR (apheresis) OR (plasma exchange) AND (sars-cov-2) OR (coronavirus) OR (COVID-19). We excluded animal studies, papers not in the English language, non-original papers and case reports (but not case series). Duplicates were identified and excluded as well. The remaining records were screened by three independent investigators and full texts were retrieved if at least two adjudicators agreed to include the paper. Differences of opinion were resolved by a discussion. Then, available manuscripts were reviewed by all investigators and included into a comprehensive assessment if three adjudicators agreed that the study results were compliant with the goals of this review. If no agreement was reached, then a fourth reviewer made a final decision. For our analysis, we retrieved the following items from the included studies: authors, year of publication, type of a study, patient’s characteristics, concomitant therapies, time of TPE initiation and cessation, dose of TPE, type of replacement fluids, adverse effects associated with TPE and outcomes (change in inflammatory biomarkers concentrations, clinical changes and survival). We used the RoB2 tool for the assessment of the risk of bias of randomized controlled trials [20].

3. Results

3.1. Included Studies

By using the search string within various medical databases (presented in the Methods Section) we identified 825 articles in total. After removing duplicates (n = 401) we screened the remaining papers by evaluating titles and abstracts (n = 424). By using the PICO criteria and the inclusion and the exclusion criteria, we distinguished 39 papers for the full-text read assessment. After excluding some of the articles for numerous reasons, the final 18 papers were included in the systematic review. The most common types of studies were case-series studies (n = 14) [21,22,23,24,25,26,27,28,29,30,31,32,33,34], then case-control studies (n = 2) [17,35] and a propensity score matched study [36]. Only one randomized controlled trial was included in the analysis [37]. Study selection process is presented on the flowchart (Figure 1). A summary of published studies is shown in Table 2. Extended data regarding the included studies is presented in the Table S1.

3.2. Quality Assessment

Only one study was a randomized controlled trial [37]. The risk of bias of the study was rated as “low” by implementing the RoB2 tool (randomization process: low risk; deviations from the intended interventions: low risk; missing outcome data: low risk; measurement of the outcome: low risk; selection of the reported results: low risk). The remaining studies (n = 17) consisted mostly of case-series and observational data of limited populations. Therefore, we collectively defined the risk of bias of those studies as “high” due to methodological reasons.

3.3. Patient Characteristics

Out of 384 patients, 220 received TPE. The number of patients in the studies varied from 3 to 90 with a median of 8 patients (IQR 5–18) [17,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37]. The mean or the median age of participants in seven studies was below 65 years [8,10,11,12,13,14,15], whereas in the remaining four studies, it was above 65 years [5,6,7,9]. Information regarding gender was available in all of the studies [17,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37], of which the majority of patients were male. Only three studies had a similar gender ratio of about 40%–60% [22,30,33]. The most common reason to qualify a patient for TPE was acute respiratory distress syndrome (ARDS) (n = 8 [21,22,23,24,32,33,35,37]). Another frequent inclusion criterion was cytokine release syndrome (CRS) [23,25,36,37]. The mean or the median SOFA score on admission was provided in nine studies and varied between 5 and 12.3 [17,23,29,30,31,34,35,36,37]. Data regarding the frequency of invasive mechanical ventilation (and intubation) were available in 17 studies and varied between 16% and 100% [17,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37].

3.4. Interventions

The information about days between the onset of COVID-19 symptoms and the initiation of TPE was provided in nine studies and varied from 6.5 to 39 days (median = 14 days) [21,22,23,24,26,29,32,33,37]. The number of TPE sessions varied between 1 and 9 [17,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37]. The information regarding single dose of TPE was provided in 11 studies and the most common dosage varied between 1 and 1.5× of the patient’s plasma volume [21,22,23,24,26,28,30,31,33,36,37]. In terms of replacement fluid, FFP was used in nine studies [21,22,28,32,33,34,35,36,37], 5% albumin solution was used in five studies (in three of those studies, together with FPP) [23,25,27,29,30] and convalescent plasma was used in two studies [24,31]. One study did not provide information regarding the type of a replacement fluid [17]. In regard to other immunomodulatory treatments along with TPE, all studies but one implemented various pharmacological treatments [17,21,22,23,24,25,26,27,28,29,30,31,32,33,35,36,37]. The most common immunomodulatory drugs were corticosteroids [17,22,23,24,26,28,29,31,32,33,36,37].

