Next Article in Journal
Severe COVID-19 Infection Management in a Patient with Mild Haemophilia—A Case Report
Previous Article in Journal
Reproductive Issues in Long-Term Surviving Patients following Therapy for Hodgkin’s Disease in the Republic of North Macedonia: Risks of Infertility According to First-Line Treatment Regimens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Cause–Effect Dilemma of Hematologic Changes in COVID-19: One Year after the Start of the Pandemic

1
Pediatric Hematology and BMT Unit, Cairo University, Giza 12613, Egypt
2
Pediatric Department, Faculty of Medicine, Cairo University, Giza 12211, Egypt
*
Author to whom correspondence should be addressed.
Current Address: Faculty of Medicine, Kasr Alainy Medical School, Cairo University, Giza 12211, Egypt.
Hematol. Rep. 2022, 14(2), 95-102; https://doi.org/10.3390/hematolrep14020014
Submission received: 26 January 2022 / Revised: 19 February 2022 / Accepted: 24 March 2022 / Published: 28 March 2022

Abstract

:
COVID-19 is a systemic infection that leads to multisystem affection, including hematological changes. On the other hand, the patients who have certain hematological diseases are more susceptible to COVID-19 infection. The aim of this review is to examine the wide spectrum of hematological changes that are reported to occur due to COVID-19 infection. Most of the studies over the past year mainly show that most of these changes are mainly non-specific, but are of prognostic value. On the other hand, the susceptibility of hematological patients to COVID-19 infection and complications remains questionable. Patients with certain hematological diseases (including malignancy) and those who are treated by aggressive immunosuppressive therapy have shown higher rates of COVID-19 infection and complications. On the other hand, for most of the patients suffering from other chronic hematological conditions, no evidence has shown a greater risk of infection, compared to the general population.

1. Introduction

COVID-19 is a systemic infection that leads to multisystem affection, including hematological changes. On the other hand, patients who have certain hematological diseases are more susceptible to COVID-19 infection [1].
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the disease named coronavirus disease-19 (COVID-19) [2], resulted in >75 million infections and >1.7 million deaths worldwide, until December 2020 [3]. Coronaviruses are a heterogeneous group of single-stranded plus-sense RNA viruses belonging to the Coronaviridae family and Nidovirales order [4]. The clinical phenotypes of COVID-19 are variable, ranging from asymptomatic up to severe illness with mortality. There are different theories about the variability of the clinical severity among the infected individuals. Those theories can be explained by genetic and epigenetic processes that include high viral load exposure; environmental conditions, such as the absence or presence of an element in the air inhaled by the patient; climate change; pollution, which may aggravate the illness; and the somatic transformation of cells, which occurs in infected human hosts. Finally, severe COVID-19 in previously healthy individuals may result from monogenic predisposition, monogenic inborn errors of immunity of Toll-like receptor 3 (TLR3), and type I IFN cell-intrinsic immunity [5,6].
COVID-19 is a systemic infection that has variable symptoms, which include many respiratory symptoms that can develop into acute respiratory distress syndrome, metabolic acidosis, septic shock, and coagulation dysfunction. Multiple organ failure, including disseminated intravascular coagulation (DIC), can further develop into the occurrence of macrophage activation syndrome (MAS), also known as secondary hemophagocytic lymphohistiocytosis (sHLH) [7,8].
The hematological presentations in COVID-19 are variable and non-specific. While in adults, lymphopenia, leukocytosis or leucopenia, and thrombocytopenia are all reported. in children, the observed hematological abnormalities are non-significant, and the full blood cell counts are normal in most of the patients [9].
In this review, we present the interrelation between hematology and COVID-19 infection in its two sides. From one side, we present the hematological presentations and its relation to the disease severity and outcome, and from the other side, we present the susceptibility of patients with hematological diseases to COVID-19 infection.

