Skip to main content

COVID-19 in patients with anemia and haematological malignancies: risk factors, clinical guidelines, and emerging therapeutic approaches

Abstract

Extensive research in countries with high sociodemographic indices (SDIs) to date has shown that coronavirus disease 2019 (COVID-19) may be directly associated with more severe outcomes among patients living with haematological disorders and malignancies (HDMs). Because individuals with moderate to severe immunodeficiency are likely to undergo persistent infections, shed virus particles for prolonged periods, and lack an inflammatory or abortive phase, this represents an overall risk of morbidity and mortality from COVID-19. In cases suffering from HDMs, further investigation is needed to achieve a better understanding of triviruses and a group of related variants in patients with anemia and HDMs, as well as their treatment through vaccines, drugs, and other methods. Against this background, the present study aimed to delineate the relationship between HDMs and the novel COVID-19, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Besides, effective treatment options for HDM cases were further explored to address this epidemic and its variants. Therefore, learning about how COVID-19 manifests in these patients, along with exploiting the most appropriate treatments, may lead to the development of treatment and care strategies by clinicians and researchers to help patients recover faster.

Video Abstract

Introduction

As of December 2019, the first confirmed cases of new COVID-19, SARS-CoV-2, were reported in Wuhan, China, and the situation has affected more than 200 countries [1]. The infection may cause severe illness with shortness of breath and some symptoms of chest pain, which can potentially develop into pneumonia [2]. On the other hand, at the same time as the spread of this virus worldwide, its detection of this virus through laboratory tests such as real-time reverse transcription polymerase chain reaction (rRT-PCR), chest CT scans, Immunoglobulin Rapid Diagnostic Tests (Ig-RDTs), Elisa-linked Immunosorbent Assays (ELISAs), and detecting seroconverted IgA, IgM and IgG antibodies were performed in serum or blood [3, 4].

Therefore, the respiratory system involved during SARS-CoV-2 could be associated with the dysregulated expression of some biomarkers [5]. In addition, lymphopenia is nothing special in this condition and high patients, which indicates anemia in cases of COVID-19. The ferritin test shows higher than normal levels, indicating an acute inflammatory response in patients or the entry of the virus and its effects on iron metabolism, which can reduce the bioavailability of iron, thereby depriving the virus of this element and leading to anemia [6, 7]. As of October 2021, there have been abundant reports of high mortality rates in HDM patients co-infected with COVID-19 [8]. Notably, HDMs caused by overproduction of blood cells are assumed as an abnormal phenomenon that leads to improper control of these cells in fighting infections or preventing serious bleeding [9, 10]. Such malignancies and their treatment may affect the human immune system and put them at high risk of contracting COVID-19 and suffering its consequences [11]. As evidenced in related studies, patients with HDM, above all those with acute lymphoblastic leukemia (ALL), essential thrombocytopenia (ET), multiple myeloma (MM), and chronic myeloid leukemia (CML), compared to people without such conditions, have experienced a higher chance of infection with COVID-19 [12]. Like other ribonucleic acid (RNA) viruses, CoV is constantly changing during the mutation process, producing new variants with a high risk of transmission and pathogenicity [13]. New variants can mainly escape the immune response caused by infection and even vaccination [14]. Considering this issue, dealing with this virus and its types in patients with anemia and HDM creates different scenarios and hypotheses or offers suggestions for preventive or therapeutic purposes that should be considered in the treatment and care of affected patients. Because this virus and its variants significantly affect all aspects of human life worldwide, measuring these risk factors among patients with COVID-19 is crucial to help treat the disease. On the other hand, the possibility of false results in diagnostic tests, especially rRT-PCR, has been abundantly observed. According to the research conducted on patients with blood malignancies, there is a higher probability of false negatives, which can depend on endogenous factors such as hematocrit, triglycerides, cholesterol, and other blood substances [15, 16]. Hence, it is necessary to identify the virus in these patients by more sensitive tests such as CRISPR [17]. Therefore, this review article can help young infectious disease specialists, hematologists, and physicians gain a deeper understanding of this condition and quickly look at recent events caused by HDMs.

COVID-19 in patients with anemia and haematological malignancies

A diverse range of the most common haematological manifestations, including lymphopenia, anemia, thrombocytopenia, hyperferritinemia, coagulopathy, and high D-dimer levels, can be closely related to COVID-19 [18, 19]. The pathogenesis of this condition is mostly attributed to the severe increase of pro-inflammatory markers such as interleukin-1 beta (IL-1β), IL-2, IL-4, IL-6, IL-10, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ) which cause an exaggerated immune response [20]. The inflammatory response also causes disturbances in iron metabolism, contributing to high hepcidin levels, decreased iron utilization, hyperferritinemia, and anemia [21] (Fig. 1). Such manifestations can be observed during the infection of COVID-19, which often disappear after the resolution of the infection [22]. Due to the rapid spread of the virus, it is necessary to take necessary precautions regarding the risk factors that make these patients more vulnerable to this disease.

Fig. 1
figure 1

Graphic overview of factors affecting some types of anemia and haematological disorders in the exacerbation of COVID-19

Beta-thalassemia

Beta (β)-thalassemias represent a group of inherited autosomal recessive anemias that occur due to a reduction or absence of beta globulin tetramers (β4), also called Hb-H [23]. In cases with β-thalassemia, some polymorphisms of the heme oxygenase-1 (HO-1) gene, especially repeat mutations in the dinucleotide (GT) promoter region, induce the HO-1 gene to generate reactive oxygen species (ROS), which protects the cell [24] ( Fig. 2). In patients with COVID-19, longer GT sequences are likely to be present, resulting in modulation of blood flow [25]. During β-thalassemia (mainly partial form), porphyrin deficiency is not observed, but ferritin and excess serum ferritin [26, 27] and iron have been confirmed as risk factors for the exacerbation of COVID-19 [24].

Fig. 2
figure 2

Abnormalities in haematological parameters of patients with anemia and RBC disorders

Recent studies have also shown that β-thalassemia patients may contribute to increased susceptibility to SARS-CoV-2 infection due to the nature of their chronic disease [28]. β thalassemias can be defined as an inherited disorder caused by a defect in hemoglobin synthesis, thus accelerating the continuous hemolysis and premature destruction of red blood cells (RBCs) in the bone marrow [29]. These patients are exposed to infectious diseases, especially bacterial infections, acute respiratory infections, and SARS-CoV-2. Therefore, during COVID-19, people with beta-thalassemia are threatened by SARS-CoV-2 infection, as most of them have underlying diseases such as diabetes, common heart diseases, liver problems, and various endocrine disorders [28] (Fig. 3).

Fig. 3
figure 3

Graphic illustration of the potential role of different types of anemia and blood disorders in the clinical manifestations of people with COVID-19, possibly influencing the pathophysiology of COVID-19

Sickle cell disease

Sickle cell disease (SCD) refers to a type of HDM that has recently been associated with SARS-CoV-2. As confirmed in previous research, people with swine flu (H1N1) commonly develop respiratory complications, including acute chest syndrome (ACS) [30]. Compared to other children with influenza, children with SCD are also referred to health centers much more often [31]. Consequently, such complications are likely to occur more rapidly in patients with comorbid SCD and COVID-19 [32]. Also, SCD leads to an increased chance of several diseases, such as pulmonary hypertension, chronic lung disease, and kidney failure [33]. In this vein, chronic pulmonary injury from thrombo-inflammation caused by COVID-19 exacerbates SCD complications and increases mortality rates [34]. Likewise, acute vaso-occlusive crisis (VOC) attributed to SCD in COVID-19 patients increases the likelihood of pulmonary embolism (PE) and ACS [35]. Thus, individuals with COVID-19 who are co-infected with SCD have a 3.5-fold increased risk of developing PE compared to patients without the disease. Furthermore, the preclinical and procoagulant status of patients with comorbid SCD and COVID-19 may contribute to milder clinical symptoms [36]. According to some clinical studies, patients with SCD and subsequently infected with COVID-19 do not face the risk of complications or mortality from the epidemic. Still, the hospitalization rate is higher in these people, which raises two different hypotheses [37]. First, patients with SCD have severe hemolysis with continuous release of heme, consisting of toll-like receptor 4 (TLR4) with proinflammatory and procoagulant states [38]. In addition, elevated levels of plasma cytokines, such as IL-1, IL-6, and TNF-α, have been reported in individuals with persistent SCD, and S protein up-regulated them for SARS-CoV-2 [39]. As reported in a study, COVID-19 patients with SCD (age > 50 years), presenting a growth in the serum D-dimer, creatinine (Cr), and lactate dehydrogenase (LDH) levels are at greater risk of mortality regardless of their genotype or gender [30]. Accordingly, the pathophysiology of SCD accounts for chronic anemia, endothelial dysfunction, chronic inflammation, immunodeficiency disease, and hypercoagulability, all as risk factors for the worst outcomes of COVID-19 in individuals with a hypercoagulable state and minor pathophysiology during Hypoxia is considered. As with renal modules, such patients could potentially be at increased risk for contracting COVID-19 [38]. Furthermore, people with SCD are likely to develop some neurological complications during their lifetime. Since SARS-CoV-2 adversely affects the central nervous system (CNS) and patients with SCD are completely immunocompromised, many concerns arise that require further investigation [38] (Figs. 2 and 3).

Iron-deficiency anemia

As iron is required for the growth and reproduction of various cells in the immune system, iron deficiency (ID) can impair the host immune response [40]. As an essential trace element for the host, iron is essential for many enzymatic and non-enzymatic reactions as well as various physiological processes [41]. For example, iron significantly contributes to mitochondrial function in adenosine triphosphate (ATP) production or synthesis, RNA and deoxyribonucleic acid (DNA) repair, cell survival, and ferroptosis [42, 43]. In addition, this valuable element is vital for the multiplication of viruses. On the other hand, IL-6 is a key mediator in post-inflammatory iron management, as hepcidin produces iron [44, 45]. As a key regulator of iron homeostasis, hepcidin further destroys the duodenum by damaging the cellular iron exporter, ferroportin (FPN1), which helps promote cell retention in macrophages and regulates cellular iron metabolism. Therefore, inflammation causes some changes in iron homeostasis due to its dysfunction [46, 47]. This deficiency is often compensated by high levels of iron in the reticuloendothelial cells and ultimately by hyperferritinemia, while low levels of iron are present in the bloodstream [48]. Subsequently, inflammation limits iron in RBCs, leading to anemia known as anemia of inflammation (AI), which is commonly seen in pregnant women with decreased red blood cell quantity and quality along with increased erythrocyte sedimentation rate (ESR). It is related to the gas exchange that occurs during the reduction of RBCs [49, 50] (Fig. 2). This can be caused by a deficiency of folate and other B vitamins. Therefore, pregnancy when infected with COVID-19, especially in IDA, makes this viral infection more visible in the third trimester because inflammatory processes occur much more often. As reported in some studies, many viruses, including SARS-CoV-2, disrupt iron homeostasis in cells, caused by hemolysis, and then enhance intercellular iron load, which steps it accelerates the multiplication of the virus and ultimately increases the severity of the disease [51, 52]. In any case, this iron overload increases serum ferritin and is further associated with rheumatoid arthritis (RA), multiple sclerosis (MS), antiphospholipid syndrome (APS), macrophage activation syndrome (MAS), adult-onset steel disease (AOSD), catastrophic APS (cAPS), and then septic shock [53, 54] (Fig. 3).

Aplastic anemia

Aplastic anemia (AA), also known as rare HDM, is characterized by central pancytopenia due to bone marrow failure [55]. Although the pathogenesis of this type of anemia is still unclear, it is hypothesized to result from the destruction of hematopoietic stem cells (HPSCs) secondary to an aberrant immune response [56]. More than 50% of AA cases are also idiopathic in nature [55]. Despite this, chemotherapy (chemo), ionizing radiation, and viral infections also contribute to this disease [57]. Accordingly, the most common infectious agents include viral hepatitis, human immunodeficiency virus (HIV) [58], cytomegalovirus (CMV) [59], parvovirus B19 (PVB19) [60], and Epstein-Barr virus (EBV) [59]. In this regard, SARS-CoV-2 mainly affects the pulmonary system, which in rarer cases leads to central neutropenia, lymphopenia, and pancytopenia and disrupts the hematopoietic system [61]. Moreover, the overproduction of inflammatory cytokines in infectious viruses, such as IL-1ß, IL-6, TNF-α, and IFN-γ [62], disrupts the bone marrow microenvironment and subsequently causes bone marrow failure [63, 64] (Fig. 3).