3.5. Outcomes

The most frequent outcome reported in the studies was mortality. The studies varied in terms of the day of mortality assessment, however, the most frequent was day 28 (n = 5) [23,24,26,35,36], followed by day 14 (n = 3) [21,25,27]. The randomized controlled trial by Faqihi et al. reported mortality at day 35 [37]. In nine studies, no specified day of mortality assessment was provided [17,22,28,29,30,31,32,33,34]. The mortality varied across the studies, ranging from 0% to 60% (median = 18.35%, IQR: 8.3%–28.6%). Four studies reported differences in mortality between patients who received TPE and those who did not receive it (8.3% vs. 58.3%, 0% vs. 35%, 8.9% vs. 38.5% and 20.9% vs. 34.1%) [17,35,36,37]. The median concentrations of pre-TPE and post-TPE biomarkers provided by the studies are presented in Table 3.
In a randomized controlled trial by Faqihi et al., the overall mortality difference was not significant (20.9% vs. 34.1%; p = 0.09); however, the median length of stay and duration of mechanical ventilation reached statistical significance. Additionally, PaO2/FiO2, Lymphocyte count, IL-6, LDH, D-Dimers, Ferritin and ADAMTS-13 activity changed significantly after the implementation of TPE (Table S1) [37].

3.6. Side Effects

Data regarding the side effects of TPE were available in 10 studies [23,24,25,26,28,29,34,35,36,37]. Most of those studies reported no side effects related to the use of TPE [23,25,28,29,34,37]. The most frequent side effect was hypotension (which occurred in five patients across all the populations from the studies) [24,26,35]. There was one episode of femoral artery puncture and thrombophlebitis [36]. There were no deaths associated with the procedure.

4. Discussion

This systematic review focused on summarizing the data concerning the role of TPE in severe COVID-19 infection (as of June 2021). Based on the papers included in this review, we cannot produce a clear message regarding the effect that TPE has on mortality. However, the surrogate endpoints such as improvements in various biomarkers (CRP, LDH, D-Dimers, Ferritin, IL-6, PaO2/FiO2 ratio, etc.) seem to be well documented and are consistent among the studies. The frequency of side effects related to TPE is low. The considerable heterogeneity within and among the studies, and the fact that the majority of them were case-series studies, limits drawing definite conclusions.
A considerable number of included studies presented a positive effect of TPE on mortality. However, the majority of those studies were case-series or case-control studies of limited populations. Such a strong effect in that many studies (even a 0% mortality rate in critically ill patients) most probably is a result of a publication bias. Indeed, the only RCT (of low risk of bias) included in this review failed to deliver significant results regarding mortality (the study might have been underpowered as it was terminated due to low patient recruitment). However, the study was able to present a significant reduction in days of mechanical ventilation or hospital length of stay.
It must be pointed out that the vast majority of patients who underwent TPE among the studies were intubated and often suffered from septic shock and progressing multi-organ failure. As intubation in COVID-19 patients is often related with the compromised prognosis, we speculate that it is possible that the implementation of TPE in such a critical condition is no longer able to restore homeostasis [38]. For example, in the PLEXIT study, TPE was initiated mostly in non-invasively ventilated patients. In that study, the patients underwent the TPE procedure on the basis of CRS recognition (higher levels of ferritin, CRP, D-dimers, LDH and lymphopenia) [36]. Therefore, we may assume that signs and symptoms of rapid deterioration in organ function, including respiratory failure, could serve as indications for TPE application. The SOFA score may be applied as an easy-to-use method of multi-organ failure assessment. Such a hypothesis could be tested in the future, well-designed studies.
In regards to biochemical improvements, the summary of those is presented in Table 3. Despite the values not being weighted, they provide a rather clear trend of changes that occurred within various biomarkers. As LDH, ferritin, IL-6, CRP and D-dimers are discussed as the biomarkers in predicting severity of the disease, one may speculate that perhaps those molecules could serve as indicators for TPE initiation [10,11,12,13,14]. However, the trigger points would still be unknown. Based on previous data, the patients may benefit the most from removing IL-6. The molecule works as a procoagulant cytokine and probably is one of the factors that accounts for microvascular thrombosis in the course of infection [39]. Interestingly, Guiaro et al. published a study in which they provided a cut-off point of 35 pg/mL of IL-6 that was associated with an increased risk of mortality and ICU admission [40]. Perhaps such a cut-off point could be discussed as one of the possible trigger points for TPE. In our review, the median value of IL-6 concentration prior to TPE initiation obtained from the included studies was 118.7 pg/mL (IQR 25.2–295.3).
Perhaps LDH removal is of particular importance as well. Its concentrations correlate with the release of proinflammatory molecules and lymphocyte count, which corresponds with the severity of the disease [41]. Increase of LDH may be harmful by production of lactate and enhancement of immune-suppressive cells and inhibition of natural killer (NK) cells and cytotoxic T-lymphocytes. The similar issue regards ferritin. Its elevated levels were found in patients with macrophage activation syndrome (MAS) and cytokine storm. Increase in ferritin concentration negatively impacts immunological condition as it plays a role in the inflammation process through its binding with the T-cell immunoglobulin and the expression of multiple proinflammatory mediators [42]. Noteworthily, severe COVID-19 patients may produce procoagulant antibodies, such as lupus anticoagulants and antibodies found in antiphospholipid syndrome [43].
TPE safety has been confirmed in the past [44,45,46]. In our analysis, adverse effects during TPE in COVID-19 were rather anecdotal, but we still need to bear in mind the limited number of observations and a considerable number of papers in which no information regarding side effects was provided. Importantly, none of the presented side effects posed a threat to a patient that could not be properly handled. The most common adverse effect in patients undergoing TPE are urticaria, hypocalcemia, rigors and headaches. More concerns were related to complications of the central venous catheter insertion site. None of them appeared in the included papers.

