Next Article in Journal
Association between Sagittal Cervical Spinal Alignment and Degenerative Cervical Spondylosis: A Retrospective Study Using a New Scoring System
Next Article in Special Issue
CRP Serum Levels Are Associated with High Cardiometabolic Risk and Clinical Disease Activity in Systemic Lupus Erythematosus Patients
Previous Article in Journal
Remote Ischaemic Preconditioning in Intra-Abdominal Cancer Surgery (RIPCa): A Pilot Randomised Controlled Trial
Previous Article in Special Issue
Cardiac Glycosides Lower C-Reactive Protein Plasma Levels in Patients with Decompensated Heart Failure: Results from the Single-Center C-Reactive Protein-Digoxin Observational Study (C-DOS)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting C-Reactive Protein by Selective Apheresis in Humans: Pros and Cons

1
Cardiovascular Center Oberallgaeu-Kempten, Clinic Association Allgaeu, 87439 Kempten, Germany
2
Pentracor GmbH, 16761 Hennigsdorf, Germany
3
Medical Clinic, Diakonissenhospital Flensburg, 24939 Flensburg, Germany
4
Medical Care Center Kempten-Allgaeu, 87439 Kempten, Germany
5
Division of Gastroenterology, Infectiology and Rheumatology, Medical Department, Charité University Medicine, 12200 Berlin, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(7), 1771; https://doi.org/10.3390/jcm11071771
Submission received: 17 February 2022 / Revised: 13 March 2022 / Accepted: 17 March 2022 / Published: 23 March 2022
(This article belongs to the Special Issue C-Reactive Protein and Cardiovascular Disease: Clinical Aspects)

Abstract

:
C-reactive protein (CRP), the prototype human acute phase protein, may be causally involved in various human diseases. As CRP has appeared much earlier in evolution than antibodies and nonetheless partly utilizes the same biological structures, it is likely that CRP has been the first antibody-like molecule in the evolution of the immune system. Like antibodies, CRP may cause autoimmune reactions in a variety of human pathologies. Consequently, therapeutic targeting of CRP may be of utmost interest in human medicine. Over the past two decades, however, pharmacological targeting of CRP has turned out to be extremely difficult. Currently, the easiest, most effective and clinically safest method to target CRP in humans may be the specific extracorporeal removal of CRP by selective apheresis. The latter has recently shown promising therapeutic effects, especially in acute myocardial infarction and COVID-19 pneumonia. This review summarizes the pros and cons of applying this novel technology to patients suffering from various diseases, with a focus on its use in cardiovascular medicine.

1. Introduction

In humans, C-reactive protein (CRP) activates the classical complement pathway via C1q [1] and stimulates macrophages via Fcγ-receptors [2,3]. Obviously, CRP utilizes the same biological structures as antibodies [4]. In further analogy, CRP may be causally involved in various human diseases by triggering (severe) ancient autoimmune reactions [5,6,7]. Although the latter hypothesis has been discussed in medical science since decades the issue has never been clarified. This is due to the fact that no drug or medical product targeting CRP has been on the market so far.
CRP synthesis and structure have extensively been reviewed elsewhere [8,9]. Here, we briefly review the role of CRP in physiology and pathophysiology with a focus on complement and macrophages. We then deal with the recent breakthrough in CRP targeting achieved by selective CRP apheresis. Pros and cons are listed in Table 1. Finally, we give an overview on current clinical trials and hypothesize on future developments. Thus, this review article may also be considered as an opinion paper.

2. CRP in Physiology

CRP is expressed in the ancient Limulus for more than 250 million years ago [10]. Although evolutionarily highly conserved, there are significant species differences in CRP function [11]. In humans, CRP activates the classical complement pathway and opsonizes biological particles for macrophages via Fcγ-receptors [1,2,3,12]. The latter seems remarkable as these functions are also antibody functions: Thus, like CRP, antibodies activate the classical complement cascade and bind to Fcγ-receptors via their Fc-region [4]. CRP has appeared earlier in evolution than antibodies and may, consequently, be the first antibody analogue in the evolution of the immune system [13]. In ancient Limulus, which survives without the benefits of adaptive immunity, CRP is vital for host defense against bacterial infection [10]. In humans, however, having developed highly sophisticated adaptive immunity, CRP may rather be a relic of evolution and emerged to have a role in tissue regeneration [6]. When apoptotic or dying cells display lysophosphatidylcholine (oxidized phosphatidylcholine) in their membranes, CRP recognizes these cells and opsonizes them for Fcγ-receptor mediated removal by macrophages [14]. Thus, in human physiology, CRP may, above all, play a major role in the process of wound healing and removal of apoptotic and necrotic cells.
CRP is probably the most commonly measured inflammatory molecule in clinical medicine. Again, as CRP activates complement via C1q and stimulates macrophages via Fcγ-receptors (in analogy to antibodies) CRP may be considered as an early primitive antibody and a pathogenic factor rather than an inflammation marker only.

3. CRP in Pathophysiology

In pathophysiology, an active contribution of CRP to initiation and progression of disease has been discussed for decades [13,15,16]. This discussion has never come to an end because a definitive proof of CRP’s causal involvement in disease in humans was lacking. Whereas the molecule’s role as a marker of activity of infectious, autoimmune, ischemic or even cardiovascular disease [17] is well established and generally accepted, it is important to note that there is no international consensus on causal contribution of CRP to the pathogenesis of any human disease. In cardiovascular disease, mendelian randomization trials strongly contradict causality and active contribution of CRP to pathogenesis [18,19,20]. This is crucial. It is, however, also crucial to realize that Mendelian randomization has to be interpreted with care and is, by far, less reliable than randomization in clinical trials. The latter has been reviewed in detail in a number of noteworthy articles [21,22,23,24]. Regulation of CRP synthesis includes not only one but a number of genes [25,26,27] and thus, a one gene/one protein genetic approach might be problematic. In addition, the issue of canalization could be relevant in a protein that is as highly conserved as CRP. Finally, cardiovascular disease is complex and each disease entity deserves detailed analysis [13]. In particular, the pathophysiological role of CRP in acute events must be considered separately from that in chronic events. The evidence in the acute setting is overwhelming (please see Section 3.3.2), whereas the evidence in the chronic inflammatory setting is still being collected. Ultimately, randomized trials might clarify these issues much better than Mendelian randomization [28]. Randomized controlled trials, however, are only possible with an available specific and efficient therapy comparing treatment group to control group. Such therapy has only very recently become reality.