2. COVID-19 Infection of Patients with Hematological Diseases

2.1. Cancer Patients and COVID-19 Infection

The highest incidence of complications among patients with hematological diseases in the era of COVID-19 are those with malignancies [1,10]. These complications include a severity of COVID-19 disease, ICU admission, the need for mechanical ventilation, and mortality [10]. The cascade of disease progression among this vulnerable subpopulation is more aggressive and life threatening. Unique pathophysiological changes are still under examination, but several studies have shown that prognostic biomarkers, such as CRP, D-dimer, prothrombin time, and serum IL-6 levels, were significantly higher in cancer patients than in non-cancer patients [11]. More specifically, the concurrence of acute myeloid leukemia in children with COVID-19 presents an extraordinary challenge, since this specific group of patients suffer from a profound and long-lasting humoral and cellular immune deficiency [11,12]. This is illustrated by the fact that the cancer institutes’ guidelines in some countries have recommended delaying the therapy for AML until symptoms resolve and PCR becomes negative [11]. Other studies show that hematopoietic stem cell transplantation (HSCT) (for hematopoietic malignancy and aplasia) or CAR-T therapy have faced unprecedented challenges for fear of infection of the donor, recipient, and healthcare workers [12]. In addition, the elevated number of leukocytes observed in patients with proliferative chronic myelomonocytic leukemia (CMML) increases the risk of leukemoid reactions and overlap syndromes (MDS/MPN). Thus, when they are subjected to COVID-19 infection, they would need a stronger form of immunosuppression, such as hydroxycarbamide, even if they are asymptomatic [13]. On the other hand, results from smaller cancer centers [14] reported successes following a non-myeloablative conditioning regimen followed by post-transplant cyclophosphamide (PTCy), because, in such cases, the successful prophylaxis against graft-versus-host disease (GVHD) could simultaneously work on the attenuation of COVID-19 disease. Moreover, another single-center study demonstrated provisional data regarding the mortality of organ transplant patients that is close to the previous rates before the pandemic, and they suggested that “such patients with an already immune suppressed immune system are not able to produce a cytokine storm and thus do not experience fulminant COVID-19 infection” [15]. What adds to the complexity of the problem, as mentioned by Papakonstantinou et al. [16], is that the molecular mechanisms governing the pathogenesis, metastasis, and relapse of cancer vary widely across the hematological malignancies. Other challenges include the deficiency of cancer research during the COVID-19 pandemic, namely, the strict public health measures taken by authorities to limit the spread of the virus negatively influenced cancer research centers due to quarantining measures, the lack of supplies, and interrupting researchers called to perform emergency hospital duties [17].

2.2. Hemoglobinopathies and COVID-19 Infection

In hemoglobinopathies patients, such as sickle cell disease and thalassemia, infection is considered as one of the leading causes of mortality [18,19]. The reason for the higher susceptibility is the comorbidities, including ineffective erythropoiesis, chronic hemolytic anemia, iron overload, and hypercoagulability, which make them vulnerable to complications of COVID-19 infection [19]. Although splenectomy (or auto-splenectomy in the case of SCD) is not known to increase the risk of severe viral infections, splenectomized patients may be at risk of severe secondary bacterial infections when infected by COVID-19 [18]. Although the experience from the 2009 H1N1 influenza pandemic has presented the H1N1 influenza virus as a trigger for acute chest syndrome (ACS) and the need for intensive-care support [18,20], most of the COVID-19 chronic hemoglobinopathy patients have a milder form. Only in a minor percentage of patients can the more severe forms of COVID-19 pneumonia be attributed to the states of hypoxia and VQ mismatch, which sometimes complicate the normal disease course [18,21]. Another consideration is the concurrent mismatch between blood demand and supply in healthcare services during the pandemic, which led to the prioritization of the needs of chronic hemolytic anemia patients [21].

2.3. Other Hematological Diseases and COVID-19 Susceptibility

Remarks about other specific hematological diseases in the era of COVID-19 have been made in different settings, but further research is needed to confirm the findings [22,23]. These observations have shown that the prevalence of COVID-19 among congenital bleeding disorders (such as Hemophilia A, von Willebrand disease (vWD)) seems to be low, when compared to the general population [24]. The reason is not yet fully known, but it might be due to the constant vigilance of patients implemented by hemophilia centers, or may be due to the spread of the home replacement therapy [21,23]. Moreover, a severe hypocoagulability state may be protective against COVID-19 hypercoagulability-related adverse effects in the absence of other co-morbidities [23]. Similarly, patients with ITP do not show increased rates of infection, even with low doses of immunosuppressants [23,25]. However, higher doses of steroids and rituximab may need to be avoided during the pandemic, and patients may benefit by attempting to replace them with thrombopoietin receptor agonist (TPO) agents and/or IVIG [25]. Moreover, a study by Quinti et al. [26] reported that certain patients with agammaglobulinemia infected by the virus had a mild clinical presentation of the disease, and, moreover, they did not require immune-modulating drug-blocking IL-6. This was explained by the hypothesis that the lack of B-cell-derived IL-6 levels resulted in an attenuation in the level of inflammation and cytokine storm, which, in return, caused a more favorable outcome than was expected. A similar observation was noted by Soresina et al. [20], who reported two COVID-19 patients with X-linked agammaglobulinemia (XLA); both recovered without the need for intensive care, which is considered as rather atypical for immunodeficiency patients. On the contrary, this cytokine inflammatory storm might not act in the favor of other hematological diseases prone to thrombosis. There are some reports that show that COVID-19-positive patients have higher levels of antiphospholipid antibodies in their blood [23,25]. When trying to quantify the types of antiphospholipid antibodies that mostly increase in this disease, Harzallah et al., from France, reported that, out of 56 patients with COVID-19, 25 cases (45%) were lupus anticoagulant (LAC) positive, in comparison to only 5 out of 50 anticardiolipin antibodies (aCL) or aβ2GPI. The scarcity of clinical follow-up information regarding the thromboembolic phenomenon in these patients [27] mandates the need for further research in this area.