5. Limitations

Firstly, a limited number of trials have been completed so far. Due to methodological heterogeneity between publications and subsequent risk of bias, we failed to prepare a meta-analysis. Case studies usually describe positive data; therefore, strong publication bias exists and the above-mentioned studies should be interpreted with caution. Only one randomized controlled trial has been performed [37]. The included studies suffer from lack of adequate reporting, namely, day of mortality assessment, side effects or median time from symptoms to TPE initiation. Furthermore, the procedure in COVID-19 has not been standardized yet. Clinical indications, time of commencement of TPE, number of sessions, time gaps between them and the type of replacement fluid varied between studies. Noteworthily, the type of a replacement fluid is debatable. Albumin solutions may cause significant and unpredictable disturbances of blood coagulation due to loss of pro- as well as anticoagulant factors [47,48]. One may then expect that convalescent plasma should be the first choice of treatment [49,50], but recent data are not so convincing [51]. Moreover, plasma transfusion may have serious side effects, including transfusion-related immunomodulation and transfusion-related lung injury [52], which are of particular importance in COVID-19 patients with respiratory failure. Moreover, the time at which various biomarkers’ concentrations were measured was different between the studies. So, reliable assessment of TPE efficacy requires the unification of procedure-related issues. In the future, this should be clarified. Lastly, TPE is never used as the sole treatment option. It is only a part of the complex patient-oriented multifactorial therapy.

6. Conclusions

Although the effect of therapeutic plasma exchange on mortality remains unclarified, the procedure seems to improve various secondary end-points such as PaO2/FiO2 ratio or biomarkers of inflammation. Therapeutic plasma exchange appears to be a safe treatment modality in COVID-19 patients in terms of side effects.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/v13081484/s1, Table S1: Detailed summary of the included studies.