3.1. CRP in Viral and Bacterial Infection

CRP is one of the most frequently determined molecules in clinical medicine. In daily practice, it is used for the non-specific initial diagnosis of viral or bacterial infection and also for monitoring the course of such infection under medical therapy [29]. Especially, the success of antibiotic therapy in bacterial infection and sepsis is usually determined by measuring CRP levels in addition to clinical evaluation. Importantly, CRP plasma levels in viral infection are usually significantly lower than CRP levels in bacterial infection [30]. This is of particular importance when looking at COVID-19 disease (please see Section 3.5). Although COVID-19 patients suffer from a viral disease, CRP levels in COVID-19 patients with a bad prognosis are surprisingly high. Plasma levels up to 400 mg/L, usually seen in severe bacterial infection or sepsis only, are common in deleterious COVID pneumonia without superinfection [31].

3.2. CRP in Autoimmune Disease

Although the association between CRP and the activity of autoimmune disease is well known and highly suggestive for a causal involvement in this heterogeneous group of diseases (like rheumatoid arthritis, ulcerative colitis, Crohn’s disease, psoriasis, giant cell arteritis etc.) [30], there are no studies on specific CRP inhibition in autoimmune disease. Notably, upstream interleukin-6 (IL-6) targeting with tocilizumab seems partly effective [32].

3.3. CRP in Cardiovascular Disease

3.3.1. Atherosclerosis

CRP plasma levels correlate with cardiovascular risk [17]. CRP accumulates in human atherosclerotic lesions [33] and exerts pro-atherogenic effects in vitro [13]. Some of the effects reported in the literature, especially on endothelial cells and vascular smooth muscle cells, however, have been shown to be caused by contamination of the used CRP preparations by either azide or lipopolysaccharide [34,35]. The latter seems reasonable as CRP interaction with ancient immune cells, i.e., macrophages, is visible in tissue specimen and seems more relevant than interaction with cells not contributing to the immune response. Mendelian trials, in awareness of their limitations, contradict the significant causal contribution of CRP to atherogenesis and its sequelae [18,19,20]. In contrast, recent large clinical trials suggest that the IL-1β/IL-6/CRP pathway is intimately involved in cardiovascular disease [36,37,38]. Specific and direct targeting of the CRP molecule, however, has never been tried yet. The only reason for this is the fact that, in spite of huge pharmacological effort, no CRP specific chemical inhibitor or antagonist has been available.

3.3.2. CRP in Myocardial Infarction

Acute myocardial infarction (AMI) implies a huge burden for the health system, since patients who recover still suffer from reduced quality of life and a high risk of severe complications later on. The risk correlates significantly with the extent of myocardial injury [39]. Especially innate immunity aggravates and extends myocardial injury [40,41]. Serum CRP concentrations during and after AMI correlate with clinical outcome and with larger infarct size [42,43,44,45]. This has been described for more than two decades now and is in line with the known pathological function of CRP: eliminating cells in the area at risk [6,46,47]. This area contains cells, which could recover after revascularization and reperfusion, but are sequentially destroyed by immune-mediated mechanisms. Numerous experimental approaches focusing specifically on AMI have shown this in detail [48,49,50,51,52,53]. Recently, it has been shown in the C-reactive protein apheresis in Acute Myocardial Infarction-1 (CAMI-1) study that the magnitude of infarct damage or reduction in cardiac output is significantly related to the amount of CRP synthesized by the patient immediately after the onset of his AMI. In the same study, significant evidence was also found that reduction in CRP levels conferred a better outcome in terms of infarct size and cardiac output. Some patients in the verum group (CRP apheresis group) even showed no scar at all (as assessed by cardiovascular magnetic resonance). These were not aborted infarctions, because aborted infarctions were treated as dropouts [5].

3.3.3. CRP in Myocarditis and Dilated Cardiomyopathy

In myocarditis, most frequently caused by viral infection [54], elevated CRP levels are common. Autoimmune myocarditis is also associated with high CRP plasma levels. Chronic myocarditis is known to trigger the development of dilated cardiomyopathy [54], a disease leading to ongoing heart failure not only in elder but also in younger patients. It is noteworthy that CRP and complement deposits have been shown to be frequently present in myocardial biopsy specimen obtained from patients suffering from dilated cardiomyopathy [55].

3.4. CRP in Neurological Disorders and Stroke

Ischemic stroke exhibits similar pathological mechanisms to AMI. To date, restoring rapid reperfusion of the brain constitutes the only established therapeutic strategy to reduce the size of the infarct and the consequences of the disease [56]. However, inflammation plays an important role in various stages of ischemic stroke. Several humoral and cellular mechanisms are set in motion by the occlusion and subsequent therapeutic reperfusion [57]. Several findings substantiate the hypothesis that CRP plays an identical pathological role as shown in AMI, facilitating the elimination of energetically challenged and compromised cells in the penumbra.
The early inflammatory response after stroke has been identified as a key prognostic factor [58,59]. Patients with favorable clinical outcome feature significantly lower levels of inflammatory parameters, especially CRP, compared to patients with poor outcome [60,61,62]. Muir et al. have shown that CRP levels measured within 72 h after stroke predict mortality over an observation period of up to 4 years [63]. Further, studies in a rat animal model have shown that infusion of human CRP enlarges cerebral infarct areas after acute occlusion via a complement-dependent mechanism [64].

3.5. CRP in COVID-19

COVID-19 is a virus-induced disease, but it also includes an important immune and autoimmune component. CRP was used early on during the pandemic as a marker for the severity and progression of the disease in patients, as it increases dramatically together with IL-6 during the clinical manifestation of COVID-19 [65,66,67,68]. The validity of CRP as a significant predictor of the outcome in COVID-19 was confirmed many times. A rapid increase in CRP allows the prognosis of ventilatory requirement of patients as well as their clinical outcome [31]. CRP levels further correlate with computed tomography (CT) findings in COVID-19 patients [69].
Corresponding to these findings, abundant amounts of CRP and complement deposits and were found in the lungs of deceased COVID-19 patients, including mainly C1q [70,71].
Severe progression occurs in roughly 14% of patients suffering from COVID-19 and in 5% this can lead to ventilator dependency with a serious prognosis [68,72,73,74]. An important therapeutic approach focuses on the treatment of acute respiratory failure—a major cause of mortality, followed by cardiac and septic complications. In the severe course of the disease, there is an initial cytokine storm, accompanied by a massive increase in the CRP concentration, followed by pulmonary fibrosis [75,76].
Intra-alveolar edema and hemorrhage are common observations in the lungs of COVID-19 patients, resulting in ischemic alveolar tissue. In COVID-19 pneumonia, there is massive damage to the alveoli as well as thrombus formation in the microcirculation. Complement binding to CRP leads to immigration of macrophages and, via increased expression of tissue factor, to thrombus formation. Both are exacerbated by high CRP levels and parallel to the underlying pathomechanism in other diseases, CRP causally enlarges destroyed tissue and contributes to irreversible tissue destruction [77].
These findings support the hypothesis stated early on that targeting CRP therapeutically can inhibit the lung deterioration and disease progression [6,7,78,79]. This innovative therapeutic approach for the early phase of severe COVID-19 is currently being used in three German hospitals. Three of the treated cases and one case series have already been published [7,80,81,82] and another publication on a case series has been submitted and can be viewed as a preprint (https://www.preprints.org/manuscript/202203.0029/v1; 16 March 2022). In the case series by Esposito et al., there is a marked improvement in COVID-19 pneumonia on imaging performed before and after CRP apheresis.