Diamond-blackfan anemia

Diamond-blackfan anemia (DBA) mainly affects the bone marrow and causes some physical abnormalities in many parts of the body. During DBA, the bone marrow is normally disrupted, resulting in reduced red blood cells to supply oxygen to the tissues [65, 66]. Actually, changes in Hb levels are predictive of worsening clinical progression in patients with COVID-19, as the bone marrow is unable to make RBCs. SARS-CoV-2 can attack the β-1 chain of Hb and detach it from iron to form perforin. Therefore, often less Hb is available to carry oxygen and carbon dioxide (CO2). Here, binding of the virus to Hb and subsequent release of ions produces free radicals that increase oxidative stress (OS) in organs and lead to hypoxia. Each Hb molecule also contains four hemes during chemical interactions, each of which binds precisely to oxygen in the lungs [67]. In addition, iron (II) and (III) ions (Fe2+ or Fe3+), as part of the toxic structure of oxyhemoglobin in a free state, augment OS in the blood. If SARS-CoV-2 binds to Hb Fe2+ and Fe3+, it may be released into blood and tissues, thus determining the main effects of the virus. In this case, the function of Hb is disturbed, the oxygen supply decreases and finally, hypoxia increases. As a result, shortness of breath and fatigue may persist even after recovery in some patients with COVID-19 [68, 69] (Fig. 3).

Hereditary spherocytosis

In patients with hereditary spherocytosis (HS), cellular stress combined with splenic clearance multiplies the chance of hemolysis [70]. Such individuals may live with chronic baseline hemolysis, sometimes requiring splenectomy to treat severe chronic anemia, or there may be intermittent major hemolysis and splenomegaly [71, 72]. Since the spleen is the site of RBC clearance in HS patients, splenectomy is often advocated as a treatment option [73]. Nonetheless, this surgical procedure does not resolve the defects in the function of the erythrocyte membrane and exposes patients to severe cellular stress and a higher chance of hemolysis [74]. Accordingly, many hemolytic markers can be considered during this emergency. For example, bilirubin (BLR) levels are a significant indicator, the increase of which can be attributed to the breakdown of the protoporphyrin IX (PPIX) ring [70]. Likewise, ferritin is another hemolytic marker, as an acute phase reactant (APR), which is increased in patients with severe COVID-19 with cytokine storm (CS) [75]. Furthermore, LDH levels in severely infected individuals with COVID-19 are compounded due to increased cytokine activity, and decreased monitoring of Hb and hemolytic markers in cases with hemolytic disorders and COVID-19 [76]. Notably, HS can have varying degrees of hemolysis and may be the first hemolytic event at the onset of COVID-19 infection [70].

Leukoerythroblastic reaction

During the leukoerythroblastic reaction (LER), immature RBCs and myeloid cells often circulate in the peripheral blood [77, 78]. This reaction is commonly reported in some disorders related to bone marrow fibrosis, including myeloproliferative disorders (MPDs) and cancer types associated with bone marrow metastatic problems [79]. LER has been identified mostly in viral infections, such as polycythemia vera (PV) and COVID-19. In severe cases, SARS-CoV-2 infection is associated with overproduction of proinflammatory cytokines such as IL-2, IL-6, IL-7, IL-8, IFN-γ, TNF-α, transforming growth factor-beta (TGF-β), C-X-C motif chemokine ligand 8 (CXCL8), CXCL10, chemokine ligand 3 (CCL3), macrophage inflammatory protein-1 alpha (MIP-1α), and -1β, known as CS. In patients infected with SARS-CoV-2, this condition results in LER, with increased production and the presence of immature myeloid cells in the circulatory system [80]. Leukoerythroblastosis can occur in children with Kawasaki disease. The exact etiology of Kawasaki disease is unknown, although an infectious agent appears to be the source of its initiation [81]. Hypersensitivity reactions or inappropriate immune responses, possibly caused by viruses or bacteria, can trigger an inflammatory process that damages blood vessels in people who are genetically predisposed to the disease. Notably, KD and COVID-19 are very similar in this respect [82,83,84] (Fig. 3).

Hemophagocytic lymphocytosis

Hemophagocytic lymphocytosis (HLH), introduced as a less common symptom in viral proinflammatory conditions, has a high consequence in most patients with COVID-19 [85]. It is an ambiguous clinical condition, followed by immune-mediated tissue damage, which occurs irregularly due to viral infections or HDM. This phenomenon may not be observed in patients with COVID-19, where phagocytosis is also observed in bone marrow aspirates, cytopenias are present, and serum ferritin elevations below ≥ 2000 ng/mL occur [86]. Also, MAS is a life-threatening proinflammatory syndrome that is likely to appear in patients with severe viral infections, such as those with EBV. There is also an interesting pathophysiological similarity between EBV infection and COVID-19 in the case of MAS. During both infections, uncontrolled and hyperactive macrophages cause hypercytokinemia, organ damage, cytopenias, and coagulopathy [85]. Besides, there are several associations between descriptions of severe forms of COVID-19 infection and secondary HLH (sHLH). For example, elevated serum ferritin and C-reactive protein (CRP) levels are commonly observed in patients with severe COVID-19 and sHLH [87]. Moreover, people with severe COVID-19 develop many complications that resemble multi-organ failure (MOF) in HLH [88]. Since COVID-19 has the same pathogenesis compared to sHLH, early diagnosis, and rapid immunosuppression before MOF are often of particular importance [89]. Therefore, all patients with severe COVID-19 should be screened with standard laboratory tests, such as HScore to detect severe inflammation [90].

Sideroblastic anemia

Sideroblastic anemia (SA) encompasses a group of inherited and acquired anemias characterized by ineffective erythropoiesis. In this type of anemia, there is an accumulation of ring sideroblasts (RS) in the bone marrow and a decrease in the production of fully developed red blood cells. These ring sideroblasts are nucleated erythroblasts that show abnormal accumulation of iron granules in the mitochondrial matrix [91]. Such mechanisms contribute to the formation of iron-rich mitochondrial complexes around erythroblast nuclei instead of the standard incorporation of iron into PPIX in the mitochondria [92]. It has then again been hypothesized that COVID-19 may induce an immune-mediated genetic defect in a hematopoietic clone, resulting in ineffective erythropoiesis and the development of RS cells [93]. Accordingly, COVID-19 likely induces a genetic change in new genes that cause SA [93]. In addition, SARS-CoV-2 interacts with Hb molecules through a cluster of differentiation 147 (CD147), CD2b, and other receptors commonly found on erythrocytes and other Hb cells, leading to Hb denaturation [94]. Considering that, hemoglobin concentration decreases and toxic heme is released, which usually causes hypoxia [94]. Furthermore, a gradual decrease in the Hb concentration may promote SA and increase erythrocyte distribution width (RDW), indicating overproduction of immature erythrocytes and an increased risk of mortality [95(Fig. 2).

Megaloblastic anemia

In some cases, megaloblastic anemia (MA), impaired nerve myelin sheath integrity, impaired immune response, neurological complications, and degenerative conditions of the spine can be caused by some effects of low cobalamin levels [96,97,98]. Following these conditions, symptoms of vitamin B12 deficiency, including elevated OS and LDH, intravascular coagulation and thrombosis, hyperhomocysteinemia, coagulation cascade, subnormal reticulocyte count, vasoconstriction, and renal failure may often accompany COVID-19 [99, 100]. As suggested, high doses of methylcobalamin could potentiate the RNA-dependent RNA polymerase (RdRp) activity of SARS-CoV-2 nonstructural protein 12 (NSP12) enzymes, which then reduces the viral infection and severity of COVID-19. Overall, methylcobalamin helps reduce the severity of COVID-19 [101]. Vitamin B12 deficiency mostly causes two conditions. Sometimes, parietal cells can't make enough vitamin B12 because people don't have a diet rich in this vitamin. In this case, the megaloblast normally forms in the cells and becomes asynchronous when the nucleus and cytoplasm are mixed, also called MA [102]. Accordingly, those suffering from MA do not have enough red blood cells to carry oxygen properly. A second scenario is that vitamin B12 is produced by the parietal cells because a person receives an adequate vitamin B12-rich diet but has difficulty absorbing it [103]. Bacteria in the large intestine also mutate so they cannot absorb vitamin B12 or have challenges in the ion channels that absorb vitamin B12 [104]. To absorb vitamin B12, folate is needed in a trivalent form, and this dihydrofolate is converted from tetrahydrofolate by these colon bacteria with the help of the dihydrofolate reductase (DHFR) enzyme. Therefore, mutations frequently occur that produce this enzyme and lead to pernicious anemia [105]. In an interesting study, vitamin B12 was identified as one of the viral proteins of SARS-CoV-2, so it can easily bind to it and reduce its effects [106]. Therefore, it is necessary to maintain its level. Furthermore, SARS-CoV-2 may interact with the metabolic activities of vitamin B12 and possibly shape the microbiological distribution in the intestine. It occurs if symptoms such as vasoconstriction, increased OS, coagulation cascade, high LDH levels, pulmonary-renal syndrome (PRS), and hyperhomocysteinemia are present. In addition, B12 deficiency can lead to some abnormalities in the CNS, gastrointestinal (GI), and respiratory systems [107]. Accordingly, a recent study has shown that extra doses of methylcobalamin may help minimize organ damage and even some symptoms associated with COVID-19. For example, a study in Singapore showed a significant reduction in existing symptoms of severe COVID-19 in patients taking magnesium, vitamin D (1000 IU), and vitamin B12 (500 μg) supplements [108]. Prenatal pancytopenia is also a rare manifestation, causing anemia, leukopenia, and thrombocytopenia with a simultaneous decline in all blood cell lineages. Vitamin B12 and folate deficiency generally present as MA, but rare manifestations of pancytopenia have been reported so far. The prevalence of vitamin B12 and folate deficiency during pregnancy is currently significantly high in developing countries due to their poor socioeconomic status and nutrition. Hemodilution with interplacental transfer of vitamin B12 further contributes to the physiological reduction of vitamin B12 levels. In addition, pancytopenia is a rare manifestation of some viral infections, including the novel COVID-19 [109].

Autoimmune and inflammatory haematological complications and COVID-19

As confirmed in related studies, COVID-19 is associated with some autoimmune diseases, including autoimmune cytopenias, cutaneous vasculitis, encephalitis, and Guillain–Barre syndrome (GBS). Among them, autoimmune hemolytic anemia (AIHA) and immune thrombocytopenic purpura (ITP) are the most common [110] (Fig. 2).

Autoimmune hemolytic anemia

In confirmed cases of COVID-19, AIHA or its reactivation has been reported so far, which can be attributed to severe anemia or rituximab treatment. During SARS-CoV-2 infection, anemia associated with elevated LDH and other hemolytic markers may be more frequently observed, and even if the anemia appears unexplained and discontinuous, AIHA is also suspected [111]. Also, molecular mimicry could be the highest factor in the development of AIHA caused by SARS-CoV-2. Immunological cross-reactivity between ankyrin 1 (ANK-1), an RBC membrane protein, and spike proteins in a virus has been implicated in the pathogenesis of AIHA among patients with COVID-19 [112]. Some researchers also believe that the induction of AIHA in children with HLH is due to OS stimulation by SARS-CoV-2. In addition, the acute phase response of COVID-19 induces the formation of aberrant complement immune complexes and complement products on the RBC surface, leading to intravascular thrombosis [113]. This could be consistent with disseminated intravascular coagulation (DIC) with MOF induced by AIHA in COVID-19 patients. Concomitantly, hypercoagulability and inflammatory responses are exacerbated and may affect red blood cells, rupture their membranes, and in such cases lead to PE and vascular coagulation. In this regard, iron and ferritin caused by hemolysis lead to OS. Accordingly, hyperferritinemia and impaired iron homeostasis contribute to endothelial damage and structural changes in red blood cells in cases of COVID-19 [113]. Besides, there are reports of AIHA in patients receiving the vaccination against this disease, particularly with influenza and diphtheria-tetanus-pertussis (DTP) vaccines, due to the induction of warm and cold anti-RBC antibodies [114]. Therefore, vaccines as infectious agents can cause HDMs by molecular mimicry, lymphocyte polyclonal activation, epitope release, and presentation of cryptic antigens [115]. On the other hand, the use of some vaccines cannot protect people with anemia and HDM against SARS-CoV-2 and lead to haematological complications (Table 1).