Author Contributions

Conceptualization, Ł.J.K.; Systematic search: Z.P., M.C., M.H.; writing—original draft preparation, Ł.J.K., Z.P., M.C., M.H.; writing—review and editing, Ł.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this article, take responsibility for the integrity of the work as a whole and have given their approval for this version to be published.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cummings, M.J.; Baldwin, M.R.; Abrams, D.; Jacobson, S.D.; Meyer, B.J.; Balough, E.M.; Aaron, J.G.; Claassen, J.; Rabbani, L.E.; Hastie, J.; et al. Epidemiology, clinical course, and outcomes of critically ill adults with COVID-19 in New York City: A prospective cohort study. Lancet 2020, 395, 1763–1770. [Google Scholar] [CrossRef]
  2. Wu, Z.; McGoogan, J.M. Characteristics of and Important Lessons From the Coronavirus Disease 2019 (COVID-19) Outbreak in China: Summary of a Report of 72,314 Cases From the Chinese Center for Disease Control and Prevention. JAMA 2020, 323, 1239–1242. [Google Scholar] [CrossRef]
  3. Wu, C.; Chen, X.; Cai, Y.; Xia, J.; Zhou, X.; Xu, S.; Huang, H.; Zhang, L.; Zhou, X.; Du, C.; et al. Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease 2019 Pneumonia in Wuhan, China. JAMA Intern. Med. 2020, 180, 934–943. [Google Scholar] [CrossRef] [Green Version]
  4. Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; HLH Across Speciality Collaboration, UK. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
  5. Giannis, D.; Ziogas, I.A.; Gianni, P. Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J. Clin. Virol. 2020, 127, 104362. [Google Scholar] [CrossRef] [PubMed]
  6. Rico-Mesa, J.S.; Rosas, D.; Ahmadian-Tehrani, A.; White, A.; Anderson, A.S.; Chilton, R. The Role of Anticoagulation in COVID-19-Induced Hypercoagulability. Curr. Cardiol. Rep. 2020, 22, 53. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, Y.; Xiao, M.; Zhang, S.; Xia, P.; Cao, W.; Jiang, W.; Chen, H.; Ding, X.; Zhao, H.; Zhang, H.; et al. Coagulopathy and Antiphospholipid Antibodies in Patients with Covid-19. N. Engl. J. Med. 2020, 382, e38. [Google Scholar] [CrossRef] [PubMed]
  8. Chang, J.C. Acute Respiratory Distress Syndrome as an Organ Phenotype of Vascular Microthrombotic Disease: Based on Hemostatic Theory and Endothelial Molecular Pathogenesis. Clin. Appl. Thromb. 2019, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial cell infection and endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  10. Teachey, D.T.; Lacey, S.F.; Shaw, P.A.; Melenhorst, J.J.; Maude, S.L.; Jeffrey, F.; Pequignot, E.; Gonzalez, V.E.; Chen, F.; Finklestein, J.; et al. Identification of Predictive Biomarkers for Cytokine Release Syndrome after Chimeric Antigen Receptor T-cell Therapy for Acute Lymphoblastic Leukemia. Cancer Discov. 2016, 6, 664–679. [Google Scholar] [CrossRef] [Green Version]
  11. Henry, B.M.; Aggarwal, G.; Wong, J.; Benoit, S.; Vikse, J.; Plebani, M.; Lippi, G. Lactate dehydrogenase levels predict coronavirus disease 2019 (COVID-19) severity and mortality: A pooled analysis. Am. J. Emerg. Med. 2020, 38, 1722–1726. [Google Scholar] [CrossRef]
  12. McGonagle, D.; Sharif, K.; O’Regan, A.; Bridgewood, C. The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease. Autoimmun. Rev. 2020, 19, 102537. [Google Scholar] [CrossRef] [PubMed]
  13. Luo, X.; Zhou, W.; Yan, X.; Guo, T.; Wang, B.; Xia, H.; Ye, L.; Xiong, J.; Jiang, Z.; Liu, Y.; et al. Prognostic Value of C-Reactive Protein in Patients With Coronavirus 2019. Clin. Infect. Dis. 2020, 71, 2174–2179. [Google Scholar] [CrossRef]
  14. Jose, R.J.P.; Manuel, A. COVID-19 cytokine storm: The interplay between inflammation and coagulation. Lancet Respir. Med. 2020, 8, e46–e47. [Google Scholar] [CrossRef]
  15. Clark, W.; Huang, S. Introduction to therapeutic plasma exchange. Transfus. Apher. Sci. 2019, 58, 228–229. [Google Scholar] [CrossRef]
  16. Padmanabhan, A.; Connelly-Smith, L.; Aqui, N.; Balogun, R.A.; Klingel, R.; Meyer, E.; Pham, H.P.; Schneiderman, J.; Witt, V.; Wu, Y.; et al. Guidelines on the Use of Therapeutic Apheresis in Clinical Practice—Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Eighth Special Issue. J. Clin. Apher. 2019, 34, 171–354. [Google Scholar] [CrossRef] [PubMed]