4. Why May CRP Apheresis Be a Breakthrough in CRP Targeting?

Over more than two decades, several approaches to target CRP have been discussed and tried by various researchers and pharmaceutical companies. These approaches include:
  • CRP inhibition by antisense technologies [83];
  • CRP inhibition by small molecular weight inhibitors [84];
  • Inhibition of hepatic CRP synthesis [85];
  • Inhibition of CRP mediated complement activation;
  • Inhibition of CRP binding to its receptors.
Each of these approaches was well justified and was also partly promising. Interestingly, however, none of them resulted in a specific substance or medication that was applicable in human clinical practice. The latter seems remarkable because all the people and companies involved were well experienced in drug development. Several reasons were causal for the problems in generating specific CRP inhibitors: Antisense technology was, up to present day, not sufficiently effective. Small molecular weight inhibitors have not yet been transferred to human application. For inhibition of hepatic CRP synthesis, no specific novel substance was identified. Blockage of CRP-mediated complement activation and its C1q binding site turned out to be difficult for steric reasons. Inhibition of CRP binding to its receptors was also impossible because CRP receptors are also antibody receptors and thus, this approach may result in severe immunosuppression. Generally spoken, CRP—in its pentameric structure—is a molecule with structural redundancies difficult to interfere with. Its synthesis is complexly regulated involving different gene loci and furthermore, its highly dynamic regulation involving an up to 10,000-fold increase in plasma levels within few hours during acute phase response counteract an efficient synthesis inhibition or targeting.
The most important clinical concern about CRP targeting is the danger of immunosuppression with consecutive bacterial or viral infection and sepsis. Interestingly, in spite of significant reduction of CRP plasma levels via CRP apheresis in both disease entities, we have not observed such effects in our patients suffering from acute myocardial infarction or COVID-19 disease [5,7,80,81,82]. Thus, either CRP is not crucial in immune defense against microbial pathogens or the remaining CRP plasma levels after CRP apheresis are still sufficient. CRP apheresis is highly specific and does not relevantly influence other inflammatory markers or medication [86]. In the clinical setting, the most relevant apheresis procedure is lipid apheresis which mainly targets lipids and is far less specific [87,88].
Very often in clinical medicine simple approaches have turned out to be the best ones. Consequently, the idea to selectively remove CRP from the human plasma by specific and highly efficient extracorporeal apheresis [52] lacking severe side effects may finally turn out to be superior to other approaches. CRP apheresis may become beneficial in clinical medicine. This potential benefit, however, needs to be proven and fostered by an additional clinical trial program. The latter is currently ongoing.

5. Clinical Trials

“First in man”-application of CRP apheresis has been published in 2018 for a ST elevation myocardial infarction (STEMI) patient [89]. In this patient, post STEMI CRP plasma levels were lowered effectively and the patient experienced no side effects from CRP apheresis. The same was published for a small cohort of STEMI patients in 2019 [90]. The first and also the only clinical study on CRP apheresis in STEMI patients published up to the present day is the C-reactive protein apheresis in Acute Myocardial Infarction-1 (CAMI-1) study [5]. CAMI-1 was a non-randomized multi-center pilot study which has investigated feasibility and safety of CRP apheresis. Although the clinical observations were promising and the significant correlation between post-infarction CRP amount and myocardial infarct size was significantly lost in the treatment group, CAMI-1 cannot be regarded as being conclusive because it was not randomized.
Clinical trials including apheresis present the issue of an adequate sham control and a double-blind design. Although a sham control is biostatistically speaking crucial in order to get valid results that are not biased by the placebo effect, including this in the apheresis procedure is ethically challenging. Few apheresis trials included adequate sham controls, one of the best examples being granulocyte/monocyte apheresis in chronic gut diseases [91,92]. Here, control patients were subjected to the same extracorporeal circuit, but blood was bypassed and did not pass the column. As patients and clinicians were blinded and only the conducting apheresis team knew which patients received the sham procedure these trials were considered double-blind. This could be a feasible design for future CRP apheresis trials. However, the underlying disease has to be taken into consideration. After STEMI, ischemic stroke and during COVID-19, patients are hospitalized and already in critical state. Submitting them to a 4–6 h extracorporeal circuit and sham procedure is ethically not justifiable. Hence, most apheresis trials do not include a sham control and have either historical controls or patients that receive standard therapy without apheresis [93].
Current clinical trials investigating the effect of CRP apheresis on the course of various human diseases are summarized on the Website of U.S. National Library of Medicine/ClinicalTrials.gov (https://www.clinicaltrials.gov/ct2/results?term=C-reactive+protein+apheresis, accessed on 16 March 2022).
Two of the studies attract special attention because, for their randomized, controlled, multi-center design, they can be considered as proof of principle trials. One of them is the “CRP Apheresis in STEMI”-trial (NCT04939805), initiated by University of Innsbruck, Austria, a randomized, multi-center interventional trial including 170 patients and comparing standard therapy of STEMI plus CRP apheresis to standard therapy alone. The study largely follows the CAMI-1 protocol, it is well-planned and well-organized. Whether it is adequately powered to finally detect statistically significant differences in a disease with heterogeneous underlying anatomy and pathology is a matter of concern. In this context, the CAMI-1 registry, a multi-center all-comer CRP apheresis in AMI registry may help to identify patient subgroups that profit best and may also help to plan another randomized trial with modified inclusion criteria.
The second study attracting special attention is the “CRP Apheresis for Attenuation of Pulmonary, Myocardial and/or Kidney Injury in COVID-19”-trial (NCT04898062), a randomized, controlled, multi-center interventional trial initiated by the University of Essen, Germany, including 50 patients and comparing standard therapy of COVID-19 plus CRP apheresis to standard therapy alone. This study is of considerable importance. It is based on the pathophysiological hypothesis that CRP, in COVID-19, triggers a fulminant innate immunity autoimmune reaction in the human body which may be the real cause for the deleterious course in subjects with severe COVID-19 (Please see Section 3.5). Like CAMI-1, it is based on published case reports and case series strongly suggesting that only few participants may power such a randomized study adequately in order to prove a therapeutic benefit of CRP apheresis in severe COVID-19 disease [7,80,82]. If so, this small trial might become a benchmark trial in demonstrating conclusively that CRP is a trigger of ischemia induced autoimmune responses in the human body.
Whether CRP apheresis is useful in atherosclerosis, myocarditis and dilated cardiomyopathy, neurological disorders and stroke, or even in autoimmune disease, requires further systematic investigation. As CRP apheresis is not yet a broadly established therapy, we propose to treat patients within a reputable scientific framework only, i.e., within a scientific registry or randomized, controlled trial.