Table 1 Summary of studies showing haematological complications of some post-injection COVID-19 vaccines

Idiopathic thrombocytopenic purpura

Depending on viruses and immune and environmental factors, idiopathic thrombocytopenic purpura (ITP) refers to a disease with isolated thrombocytopenia and platelets less than 100 × 109/L, the causes of which are still unknown. Accordingly, autoantibodies reduce platelet synthesis, antibodies against platelet membrane antigens, increase platelet secretion, and prolong life, while platelet production in the bone marrow is reduced due to thrombocytopenia [122]. Acute ITP is usually initiated by a viral infection, and platelet levels usually improve independently after a few weeks or months. Of note, acute ITP lasts more than a year if thrombocytopenia persists (Fig. 2). The most potential viruses as triggers are cytomegalovirus, hepatitis C virus (HCV), herpes simplex virus (HSV), varicella-zoster virus (VZV), rubella virus, EBV, influenza virus, HIV, and SARS-CoV. Furthermore, molecular mimicry between virus-specific antibodies and host proteins may cause virus-mediated ITP [123,124,125]. The main cause of increased mortality in SARS-CoV-2 cases is thrombocytopenia, which can be caused by DIC, thrombosis, septicemia, or drugs. ITP most often occurs during and up to ten days after a COVID-19 infection. In this regard, antibodies directed against viral glycoproteins can interact with platelet surface integrins, such as glycoprotein IIb/IIIa (GP IIb/IIIa) or GPIb-IX-V, which accounts for approximately 5–10% of cases of ITP caused by SARS-CoV-2 [126]. Therefore, patients with COVID-19 and ITP can present with increased thrombocytopenia and excessive bleeding, mainly in the second stage of the disease [127]. According to this, ITP thrombocytopenia is one of the mechanisms that is at the top of the decrease in the number of platelets in patients with COVID-19 [115]. Until now, this phenomenon has been explained for some reasons, especially virus-induced autoimmunity. Therefore, molecular mimicry along with the expression of cryptic antigens or the release of epitopes can clarify this immune disorder. In most cases, ITP can appear two to three weeks after COVID-19 infection and even before vaccination [128] (Fig. 3). On the other hand, it is noteworthy that patients with previous SARS-CoV-2 infection may have excessive procoagulation factors that can lead to thrombosis and thrombocytopenia. However, currently its pathophysiology is unknown. Experimental studies currently show that a type of soluble adenoviral spike protein leads to the formation of thrombosis, which ultimately results from graft events creates significant endothelial inflammatory events, and leads to binding with endothelial cells expressing ACE2 [129].

Haematological malignancies and COVID-19

Patients living with HDM are at increased risk of contracting COVID-19 compared to patients without such symptoms [130]. Such malignancies can affect the production and function of blood cells to fight viral infections [131]. For example, HDM cases often have multiple abnormalities in the innate and adaptive immune system, including low levels of immunoglobulin G (IgG) in patients with chronic lymphocytic leukemia (CLL) or other B-cell neoplasms, as well as immature or neoplastic dysfunctions [132, 133]. Therefore, such immune disorders could make people with HDM susceptible to COVID-19 [134] (Figs. 4 (some are hypotheses and are listed in Tables 4 and 5) and 5 (hypothesis)).

Fig. 4
figure 4

Graphical overview of the effectiveness of different treatments on the mechanisms of patients with haematological malignancies and severe COVID-19. Created with BioRender.com

Fig. 5
figure 5

Plausible host miRNA action modes in SARS-CoV-2 infection. Host miRNAs may regulate COVID-19 infection in patients with haematological malignancies. Created with BioRender.com

Chronic lymphocytic leukemia

As a malignancy, chronic lymphocytic leukemia (CLL) is characterized by an increase in monoclonal CD5+ B lymphocytes, leading to intrinsic and extrinsic triggering events [135]. For example, the function of various elements in the immune system during viral infections can determine the onset of CLL [136]. Certain factors such as high levels of markers of immune activation such as IL-4, IL-10, and TNF-α, or cytokine release syndrome (CRS) in patients with COVID-19 and high levels of granulocyte colony-stimulating factor (G-CSF), IL-6, IL-7, IL-8, IL-10, IL-1Rα, IFN-γ, TNF-α, granulocyte–macrophage (GM)-CSF, and monocyte chemoattractant protein-1 (MCP-1) are of great importance in this malignancy. Such cytokines can lead to a rapid increase in the clonal expansion of lymphocytes in COVID-19 patients, potentially increasing the chance of malignancy (Fig. 4) [137]. After cancer patients were infected with SARS-CoV-2, CS could effectively induce a severe form of the disease. In this respect, if the patient is healthy enough, the CS will end and the cancer treatment process will not be interrupted [138]. Therefore, activated signaling pathways may negatively affect the therapeutic response and survival rate in cancer patients just at the beginning and before the end of CS. Accordingly, early detection of CS in such patients (such as patients with CLL) with COVID-19 is critical to multiply the effectiveness of targeted therapy [139, 140]. During acute inflammation, this condition may be caused by high endogenous hormone levels, but additional processes may be beneficial that require further investigation [140]. Moreover, it is not known whether the growth of lymphocyte count is a prognostic marker in patients with severe type of COVID-19 and untreated CLL. Furthermore, similar results are unavailable for CLL subjects who have never been treated. For this reason, treatment for people with COVID-19 and CLL poses great challenges [141]. It also gives the impression that the immune system is ineffective in CLL patients and that lymphocytes do not respond strongly to viral infection. Accordingly, such an agent is likely to help protect these patients against CRS and its subsequent damage and MOF [142]. However, chemotherapy in CLL and COVID-19 cases remains controversial, as it may increase the risk of cardiotoxicity, SARS-CoV-2-induced immunodeficiency, and prognosis [143]. To minimize treatment-induced immunodeficiency and drug interactions, it is therefore best to avoid chemotherapy in patients with comorbid CLL and COVID-19 [144, 145].

Acute lymphocytic leukemia

Among the most common types of cancer recognized as the leading cause of death in young adults is acute lymphocytic leukemia (ALL) [146]. Thus, disruption of transcription factors that contribute to direct lymphocyte growth [147, 148], abnormal activation of key signaling pathways, and loss of tumor suppressor genes required for cell cycle regulation [149] are commonly associated with ALL pathogenesis. Furthermore, this condition is often implicated in gene mutations that provide epigenetic regulatory codes [149]. Notably, most cases of ALL and COVID-19 infection have so far not been reported during the pandemic, and the disease progresses slowly in ALL patients with or without clinical symptoms. Therefore, systemic therapy should be delayed in SARS-CoV-2-positive patients (e.g., following the absence of primary hyperleukocytosis). Then, some symptoms such as dry cough, high temperature, anosmia and gastrointestinal problems should be carefully evaluated. If a diagnostic test for SARS-CoV-2 is not possible, a CT scan of the chest should be performed. Furthermore, serological tests should be performed on all patients as soon as they are available [150]. In this line, some studies have further emphasized the abnormally expressed micro (mi)RNAs in ALL patients, as they seem to play a central role in controlling carcinogenesis and drug resistance [151]. Therefore, the etiopathogenesis of HDMs is related to many members of this family, namely miRNA-181a and -181b. Since the expression of miRNA-181a and -181b is much higher in ALL patients than in healthy patients, these findings raise the possibility of using miRNA-181a and miRNA-181b as biomarkers [152]. Researchers have similarly found that all patients living with COVID-19 showed significantly higher levels of miRNA-181a expression, indicating the pathogenic function and prognostic significance of miRNA-181 in patients with comorbid ALL and COVID-19 [153] (Fig. 5 and Table 2).

Table 2 COVID-19-associated miRNAs involved in immune response

Chronic myeloid leukemia

Chronic myeloid leukemia (CML) is usually initiated by BCR-ABL1 as a hybrid gene in cells with innate or acquired biological potential [177]. This type of cancer can initiate complications in patients with COVID-19 [178]. Furthermore, drug-drug interactions between tyrosine kinase inhibitors (TKIs) for the treatment of this malignancy and those targeting COVID-19 infection may be very hazardous [179]. Also, the side effects of TKI are unbearable for patients with SARS-CoV-2. TKI is often used as an initial treatment for patients with CML and has resulted in a good prognosis and significant improvement [180]. Therefore, treatment with TKIs in cases with CML, which shows a slight increase in the risk of infection, may be due to off-target inhibition of immune-related kinases [181]. Therefore, the decision to withhold or continue TKI-based treatment during the period of COVID-19 seems challenging and needs further investigation. Some studies have also concluded that TKIs help control the immune response to infection [181]. Anti-immune genes such as CD28, CCL55, and IFN-γ and low expression of some such as arginase 1 (ARG1) and fucosyltransferase 4 (FUT4) have been observed so far [182]. Overall, the mortality rate of COVID-19 in Latin American patients with CML has been higher than in the general population. Accordingly, this type of leukemia can cause problems during SARS-CoV-2. Drug interactions between TKIs and COVID-19 treatments can be more dangerous and require careful monitoring [183].

Acute myeloid leukemia

Acute myeloid leukemia (AML), as one of the most common HDMs, can have adverse effects on blood, bone marrow, and other tissues [184]. This condition is characterized by abnormal proliferation or differentiation of clonal cells and a weakened immune system [185]. Currently, several effective treatments are available for AML, especially for young adults [185]. Therefore, infections, including viral infections, can be a major complication of AML treatment, as in other HDMs. Treatment regimens used for patients with AML can also lead to severe granulocytopenia and an increased risk of serious infections. Therefore, a person with AML and SARS-CoV-2 is at high risk of respiratory failure, which requires a reduction in drug dosage and the fact that antiviral drugs [186].

Besides, it is hypothesized that AML patients with COVID-19 undergo a more severe form of the disease. Some even expire due to these conditions, experience significant progression to recurrence of the malignancy, or become resistant to therapeutic drugs, especially if they harbor FMS-like intronic tyrosine kinase-3 tandem mutations (FLT3-ITD). Based on the theory developed by Zalpoor et al. the pharmacological targeting of autophagy and hypoxia-inducible factor 1 alpha (HIF-1α) may be a potential treatment for FLT3-ITD mutations with COVID-19 and risk of mortality, development of HDMs, and drug resistance [187]. In a remarkable study, Deeb et al. established that cytoplasmic expression of HIF-1α was associated with poor prognosis following conventional therapy in older AML patients with normal karyotype [188]. Therefore, they suggested that stimulation of autophagy and HIF-1α by COVID-19 may be a marker for AML patients, especially those with FLT3-ITD mutations. They also hypothesized that autophagy associated with COVID-19, FLT3-ITD, and overexpression of HIF-1α may cause leukemia and drug resistance in these patients. It probably increases the severity of COVID-19 [188]. However, more studies are still needed to support it. Furthermore, autophagy-related drugs have recently been proposed as potential SARS-CoV-2 treatments based on some in vitro and in vivo studies [189]. Accordingly, it has been hypothesized that autophagy induced by COVID-19 may contribute to cancer growth, chemotherapy resistance, and tumor recurrence in patients. These data also suggest that COVID-19 can induce autophagy due to various factors [190]. In addition to being an antiviral therapeutic strategy, targeting autophagy may be a viable option for treating cancer patients with COVID-19 to reduce the risk of mortality, progression, chemotherapy resistance, and tumor recurrence in a variety of cancers [191] (Fig. 4).