3. Hematological Presentations of COVID-19 Infection

3.1. Hypercoagulability and the D-Dimer

Arterial and venous thrombosis were correlated with COVID-19 infection. The elevation in D-dimer levels [28], prolongation of prothrombin time (PT), and thrombocytopenia [29] are considered as important markers to assess the severity and outcome of COVID-19 infection. However, these markers may remain normal in many patients, suggesting an unusual prothrombotic state that is distinct from the sepsis-induced coagulopathy [30]. Cytokine storm, hypoxic vaso-occlusion, and the direct activation of immune and vascular cells by viral infection can be potential causes of the macro/micro vascular complications. Additionally, cellular remnants from the neutrophil extracellular traps (NETs) witnessed in many hospitalized patients [31,32] may contribute to the prothrombotic cascade [33]. Furthermore, there are findings that suggest that half of the patients hospitalized with COVID-19 become at least transiently positive for aPL antibodies, including anticardiolipin, anti-β2 glycoprotein I, and anti-phosphatidylserine/prothrombin (aPS/PT), which are potentially pathogenic [34]. The dysregulation of the renin–angiotensin–aldosterone system (RAAS), which induces acute lung injury, further induces endothelial dysfunction causing widespread immune thrombosis and multi-system organ damage [28].
Hence, several risk assessments models (RAMs) and antithrombotic therapeutic options have been tried, and one of the earliest evidence-based WHO guidelines [35] recommended the use of low molecular weight heparin LMWH, which improved 28-day overall survival (one of the earliest applications in a Chinese study [36]). On the other hand, less evidence is found concerning vaccine-induced immune thrombotic thrombocytopenia (VITT), and the issue of anticoagulation with or without the use of IVIG still remains controversial [7]. Some authors have even argued that the addition of fibrinolytic agents to heparin can increase the incidence of bleeding and, in this case, the improvement in the D-dimer can exceptionally be used as a biomarker of fibrinolysis suppression in high-risk patients [37].
Studies demonstrated other changes in coagulation (apart from D-dimer elevation), such as the prolongation of prothrombin time and activated thromboplastin time, which indicate laboratory coagulopathy that progresses to clinically evident disseminated intravascular coagulopathy (DIC) (defined by the International Society on Thrombosis and Haemostasis Score). The progression to DIC in infected patient predicted a poor prognosis, occurring in 71.4% of all non-survivors vs. 0.6% of survivors [28].

3.2. Complete Blood Counts and Other Laboratory Markers

During the incubation period and the early phase of the disease, peripheral blood leukocyte and lymphocyte counts were normal or slightly reduced. Following viremia, COVID-19 primarily affects the tissues expressing high levels of ACE2; with a pronounced systemic increase in inflammatory mediators and cytokines, significant lymphopenia becomes evident [38]. Thrombocytopenia and leucopenia were also described in several studies, highlighting the association between lymphopenia, the need of ICU admission, and the development of acute respiratory distress syndrome (ARDS) [38,39].
Several theories explained the cause of this lymphopenia and included the presence of the ACE2 receptor on the lymphocytes’ surface, leading to their lysis [40]; Bo Diao et.al reported that the decrease in the numbers of T-lymphocyte (CD3+, CD4+, and CD8+) subsets was inversely proportionate to the inflammatory cytokines, including IL-6, IL-10, and TNF-a, which promote lymphocyte apoptosis [41]. Furthermore, the coexistence of lactic acid acidosis may also be an adding factor that inhibits lymphocyte proliferation [42] Thrombocytopenia is also significantly associated with the intensity of the COVID-19 disease [38]. Leukocytosis with neutrophilia were significantly reported in patients with myocardial injury admitted in ICU, compared to the others [43,44]. In a recent study conducted by Urbano et al. [45] in 2022, interesting results reported a 16-times-higher risk of mortality in patients with a neutrophil count of 5.91 × 109/L or higher, compared to normal levels. Nevertheless, both can be an indication of the coexistence of bacterial infection [38,46]. Additionally, neutrophilia could be an expression of the cytokine storm and hyper-inflammatory state [47,48,49,50]. A serial assessment of the counts level presents a predictive value; the greater the change in the counts over time, the more severe the disease, resulting in deterioration and the necessity of ICU hospitalization [39,48,51,52].