6. Discussion and Future Developments

CRP has been the first antibody-like molecule in the evolution of the immune system. Surprisingly, although it appeared earlier in evolution than nowadays antibodies, CRP utilizes the same biological structures (C1q, FcγRs) and, by doing this, has similar functions as modern antibodies (activation of classical complement cascade, opsonization of biological particles for macrophages). Notably, like antibodies, CRP may cause autoimmune reactions in the human body. First, clinical evidence for this comes from a clinical pilot study on using C-reactive protein apheresis as an add on-treatment of myocardial infarction (CAMI-1) and from case reports on successful use of C-reactive protein apheresis in COVID-19 disease.
A definitive proof of principle, however, is still lacking, With the initiation of “CRP Apheresis in STEMI” and “CRP Apheresis for Attenuation of Pulmonary, Myocardial and/or Kidney Injury in COVID-19”, two randomized multicenter trials on C-reactive protein apheresis in STEMI on the one hand and COVID-19 on the other hand, a big step forward is to be expected soon after completion. These randomized trials are flanked by a number of clinical registries that may help to identify patient subgroups that strongly benefit from CRP apheresis. Finally, patients suffering from other acute and chronic diseases, in which CRP levels inversely correlate with prognosis (f. e. stroke, ulcerative colitis, Crohn’s disease, pancreatitis, chronic polyarthritis, atherosclerosis etc.) might benefit from CRP apheresis in the future.

Author Contributions

J.T., P.B. and A.S. wrote the first draft, conceptualized and finalized the manuscript. C.D.G., W.R., S.K. and F.H. made substantial corrections. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was applied to writing this review.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

Ahmed Sheriff is Founder and Shareholder of Pentracor GmbH. Patrizia Brunner and Stefan Kayser are employees of Pentracor GmbH.