Multiple myeloma

Another type of HDM is multiple myeloma (MM), which affects plasma cells in the bone marrow [192]. In cases with MM, the immune system is often compromised by various factors making people with this malignancy susceptible to infection [193]. People with a mean age of 65 years have more underlying diseases, so they are at risk of infection [194]. CD4 depletion, lymphopenia, and loss of functional immunoglobulins can increase the chance of viral, bacterial, and fungal infections [195]. Thus, immunosuppressive drugs advocated in this regard can lead to neutropenia, thereby increasing the risk of contracting COVID-19, as the virus exacerbates the cause of abnormally low concentrations of neutrophils in the blood [196]. Notably, new apheresis testing using autologous stem cell transplantation (ASCT) and polymerase chain reaction (PCR) is required before hospitalization in epidemic-affected countries [197]. While living with this anemia, these patients receive treatments that cause some changes in immune system function, such as humoral immunodeficiency, hypogammaglobulinemia, and impaired B-lymphocyte response to SARS-COV-2. Management of MM in the era of COVID-19 accordingly calls for a thorough assessment of patient- and disease-related variables in order to reduce the risk of developing MM through effective treatment [198].

Myeloproliferative neoplasm

In myeloproliferative neoplasm (MPN), platelets, RBCs, and leukocytes are continuously activated from clonal progenitor cells to hematopoietic cells [199]. PV, ET, and myelofibrosis are also among the leading active neoplasms that can affect mortality [200]. In ET, it is often associated with a persistent increase in the number of platelets that have a propensity for thrombosis, bleeding, and activation of inflammatory mechanisms [201]. In patients with COVID-19 and this malignancy, the lungs may be involved first and then adverse effects on different organs may be observed. In various reports, thrombosis has been presented with some complications of COVID-19 [202]. Therefore, virus-induced thrombosis is a very important genetic thrombosis mechanism in this disease. Accordingly, patients with MPN and COVID-19 are more prone to thrombotic complications and higher mortality [203].

Hodgkin’s lymphoma

Known as a curable malignancy, Hodgkin’s lymphoma (HL) is probably associated with EBV [204]. First, COVID-19 infection may play a significant role in the transient improvement of HL [205]. Decreased peripheral blood lymphocytes (PBLs) and natural killer (NK) cells can be observed in COVID-19 patients [206, 207]. As well, the total number of lymphocytes (here, the CD4+ and CD8+ cells) decreases in severe forms of the disease, more than in mild cases [208]. Second, inflammatory microenvironments may minimize the effective function of NK cells. The high levels of IL-6 and IL-10 in these patients therefore add to the capacity to reduce the cytotoxic process and increase the expression of NKG2A in killing virus-infected cells [205]. In these patients, it also binds to angiotensin-converting enzyme 2 (ACE2) in NK cells and suppresses their function [205]. SARS-CoV-2 and subsequent immune cell inflammatory responses inhibit NK cell cytotoxicity, induce CRS, and amplify inadequate immune responses [209]. Third, individuals with EBV-positive HL is that the NSP10/NSP7/3CLpro/major protease (Mpro) and SARS-CoV-2 S proteins bind to the tumor necrosis factor receptor type 1-associated death domain protein (TRADD) at the binding site of latent membrane protein 1 (LMP1), blocking LMP1 binds to TRADD. This interaction may therefore inhibit LMP1-mediated nuclear factor kappa B (NF-κB) signaling to induce remission [206] (Figs. 4 and 5).

Non- Hodgkin's lymphoma

The most common type of cancer in HIV-infected individuals is non-Hodgkin's lymphoma (NHL), with an increased incidence of B-cell aggressive NHL [210]. Factors affecting the emergence and development of NHL include HIV infection with high viremia, the presence of EBV, and possibly SARS-CoV-2 infection [210]. Recent data also suggest that patients with HDM, including patients with B-cell NHL, are at high risk of severe COVID-19 and may act as a persistent viral reservoir that gives rise to new and potentially more aggressive mutations. Therefore, prevention of COVID-19 or at least modulating its severity in these patients is of great importance [211]. Moreover, patients with B-cell NHL have lower rates of seroconversion and antibody levels compared to other subjects with HDM. Patients with B-cell NHL are also at increased risk of complications and mortality from SARS-CoV-2 [212]. Vaccination against SARS-CoV-2 reduces COVID-19-related deaths and hospitalizations. However, NHL cases experience suboptimal antibody responses to COVID-19 vaccines before and after B-cell-targeted therapies, such as the rituximab anti-CD20 antibody therapy [213] (Fig. 4).

Overview of therapeutic candidates for COVID- 19 infection and related variants

The current pandemic of SARS-CoV-2 and COVID-19 has so far resulted in high rates of mortality and morbidity worldwide. Hematology societies are therefore suggested to conduct prospective and multicenter studies to clarify the effects of this virus and even measure disease severity in patients with anemia and HDMs [214]. In this regard, various blood markers that act as prognostic markers in the severity of the disease have been investigated in previous researches. In this case, patients with HDM are at a higher risk of contracting various infections, including SARS-CoV-2 [215] (Table 3).

Table 3 Summary of studies evaluating the impacts of COVID-19 in patients with anemia and haematological malignancies

On the other hand, this study is very important in the management of patients with blood cancers in the face of SARS-COV-2 and its variants. It also emphasizes the priority of these patients in receiving vaccines and many other treatments. Also, various vaccines and treatment methods SARS-COV-2 and its variants have been considered in different patients (Tables 4 and 5).

Table 4 Potential therapeutic candidates for immunomodulation in COVID-19 infection
Table 5 SARS-CoV-2 variants in patients with haematological malignancies: summary of received treatments and hypothesis for haematological malignancies

Conclusion and future directions

In conclusion, COVID-19 has presented unique and significant challenges for patients with anemia and hematological malignancies, who face a higher risk of severe illness and mortality. This review article has examined the risk factors, clinical guidelines, and emerging therapeutic approaches for managing COVID-19 in this patient population. While much progress has been made in understanding COVID-19 in this context, there are still many areas that require further research.

Prospective type comparative studies using different vaccines, drugs, or combinations against SARS-CoV-2 and its multiple variants in HDM cases are necessary to discover the best options for this specific scenario. Considering the effects of anemia and HDMs on the quality of human life, this issue cannot be ignored, especially during the COVID-19 pandemic, and even because of the high costs, side effects, and shortage of blood. It is hoped that knowing the types of anemia and HDM in the face of this virus, as well as the vaccines and drugs used for the virus itself and related types, will help reduce the clinical burden of COVID-19 and its variants in terms of treatment and care.

For future directions, researchers must focus on uncovering the long-term effects of COVID-19 on patients with anemia and hematological malignancies. Specifically, understanding the cellular and molecular mechanisms of SARS-CoV-2 in potential long-term effects such as chemotherapy resistance, metastasis, and recurrence can open new avenues for developing therapeutic and preventive strategies. In addition, future studies should evaluate the efficacy and potential side effects of vaccination and emerging therapeutic approaches, with a focus on developing new vaccines and drugs that are more suited to the clinical, cellular, and molecular conditions of these diseases to improve efficacy and reduce side effects.

This study provides a potent foundation for preparing for future outbreaks of newly emerged coronaviruses. By examining the risk factors, clinical guidelines, and emerging therapeutic approaches for managing COVID-19 in this patient population, we can gain valuable insights into the challenges of managing patients with underlying health conditions during a possible upcoming new pandemic. This information can be used to guide the development of clinical guidelines and protocols for managing patients with newly emerged coronaviruses in the future, as well as to inform the development of therapeutic approaches and vaccination strategies. Overall, this review article on COVID-19 in patients with anemia and hematological malignancies is an important tool in preparing for and managing future outbreaks of newly emerged coronaviruses.

Availability of data and materials

Not applicable.

Abbreviations

ACS:

Acute chest syndrome

ALL:

Acute lymphoid leukemia

AML:

Acute myeloid leukemia

AI:

Anemia of inflammation

APS:

Antiphospholipid syndrome

AA:

Aplastic anemia

AIHA:

Autoimmune hemolytic anemia

CNS:

Central nervous system

CAR:

Chimeric antigen receptor

CLL:

Chronic lymphocytic leukemia

CML:

Chronic myeloid leukemia

CSF:

Colony-stimulating factor

COVID-19:

Coronavirus disease 2019

CRS:

Cytokine release syndrome

CS:

Cytokine storm

DBA:

Diamond-blackfan anemia

DIC:

Disseminated intravascular coagulation

EBV:

Epstein-Barr virus

ET:

Essential thrombocytopenia

FLT3-ITD:

FMS-like tyrosine kinase-3 internal tandem duplication

GI:

Gastrointestinal

HDMs:

Haematological disorders and malignancies

Hb:

Hemoglobin

HLH:

Hemophagocytic lymphocytosis

HS:

Hereditary spherocytosis

HL:

Hodgkin’s lymphoma

hATG:

Horse antithymocyte globulin

HIV:

Human immunodeficiency virus

HIF-1α:

Hypoxia-inducible factor 1 alpha

IFN-γ:

Interferon gamma

IL-1β:

Interleukin-1 beta

IgG:

Immunoglobulin G

ITP:

Immune thrombocytopenic purpura

IL:

Interleukin

KD:

Kawasaki disease

LER:

Leukoerythroblastic reaction

MAS:

Macrophage activation syndrome

MCL:

Mantle cell lymphoma

MAbs:

Monoclonal antibodies

MM:

Multiple myeloma

MOF:

Multiple-organ failure

MPN:

Myeloproliferative neoplasm

NK:

Natural killer

NHL:

Non-hodgkin’s lymphoma

ORF:

Open reading frames

OS:

Oxidative stress

PV:

Polycythemia vera

PPIX:

Protoporphyrin IX

ROS:

Reactive oxygen species

RBCs:

Red blood cells

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus 2

SCD:

Sickle cell disease

SLL:

Small lymphocytic lymphoma

SDIs:

Sociodemographic indices

TNF-α:

Tumor necrosis factor alpha

TKIs:

Tyrosine kinase inhibitors

References

  1. Costa BA, da Luz KV, Campos SEV, Lopes GS, Leitão JP, Duarte FB (2022) Can SARS-CoV-2 induce hematologic malignancies in predisposed individuals? A case series and review of the literature. Hematol Transfus Cell Ther 44:26–31

    Article  PubMed  PubMed Central  Google Scholar 

  2. Aggarwal A, Shrivastava A, Kumar A, Ali A (2020) Clinical and epidemiological features of SARS-CoV-2 patients in SARI ward of a tertiary care centre in New Delhi. J Assoc Physicians India. 68:19–26

    PubMed  Google Scholar 

  3. GenScript Rare Codon Analysis Tool, http://www.genscript.com/cgi-bin/tools/rare_codon_analysis. 2014.

  4. Zhai P, Ding Y, Wu X, Long J, Zhong Y, Li Y (2020) The epidemiology, diagnosis and treatment of COVID-19. Int J Antimicrob Agents 55:105955

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zhang L, Guo H (2020) Biomarkers of COVID-19 and technologies to combat SARS-CoV-2. Adv Biomark Sci Technol 2:1–23

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  6. Algassim AA, Elghazaly AA, Alnahdi AS, Mohammed-Rahim OM, Alanazi AG, Aldhuwayhi NA, Alanazi MM, Almutairi MF, Aldeailej IM, Kamli NA (2021) Prognostic significance of hemoglobin level and autoimmune hemolytic anemia in SARS-CoV-2 infection. Ann Hematol 100:37–43

    Article  CAS  PubMed  Google Scholar 

  7. Taneri PE, Gómez-Ochoa SA, Llanaj E, Raguindin PF, Rojas LZ, Roa-Díaz ZM, Salvador D, Groothof D, Minder B, Kopp-Heim D (2020) Anemia and iron metabolism in COVID-19: a systematic review and meta-analysis. Eur J Epidemiol 35:763–773

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Taenaka R, Obara T, Kohno K, Aoki K, Ogawa R (2022) Infections with the SARS-CoV-2 Omicron variant show a similar outcome as infections with the previous variants in patients with hematologic malignancies. Ann Hematol 101:1877–1878

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Miller MA, Zachary JF. Mechanisms and morphology of cellular injury, adaptation, and death. Pathol Basis Vet Dis. 2017;2–43.e19.