3.3. The Biomarkers Procalcitonin, Ferritin, and C-Reactive Protein, IL6

  • C-reactive protein (CRP) is an acute phase reactant. Its high level is associated with admissions to the ICU, ARDS development, and not significantly associated with mortality [43]. On the other hand, Deng Y and his colleagues reported that it was significantly higher in the death group than the recovered group [53]. It could also indicate the associated bacterial infection.
  • Procalcitonin is a precursor of calcitonin, a hormone that plays a pivotal role in calcium homeostasis. In severe COVID-19 infections, which requires ICU admission, high levels of procalcitonin were observed [54], which could be explained by the association of bacterial infection.
  • IL-6: COVID-19 infections can cause cytokine storm and macrophage activation syndrome (MAS), due to the hyper-activated T lymphocytes with the release of different inflammatory cytokines, including IL-6 [55,56]. Based on this pathophysiology, Tocliziumab, which is a recombinant humanized monoclonal anti-IL-6 receptor antibody, inhibiting IL-6, which has been used in auto-inflammatory diseases [56], can be tried. Tocliziumab is the best-studied drug, as along with sarilumab, among IL-6 inhibitors in COVID-19 patients, showing promising results among critically ill patient with MAS [57,58].
Unlike in the case of adults, the radiological and laboratory findings were non-specific in children, and the full blood cell counts were normal in the majority of patients, while the CRP and pro-calcitonin were abnormal in almost one third of the patients [8]. In pediatric inflammatory multisystem syndrome temporally associated with COVID-19 (PIMS-TS) or multisystem inflammatory syndrome in children (MIS-C), there were neutrophilia, abnormal levels of CRP, ESR, ferritin, and D-dimers. The studies supported that high WBCs, neutrophils, CRPs, low lymphocytes, anemia, low platelets, high fibrinogen levels, procalcitonin, ALTs, and Troponin are more commonly associated with MIS-C rather than Kawasaki disease (KD), while the D-dimer level was similar between MIS-C and KD [59,60,61]. Despite the high levels of D-dimers among the hospitalized children, the studies did not point to the use of an anticoagulant in these children with a very good outcome and discharge from the hospitals [59,60,61].