  17. Gucyetmez, B.; Atalan, H.K.; Sertdemir, I.; Cakir, U.; Telci, L.; COVID-19 Study Group. Therapeutic plasma exchange in patients with COVID-19 pneumonia in intensive care unit: A retrospective study. Crit. Care 2020, 24, 492. [Google Scholar] [CrossRef] [PubMed]
  18. Keith, P.; Day, M.; Perkins, L.; Moyer, L.; Hewitt, K.; Wells, A. A novel treatment approach to the novel coronavirus: An argument for the use of therapeutic plasma exchange for fulminant COVID-19. Crit. Care 2020, 24, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gøtzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: Explanation and elaboration. BMJ 2009, 339, b2700. [Google Scholar] [CrossRef] [Green Version]
  20. Sterne, J.A.C.; Savović, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.-Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef] [Green Version]
  21. Zhang, L.; Zhai, H.; Ma, S.; Chen, J.; Gao, Y. Efficacy of therapeutic plasma exchange in severe COVID-19 patients. Br. J. Haematol. 2020, 190, 181–183. [Google Scholar] [CrossRef] [PubMed]
  22. Morath, C.; Weigand, M.A.; Zeier, M.; Speer, C.; Tiwari-Heckler, S.; Merle, U. Plasma exchange in critically ill COVID-19 patients. Crit. Care 2020, 24, 481. [Google Scholar] [CrossRef] [PubMed]
  23. Faqihi, F.; Alharthy, A.; Alodat, M.; Kutsogiannis, D.J.; Brindley, P.G.; Karakitsos, D. Therapeutic plasma exchange in adult critically ill patients with life-threatening SARS-CoV-2 disease: A pilot study. J. Crit. Care 2020, 60, 328–333. [Google Scholar] [CrossRef] [PubMed]
  24. Jaiswal, V.; Nasa, P.; Raouf, M.; Gupta, M.; Dewedar, H.; Mohammad, H.; Al Rais, Z.; Baqer, M.A.; Alsabbah, A.; Ibrahim, Y.; et al. Therapeutic plasma exchange followed by convalescent plasma transfusion in critical COVID-19—An exploratory study. Int. J. Infect. Dis. 2021, 102, 332–334. [Google Scholar] [CrossRef]
  25. Gluck, W.L.; Callahan, S.P.; Brevetta, R.A.; Stenbit, A.E.; Smith, W.M.; Martin, J.C.; Blenda, A.V.; Arce, S.; Edenfield, W.J. Efficacy of therapeutic plasma exchange in the treatment of penn class 3 and 4 cytokine release syndrome complicating COVID-19. Respir. Med. 2020, 175, 106188. [Google Scholar] [CrossRef]
  26. Fernandez, J.; Ginès, J.G.; Olivas, P.; Costa, M.; Nieto, S.; Mateo, D.; Sánchez, M.B.; Aguilar, F.; Bassegoda, O.; Covid Clinic Critical Care (CCCC) Group; et al. Plasma Exchange: An Effective Rescue Therapy in Critically Ill Patients With Coronavirus Disease 2019 Infection. Crit. Care Med. 2020, 48, e1350–e1355. [Google Scholar] [CrossRef]
  27. Dogan, L.; Kaya, D.; Sarıkaya, Z.T.; Zengin, R.; Dincer, A.; Akinci, I.O.; Afsar, N. Plasmapheresis treatment in COVID-19–related autoimmune meningoencephalitis: Case series. Brain Behav. Immun. 2020, 87, 155–158. [Google Scholar] [CrossRef]
  28. Adeli, S.H.; Asghari, A.; Tabarraii, R.; Shajari, R.; Afshari, S.; Kalhor, N.; Vafaeimanesh, J. Therapeutic plasma exchange as a rescue therapy in patients with coronavirus disease 2019: A case series. Pol. Arch. Intern. Med. 2020, 130, 455–458. [Google Scholar] [CrossRef] [PubMed]
  29. de Prost, N.; Bastard, P.; Arrestier, R.; Fourati, S.; Mahévas, M.; Burrel, S.; Dorgham, K.; Gorochov, G.; Tandjaoui-Lambiotte, Y.; Azzaoui, I.; et al. Plasma Exchange to Rescue Patients with Autoantibodies Against Type I Interferons and Life-Threatening COVID-19 Pneumonia. J. Clin. Immunol. 2021, 41, 536–544. [Google Scholar] [CrossRef]
  30. Hashemian, S.M.; Shafigh, N.; Afzal, G.; Jamaati, H.; Tabarsi, P.; Marjani, M.; Malekmohammad, M.; Mortazavi, S.M.; Khoundabi, B.; Mansouri, D.; et al. Plasmapheresis reduces cytokine and immune cell levels in COVID-19 patients with acute respiratory distress syndrome (ARDS). Pulmonology 2020. [Google Scholar] [CrossRef] [PubMed]
  31. Keith, P.D.; Scott, L.K.; Weaver, K.E.; Day, M.; Choe, C.; Perkins, L.; Moyer, L.; Hays, E.; French, M.; Hewitt, K.; et al. Treatment of Critically Ill Coronavirus Disease 2019 Patients With Adjunct Therapeutic Plasma Exchange: A Single-Center Retrospective Case Series. Crit. Care Explor. 2020, 2, e0223. [Google Scholar] [CrossRef]