References

  1. Kaplan, M.H.; Volanakis, J.E. Interaction of C-reactive protein complexes with the complement system. I. Consumption of human complement associated with the reaction of C-reactive protein with pneumococcal C-polysaccharide and with the choline phosphatides, lecithin and sphingomyelin. J. Immunol. 1974, 112, 2135–2147. [Google Scholar] [PubMed]
  2. Bharadwaj, D.; Stein, M.P.; Volzer, M.; Mold, C.; Du Clos, T.W. The major receptor for C-reactive protein on leukocytes is fcgamma receptor II. J. Exp. Med. 1999, 190, 585–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Manolov, D.E.; Röcker, C.; Hombach, V.; Nienhaus, G.U.; Torzewski, J. Ultrasensitive Confocal Fluorescence Microscopy of C-Reactive Protein Interacting With FcγRIIa. Arter. Thromb. Vasc. Biol. 2004, 24, 2372–2377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Daëron, M. Fc Receptor Biology. Annu. Rev. Immunol. 1997, 15, 203–234. [Google Scholar] [CrossRef]
  5. Ries, W.; Torzewski, J.; Heigl, F.; Pfluecke, C.; Kelle, S.; Darius, H.; Ince, H.; Mitzner, S.; Nordbeck, P.; Butter, C.; et al. C-Reactive Protein Apheresis as Anti-inflammatory Therapy in Acute Myocardial Infarction: Results of the CAMI-1 Study. Front. Cardiovasc. Med. 2021, 8, 591714. [Google Scholar] [CrossRef]
  6. Sheriff, A.; Kayser, S.; Brunner, P.; Vogt, B. C-Reactive Protein Triggers Cell Death in Ischemic Cells. Front. Immunol. 2021, 12, 630430. [Google Scholar] [CrossRef]
  7. Torzewski, J.; Heigl, F.; Zimmermann, O.; Wagner, F.; Schumann, C.; Hettich, R.; Bock, C.; Kayser, S.; Sheriff, A. First-in-Man: Case Report of Selective C-Reactive Protein Apheresis in a Patient with SARS-CoV-2 Infection. Am. J. Case Rep. 2020, 21, e925020. [Google Scholar] [CrossRef]
  8. Pepys, M.B.; Hirschfield, G. C-reactive protein: A critical update. J. Clin. Investig. 2003, 111, 1805–1812. [Google Scholar] [CrossRef]
  9. Szalai, A.J.; Agrawal, A.; Greenhough, T.J.; Volanakis, J.E. C-reactive protein: Structural biology, gene expression, and host defense function. Immunol. Res. 1997, 16, 127–136. [Google Scholar] [CrossRef]
  10. Nguyen, N.Y.; Suzuki, A.; Cheng, S.M.; Zon, G.; Liu, T.Y. Isolation and characterization of Limulus C-reactive protein genes. J. Biol. Chem. 1986, 261, 10450–10455. [Google Scholar] [CrossRef]
  11. Torzewski, M.; Waqar, A.B.; Fan, J. Animal Models of C-Reactive Protein. Mediat. Inflamm. 2014, 2014, 683598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Mortensen, R.F.; Osmand, A.P.; Lint, T.F.; Gewurz, H. Interaction of C-reactive protein with lymphocytes and monocytes: Complement-dependent adherence and phagocytosis. J. Immunol. 1976, 117, 774–781. [Google Scholar] [PubMed]
  13. Zimmermann, O.; Li, K.; Zaczkiewicz, M.; Graf, M.; Liu, Z.; Torzewski, J. C-Reactive Protein in Human Atherogenesis: Facts and Fiction. Mediat. Inflamm. 2014, 2014, 561428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sproston, N.R.; Ashworth, J.J. Role of C-Reactive Protein at Sites of Inflammation and Infection. Front. Immunol. 2018, 9, 754. [Google Scholar] [CrossRef]
  15. Pepys, M.B. C-reactive protein is neither a marker nor a mediator of atherosclerosis. Nat. Clin. Pract. Nephrol. 2008, 4, 234–235. [Google Scholar] [CrossRef]
  16. Schunkert, H.; Samani, N.J. Elevated C-Reactive Protein in Atherosclerosis—Chicken or Egg? N. Engl. J. Med. 2008, 359, 1953–1955. [Google Scholar] [CrossRef] [Green Version]
  17. Pearson, T.A.; Mensah, G.A.; Alexander, R.W.; Anderson, J.L.; Cannon, R.O., III; Criqui, M.; Fadl, Y.Y.; Fortmann, S.P.; Hong, Y.; Myers, G.L.; et al. Markers of Inflammation and Cardiovascular Disease: Application to Clinical and Public Health Practice: A Statement for Healthcare Professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation 2003, 107, 499–511. [Google Scholar] [CrossRef]
  18. Wensley, F.; Gao, P.; Burgess, S.; Kaptoge, S.; di Angelantonio, E.; Shah, T.; Engert, J.C.; Clarke, R.; Davey-Smith, G.; Nordestgaard, B.G.; et al. Association between C reactive protein and coronary heart disease: Mendelian randomisation analysis based on individual participant data. BMJ 2011, 342, d548. [Google Scholar] [CrossRef] [Green Version]
  19. Elliott, P.; Chambers, J.C.; Zhang, W.; Clarke, R.; Hopewell, J.C.; Peden, J.F.; Erdmann, J.; Braund, P.; Engert, J.C.; Bennett, D.; et al. Genetic Loci Associated With C-Reactive Protein Levels and Risk of Coronary Heart Disease. JAMA 2009, 302, 37–48. [Google Scholar] [CrossRef] [Green Version]
  20. Zacho, J.; Tybjaerg-Hansen, A.; Jensen, J.S.; Grande, P.; Sillesen, H.; Nordestgaard, B.G. Genetically Elevated C-Reactive Protein and Ischemic Vascular Disease. N. Engl. J. Med. 2008, 359, 1897–1908. [Google Scholar] [CrossRef]
  21. Glynn, R.J. Promises and Limitations of Mendelian Randomization for Evaluation of Biomarkers. Clin. Chem. 2010, 56, 388–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Morita, H.; Nagai, R. Genetically elevated C-reactive protein and vascular disease. N. Engl. J. Med. 2009, 360, 934, author reply 934–935. [Google Scholar] [PubMed]
  23. Smith, G.D.; Ebrahim, S. “Mendelian randomization”: Can genetic epidemiology contribute to understanding environmental determinants of disease? Int. J. Epidemiol. 2003, 32, 1–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Smith, G.D.; Ebrahim, S. Mendelian randomization: Prospects, potentials, and limitations. Int. J. Epidemiol. 2004, 33, 30–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Agrawal, A.; Cha-Molstad, H.; Samols, D.; Kushner, I. Transactivation of C-Reactive Protein by IL-6 Requires Synergistic Interaction of CCAAT/Enhancer Binding Protein β (C/EBPβ) and Rel p50. J. Immunol. 2001, 166, 2378–2384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Singh, P.P.; Voleti, B.; Agrawal, A. A Novel RBP-Jκ-Dependent Switch from C/EBPβ to C/EBPζ at the C/EBP Binding Site on the C-Reactive Protein Promoter. J. Immunol. 2007, 178, 7302–7309. [Google Scholar] [CrossRef] [Green Version]