4. Conclusions

Patients with certain hematological diseases (including malignancy) and those who are treated by aggressive immunosuppressive therapy have shown higher rates of COVID-19 infection and complications. On the other hand, for most of the patients suffering from other chronic hematological conditions, no evidence has shown a greater risk of infection, compared to the general population. However, COVID-19 leads to a wide spectrum of hematological changes that are non-specific, but are of prognostic value.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No data reporting.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liguoro, I.; Pilotto, C.; Bonanni, M.; Ferrari, M.E.; Pusiol, A.; Nocerino, A.; Vidal, E.; Cogo, P. SARS-CoV-2 infection in children and newborns: A systematic review. Eur. J. Pediatr. 2020, 179, 1029–1046. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int (accessed on 18 February 2022).
  3. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef] [PubMed]
  4. Lauber, C.; Ziebuhr, J.; Junglen, S.; Drosten, C.; Zirkel, F.; Nga, P.T.; Morita, K.; Snijder, E.J.; Gorbalenya, A.E. Mesoniviridae: A proposed new family in the order Nidovirales formed by a single species of mosquito-borne viruses. Arch Virol. 2012, 157, 1623–1628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Zhang, S.-Y.; Zhang, Q.; Casanova, J.-L.; Su, H.C. Severe COVID-19 in the young and healthy: Monogenic inborn errors of immunity? Nat. Rev. Immunol. 2020, 20, 455–456. [Google Scholar] [CrossRef]
  6. Zhang, Q.; Bastard, P.; Liu, Z.; Le Pen, J.; Moncada-Velez, M.; Chen, J.; Ogishi, M.; Sabli, I.K.D.; Hodeib, S.; Korol, C.; et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 2020, 370, eabd4570. [Google Scholar] [CrossRef]
  7. Rizk, J.G.; Gupta, A.; Sardar, P.; Henry, B.M.; Lewin, J.C.; Lippi, G.; Lavie, C.J. Clinical Characteristics and Pharmacological Management of COVID-19 Vaccine–Induced Immune Thrombotic Thrombocytopenia with Cerebral Venous Sinus Thrombosis: A Review. JAMA Cardiol. 2021, 6, 1451–1460. [Google Scholar] [CrossRef]
  8. Crayne, C.B.; Albeituni, S.; Nichols, K.E.; Cron, R.Q. The Immunology of Macrophage Activation Syndrome. Front. Immunol. 2019, 10, 119. [Google Scholar] [CrossRef] [Green Version]
  9. ElGohary, G.M.; Hashmi, S.; Styczynski, J.; Kharfan-Dabaja, M.A.; Alblooshi, R.M.; de la Cámara, R.; Mohmed, S.; Alshaibani, A.; Cesaro, S.; El-Aziz, N.A.; et al. The risk and prognosis of COVID-19 infection in cancer patients: A systematic review and meta-analysis. Hematol. Oncol. Stem Cell Ther. 2020, in press. [Google Scholar] [CrossRef]
  10. Khan, A.M.; Ajmal, Z.; Raval, M.; Tobin, E. Concurrent Diagnosis of Acute Myeloid Leukemia and COVID-19: A Management Challenge. Cureus 2020, 12, e9629. [Google Scholar] [CrossRef]
  11. Brissot, E.; Labopin, M.; Baron, F.; Bazarbachi, A.; Bug, G.; Ciceri, F.; Esteve, J.; Giebel, S.; Gilleece, M.H.; Gorin, N.-C.; et al. Management of patients with acute leukemia during the COVID-19 outbreak: Practical guidelines from the acute leukemia working party of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant. 2020, 56, 532–535. [Google Scholar] [CrossRef]
  12. Patnaik, M.M.; Lasho, T.; Padron, E.; McCullough, K.; Al-Kali, A.; Tefferi, A.; Zeidan, A.M.; Gangat, N.; Savona, M.; Steensma, D.P.; et al. Special considerations in the management of patients with myelodysplastic myndrome / myeloproliferative neoplasm overlap syndromes during the SARS-CoV-2 pandemic. Am. J. Hematol. 2020, 95, E203–E208. [Google Scholar] [CrossRef] [PubMed]
  13. Ljungman, P.; Mikulska, M.; De La Camara, R.; Basak, G.W.; Chabannon, C.; Corbacioglu, S.; Duarte, R.; Dolstra, H.; Lankester, A.C.; Mohty, M.; et al. The challenge of COVID-19 and hematopoietic cell transplantation; EBMT recommendations for management of hematopoietic cell transplant recipients, their donors, and patients undergoing CAR T-cell therapy. Bone Marrow Transplant. 2020, 55, 2071–2076. [Google Scholar] [CrossRef] [PubMed]