  32. Matsushita, Y.; Kusaoi, M.; Hiki, M.; Murayama, G.; Abe, Y.; Nozawa, K.; Takahashi, K.; Yamaji, K.; Tamura, N.; Naito, T. Combination therapy with plasma exchange and glucocorticoid may be effective for severe COVID-19 infection: A retrospective observational study. Ther. Apher. Dial. 2021, 25, 390–400. [Google Scholar] [CrossRef] [PubMed]
  33. Roshandel, E.; Sankanian, G.; Salimi, M.; Jalili, A.; Salari, S.; Sadeghi, A.; Hashemian, S.M.; Moshari, M.R.; Pirsalehi, A.; Hajifathali, A. Plasma exchange followed by convalescent plasma transfusion in COVID-19 patients. Transfus. Apher. Sci. 2021, 103141. [Google Scholar] [CrossRef]
  34. Truong, A.D.; Auld, S.C.; Barker, N.A.; Friend, S.; Wynn, A.T.; Cobb, J.; Sniecinski, R.M.; Tanksley, C.; Polly, D.M.; Gaddh, M.; et al. Therapeutic plasma exchange for COVID-19-associated hyperviscosity. Transfusion 2021, 61, 1029–1034. [Google Scholar] [CrossRef]
  35. Khamis, F.; Al-Zakwani, I.; Al Hashmi, S.; Al Dowaiki, S.; Al Bahrani, M.; Pandak, N.; Al Khalili, H.; Memish, Z. Therapeutic plasma exchange in adults with severe COVID-19 infection. Int. J. Infect. Dis. 2020, 99, 214–218. [Google Scholar] [CrossRef] [PubMed]
  36. Kamran, S.M.; Mirza, Z.-E.-H.; Naseem, A.; Liaqat, J.; Fazal, I.; Alamgir, W.; Saeed, F.; Saleem, S.; Nisar, S.; Yousaf, M.A.; et al. Therapeutic plasma exchange for coronavirus disease-2019 triggered cytokine release syndrome; a retrospective propensity matched control study. PLoS ONE 2021, 16, e0244853. [Google Scholar] [CrossRef] [PubMed]
  37. Faqihi, F.; Alharthy, A.; Abdulaziz, S.; Balhamar, A.; Alomari, A.; AlAseri, Z.; Tamim, H.; Alqahtani, S.A.; Kutsogiannis, D.J.; Brindley, P.G.; et al. Therapeutic plasma exchange in patients with life-threatening COVID-19: A randomised controlled clinical trial. Int. J. Antimicrob. Agents 2021, 57, 106334. [Google Scholar] [CrossRef]
  38. Papoutsi, E.; Giannakoulis, V.G.; Xourgia, E.; Routsi, C.; Kotanidou, A.; Siempos, I.I. Effect of timing of intubation on clinical outcomes of critically ill patients with COVID-19: A systematic review and meta-analysis of non-randomized cohort studies. Crit. Care 2021, 25, 121. [Google Scholar] [CrossRef]
  39. McGonagle, D.; O’Donnell, J.S.; Sharif, K.; Emery, P.; Bridgewood, C. Immune mechanisms of pulmonary intravascular coagulopathy in COVID-19 pneumonia. Lancet Rheumatol. 2020, 2, e437–e445. [Google Scholar] [CrossRef]
  40. Guirao, J.J.; Cabrera, C.M.; Jiménez, N.; Rincón, L.; Urra, J.M. High serum IL-6 values increase the risk of mortality and the severity of pneumonia in patients diagnosed with COVID-19. Mol. Immunol. 2020, 128, 64–68. [Google Scholar] [CrossRef]
  41. Han, Y.; Zhang, H.; Mu, S.; Wei, W.; Jin, C.; Tong, C.; Song, Z.; Zha, Y.; Xue, Y.; Gu, G. Lactate dehydrogenase, an independent risk factor of severe COVID-19 patients: A retrospective and observational study. Aging 2020, 12, 11245–11258. [Google Scholar] [CrossRef] [PubMed]
  42. Kernan, K.F.; Carcillo, J.A. Hyperferritinemia and inflammation. Int. Immunol. 2017, 29, 401–409. [Google Scholar] [CrossRef] [PubMed]
  43. Bowles, L.; Platton, S.; Yartey, N.; Dave, M.; Lee, K.; Hart, D.P.; Macdonald, V.; Green, L.; Sivapalaratnam, S.; Pasi, K.J.; et al. Lupus Anticoagulant and Abnormal Coagulation Tests in Patients with Covid-19. N. Engl. J. Med. 2020, 383, 288–290. [Google Scholar] [CrossRef] [PubMed]
  44. Shemin, D.; Briggs, D.; Greenan, M. Complications of therapeutic plasma exchange: A prospective study of 1727 procedures. J. Clin. Apher. 2007, 22, 270–276. [Google Scholar] [CrossRef] [PubMed]
  45. Basic-Jukic, N.; Kes, P.; Glavas-Boras, S.; Brunetta, B.; Bubic-Filipi, L.; Puretic, Z. Complications of Therapeutic Plasma Exchange: Experience with 4857 Treatments. Ther. Apher. Dial. 2005, 9, 391–395. [Google Scholar] [CrossRef]
  46. Gala-Błądzińska, A.; Mazur, K.; Dębiec, A.; Gargasz, K.; Bartosik-Psujek, H. Safety and tolerability of therapeutic plasma exchange in autoimmune neurological diseases—A retrospective single-centre analysis. Neurol. Neurochir. Polska 2020, 54, 344–349. [Google Scholar] [CrossRef]
  47. Tabibi, S.; Tabibi, T.; Conic, R.R.Z.; Banisaeed, N.; Streiff, M.B. Therapeutic Plasma Exchange: A potential Management Strategy for Critically Ill COVID-19 Patients. J. Intensiv. Care Med. 2020, 35, 827–835. [Google Scholar] [CrossRef]