  27. Young, D.P.; Kushner, I.; Samols, D. Binding of C/EBPβ to the C-Reactive Protein (CRP) Promoter in Hep3B Cells Is Associated with Transcription of CRP mRNA. J. Immunol. 2008, 181, 2420–2427. [Google Scholar] [CrossRef] [Green Version]
  28. Torzewski, J.; Fan, J.; Schunkert, H.; Szalai, A.J.; Torzewski, M. C-Reactive Protein and Arteriosclerosis. Mediat. Inflamm. 2014, 2014, 646817. [Google Scholar] [CrossRef]
  29. Fernandez-Carballo, B.L.; Escadafal, C.; MacLean, E.; Kapasi, A.J.; Dittrich, S. Distinguishing bacterial versus non-bacterial causes of febrile illness—A systematic review of host biomarkers. J. Infect. 2021, 82, 1–10. [Google Scholar] [CrossRef]
  30. Du Clos, T.W. C-reactive protein as a regulator of autoimmunity and inflammation. Arthritis Care Res. 2003, 48, 1475–1477. [Google Scholar] [CrossRef]
  31. Smilowitz, N.R.; Kunichoff, D.; Garshick, M.; Shah, B.; Pillinger, M.; Hochman, J.S.; Berger, J.S. C-reactive protein and clinical outcomes in patients with COVID-19. Eur. Heart J. 2021, 42, 2270–2279. [Google Scholar] [CrossRef] [PubMed]
  32. Serling-Boyd, N.; Wallace, Z.S. Management of primary vasculitides with biologic and novel small molecule medications. Curr. Opin. Rheumatol. 2020, 33, 8–14. [Google Scholar] [CrossRef] [PubMed]
  33. Torzewski, J.; Torzewski, M.; Bowyer, D.E.; Fröhlich, M.; Koenig, W.; Waltenberger, J.; Fitzsimmons, C.; Hombach, V. C-Reactive Protein Frequently Colocalizes With the Terminal Complement Complex in the Intima of Early Atherosclerotic Lesions of Human Coronary Arteries. Arter. Thromb. Vasc. Biol. 1998, 18, 1386–1392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Berg, C.W.V.D.; Taylor, K.E.; Lang, D. C-Reactive Protein-Induced In Vitro Vasorelaxation Is an Artefact Caused by the Presence of Sodium Azide in Commercial Preparations. Arter. Thromb. Vasc. Biol. 2004, 24, e168–e171. [Google Scholar] [CrossRef]
  35. Taylor, K.E.; Giddings, J.C.; Berg, C.W.V.D. C-Reactive Protein–Induced In Vitro Endothelial Cell Activation Is an Artefact Caused by Azide and Lipopolysaccharide. Arter. Thromb. Vasc. Biol. 2005, 25, 1225–1230. [Google Scholar] [CrossRef] [Green Version]
  36. Ridker, P.M.; Danielson, E.; Fonseca, F.A.; Genest, J.; Gotto, A.M., Jr.; Kastelein, J.J.; Koenig, W.; Libby, P.; Lorenzatti, A.J.; MacFadyen, J.G.; et al. Rosuvastatin to Prevent Vascular Events in Men and Women with Elevated C-Reactive Protein. N. Engl. J. Med. 2008, 359, 2195–2207. [Google Scholar] [CrossRef] [Green Version]
  37. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef]
  38. Tardif, J.-C.; Kouz, S.; Waters, D.D.; Bertrand, O.F.; Diaz, R.; Maggioni, A.P.; Pinto, F.J.; Ibrahim, R.; Gamra, H.; Kiwan, G.S.; et al. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N. Engl. J. Med. 2019, 381, 2497–2505. [Google Scholar] [CrossRef]
  39. De Waha, S.; Patel, M.R.; Granger, C.B.; Ohman, E.M.; Maehara, A.; Eitel, I.; Ben-Yehuda, O.; Jenkins, P.; Thiele, H.; Stone, G.W. Relationship between microvascular obstruction and adverse events following primary percutaneous coronary intervention for ST-segment elevation myocardial infarction: An individual patient data pooled analysis from seven randomized trials. Eur. Heart J. 2017, 38, 3502–3510. [Google Scholar] [CrossRef]
  40. Frangogiannis, N.; Smith, C.; Entman, M.L. The inflammatory response in myocardial infarction. Cardiovasc. Res. 2002, 53, 31–47. [Google Scholar] [CrossRef]
  41. Ong, S.-B.; Hernández-Reséndiz, S.; Crespo-Avilan, G.E.; Mukhametshina, R.T.; Kwek, X.-Y.; Cabrera-Fuentes, H.A.; Hausenloy, D.J. Inflammation following acute myocardial infarction: Multiple players, dynamic roles, and novel therapeutic opportunities. Pharmacol. Ther. 2018, 186, 73–87. [Google Scholar] [CrossRef] [PubMed]
  42. Beranek, J.T. C-reactive protein and complement in myocardial infarction and postinfarction heart failure. Eur. Heart J. 1997, 18, 1834–1835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Liu, D.; Qi, X.; Li, Q.; Jia, W.; Wei, L.; Huang, A.; Liu, K.; Li, Z. Increased complements and high-sensitivity C-reactive protein predict heart failure in acute myocardial infarction. Biomed. Rep. 2016, 5, 761–765. [Google Scholar] [CrossRef] [PubMed]
  44. Mani, P.; Puri, R.; Schwartz, G.G.; Nissen, S.E.; Shao, M.; Kastelein, J.J.P.; Menon, V.; Lincoff, A.M.; Nicholls, S. Association of Initial and Serial C-Reactive Protein Levels with Adverse Cardiovascular Events and Death after Acute Coronary Syndrome: A Secondary Analysis of the VISTA-16 Trial. JAMA Cardiol. 2019, 4, 314–320. [Google Scholar] [CrossRef] [Green Version]
  45. Suleiman, M.; Khatib, R.; Agmon, Y.; Mahamid, R.; Boulos, M.; Kapeliovich, M.; Levy, Y.; Beyar, R.; Markiewicz, W.; Hammerman, H.; et al. Early Inflammation and Risk of Long-Term Development of Heart Failure and Mortality in Survivors of Acute Myocardial Infarction: Predictive Role of C-Reactive Protein. J. Am. Coll. Cardiol. 2006, 47, 962–968. [Google Scholar] [CrossRef]
  46. Kayser, S.; Brunner, P.; Althaus, K.; Dorst, J.; Sheriff, A. Selective Apheresis of C-Reactive Protein for Treatment of Indications with Elevated CRP Concentrations. J. Clin. Med. 2020, 9, 2947. [Google Scholar] [CrossRef]
  47. Kunze, R. C-Reactive Protein: From Biomarker to Trigger of Cell Death? Ther. Apher. Dial. 2019, 23, 494–496. [Google Scholar] [CrossRef]
  48. Barrett, T.D.; Hennan, J.K.; Marks, R.M.; Lucchesi, B.R. C-Reactive-Protein-Associated Increase in Myocardial Infarct Size After Ischemia/Reperfusion. J. Pharmacol. Exp. Ther. 2002, 303, 1007–1013. [Google Scholar] [CrossRef] [Green Version]
  49. Griselli, M.; Herbert, J.; Hutchinson, W.; Taylor, K.; Sohail, M.; Krausz, T.; Pepys, M. C-Reactive Protein and Complement Are Important Mediators of Tissue Damage in Acute Myocardial Infarction. J. Exp. Med. 1999, 190, 1733–1740. [Google Scholar] [CrossRef] [Green Version]
  50. Lagrand, W.K.; Niessen, H.W.; Wolbink, G.-J.; Jaspars, L.H.; Visser, C.A.; Verheugt, F.W.; Meijer, C.J.; Hack, C.E. C-Reactive Protein Colocalizes With Complement in Human Hearts During Acute Myocardial Infarction. Circulation 1997, 95, 97–103. [Google Scholar] [CrossRef] [Green Version]