  14. Kanellopoulos, A.; Ahmed, M.Z.; Kishore, B.; Lovell, R.; Horgan, C.; Paneesha, S.; Lloyd, R.; Salhan, B.; Giles, H.; Chauhan, S.; et al. COVID-19 in bone marrow transplant recipients: Reflecting on a single centre experience. Br. J. Haematol. 2020, 190, e67–e70. [Google Scholar] [CrossRef] [PubMed]
  15. Alfishawy, M.; Elbendary, A.; Mohamed, M.; Nassar, M. COVID-19 Mortality in Transplant Recipients. Int. J. Organ Transplant. Med. 2020, 11, 145–162. [Google Scholar] [PubMed]
  16. Papakonstantinou, E.; Dragoumani, K.; Efthimiadou, A.; Palaiogeorgou, A.M.; Pierouli, K.; Mitsis, T.; Chrousos, G.P.; Bacopoulou, F.; Vlachakis, D. Haematological malignancies implications during the times of the COVID-19 pandemic (Review). Oncol. Lett. 2021, 22, 856. [Google Scholar] [CrossRef] [PubMed]
  17. Moujaess, E.; Kourie, H.R.; Ghosn, M. Cancer patients and research during COVID-19 pandemic: A systematic review of current evidence. Crit. Rev. Oncol. 2020, 150, 102972. [Google Scholar] [CrossRef]
  18. Ali, M.; Okar, L.; Omar, N.E.; Parengal, J.; Soliman, A.; Yassin, M.A. Sickle Cell Anemia Presenting with Vaso-Occlusive Pain: Should We Screen for COVID-19? DMJ 2021, 4, 36–39. [Google Scholar] [CrossRef]
  19. Saliba, A.N.; Atoui, A.; Labban, M.; Hamade, H.; Bou-Fakhredin, R.; Mufarrij, A.; Taher, A.T. Thalassemia in the emergency department: Special considerations for a rare disease. Ann. Hematol. 2020, 99, 1967–1977. [Google Scholar] [CrossRef]
  20. Bundy, D.G.; Strouse, J.J.; Casella, J.F.; Miller, M.R. Burden of Influenza-Related Hospitalizations Among Children With Sickle Cell Disease. Pediatrics 2010, 125, 234–243. [Google Scholar] [CrossRef] [Green Version]
  21. Stanworth, S.J.; New, H.V.; Apelseth, T.O.; Brunskill, S.; Cardigan, R.; Doree, C.; Germain, M.; Goldman, M.; Massey, E.; Prati, D.; et al. Effects of the COVID-19 pandemic on supply and use of blood for transfusion. Lancet Haematol. 2020, 7, e756–e764. [Google Scholar] [CrossRef]
  22. Noun, P.; Ibrahim, A.; Hodroj, M.H.; Bou-Fakhredin, R.; Taher, A.T. COVID-19 in benign hematology: Emerging challenges and special considerations for healthcare professionals. Expert Rev. Hematol. 2020, 13, 1081–1092. [Google Scholar] [CrossRef] [PubMed]
  23. Finelli, C.; Parisi, S. The clinical impact of COVID-19 epidemic in the hematologic setting. Adv. Biol. Regul. 2020, 77, 100742. [Google Scholar] [CrossRef] [PubMed]
  24. Naderi, M.; Malek, F.; Aliabad, G.M.; Behnampoor, M.; De Sanctis, V.; Karimi, M. Congenital Bleeding Disorders amid the COVID-19 pandemic: Open questions and recommendations. Acta Biomed. 2020, 91, e2020028. [Google Scholar] [PubMed]
  25. Sahu, K.K.; Cerny, J. A review on how to do hematology consults during COVID-19 pandemic. Blood Rev. 2020, 47, 100777. [Google Scholar] [CrossRef]
  26. Quinti, I.; Lougaris, V.; Milito, C.; Cinetto, F.; Pecoraro, A.; Mezzaroma, I.; Mastroianni, C.M.; Turriziani, O.; Bondioni, M.P.; Filippini, M.; et al. A possible role for B cells in COVID-19? Lesson from patients with agammaglobulinemia. J. Allergy Clin. Immunol. 2020, 146, 211–213.e4. [Google Scholar] [CrossRef]
  27. Soresina, A.; Moratto, D.; Chiarini, M.; Paolillo, C.; Baresi, G.; Focà, E.; Bezzi, M.; Baronio, B.; Giacomelli, M.; Badolato, R. Two X-linked agammaglobulinemia patients develop pneumonia as COVID-19 manifestation but recover. Pediatr. Allergy Immunol. 2020, 31, 565–569. [Google Scholar] [CrossRef] [Green Version]
  28. Harzallah, I.; Debliquis, A.; Drénou, B. Lupus anticoagulant is frequent in patients with Covid-19. J. Thromb. Haemost. 2020, 18, 2064–2065. [Google Scholar] [CrossRef]
  29. Wu, H.; Zhu, H.; Yuan, C.; Yao, C.; Luo, W.; Shen, X.; Wang, J.; Shao, J.; Xiang, Y. Clinical and Immune Features of Hospitalized Pediatric Patients With Coronavirus Disease 2019 (COVID-19) in Wuhan, China. JAMA Netw. Open 2020, 3, e2010895. [Google Scholar] [CrossRef]
  30. Abers, M.S.; Delmonte, O.M.; Ricotta, E.E.; Fintzi, J.; Fink, D.L.; de Jesus, A.A.A.; Zarember, K.A.; Alehashemi, S.; Oikonomou, V.; Desai, J.V.; et al. An immune-based biomarker signature is associated with mortality in COVID-19 patients. JCI Insight 2021, 6, 144455. [Google Scholar] [CrossRef]