  10. Habib HM, Ibrahim S, Zaim A, Ibrahim WH (2021) The role of iron in the pathogenesis of COVID-19 and possible treatment with lactoferrin and other iron chelators. Biomed Pharmacother 136:111228

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vivarelli S, Falzone L, Grillo CM, Scandurra G, Torino F, Libra M (2020) Cancer management during COVID-19 pandemic: is immune checkpoint inhibitors-based immunotherapy harmful or beneficial? Cancers 12:2237

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang Q, Berger NA, Xu R (2021) When hematologic malignancies meet COVID-19 in the United States: infections, death and disparities. Blood Rev 47:100775

    Article  CAS  PubMed  Google Scholar 

  13. Nguyen KV (2022) Containing the spread of COVID-19 virus facing to its high mutation rate: approach to intervention using a nonspecific way of blocking its entry into the cells. Nucleosides Nucleotides Nucleic Acids 41:778–814

    Article  CAS  PubMed  Google Scholar 

  14. Castro Dopico X, Ols S, Loré K, Karlsson Hedestam GB (2022) Immunity to SARS-CoV-2 induced by infection or vaccination. J Intern Med 291:32–50

    Article  CAS  PubMed  Google Scholar 

  15. Mouliou DS (2022) Managing viral emerging infectious diseases via current molecular diagnostics in the emergency department: the tricky cases. Expert Rev Anti Infect Ther 20:1163–1169

    Article  CAS  PubMed  Google Scholar 

  16. Mouliou DS, Gourgoulianis KI (2021) False-positive and false-negative COVID-19 cases: respiratory prevention and management strategies, vaccination, and further perspectives. Expert Revi Respir Med 15:993–1002

    Article  CAS  Google Scholar 

  17. Ibrahim M, Vegel A, Niu A, Panse K, Chen R, Safah H, Socola F, Luk A, Saba NS (2021) Reinfection versus failure of viral clearance in a COVID-19 patient with hematologic malignancy. Leukemia Res 101:106514

    Article  CAS  Google Scholar 

  18. Agbuduwe C, Basu S (2020) Haematological manifestations of COVID-19: from cytopenia to coagulopathy. Eur J Haematol 105:540–546

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Korompoki E, Gavriatopoulou M, Fotiou D, Ntanasis-Stathopoulos I, Dimopoulos MA, Terpos E (2022) Late-onset hematological complications post COVID-19: an emerging medical problem for the hematologist. Am J Hematol 97:119–128

    Article  CAS  PubMed  Google Scholar 

  20. Gomes SM, Brito AC, Manfro WF, Ribeiro-Alves M, Ribeiro RS, da Cal MS, Lisboa VD, Abreu DP, Castilho LD, Porto LC (2023) High levels of pro-inflammatory SARS-CoV-2-specific biomarkers revealed by in vitro whole blood cytokine release assay (CRA) in recovered and long-COVID-19 patients. Plos one 18:e0283983

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Camaschella C, Nai A, Silvestri L (2020) Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 105:260

    Article  PubMed  PubMed Central  Google Scholar 

  22. Mancilha EMB, Oliveira JS (2021) SARS-CoV-2 association with hemoglobin and iron metabolism. Revista da Associação Médica Brasileira 67:1349–1352

    Article  PubMed  Google Scholar 

  23. Urbinati F, Madigan C, Malik P (2006) Pathophysiology and therapy for haemoglobinopathies; part II: thalassaemias. Expert Rev Mol Med 8:1–26

    Article  PubMed  Google Scholar 

  24. Lansiaux E, Drouin E, Bolm C (2023) Beta-thalassemia minor and SARS-CoV-2: physiopathology, prevalence, severity, morbidity, and mortality. Thalassemia Rep 13:21–32

    Article  CAS  Google Scholar 

  25. Fakhouri EW, Peterson SJ, Kothari J, Alex R, Shapiro JI, Abraham NG (2020) Genetic polymorphisms complicate COVID-19 therapy: pivotal role of HO-1 in cytokine storm. Antioxidants 9:636

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kong Y, Zhou S, Kihm AJ, Katein AM, Yu X, Gell DA, Mackay JP, Adachi K, Foster-Brown L, Louden CS (2004) Loss of α-hemoglobin–stabilizing protein impairs erythropoiesis and exacerbates β-thalassemia. J Clin Investig 114:1457–1466

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Stockman JA, Weiner LS, Simon GE, Stuart MJ, Oski FA (1975) The measurement of free erythrocyte porphyrin (FEP) as a simple means of distinguishing iron deficiency from beta-thalassemia trait in subjects with microcytosis. J Lab Clin Med 85:113–119

    CAS  PubMed  Google Scholar 

  28. Sotiriou S, Samara AA, Lachanas KE, Vamvakopoulou D, Vamvakopoulos KO, Vamvakopoulos N, Janho MB, Perivoliotis K, Donoudis C, Daponte A, Gourgoulianis KI (2022) Vulnerability of β-thalassemia heterozygotes to COVID-19: results from a cohort study. J Pers Med 12:352

  29. Marengo-Rowe AJ (2007) The thalassemias and related disorders. Proc (Bayl Univ Med Cent) 20:27–31

    PubMed  Google Scholar 

  30. Teulier M, Elabbadi A, Gerotziafas G, Lionnet F, Voiriot G, Fartoukh M (2021) Severe COVID-19 with acute respiratory distress syndrome (ARDS) in a sickle cell disease adult patient: case report. BMC Pulm Med 21:1–5

  31. Alsayegh F, Mousa SA (2020) Challenges in the management of sickle cell disease during SARS-CoV-2 pandemic. Clin Appl Thromb Hemost 26:1–8

    Article  Google Scholar 

  32. Umar Z, Ilyas U, Nso N (2022) Sickle cell disease and COVID-19 infection: importance of COVID-19 testing and approach to management. Cureus 14:e23604

    PubMed  PubMed Central  Google Scholar 

  33. Machado RF, Gladwin MT (2005) Chronic sickle cell lung disease: new insights into the diagnosis, pathogenesis and treatment of pulmonary hypertension. Br J Haematol 129:449–464

    Article  CAS  PubMed  Google Scholar 

  34. Chiang KC, Gupta A, Sundd P, Krishnamurti L (2023) Thrombo-inflammation in COVID-19 and sickle cell disease: two faces of the same coin. Biomedicines 11:338

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sivalingam T, Inusa B, Doyle P, Oteng-Ntim E (2020) COVID-19 and the pulmonary complications of sickle cell disease. EJHaem 1:545–547

    Article  PubMed  PubMed Central  Google Scholar 

  36. Alhazmi A, Moafa WA, Madkhali J, Saifain O, Alyahyawi F, Adhabi O, Alharbi AA (2022) Coronavirus disease 2019 in patients with sickle cell disease: a cross-sectional study from Jazan Province, Saudi Arabia. J Nat Sci Med 5:199

    Article  Google Scholar 

  37. Alyammahi SK, Abdin SM, Alhamad DW, Elgendy SM, Altell AT, Omar HA (2021) The dynamic association between COVID-19 and chronic disorders: an updated insight into prevalence, mechanisms and therapeutic modalities. Infect Genet Evol 87:104647

    Article  CAS  PubMed  Google Scholar 

  38. Hoogenboom WS, Alamuri TT, McMahon DM, Balanchivadze N, Dabak V, Mitchell WB, Morrone KB, Manwani D, Duong TQ (2022) Clinical outcomes of COVID-19 in patients with sickle cell disease and sickle cell trait: a critical appraisal of the literature. Blood Rev 53:100911

    Article  CAS  PubMed  Google Scholar 

  39. Kakavandi S, Zare I, VaezJalali M, Dadashi M, Azarian M, Akbari A, Ramezani Farani M, Zalpoor H, Hajikhani B (2023) Structural and non-structural proteins in SARS-CoV-2: potential aspects to COVID-19 treatment or prevention of progression of related diseases. Cell Commun Signal 21:110

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ward RJ, Crichton RR, Taylor DL, Corte LD, Srai SK, Dexter DT (2011) Iron and the immune system. J Neural Transm 118:315–328

    Article  CAS  PubMed  Google Scholar 

  41. Jiang X, Stockwell BR, Conrad M (2021) Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 22:266–282

    Article  PubMed  PubMed Central  Google Scholar 

  42. Rodriguez R, Schreiber SL, Conrad M (2022) Persister cancer cells: Iron addiction and vulnerability to ferroptosis. Mol Cell 82:728–740

    Article  CAS  PubMed  Google Scholar 

  43. Panda SP, Kesharwani A (2023) Micronutrients/miRs/ATP networking in mitochondria: clinical intervention with ferroptosis, cuproptosis, and calcium burden. Mitochondrion 71:1–16

    Article  Google Scholar 

  44. McGonagle D, Sharif K, O’Regan A, Bridgewood C (2020) The role of cytokines including interleukin-6 in COVID-19 induced pneumonia and macrophage activation syndrome-like disease. Autoimmun Rev 19:102537

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nemeth E, Ganz T (2006) Regulation of iron metabolism by hepcidin. Annu Rev Nutr 26:323–342

    Article  CAS  PubMed  Google Scholar 

  46. Wlazlo E, Mehrad B, Morel L, Scindia Y (2021) Iron metabolism: an under investigated driver of renal pathology in lupus nephritis. Frontiers in Medicine. 8:643686

    Article  PubMed  PubMed Central  Google Scholar 

  47. Vogt A-CS, Arsiwala T, Mohsen M, Vogel M, Manolova V, Bachmann MF (2021) On iron metabolism and its regulation. Int J Mol Sci 22:4591

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bellmann-Weiler R, Lanser L, Barket R, Rangger L, Schapfl A, Schaber M, Fritsche G, Wöll E, Weiss G (2020) Prevalence and predictive value of anemia and dysregulated iron homeostasis in patients with COVID-19 infection. J Clin Med 9:2429

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lv Y, Chen L, Liang X, Liu X, Gao M, Wang Q, Wei Q, Liu L (2021) Association between iron status and the risk of adverse outcomes in COVID-19. Clin Nutr 40:3462–3469

    Article  CAS  PubMed  Google Scholar 

  50. Shi L, Wang Y, Yang H, Duan G, Wang Y (2020) Laboratory abnormalities in pregnant women with novel coronavirus disease 2019. Am J Perinatol 37:1070–1073

    Article  PubMed  PubMed Central  Google Scholar 

  51. Ward JL, Torres-Gonzalez M, Ammons MCB (2022) The influence of viral infections on iron homeostasis and the potential for lactoferrin as a therapeutic in the age of the SARS-CoV-2 pandemic. Nutrients 14:3090

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kronstein-Wiedemann R, Stadtmüller M, Traikov S, Georgi M, Teichert M, Yosef H, Wallenborn J, Karl A, Schütze K, Wagner M (2022) SARS-CoV-2 Infects red blood cell progenitors and dysregulates hemoglobin and iron metabolism. Stem Cell Rev Rep 18:1809–1821

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Rosário C, Zandman-Goddard G, Meyron-Holtz EG, D’Cruz DP, Shoenfeld Y (2013) The hyperferritinemic syndrome: macrophage activation syndrome, Still’s disease, septic shock and catastrophic antiphospholipid syndrome. BMC Med 11:1–11

    Article  Google Scholar 

  54. Giemza-Stokłosa J, Islam MA, Kotyla PJ (2019) Hyperferritinaemia: an iron sword of autoimmunity. Curr Pharm Des 25:2909–2918

    Article  PubMed  Google Scholar 

  55. Sumbly V, Siddiqui R, Alshamam M, Kurbanova T, Rizzo V (2021) New onset aplastic anemia after a COVID-19 infection: a case report. Am J Med Case Rep 9:451–455

    Google Scholar 

  56. Shallis RM, Ahmad R, Zeidan AM (2018) Aplastic anemia: etiology, molecular pathogenesis, and emerging concepts. Eur J Haematol 101:711–720

    Article  CAS  PubMed  Google Scholar 

  57. Khalid I, Abbas F, Mustafa Z, Kamran M (2022) Association of various etiological factors with idiopathic acquired aplastic anemia. J Haematol Stem Cell Res 2:19–25