  51. Nijmeijer, R.; Lagrand, W.K.; Lubbers, Y.T.; Visser, C.A.; Meijer, C.J.; Niessen, H.W.; Hack, C.E. C-Reactive Protein Activates Complement in Infarcted Human Myocardium. Am. J. Pathol. 2003, 163, 269–275. [Google Scholar] [CrossRef] [Green Version]
  52. Sheriff, A.; Schindler, R.; Vogt, B.; Abdel-Aty, H.; Unger, J.K.; Bock, C.; Gebauer, F.; Slagman, A.; Jerichow, T.; Mans, D.; et al. Selective apheresis of C-reactive protein: A new therapeutic option in myocardial infarction? J. Clin. Apher. 2014, 30, 15–21. [Google Scholar] [CrossRef] [PubMed]
  53. Valtchanova-Matchouganska, A.; Gondwe, M.; Nadar, A. The role of C-reactive protein in ischemia/reperfusion injury and preconditioning in a rat model of myocardial infarction. Life Sci. 2004, 75, 901–910. [Google Scholar] [CrossRef] [PubMed]
  54. Tschöpe, C.; Ammirati, E.; Bozkurt, B.; Caforio, A.L.P.; Cooper, L.T.; Felix, S.B.; Hare, J.M.; Heidecker, B.; Heymans, S.; Hübner, N.; et al. Myocarditis and inflammatory cardiomyopathy: Current evidence and future directions. Nat. Rev. Cardiol. 2020, 18, 169–193. [Google Scholar] [CrossRef] [PubMed]
  55. Zimmermann, O.; Bienek-Ziolkowski, M.; Wolf, B.; Vetter, M.; Baur, R.; Mailänder, V.; Hombach, V.; Torzewski, J. Myocardial inflammation and non-ischaemic heart failure: Is there a role for C-reactive protein? Basic Res. Cardiol. 2009, 104, 591–599. [Google Scholar] [CrossRef] [PubMed]
  56. Catanese, L.; Tarsia, J.; Fisher, M. Acute Ischemic Stroke Therapy Overview. Circ. Res. 2017, 120, 541–558. [Google Scholar] [CrossRef] [PubMed]
  57. Anrather, J.; Iadecola, C. Inflammation and Stroke: An Overview. Neurotherapeutics 2016, 13, 661–670. [Google Scholar] [CrossRef]
  58. Montaner, J.; Fernandez-Cade, I.; Molina, C.A.; Ribo, M.; Huertas, R.; Rosell, A.; Penalba, A.; Ortega, L.; Chacón, P.; Alvarez-Sabin, J. Poststroke C-Reactive Protein Is a Powerful Prognostic Tool Among Candidates for Thrombolysis. Stroke 2006, 37, 1205–1210. [Google Scholar] [CrossRef] [Green Version]
  59. Winbeck, K.; Poppert, H.; Etgen, T.; Conrad, B.; Sander, D. Prognostic Relevance of Early Serial C-Reactive Protein Measurements After First Ischemic Stroke. Stroke 2002, 33, 2459–2464. [Google Scholar] [CrossRef]
  60. Di Napoli, M.; Papa, F.; Bocola, V. Prognostic Influence of Increased C-Reactive Protein and Fibrinogen Levels in Ischemic Stroke. Stroke 2001, 32, 133–138. [Google Scholar] [CrossRef]
  61. Di Napoli, M.; Schwaninger, M.; Cappelli, R.; Ceccarelli, E.; Di Gianfilippo, G.; Donati, C.; Emsley, H.; Forconi, S.; Hopkins, S.; Masotti, L.; et al. Evaluation of C-Reactive Protein Measurement for Assessing the Risk and Prognosis in Ischemic Stroke: A statement for health care professionals from the CRP Pooling Project members. Stroke 2005, 36, 1316–1329. [Google Scholar] [CrossRef] [PubMed]
  62. Elkind, M.S.V.; Tai, W.; Coates, K.; Paik, M.C.; Sacco, R.L. High-Sensitivity C-Reactive Protein, Lipoprotein-Associated Phospholipase A2, and Outcome After Ischemic Stroke. Arch. Intern. Med. 2006, 166, 2073–2080. [Google Scholar] [CrossRef] [PubMed]
  63. Muir, K.W.; Weir, C.; Alwan, W.; Squire, I.B.; Lees, K.R. C-Reactive Protein and Outcome After Ischemic Stroke. Stroke 1999, 30, 981–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Gill, R.; Kemp, J.A.; Sabin, C.; Pepys, M.B. Human C-Reactive Protein Increases Cerebral Infarct Size after Middle Cerebral Artery Occlusion in Adult Rats. J. Cereb. Blood Flow Metab. 2004, 24, 1214–1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, F.; Li, L.; Xu, M.; Wu, J.; Luo, D.; Zhu, Y.; Li, B.; Song, X.; Zhou, X. Prognostic value of interleukin-6, C-reactive protein, and procalcitonin in patients with COVID-19. J. Clin. Virol. 2020, 127, 104370. [Google Scholar] [CrossRef]
  66. Mueller, A.A.; Tamura, T.; Crowley, C.P.; DeGrado, J.R.; Haider, H.; Jezmir, J.L.; Keras, G.; Penn, E.H.; Massaro, A.F.; Kim, E.Y. Inflammatory Biomarker Trends Predict Respiratory Decline in COVID-19 Patients. Cell Rep. Med. 2020, 1, 100144. [Google Scholar] [CrossRef]
  67. Ruan, Q.; Yang, K.; Wang, W.; Jiang, L.; Song, J. Clinical predictors of mortality due to COVID-19 based on an analysis of data of 150 patients from Wuhan, China. Intensive Care Med. 2020, 46, 846–848. [Google Scholar] [CrossRef] [Green Version]
  68. Shang, W.; Dong, J.; Ren, Y.; Tian, M.; Li, W.; Hu, J.; Li, Y. The value of clinical parameters in predicting the severity of COVID-19. J. Med. Virol. 2020, 92, 2188–2192. [Google Scholar] [CrossRef]
  69. Tan, C.; Huang, Y.; Shi, F.; Tan, K.; Ma, Q.; Chen, Y.; Jiang, X.; Li, X. C-reactive protein correlates with computed tomographic findings and predicts severe COVID-19 early. J. Med. Virol. 2020, 92, 856–862. [Google Scholar] [CrossRef] [Green Version]
  70. Nienhold, R.; Ciani, Y.; Koelzer, V.H.; Tzankov, A.; Haslbauer, J.D.; Menter, T.; Schwab, N.; Henkel, M.; Frank, A.; Zsikla, V.; et al. Two distinct immunopathological profiles in autopsy lungs of COVID-19. Nat. Commun. 2020, 11, 5086. [Google Scholar] [CrossRef]
  71. Torzewski, M. C-reactive Protein: Friend or Foe? Evidence from Phylogeny. Front. Cardiovasc. Med. 2022; in press. [Google Scholar]
  72. Smilowitz, N.R.; Nguy, V.; Aphinyanaphongs, Y.; Newman, J.D.; Xia, Y.; Reynolds, H.R.; Hochman, J.S.; Fishman, G.I.; Berger, J.S. Multiple Biomarker Approach to Risk Stratification in COVID-19. Circulation 2021, 143, 1338–1340. [Google Scholar] [CrossRef] [PubMed]
  73. Velavan, T.P.; Meyer, C.G. Mild versus severe COVID-19: Laboratory markers. Int. J. Infect. Dis. 2020, 95, 304–307. [Google Scholar] [CrossRef] [PubMed]
  74. 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]
  75. Bhatraju, P.K.; Ghassemieh, B.J.; Nichols, M.; Kim, R.; Jerome, K.R.; Nalla, A.K.; Greninger, A.L.; Pipavath, S.; Wurfel, M.M.; Evans, L.; et al. COVID-19 in Critically Ill Patients in the Seattle Region—Case Series. N. Engl. J. Med. 2020, 382, 2012–2022. [Google Scholar] [CrossRef]