  31. Morais-Almeida, M.; Aguiar, R.; Martin, B.; Ansotegui, I.J.; Ebisawa, M.; Arruda, L.K.; Caminati, M.; Canonica, G.W.; Carr, T.; Chupp, G.; et al. COVID-19, asthma, and biological therapies: What we need to know. World Allergy Organ. J. 2020, 13, 100126. [Google Scholar] [CrossRef]
  32. Pouletty, M.; Borocco, C.; Ouldali, N.; Caseris, M.; Basmaci, R.; Lachaume, N.; Bensaid, P.; Pichard, S.; Kouider, H.; Morelle, G.; et al. Paediatric multisystem inflammatory syndrome temporally associated with SARS-CoV-2 mimicking Kawasaki disease (Kawa-COVID-19): A multicentre cohort. Ann. Rheum. Dis. 2020, 79, 999–1006. [Google Scholar] [CrossRef] [PubMed]
  33. Dulek, D.E.; Fuhlbrigge, R.C.; Tribble, A.C.; Connelly, J.A.; Loi, M.M.; El Chebib, H.; Chandrakasan, S.; Otto, W.R.; Diorio, C.; Keim, G.; et al. Multidisciplinary Guidance Regarding the Use of Immunomodulatory Therapies for Acute Coronavirus Disease 2019 in Pediatric Patients. J. Pediatr. Infect. Dis. Soc. 2020, 9, 716–737. [Google Scholar] [CrossRef] [PubMed]
  34. Terpos, E.; Ntanasis-Stathopoulos, I.; Elalamy, I.; Kastritis, E.; Sergentanis, T.N.; Politou, M.; Psaltopoulou, T.; Gerotziafas, G.; Dimopoulos, M.A. Hematological findings and complications of COVID-19. Am. J. Hematol. 2020, 95, 834–847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Guan, W.J.; Ni, Z.Y.; Hu, Y.; Liang, W.H.; Qu, C.Q.; He, J.X.; Liu, L.; Shan, H.; Lei, C.L.; Hui, D.S.C.; et al. China medical treatment expert group for COVID-19 2020. Clinical Characteristics of coronavirus disease in China. N. Engl. J. Med. 2020, 382, 1708–1720. [Google Scholar] [CrossRef] [PubMed]
  36. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  37. Manzoor, D.; Bui, C.; Makhoul, E.; Luthringer, D.; Marchevsky, A.; Volod, O. Improvement in plasma D-dimer level in severe SARS-CoV-2 infection can be an indicator of fibrinolysis suppression. Medicine 2021, 100, e25255. [Google Scholar] [CrossRef]
  38. Chen, N.; Zhou, M.; Dong, X.; Qu, J.; Gong, F.; Han, Y.; Qiu, Y.; Wang, J.; Liu, Y.; Wei, Y.; et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020, 395, 507–513. [Google Scholar] [CrossRef] [Green Version]
  39. Diao, B.; Wang, C.; Tan, Y.; Chen, X.; Liu, Y.; Ning, L.; Chen, L.; Li, M.; Liu, Y.; Wang, G.; et al. Reduction and Functional Exhaustion of T Cells in Patients With Coronavirus Disease 2019 (COVID-19). Front. Immunol. 2020, 11, 827. [Google Scholar] [CrossRef]
  40. Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell–derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
  41. Lippi, G.; Plebani, M.; Henry, B.M. Thrombocytopenia is associated with severe coronavirus disease 2019 (COVID-19) infections: A meta-analysis. Clin. Chim. Acta 2020, 506, 145–148. [Google Scholar] [CrossRef]
  42. Shi, S.; Qin, M.; Shen, B.; Cai, Y.; Liu, T.; Yang, F.; Gong, W.; Liu, X.; Liang, J.; Zhao, Q.; et al. Association of Cardiac Injury With Mortality in Hospitalized Patients with COVID-19 in Wuhan, China. JAMA Cardiol. 2020, 5, 802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Guo, T.; Fan, Y.; Chen, M.; Wu, X.; Zhang, L.; He, T.; Wang, H.; Wan, J.; Wang, X.; Lu, Z. Cardiovascular Implications of Fatal Outcomes of Patients with Coronavirus Disease 2019 (COVID-19). JAMA Cardiol. 2020, 5, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Lippi, G.; Plebani, M. The critical role of laboratory medicine during coronavirus disease 2019 (COVID-19) and other viral outbreaks. Clin. Chem. Lab. Med. 2020, 58, 1063–1069. [Google Scholar] [CrossRef] [Green Version]
  45. Urbano, M.; Costa, E.; Geraldes, C. Hematological Changes in SARS-CoV-2 Positive Patients. Hematology, Transfusion and Cell Therapy. Available online: https://www.sciencedirect.com/science/article/pii/S2531137922000128 (accessed on 19 February 2022).
  46. 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]
  47. Qin, C.; Zhou, L.; Hu, Z.; Zhang, S.; Yang, S.; Tao, Y.; Xie, C.; Ma, K.; Shang, K.; Wang, W.; et al. Dysregulation of Immune Response in Patients with Coronavirus 2019 (COVID-19) in Wuhan, China. Clin. Infect. Dis. 2020, 71, 762–768. [Google Scholar] [CrossRef] [PubMed]