  48. Thölking, G.; Mesters, R.; Dittrich, R.; Pavenstädt, H.; Kümpers, P.; Reuter, S. Assessment of Hemostasis after Plasma Exchange Using Rotational Thrombelastometry (ROTEM). PLoS ONE 2015, 10, e0130402. [Google Scholar] [CrossRef] [Green Version]
  49. Singhania, N.; Bansal, S.; Nimmatoori, D.P.; Ejaz, A.A.; McCullough, P.A.; Singhania, G. Current Overview on Hypercoagulability in COVID-19. Am. J. Cardiovasc. Drugs 2020, 20, 393–403. [Google Scholar] [CrossRef]
  50. Spiezia, L.; Boscolo, A.; Poletto, F.; Cerruti, L.; Tiberio, I.; Campello, E.; Navalesi, P.; Simioni, P. COVID-19-Related Severe Hypercoagulability in Patients Admitted to Intensive Care Unit for Acute Respiratory Failure. Thromb. Haemost. 2020, 120, 998–1000. [Google Scholar] [CrossRef]
  51. Simonovich, V.A.; Pratx, L.D.B.; Scibona, P.; Beruto, M.V.; Vallone, M.G.; Vázquez, C.; Savoy, N.; Giunta, D.H.; Pérez, L.G.; PlasmAr Study Group; et al. A Randomized Trial of Convalescent Plasma in Covid-19 Severe Pneumonia. N. Engl. J. Med. 2021, 384, 619–629. [Google Scholar] [CrossRef] [PubMed]
  52. Pluta, M.; Dziech, M.; Jaworski, T.; Krzych, Ł. Is this TRALI, TACO, or just pneumonia?—A case report of acute respiratory failure. Anestezjol. Intensywna Ter. 2019, 51, 414–416. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structured search strategy.
Figure 1. Structured search strategy.
Viruses 13 01484 g001
Table 1. The PICO criteria used in the study.
Table 1. The PICO criteria used in the study.
ParticipantsPatients with severe course of COVID-19.
InterventionsTherapeutic plasma exchange (any type) as an adjunctive treatment.
ControlDue to limited number of studies and their methodological type, a control group was not required to include in the study.
OutcomesMortality and changes in various biomarkers, along with which additional attention was given to safety issues in retrieved papers.
Table 2. Summary of the included studies.
Table 2. Summary of the included studies.
AuthorStudy TypePopulationInterventionMedian Time from First Symptoms to TPE InitiationReplacement FluidAdverse Effects of TPEOutcome
Zhang et al. [21]Case-series3 severely ill patients1 TPE session15 daysFFPN/AMortality (day 14): 0%
Morath et al. [22]Case-series5 patients with COVID-19-induced multi-organ failure and ARDSAll patients received 1–2 TPE sessions 12 daysFFPN/AMortality: 20%
Faqihi et al. [23]Case-series10 patients with ARDS, APACHE II score >20, septic shock or cytokine release syndromeAll patients received 5–7 TPE6.5 days5% albumin or FFPNoneMortality (day 28): 10%
Gucyemetz et al. [17]Case-control73 patients with COVID-19-related pneumonia18 patients received 3 TPE sessionsN/AN/AN/AMortality (non-TPE vs. TPE): 58.3% vs. 8.3% *
Khamis et al. [35]Case-control31 critically ill patients with COVID-19-related ARDS, severe pneumonia, septic shock or multiple organ dysfunction syndrome11 patients underwent 5 TPE sessionsN/AFFPOne hypotension episode treated with fluid bolus and hydrocortisoneMortality (non-TPE vs. TPE, day 28): 35% vs. 0% *
Jaiswal et al. [24]Case-series14 patients with severe COVID-19 infection according to WHO classificationAll patients received 1 TPE session9 daysConvalescent Plasma3 cases of hypotension treated with fluid bolusMortality (day 28): 28.6%
Gluck et al. [25]Case-series10 patients with COVID-19 and Penn class 3 and 4 cytokine release syndromeAll patients received 5 TPE sessionsN/A5% albumin or FFPNoneMortality (day 14): 0%
Karman et al. [36]PSM90 patients with severe COVID-19 infection and cytokine release syndrome45 patients received one TPE until resolution of the diseaseN/AFFP and normal saline in 2:1 ratio1 femoral artery puncture and thrombophlebitis treated accordinglyMortality (non-TPE vs. TPE, day 28): 38.5% vs. 8.9% *
Fernandez et al. [26]Case-series4 critically ill patients with COVID-192–6 plasma exchange sessions 20 days5% albumin + FFP1 episode of hypotension and tachycardiaMortality (day 28): 0%
Dogan et al. [27]Case-series6 patients with COVID-19–related autoimmune meningoencephalitis1–9 plasma exchange sessionsN/A5% albuminN/AMortality (day 14): 16.7%