    Google Scholar 

  58. Shehi E, Ghazanfar H, Fortuzi K, Gonzalez E, Zeana C (2020) A rare case of parvovirus B19 infection manifesting as chronic aplastic anemia and neutropenia in a human immunodeficiency virus-infected patient. Cureus 12:e12174

    PubMed  PubMed Central  Google Scholar 

  59. Scheinberg P, Fischer SH, Li L, Nunez O, Wu CO, Sloand EM, Cohen JI, Young NS, John Barrett A (2007) Distinct EBV and CMV reactivation patterns following antibody-based immunosuppressive regimens in patients with severe aplastic anemia. Blood 109:3219–3224

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kerr JR (2015) A review of blood diseases and cytopenias associated with human parvovirus B19 infection. Rev Med Virol 25:224–240

    Article  PubMed  Google Scholar 

  61. Nasiri K, Mohammadzadehsaliani S, Kheradjoo H, Shabestari AM, Eshaghizadeh P, Pakmehr A, Alsaffar MF, Al-Naqeeb BZT, Yasamineh S, Gholizadeh O (2023) Spotlight on the impact of viral infections on Hematopoietic Stem Cells (HSCs) with a focus on COVID-19 effects. Cell Commun Signal 21:1–15

    Article  Google Scholar 

  62. Karki R, Sharma BR, Tuladhar S, Williams EP, Zalduondo L, Samir P, Zheng M, Sundaram B, Banoth B, Malireddi RS (2021) Synergism of TNF-α and IFN-γ triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell 184:149–168

    Article  CAS  PubMed  Google Scholar 

  63. Kitagawa M, Saito I, Kuwata T, Yoshida S, Yamaguchi S, Takahashi M, Tanizawa T, Kamiyama R, Hirokawa K (1997) Overexpression of tumor necrosis factor (TNF)-α and interferon (IFN)-γ by bone marrow cells from patients with myelodysplastic syndromes. Leukemia 11:2049–2054

    Article  CAS  PubMed  Google Scholar 

  64. Zheng S, Vrindts Y, Lopez M, De Groote D, Zangerlé P-F, Collette J, Franchimont N, Geenen V, Albert A, Reginster J-Y (1997) Increase in cytokine production (IL-1β, IL-6, TNF-α but not IFN-γ, GM-CSF or LIF) by stimulated whole blood cells in postmenopausal osteoporosis. Maturitas 26:63–71

    Article  CAS  PubMed  Google Scholar 

  65. Iskander D, Wang G, Heuston EF, Christodoulidou C, Psaila B, Ponnusamy K, Ren H, Mokhtari Z, Robinson M, Chaidos A (2021) Single-cell profiling of human bone marrow progenitors reveals mechanisms of failing erythropoiesis in Diamond-Blackfan anemia. Sci Transl Med 13:eabf0113

    Article  CAS  PubMed  Google Scholar 

  66. Barber C (2021) Rare health conditions 44: ataxia, Diamond Blackfan anaemia, sexual identities. Br J Healthcare Assist 15:78–83

    Article  MathSciNet  Google Scholar 

  67. Wenzhong, Liu, and Li Hualan (2020) COVID-19: attacks the 1-beta chain of hemoglobin and captures the porphyrin to inhibit human heme metabolism. ChemRxiv Preprint

  68. Dutt S, Hamza I, Bartnikas TB (2022) Molecular mechanisms of iron and heme metabolism. Annu Rev Nutr 42:311–35

  69. Lee JX, Chieng WK, Lau SC, Tan CE (2021) COVID-19 and hemoglobinopathies: a systematic review of clinical presentations, investigations, and outcomes. Front Med 8:757510

  70. Severance T, Rahim M, French J, Baker R, Shriner A, Khaitan A, Overholt K (2021) COVID-19 and hereditary spherocytosis: a recipe for hemolysis. Pediatr Blood Cancer 68:e28548

    Article  CAS  PubMed  Google Scholar 

  71. Rothman JA, Stevens JL, Gray FL, Kalfa TA (2020) How I approach hereditary hemolytic anemia and splenectomy. Pediatr Blood Cancer 67:e28337

    Article  PubMed  Google Scholar 

  72. Schwartz SI, Bernard RP, Adams JT, Bauman AW (1970) Splenectomy for hematologic disorders. Arch Surg 101:338–347

    Article  CAS  PubMed  Google Scholar 

  73. Buesing KL, Tracy ET, Kiernan C, Pastor AC, Cassidy LD, Scott JP, Ware RE, Davidoff AM, Rescorla FJ, Langer JC (2011) Partial splenectomy for hereditary spherocytosis: a multi-institutional review. J Pediatr Surg 46:178–183

    Article  PubMed  Google Scholar 

  74. Manciu S, Matei E, Trandafir B (2017) Hereditary spherocytosis-diagnosis, surgical treatment and outcomes. A literature review. Chirurgia (Bucur) 112:110–116

    Article  PubMed  Google Scholar 

  75. Severance T, Rahim M, French J, Baker R, Shriner A, Khaitan A, Overholt K (2020) COVID-19 and hereditary spherocytosis: a recipe for hemolysis. Authorea Preprints

  76. Cavezzi A, Menicagli R, Troiani E, Corrao S (2022) COVID-19, cation dysmetabolism, sialic acid, CD147, ACE2, viroporins, hepcidin and ferroptosis: a possible unifying hypothesis. F1000Research 11:102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mitra A, Dwyre DM, Schivo M, Thompson GR III, Cohen SH, Ku N, Graff JP (2020) Leukoerythroblastic reaction in a patient with COVID-19 infection. Am J Hematol 95:999–1000

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Tabares Calvache E, Tabares Calvache AD, Faulhaber GAM (2020) Systematic review about etiologic association to the leukoerythroblastic reaction. Int J Lab Hematol 42:495–500

    Article  PubMed  Google Scholar 

  79. Wolf BC, Neiman RS (1985) Myelofibrosis with myeloid metaplasia: pathophysiologic implications of the correlation between bone marrow changes and progression of splenomegaly. Blood 65:803–809

    Article  CAS  PubMed  Google Scholar 

  80. Costela-Ruiz VJ, Illescas-Montes R, Puerta-Puerta JM, Ruiz C, Melguizo-Rodríguez L (2020) SARS-CoV-2 infection: the role of cytokines in COVID-19 disease. Cytokine Growth Factor Rev 54:62–75

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Lee WS, Margolskee E (2020) Leukoerythroblastosis and plasmacytoid lymphocytes in a child with SARS-CoV-2–associated multisystem inflammatory syndrome. Blood 136(7):914

  82. Rowley AH, Shulman ST (2010) Pathogenesis and management of Kawasaki disease. Expert Rev Anti Infect Ther 8:197–203

    Article  PubMed  PubMed Central  Google Scholar 

  83. Dajani AS, Taubert KA, Gerber MA, Shulman ST, Ferrieri P, Freed M, Takahashi M, Bierman FZ, Karchmer AW, Wilson W (1993) Diagnosis and therapy of Kawasaki disease in children. Circulation 87:1776–1780

    Article  CAS  PubMed  Google Scholar 

  84. Esper F, Shapiro ED, Weibel C, Ferguson D, Landry ML, Kahn JS (2005) Association between a novel human coronavirus and Kawasaki disease. J Infect Dis 191:499–502

    Article  CAS  PubMed  Google Scholar 

  85. Soy M, Atagündüz P, Atagündüz I, Sucak GT (2021) Hemophagocytic lymphohistiocytosis: a review inspired by the COVID-19 pandemic. Rheumatol Int 41:7–18

    Article  CAS  PubMed  Google Scholar 

  86. Loscocco GG (2020) Secondary hemophagocytic lymphohistiocytosis, HScore and COVID-19. Int J Hematol 112:125–126

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kiran KS, Yasitha K, Panakala S, Pitcairn N, Tiwari RV, Sharma M, Tiwari H (2021) Role of C-reactive protein, serum ferritin and D-Dimer in Covid cases: systematic review & meta analysis. Ann Romanian Soc Cell Biol 25:10807–10816

    Google Scholar 

  88. Hsu R-J, Yu W-C, Peng G-R, Ye C-H, Hu S, Chong PCT, Yap KY, Lee JYC, Lin W-C, Yu S-H (2022) The role of cytokines and chemokines in severe acute respiratory syndrome coronavirus 2 infections. Front Immunol 13:832394

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Kim JS, Lee JY, Yang JW, Lee KH, Effenberger M, Szpirt W, Kronbichler A, Shin JI (2021) Immunopathogenesis and treatment of cytokine storm in COVID-19. Theranostics 11:316

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Bordbar M, Sanaei Dashti A, Amanati A, Shorafa E, Mansoori Y, Dehghani SJ, Molavi Vardanjani H (2021) Assessment of the HScore as a predictor of disease outcome in patients with COVID-19. BMC Pulm Med 21:1-9

  91. Abu-Zeinah G, DeSancho MT (2020) Understanding sideroblastic anemia: an overview of genetics, epidemiology, pathophysiology and current therapeutic options. J Blood Med 25:305-18

    Google Scholar 

  92. Brissot P, Bernard DG, Brissot E, Loréal O, Troadec M-B (2018) Rare anemias due to genetic iron metabolism defects. Mutat Res Rev Mutat Res 777:52–63

    Article  CAS  PubMed  Google Scholar 

  93. Mukhi N, Soto LR, Vuppala A (2022) Transient sideroblastic anemia post-COVID-19 infection. Cureus 14:e30275

    PubMed  PubMed Central  Google Scholar 

  94. El-Sayed EM, Ibrahim KS (2022) Ameliorating effects of probiotics on alterations in iron homeostasis and inflammation in COVID-19. Mol Biol R 49:5153–5163

    Article  CAS  Google Scholar 

  95. Ayyashi M, Darbashi H, Hakami A, Sharahili F, Sharahili F Sr (2023) Evaluation of remdesivir utilization pattern in critically Ill patients with COVID-19 in Jazan Province. Cureus 15:e36247

    PubMed  PubMed Central  Google Scholar 

  96. Wolffenbuttel BH, Wouters HJ, Heiner-Fokkema MR, van der Klauw MM (2019) The many faces of cobalamin (vitamin B12) deficiency. Mayo Clin Proc Innov Qual Outcomes 3:200–214

    Article  PubMed  PubMed Central  Google Scholar 

  97. Allen LH. Efficacy and safety of vitamin B12 fortification. In: Food Fortification in a Globalized World. Elsevier. 2018:255-261.

  98. Fath MK, Naderi M, Hamzavi H, Ganji M, Shabani S, Khalesi B, Pourzardosht N, Hashemi ZS, Khalili S (2022) Molecular Mechanisms and therapeutic effects of different vitamins and minerals in COVID-19 patients. J Trace Elem Med Biol 73:127044

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Sabry W, Elemary M, Burnouf T, Seghatchian J, Goubran H (2020) Vitamin B12 deficiency and metabolism-mediated thrombotic microangiopathy (MM-TMA). Transfus Apher Sci 59:102717

    Article  PubMed  Google Scholar 

  100. Stokes MB, Zviti R, Lin F, D’Agati VD (2016) An unusual cause of hypertension with hematuria and proteinuria: answers. Pediatr Nephrol 31:2265–2270

    Article  PubMed  Google Scholar 

  101. Tan CW, Ho LP, Kalimuddin S, Cherng BPZ, Teh YE, Thien SY, Wong HM, Tern PJW, Chandran M, Chay JWM (2020) Cohort study to evaluate the effect of vitamin D, magnesium, and vitamin B12 in combination on progression to severe outcomes in older patients with coronavirus (COVID-19). Nutrition 79:111017

    Article  PubMed  PubMed Central  Google Scholar 

  102. Stabler SP (2013) Vitamin B12 deficiency. N Engl J Med 368:149–160

    Article  CAS  PubMed  Google Scholar 

  103. Green R, Allen LH, Bjørke-Monsen A-L, Brito A, Guéant J-L, Miller JW, Molloy AM, Nexo E, Stabler S, Toh B-H (2017) Vitamin B12 deficiency. Nat Rev Dis Primers 3:1–20

    Google Scholar 

  104. Guéant J-L, Guéant-Rodriguez R-M, Alpers DH (2022) Vitamin B12 absorption and malabsorption. Vitam Horm 119:241–274 (Elsevier)

    Article  PubMed  Google Scholar 

  105. Kuijpers TW, de Vries AC, van Leeuwen EM, Ermens AT, de Pont S, Smith DE, Wamelink MM, Mensenkamp AR, Nelen MR, Lango Allen H, Pals ST (2022) Megalobastic anemia, infantile leukemia, and immunodeficiency caused by a novel homozygous mutation in the DHFR gene. Blood Adv 6:5829-34

  106. Batista KS, Cintra VM, Lucena PA, Manhães-de-Castro R, Toscano AE, Costa LP, Queiroz ME, de Andrade SM, Guzman-Quevedo O (2022) Aquino JdS: The role of vitamin B12 in viral infections: a comprehensive review of its relationship with the muscle–gut–brain axis and implications for SARS-CoV-2 infection. Nutr Rev 80:561–578

    Article  PubMed  Google Scholar 

  107. dos Santos LMJ (2020) Can vitamin B12 be an adjuvant to COVID-19 treatment? GSC Biol Pharm Sci 11:001–005

    Article  Google Scholar 

  108. Babar Q, Ali A, Saeed A, Tahir MF: Novel treatment strategy against COVID-19 through anti-inflammatory, antioxidant and immunostimulatory properties of the b vitamin complex. In: B-Complex Vitamins-Sources, Intakes and Novel Applications. Intechopen. 2021.