  76. Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J.; on behalf of the HLH Across Speciality Collaboration, UK. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
  77. Mosquera-Sulbaran, J.A.; Pedreañez, A.; Carrero, Y.; Callejas, D. C-reactive protein as an effector molecule in COVID-19 pathogenesis. Rev. Med. Virol. 2021, 31, e2221. [Google Scholar] [CrossRef]
  78. Kayser, S.; Kunze, R.; Sheriff, A. Selective C-reactive protein apheresis for COVID-19 patients suffering from organ damage. Ther. Apher. Dial. 2020, 25, 251–252. [Google Scholar] [CrossRef]
  79. Pepys, M.B. C-reactive protein predicts outcome in COVID-19: Is it also a therapeutic target? Eur. Heart J. 2021, 42, 2280–2283. [Google Scholar] [CrossRef]
  80. Ringel, J.; Ramlow, A.; Bock, C.; Sheriff, A. Case Report: C-Reactive Protein Apheresis in a Patient With COVID-19 and Fulminant CRP Increase. Front. Immunol. 2021, 12, 708101. [Google Scholar] [CrossRef]
  81. Schumann, C.; Heigl, F.; Rohrbach, I.J.; Sheriff, A.; Wagner, L.; Wagner, F.; Torzewski, J. A Report on the First 7 Sequential Patients Treated Within the C-Reactive Protein Apheresis in COVID (CACOV) Registry. Am. J. Case Rep. 2021, 23, e935263. [Google Scholar] [CrossRef] [PubMed]
  82. Torzewski, J.; Zimmermann, O.; Kayser, S.; Heigl, F.; Wagner, F.; Sheriff, A.; Schumann, C. Successful Treatment of a 39-Year-Old COVID-19 Patient with Respiratory Failure by Selective C-Reactive Protein Apheresis. Am. J. Case Rep. 2021, 22, e932964. [Google Scholar] [CrossRef] [PubMed]
  83. Szalai, A.J.; McCrory, M.A.; Xing, N.; Hage, F.G.; Miller, A.; Oparil, S.; Chen, Y.-F.; Mazzone, M.; Early, R.; Henry, S.P.; et al. Inhibiting C-Reactive Protein for the Treatment of Cardiovascular Disease: Promising Evidence from Rodent Models. Mediat. Inflamm. 2014, 2014, 353614. [Google Scholar] [CrossRef] [PubMed]
  84. Pepys, M.B.; Hirschfield, G.; Tennent, G.A.; Gallimore, J.R.; Kahan, M.C.; Bellotti, V.; Hawkins, P.N.; Myers, R.M.; Smith, M.D.; Polara, A.; et al. Targeting C-reactive protein for the treatment of cardiovascular disease. Nature 2006, 440, 1217–1221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kolkhof, P.; Geerts, A.; Schäfer, S.; Torzewski, J. Cardiac glycosides potently inhibit C-reactive protein synthesis in human hepatocytes. Biochem. Biophys. Res. Commun. 2010, 394, 233–239. [Google Scholar] [CrossRef] [PubMed]
  86. Mattecka, S.; Brunner, P.; Hähnel, B.; Kunze, R.; Vogt, B.; Sheriff, A. PentraSorb C-Reactive Protein: Characterization of the Selective C-Reactive Protein Adsorber Resin. Ther. Apher. Dial. 2019, 23, 474–481. [Google Scholar] [CrossRef] [Green Version]
  87. Heigl, F.; Pflederer, T.; Klingel, R.; Hettich, R.; Lotz, N.; Reeg, H.; Schettler, V.J.; Roeseler, E.; Grützmacher, P.; Hohenstein, B.; et al. Lipoprotein apheresis in Germany—Still more commonly indicated than implemented. How can patients in need access therapy? Atheroscler. Suppl. 2019, 40, 23–29. [Google Scholar] [CrossRef]
  88. Grazia, Z.M.; Claudia, S. Effects of selective H.E.L.P. LDL-apheresis on plasma inflammatory markers concentration in severe dyslipidemia: Implication for anti-inflammatory response. Cytokine 2011, 56, 850–854. [Google Scholar] [CrossRef]
  89. Ries, W.; Sheriff, A.; Heigl, F.; Zimmermann, O.; Garlichs, C.D.; Torzewski, J. “First in Man”: Case Report of Selective C-Reactive Protein Apheresis in a Patient with Acute ST Segment Elevation Myocardial Infarction. Case Rep. Cardiol. 2018, 2018, 4767105. [Google Scholar] [CrossRef]
  90. Ries, W.; Heigl, F.; Garlichs, C.; Sheriff, A.; Torzewski, J. Selective C-Reactive Protein-Apheresis in Patients. Ther. Apher. Dial. 2019, 23, 570–574. [Google Scholar] [CrossRef]
  91. Sands, B.E.; Katz, S.; Wolf, D.C.; Feagan, B.G.; Wang, T.; Gustofson, L.-M.; Wong, C.; Vandervoort, M.K.; Hanauer, S. A randomised, double-blind, sham-controlled study of granulocyte/monocyte apheresis for moderate to severe Crohn’s disease. Gut 2012, 62, 1288–1294. [Google Scholar] [CrossRef] [PubMed]
  92. Sands, B.E.; Sandborn, W.J.; Feagan, B.; Löfberg, R.; Hibi, T.; Wang, T.; Gustofson, L.; Wong, C.J.; Vandervoort, M.K.; Hanauer, S. A Randomized, Double-Blind, Sham-Controlled Study of Granulocyte/Monocyte Apheresis for Active Ulcerative Colitis. Gastroenterology 2008, 135, 400–409. [Google Scholar] [CrossRef] [PubMed]
  93. Bosch, T. Recent advances in therapeutic apheresis. J. Artif. Organs 2003, 6, 1–8. [Google Scholar] [CrossRef] [PubMed]
Table 1. Pros and cons for CRP apheresis.
Table 1. Pros and cons for CRP apheresis.
ProsCons
efficient and fast removal of large amounts of CRP within hoursblood plasma needs to be supplied to the adsorber instead of whole blood
regenerable immune adsorber→nearly unlimited capacity the treatment takes approximately 5 h and needs to be repeated on successive days depending on the indication
approved by CE certification for removal of CRP additional anticoagulation maybe critical in some patients
specific for CRP (minimally) invasive procedure requiring peripheral venous access or Shaldon catheter
no removal of other molecules or medicationimmunosuppression (?)
reusable adsorber
apheresis is an established technique
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Torzewski, J.; Brunner, P.; Ries, W.; Garlichs, C.D.; Kayser, S.; Heigl, F.; Sheriff, A. Targeting C-Reactive Protein by Selective Apheresis in Humans: Pros and Cons. J. Clin. Med. 2022, 11, 1771. https://doi.org/10.3390/jcm11071771

AMA Style

Torzewski J, Brunner P, Ries W, Garlichs CD, Kayser S, Heigl F, Sheriff A. Targeting C-Reactive Protein by Selective Apheresis in Humans: Pros and Cons. Journal of Clinical Medicine. 2022; 11(7):1771. https://doi.org/10.3390/jcm11071771

Chicago/Turabian Style

Torzewski, Jan, Patrizia Brunner, Wolfgang Ries, Christoph D. Garlichs, Stefan Kayser, Franz Heigl, and Ahmed Sheriff. 2022. "Targeting C-Reactive Protein by Selective Apheresis in Humans: Pros and Cons" Journal of Clinical Medicine 11, no. 7: 1771. https://doi.org/10.3390/jcm11071771

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