Adeli et al. [28]Case-series8 patients3–5 plasma exchange sessionsN/AFFP + albumin solution + calcium gluconateNoneMortality (no specified day): 12.5%
De Prost et al. [29]Case-series4 critically-ill patients with high blood concentrations of neutralizing autoantibodies against type I interferons3–4 plasma exchange sessions18 days5% albumin solutionNoneMortality (no specified day): 50%
Faqihi et al. [37]RCT87 intubated patients with either ARDS, APACHE II score >20 pts, septic shock or cytokine release syndrome43 patients received 1–5 (median 3) plasma exchange sessions8 daysFFPNoneMortality (TPE vs non-TPE, day 35):
20.9% vs. 34.1 % (p = 0.09)
Hashemian et al. [30]Case-series15 patients1–3 TPE sessionsN/A5% albumin solution + 0.9% NaCl/convalescent plasmaN/AMortality (no specified day): 40%
Keith et al. [31]Case-series8 patients1–7 plasma exchange sessionsN/AFFPN/AMortality (no specified day): 25%
Matsushita et al. [32]Case-series5 patients with PaO2/FiO2 ratio of less than 200 mmHg and/or labored respiration and/or tracheal intubation3–7 plasma exchange sessions14 daysFFPN/AMortality (no specified day): 60%
Roshandel et al. [33]Case-series5 COVID-19 patients with respiratory failure2 standard plasma exchange sessions39 daysFFP + 5% albumin,
then 0.9% NaCl/convalescent plasma
N/AMortality (no specified day): 20%
Truong et al. [34]Case-series6 critically ill patients with plasma hyperviscosity2–3 plasma exchange sessionsN/AFFPNoneMortality (no specified day): 50%
Results presented in the “outcome” column are median. * results that were statistically significant. Absence of “*” means that the result was either not significant or the significance was not calculated; PaO2: partial pressure of oxygen; FiO2: fraction of inspired oxygen; ARDS: Acute Respiratory Distress Syndrome; SOFA—Sequential Organ Failure Assessment; APACHE II—Acute Physiology and Chronic Health Evaluation II; TPE—therapeutic plasma exchange; RCT–randomized controlled trial; PSM: propensity score matching; N/A: information not available.
Table 3. Median and interquartile ranges of values of various biomarkers measured before and after TPE.
Table 3. Median and interquartile ranges of values of various biomarkers measured before and after TPE.
ParameterValues: Median (IQR)
pre-TPE PaO2/FiO2 (mmHg)132 (112.5–153.5) [21,23,24,25,29,30,35,37]
post-TPE PaO2/FiO2 (mmHg)224 (216.5–300) [21,23,24,29,30,35,37]
pre-TPE CRP (mg/L)132 (79–168.5) [17,21,22,23,24,25,26,27,29,30,31,33,34,35,36,37]
post-TPE CRP (mg/L)28.5 (11.1–47.5) [17,21,22,23,24,25,30,31,33,34,35,37]
pre-TPE Lymphocytes (109/L)0.7 (0.58–1.0) [17,21,23,24,25,26,29,35,37]
post-TPE Lymphocytes (109/L)1.04 (1.0–1.5) [17,21,23,24,35,37]
pre-TPE IL-6 (pg/mL)118.7 (25.2–295.3) [17,21,22,23,25,26,27,32,35,36,37]
post-TPE IL-6 (pg/mL)18.5 (5.7–35) [17,21,22,23,26,30,33,35]
pre-TPE LDH (U/L)576.5 (492.5–849.5) [17,21,22,23,26,27,33,36,37]
post-TPE LDH (U/L)245.5 (236–440) [17,22,23,26,33,37]
pre-TPE D-Dimers (mg/L)6.05 (4.5–7.6) [17,22,23,24,25,26,27,28,29,31,34,35,36,37]
post-TPE D-Dimers (mg/L)2.6 (1.3–4.0) [17,22,23,24,26,31,33,34,35,37]
pre-TPE Ferritin (ug/L)1332 (1125–1444) [17,22,23,24,26,27,30,31,35,36,37]
post-TPE Ferritin (ug/L)494 (352–842) [17,22,23,24,26,30,31,35,37]
The values presented on the table are not weighted. The values included in the table are provided in the Table S1.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Krzych, Ł.J.; Putowski, Z.; Czok, M.; Hofman, M. What Is the Role of Therapeutic Plasma Exchange as an Adjunctive Treatment in Severe COVID-19: A Systematic Review. Viruses 2021, 13, 1484. https://doi.org/10.3390/v13081484

AMA Style

Krzych ŁJ, Putowski Z, Czok M, Hofman M. What Is the Role of Therapeutic Plasma Exchange as an Adjunctive Treatment in Severe COVID-19: A Systematic Review. Viruses. 2021; 13(8):1484. https://doi.org/10.3390/v13081484

Chicago/Turabian Style

Krzych, Łukasz J., Zbigniew Putowski, Marcelina Czok, and Mariusz Hofman. 2021. "What Is the Role of Therapeutic Plasma Exchange as an Adjunctive Treatment in Severe COVID-19: A Systematic Review" Viruses 13, no. 8: 1484. https://doi.org/10.3390/v13081484

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