  109. Agarwal N, Khatri N, Singh P (2021) Pancytopenia in pregnant patients with COVID-19 infection and vitamin B12 deficiency: a case report study. JIDHealth 4:415-8

  110. Tang K-T, Hsu B-C, Chen D-Y (2021) Autoimmune and rheumatic manifestations associated with COVID-19 in adults: an updated systematic review. Front Immunol 12:645013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Fattizzo B, Pasquale R, Bellani V, Barcellini W, Kulasekararaj AG (2021) Complement mediated hemolytic anemias in the COVID-19 era: case series and review of the literature. Front Immunol 12:791429.

  112. Dotan A, Muller S, Kanduc D, David P, Halpert G, Shoenfeld Y (2021) The SARS-CoV-2 as an instrumental trigger of autoimmunity. Autoimmun Rev 20:102792.

  113. Al-Kuraishy HM, Al-Gareeb AI, Kaushik A, Kujawska M (2022) Batiha GE-S: Hemolytic anemia in COVID-19. Ann Hematol 101:1887–1895

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Jafarzadeh A, Jafarzadeh S, Pardehshenas M, Nemati M, Mortazavi SMJ (2023) Development and exacerbation of autoimmune hemolytic anemia following COVID-19 vaccination: a systematic review. Int J Lab Hematol 45:145–155

    Article  PubMed  Google Scholar 

  115. González-López TJ, Bárez A, Bernardo-Gutiérrez A, Bernat S, Canaro-Hirnyk M, Entrena-Ureña L, Fernández-Fuertes F, de GuineaCastro JM, Jiménez-Bárcenas R, Pascual-Izquierdo C (2023) Recommendations on the management of patients with immune thrombocytopenia (ITP) in the context of SARS-CoV-2 infection and vaccination: consensus guidelines from a Spanish ITP expert group. Infect Dis Ther 12:303–315

    Article  PubMed  Google Scholar 

  116. Sridhara S, Nair R, Stanek M (2022) Severe aplastic anemia after receiving SARS-CoV-2 Moderna mRNA vaccination. J Hematol 11:34

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Murdych TM (2022) A case of severe autoimmune hemolytic anemia after a receipt of a first dose of SARS-CoV-2 vaccine. Int J Lab Hematol 44:e10

    Article  PubMed  Google Scholar 

  118. Osmanodja B, Schreiber A, Schrezenmeier E, Seelow E (2021) First diagnosis of thrombotic thrombocytopenic purpura after SARS-CoV-2 vaccine–case report. BMC Nephrol 22:1–5

    Article  Google Scholar 

  119. Okuno S, Hashimoto K, Shimizu R, Takagi E, Kajiguchi T (2021) Development of autoimmune hemolytic anemia after BNT162b2 mRNA COVID-19 vaccination. Jpn J Clin Hematol 62:1510–1514

    Google Scholar 

  120. Waqar SHB, Khan AA, Memon S (2021) Thrombotic thrombocytopenic purpura: a new menace after COVID bnt162b2 vaccine. Int J Hematol 114:626–629

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Tabata S, Hosoi H, Murata S, Takeda S, Mushino T, Sonoki T (2022) Severe aplastic anemia after COVID-19 mRNA vaccination: causality or coincidence? J Autoimmun 126:102782

    Article  CAS  PubMed  Google Scholar 

  122. Julian JA, Mathern DR, Fernando D (2021) Idiopathic thrombocytopenic purpura and the Moderna COVID-19 vaccine. Ann Emerg Med 77:654–656

    Article  PubMed  PubMed Central  Google Scholar 

  123. Liebman HA, Weitz IC (2017) Autoimmune hemolytic anemia. Med Clin 101:351–359

    Google Scholar 

  124. Hill A, Hill QA. Autoimmune hemolytic anemia. Hematology 2014, the American Society of Hematology Education Program Book. 2018:382-389.

  125. Barcellini W, Zaninoni A, Giannotta JA, Fattizzo B (2020) New insights in autoimmune hemolytic anemia: from pathogenesis to therapy. J Clin Med 9:3859

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Mahévas M, Moulis G, Andres E, Riviere E, Garzaro M, Crickx E, Guillotin V, Malphettes M, Galicier L, Noel N, Darnige L (2020) Clinical characteristics, management and outcome of COVID‐19‐associated immune thrombocytopenia: a French multicentre series. Br J Haematol 190:e224-9

  127. Mahévas M, Moulis G, Andres E, Riviere E, Garzaro M, Crickx E, Guillotin V, Malphettes M, Galicier L, Noel N (2020) Clinical characteristics, management and outcome of COVID-19-associated immune thrombocytopenia: a French multicentre series. Br J Haematol 190:e224–e229

    Article  PubMed  PubMed Central  Google Scholar 

  128. Chou S-C, Chang Y-C, Liao C-K, Chen T-C, Sun K-J, Huang W-H, Wu Y-F (2022) New presentations and exacerbations of immune thrombocytopenia after coronavirus disease 2019 vaccinations: the Taiwan experience. Platelets 33:531–535

    Article  CAS  PubMed  Google Scholar 

  129. Mouliou DS, Dardiotis E (2022) Current evidence in SARS-CoV-2 mRNA vaccines and post-vaccination adverse reports: knowns and unknowns. Diagnostics 12:1555

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Tian J, Yuan X, Xiao J, Zhong Q, Yang C, Liu B, Cai Y, Lu Z, Wang J, Wang Y (2020) Clinical characteristics and risk factors associated with COVID-19 disease severity in patients with cancer in Wuhan, China: a multicentre, retrospective, cohort study. Lancet Oncol 21:893–903

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Pascutti MF, Erkelens MN, Nolte MA (2016) Impact of viral infections on hematopoiesis: from beneficial to detrimental effects on bone marrow output. Front Immunol 7:364

    Article  PubMed  PubMed Central  Google Scholar 

  132. Griggio V, Perutelli F, Salvetti C, Boccellato E, Boccadoro M, Vitale C, Coscia M (2020) Immune dysfunctions and immune-based therapeutic interventions in chronic lymphocytic leukemia. Front Immunol 11:594556

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Allegra A, Tonacci A, Musolino C, Pioggia G, Gangemi S (2021) Secondary immunodeficiency in hematological malignancies: focus on multiple myeloma and chronic lymphocytic leukemia. Front Immunol 12:738915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Martínez JC, Sica RA, Stockerl-Goldstein K, Rubinstein SM (2022) COVID-19 in patients with hematologic malignancies: outcomes and options for treatments. Acta Haematologica 145:244–256

    Article  PubMed  Google Scholar 

  135. Cutrona G, Tripodo C, Matis S, Recchia AG, Massucco C, Fabbi M, Colombo M, Emionite L, Sangaletti S, Gulino A, Reverberi D (2018) Microenvironmental regulation of the IL-23R/IL-23 axis overrides chronic lymphocytic leukemia indolence. Sci Transl Med 10:eaal1571

  136. Ye X, Xiao X, Li B, Zhu W, Li Y, Wu J, Huang X, Jin J, Chen D, Jin J, Huang J (2020) Low humoral immune response and ineffective clearance of SARS-Cov-2 in a COVID-19 patient with CLL during a 69-day follow-up. Front Oncol 10:1272

  137. Largeaud L, Ribes A, Dubois-Galopin F, Memier V, Rolland Y, Gaudin C, Rousset D, Geeraerts T, Noel-Savina E, Rieu JB, Vergez F (2021) Major rise of a chronic lymphoid leukemia clone during the course of COVID-19. Int J Lab Hematol 43:e82–3

  138. Allegra A, Pioggia G, Tonacci A, Musolino C, Gangemi S (2020) Cancer and SARS-CoV-2 infection: diagnostic and therapeutic challenges. Cancers 12:1581

  139. Li X, Shao M, Zeng X, Qian P, Huang H (2021) Signaling pathways in the regulation of cytokine release syndrome in human diseases and intervention therapy. Signal Transduct Target Ther 6:367

  140. Dettorre GM, Patel M, Gennari A, Pentheroudakis G, Romano E, Cortellini A, Pinato DJ (2021) The systemic pro-inflammatory response: targeting the dangerous liaison between COVID-19 and cancer. ESMO open 6:100123

  141. Ujjani CS, Gooley TA, Spurgeon SE, Stephens DM, Lai C, Broome CM, O'Brien SM, Zhu H, Laing KJ, Winter AM. Diminished humoral and cellular responses to SARS-CoV-2 Vaccines in patients with chronic lymphocytic leukemia. Blood Adv. 2022:bloodadvances. 2022009164.

  142. Moore JB, June CH (2020) Cytokine release syndrome in severe COVID-19. Science 368:473–474

    Article  CAS  PubMed  ADS  Google Scholar 

  143. Roeker LE, Knorr DA, Pessin MS, Ramanathan LV, Thompson MC, Leslie LA, Zelenetz AD, Mato AR (2020) Anti-SARS-CoV-2 antibody response in patients with chronic lymphocytic leukemia. Leukemia 34:3047–3049

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Heydarian M, Mohammadtaghizadeh M, Shojaei M, Babazadeh M, Abbasian S, Amrovani M (2022) The effect of COVID-19 derived cytokine storm on cancer cells progression: double-edged sword. Mol Biol Rep 49:605–615

    Article  CAS  PubMed  Google Scholar 

  145. Gandomi-Mohammadabadi A, Divsalar F, Taram S, Kateb Z, Montazer F, Farbod Y (2021) COVID-19 in a patient with newly diagnosed chronic lymphocytic leukemia (CLL): a case report. J Res Appl Basic Med Sci 7:26–30

    Article  CAS  Google Scholar 

  146. Bernar B, Kropshofer G, Crazzolara R, Kapelari K, Griesmacher A, Müller T, Scholl-Bürgi S (2020) SARS-CoV-2 infection in a 7-year-old girl with pancytopenia during acute lymphocytic leukemia maintenance therapy. Pediatr Blood Cancer 67:e28391

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Somasundaram R, Prasad MA, Ungerbäck J, Sigvardsson M (2015) Transcription factor networks in B-cell differentiation link development to acute lymphoid leukemia. Blood J Am Soc Hematol 126:144–152

    CAS  Google Scholar 

  148. Sanda T, Leong WZ (2017) TAL1 as a master oncogenic transcription factor in T-cell acute lymphoblastic leukemia. Exp Hematol 53:7–15

    Article  CAS  PubMed  Google Scholar 

  149. Montaño A, Forero-Castro M, Marchena-Mendoza D, Benito R, Hernández-Rivas JM (2018) New challenges in targeting signaling pathways in acute lymphoblastic leukemia by NGS approaches: an update. Cancers 10:110