  48. 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, Erratum in JAMA Intern. Med. 2020, 180, 1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Deng, Y.; Liu, W.; Liu, K.; Fang, Y.-Y.; Shang, J.; Zhou, L.; Wang, K.; Leng, F.; Wei, P.-F.; Chen, L.; et al. Clinical characteristics of fatal and recovered cases of coronavirus disease 2019 in Wuhan, China: A retrospective study. Chin. Med. J. 2020, 133, 1261–1267. [Google Scholar] [CrossRef]
  50. Lippi, G.; Plebani, M. Laboratory abnormalities in patients with COVID-2019 infection. Clin. Chem. Lab. Med. 2020, 58, 1131–1134. [Google Scholar] [CrossRef] [Green Version]
  51. Wang, D.; Hu, B.; Hu, C.; Zhu, F.; Liu, X.; Zhang, J.; Wang, B.; Xiang, H.; Cheng, Z.; Xiong, Y.; et al. Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus—Infected Pneumonia in Wuhan, China. JAMA 2020, 323, 1061–1069. [Google Scholar] [CrossRef]
  52. Xu, H.; Zhong, L.; Deng, J.; Peng, J.; Dan, H.; Zeng, X.; Li, T.; Chen, Q. High expression of ACE2 receptor of 2019-nCoV on the epithelial cells of oral mucosa. Int. J. Oral Sci. 2020, 12, 8. [Google Scholar] [CrossRef]
  53. Al-Samkari, H.; Berliner, N. Hemophagocytic Lymphohistiocytosis. Annu. Rev. Pathol. 2018, 13, 27–49. [Google Scholar] [CrossRef] [PubMed]
  54. Ding, C.; Jones, G. Anti-interleukin-6 receptor antibody treatment in inflammatory autoimmune diseases. Rev. Recent Clin. Trials 2006, 1, 193–200. [Google Scholar] [CrossRef] [PubMed]
  55. Alzghari, S.K.; Acuña, V.S. Supportive Treatment with Tocilizumab for COVID-19: A Systematic Review. J. Clin. Virol. 2020, 127, 104380. [Google Scholar] [CrossRef] [PubMed]
  56. Wiersinga, W.J.; Rhodes, A.; Cheng, A.C.; Peacock, S.J.; Prescott, H.C. Pathophysiology, Transmission, Diagnosis, and Treatment of Coronavirus Disease 2019 (COVID-19): A Review. Available online: https://pubmed.ncbi.nlm.nih.gov/32648899/ (accessed on 19 February 2022).
  57. Lippi, G.; Favaloro, E.J. D-dimer is Associated with Severity of Coronavirus Disease 2019: A Pooled Analysis. Thromb. Haemost. 2020, 120, 876–878. [Google Scholar] [CrossRef] [Green Version]
  58. Henry, B.M.; de Oliveira, M.H.S.; Benoit, S.; Plebani, M.; Lippi, G. Hematologic, biochemical and immune biomarker abnormalities associated with severe illness and mortality in coronavirus disease 2019 (COVID-19): A meta-analysis. Clin. Chem. Lab. Med. 2020, 58, 1021–1028. [Google Scholar] [CrossRef] [Green Version]
  59. Iba, T.; Di Nisio, M.; Thachil, J.; Wada, H.; Asakura, H.; Sato, K.; Saitoh, D. A Proposal of the Modification of Japanese Society on Thrombosis and Hemostasis (JSTH) Disseminated Intravascular Coagulation (DIC) Diagnostic Criteria for Sepsis-Associated DIC. Clin. Appl. Thromb Hemost. 2018, 24, 439–445. [Google Scholar] [CrossRef] [Green Version]
  60. Shi, H.; Zuo, Y.; Yalavarthi, S.; Gockman, K.; Zuo, M.; Madison, J.A.; Blair, C.; Woodward, W.; Lezak, S.P.; Lugogo, N.L.; et al. Neutrophil calprotectin identifies severe pulmonary disease in COVID-19. J. Leukoc. Biol. 2020, 109, 67–72. [Google Scholar] [CrossRef]
  61. Ramcharan, T.; Nolan, O.; Lai, C.Y.; Prabhu, N.; Krishnamurthy, R.; Richter, A.G.; Jyothish, D.; Kanthimathinathan, H.K.; Welch, S.B.; Hackett, S.; et al. Paediatric Inflammatory Multisystem Syndrome: Temporally Associated with SARS-CoV-2 (PIMS-TS): Cardiac Features, Management and Short-Term Outcomes at a UK Tertiary Paediatric Hospital. Pediatr. Cardiol. 2020, 41, 1391–1401. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Youssry, I.; Abd Elaziz, D.; Ayad, N.; Eyada, I. The Cause–Effect Dilemma of Hematologic Changes in COVID-19: One Year after the Start of the Pandemic. Hematol. Rep. 2022, 14, 95-102. https://doi.org/10.3390/hematolrep14020014

AMA Style

Youssry I, Abd Elaziz D, Ayad N, Eyada I. The Cause–Effect Dilemma of Hematologic Changes in COVID-19: One Year after the Start of the Pandemic. Hematology Reports. 2022; 14(2):95-102. https://doi.org/10.3390/hematolrep14020014

Chicago/Turabian Style

Youssry, Ilham, Dalia Abd Elaziz, Nardeen Ayad, and Iman Eyada. 2022. "The Cause–Effect Dilemma of Hematologic Changes in COVID-19: One Year after the Start of the Pandemic" Hematology Reports 14, no. 2: 95-102. https://doi.org/10.3390/hematolrep14020014

Article Metrics

Back to TopTop