    Article  PubMed  PubMed Central  Google Scholar 

  150. Baruchel A, Bertrand Y, Boissel N, Brethon B, Ducassou S, Gandemer V, Halfon-Domenech C, Leblanc T, Leverger G, Michel G (2020) COVID-19 and acute lymphoblastic leukemias of children and adolescents: First recommendations of the Leukemia committee of the French Society for the fight against cancers and leukemias in children and adolescents (SFCE). Bulletin Du Cancer 107:629–632

    Article  PubMed  PubMed Central  Google Scholar 

  151. Huang FL, Liao EC, Li CL, Yen CY, Yu SJ (2020) Pathogenesis of pediatric B-cell acute lymphoblastic leukemia: molecular pathways and disease treatments. Oncol Lett 20:448–454

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Yan W, Song L, Wang H, Yang W, Hu L, Yang Y (2021) Extracellular vesicles carrying miRNA-181b-5p affects the malignant progression of acute lymphoblastic leukemia. J Transl Med 19:1–9

    Article  Google Scholar 

  153. Iravani Saadi M, Ramzi M, Hesami Z, Kheradmand N, Owjfard M, Nabi Abdolyousefi E, Karimi Z (2021) MiR-181a and-b expression in acute lymphoblastic leukemia and its correlation with acute graft-versus-host disease after hematopoietic stem cell transplantation, COVID-19 and torque teno viruses. Virusdisease 32:727–736

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Baghbani E, Khaze V, Sadreddini S, Mokhtarzadeh A, Mansoori B, Mohammadi A, Vatankhahan V, Toosi P, Baradaran B (2018) PTPN22 silencing in human acute T-cell leukemia cell line (Jurkat cell) and its effect on the expression of miR-181a and miR-181b. Adv Pharm Bull 8:277

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Arisan ED, Dart A, Grant GH, Arisan S, Cuhadaroglu S, Lange S, Uysal-Onganer P (2020) The prediction of miRNAs in SARS-CoV-2 genomes: hsa-miR databases identify 7 key miRs linked to host responses and virus pathogenicity-related KEGG pathways significant for comorbidities. Viruses 12:614

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Amini-Farsani Z, Yadollahi-Farsani M, Arab S, Forouzanfar F, Yadollahi M, Asgharzade S (2021) Prediction and analysis of microRNAs involved in COVID-19 inflammatory processes associated with the NF-kB and JAK/STAT signaling pathways. Int immunopharmacol 100:108071

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Badreldein M, Elsorady M, Alhadidi A, Gallab O, Eldabah N (2021) ALL-443: diagnostic and prognostic significance of miRNA-511 and miRNA-16 expression in adult B-Acute lymphoblastic leukemia patients. Clin Lymphoma Myeloma Leuk 21:S277

    Article  Google Scholar 

  158. Li C-X, Chen J, Lv S-K, Li J-H, Li L-L, Hu X (2021) Whole-transcriptome RNA sequencing reveals significant differentially expressed mRNAs, miRNAs, and lncRNAs and related regulating biological pathways in the peripheral blood of COVID-19 patients. Mediators Inflamm 6635925:22

    Google Scholar 

  159. Witten L, Slack FJ (2020) miR-155 as a novel clinical target for hematological malignancies. Carcinogenesis 41:2–7

    Article  CAS  PubMed  Google Scholar 

  160. Haroun RA-H, Osman WH, Amin RE, Hassan AK, Abo-Shanab WS, Eessa AM (2022) Circulating plasma miR-155 is a potential biomarker for the detection of SARS-CoV-2 infection. Pathology 54:104–110

    Article  CAS  PubMed  Google Scholar 

  161. Di Martino MT, Leone E, Amodio N, Foresta U, Lionetti M, Pitari MR, Cantafio MEG, Gullà A, Conforti F, Morelli E (2012) Synthetic miR-34a Mimics as a Novel Therapeutic Agent for Multiple Myeloma: In Vitro and In Vivo EvidenceAntitumor Activity of miR-34a in Multiple Myeloma. Clin Cancer Res 18:6260–6270

    Article  PubMed  PubMed Central  Google Scholar 

  162. Sagulkoo P, Chuntakaruk H, Rungrotmongkol T, Suratanee A, Plaimas K (2022) Multi-level biological network analysis and drug repurposing based on leukocyte transcriptomics in severe COVID-19: in silico systems biology to precision medicine. J Personal Med 12:1030

    Article  Google Scholar 

  163. Liu A-F, Wang J-X, Li F-L, Chen Y-J (2017) Research progress on miR-125 family in malignant hematologic diseases-review. J Exp Hematol 25:1842–1846

    Google Scholar 

  164. Zhang S, Amahong K, Sun X, Lian X, Liu J, Sun H, Lou Y, Zhu F, Qiu Y (2021) The miRNA: a small but powerful RNA for COVID-19. Brief Bioinform 22:1137–1149

    Article  CAS  PubMed  Google Scholar 

  165. Haneklaus M, Gerlic M, O’Neill LA, Masters S (2013) miR-223: infection, inflammation and cancer. J Intern Med 274:215–226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Leoncini PP, Bertaina A, Papaioannou D, Flotho C, Masetti R, Bresolin S, Menna G, Santoro N, Zecca M, Basso G (2015) MicroRNA-150 regulates STAT5b levels in juvenile myelomonocytic leukemia (JMML). Blood 126:2851

    Article  Google Scholar 

  167. Akula SM, Bolin P, Cook PP (2022) Cellular miR-150-5p may have a crucial role to play in the biology of SARS-CoV-2 infection by regulating nsp10 gene. RNA Biol 19:1–11

    Article  CAS  PubMed  Google Scholar 

  168. Lee JH, Choi YS, Park JH, Kim H, Lee I, Won YB, Yun BH, Park JH, Seo SK, Lee BS (2019) miR-150-5p may contribute to pathogenesis of human leiomyoma via regulation of the Akt/p27Kip1 pathway in vitro. Int J Mol Sci 20:2684

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Derda AA, Garg A, Bär C, Thum T (2021) Reply to ‘COVID-19 severity, miR-21 targets, and common human genetic variation.’ Eur J Heart Fail 23:1987

    Article  CAS  PubMed  Google Scholar 

  170. Ma X, Choudhury SN, Hua X, Dai Z, Li Y (2013) Interaction of the oncogenic miR-21 microRNA and the p53 tumor suppressor pathway. Carcinogenesis 34:1216–1223

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Leone E, Morelli E, Di Martino MT, Amodio N, Foresta U, Gullà A, Rossi M, Neri A, Giordano A, Munshi NC (2013) Targeting miR-21 inhibits in vitro and in vivo multiple myeloma cell growthantitumor activity of mir-21 inhibitors in multiple myeloma. Clin Cancer Res 19:2096–2106

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Askari N, Hadizadeh M, Rashidifar M (2022) A new insight into sex-specific non-coding RNAs and networks in response to SARS-CoV-2. Infect Genet Evol 97:105195

    Article  CAS  PubMed  Google Scholar 

  173. Sevcikova S, Kubiczkova L, Sedlarikova L, Slaby O, Hajek R (2013) Serum miR-29a as a marker of multiple myeloma. Leuk Lymphoma 54:189–191

    Article  CAS  PubMed  Google Scholar 

  174. Fiserovaa B, Kubiczkova L, Sedlarikova L, Hajek R, Sevcikova S (2015) The miR-29 family in hematological malignancies. Biomed Pap Med Fac Palacky Univ Olomouc 159:184–191

    Article  Google Scholar 

  175. Hershkovitz Rokah O, Granot G, Ovcharenko A, Modai S, Pasmanik-Chor M, Toren A, Shomron N, Shpilberg O (2012) Downregulation of miR-31, miR-155, and miR-564 in chronic myeloid leukemia cells. PloS one 7:e35501

    Article  PubMed Central  ADS  Google Scholar 

  176. Farr RJ, Rootes CL, Rowntree LC, Nguyen TH, Hensen L, Kedzierski L, Cheng AC, Kedzierska K, Au GG, Marsh GA (2021) Altered microRNA expression in COVID-19 patients enables identification of SARS-CoV-2 infection. PLoS Pathogens 17:e1009759

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Zhou T, Medeiros LJ, Hu S (2018) Chronic myeloid leukemia: beyond BCR-ABL1. Curr Hematol Malig Rep 13:435–445

    Article  CAS  PubMed  Google Scholar 

  178. Delgado N, Torres A (2022) What do we currently know about chronic myeloid leukemia (CML) and COVID-19? Curr Oncol Rep 24:645–650

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Eşkazan AE (2020) Chronic myeloid leukaemia and the use of tyrosine kinase inhibitors in the days of COVID-19 pandemic. Br J Clin Pharmacol 86:1790

    Article  PubMed  PubMed Central  Google Scholar 

  180. García-Gutiérrez V, Hernández-Boluda JC (2019) Tyrosine kinase inhibitors available for chronic myeloid leukemia: efficacy and safety. Front Oncol 9:603

    Article  PubMed  PubMed Central  Google Scholar 

  181. Alves R, McArdle SE, Vadakekolathu J, Gonçalves AC, Freitas-Tavares P, Pereira A, Almeida AM, Sarmento-Ribeiro AB, Rutella S (2020) Flow cytometry and targeted immune transcriptomics identify distinct profiles in patients with chronic myeloid leukemia receiving tyrosine kinase inhibitors with or without interferon-α. J Transl Med. 18:1–15

    Article  Google Scholar 

  182. Galimberti S, Petrini M, Baratè C, Ricci F, Balducci S, Grassi S, Guerrini F, Ciabatti E, Mechelli S, Di Paolo A (2020) Tyrosine kinase inhibitors play an antiviral action in patients affected by chronic myeloid leukemia: a possible model supporting their use in the fight against SARS-CoV-2. Front Oncol 10:1428

    Article  PubMed  PubMed Central  Google Scholar 

  183. Pagnano KB, Peralta EH, Navarro JR, David Salas LDR, Delgado N, Moiraghi B, Toreli ACM, Perobelli LM, Fechio L, Quixada AT (2021) COVID-19 in chronic myeloid leukemia patients in Latin America. Leuk Lymphoma 62:3212–3218

    Article  CAS  PubMed  Google Scholar 

  184. Chiarini F, Lonetti A, Evangelisti C, Buontempo F, Orsini E, Evangelisti C, Cappellini A, Neri LM, McCubrey JA, Martelli AM (2016) Advances in understanding the acute lymphoblastic leukemia bone marrow microenvironment: from biology to therapeutic targeting. Biochim Biophys Acta 1863:449–463

    Article  CAS  PubMed  Google Scholar 

  185. Grimwade D, Ivey A, Huntly BJ (2016) Molecular landscape of acute myeloid leukemia in younger adults and its clinical relevance. Blood 127:29-41

  186. Ferrara F, Zappasodi P, Roncoroni E, Borlenghi E, Rossi G (2020) Impact of COVID-19 on the treatment of acute myeloid leukemia. Leukemia 34:2254-6

  187. Zalpoor H, Bakhtiyari M, Akbari A, Aziziyan F, Shapourian H, Liaghat M, Zare-Badie Z, Yahyazadeh S, Tarhriz V, Ganjalikhani-Hakemi M (2022) Potential role of autophagy induced by FLT3-ITD and acid ceramidase in acute myeloid leukemia chemo-resistance: new insights. Cell Commun Signal 20:172

  188. Deeb G, Vaughan MM, McInnis I, Ford LA, Sait SN, Starostik P, Wetzler M, Mashtare T, Wang ES (2011) Hypoxia-inducible factor-1α protein expression is associated with poor survival in normal karyotype adult acute myeloid leukemia. Leuk Res 35:579–584

    Article  CAS  PubMed  Google Scholar 

  189. Sargazi S, Sheervalilou R, Rokni M, Shirvaliloo M, Shahraki O, Rezaei N (2021) The role of autophagy in controlling SARS-CoV-2 infection: An overview on virophagy-mediated molecular drug targets. Cell Biol Int 45:1599–1612

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Bartlett DL, Howe JR, Chang G, Crago A, Hogg M, Karakousis G, Levine E, Maker A, Mamounas E, McGuire K (2020) Management of cancer surgery cases during the COVID-19 pandemic: considerations. Ann Surg Oncol 27:1717–1720

    Article  PubMed  PubMed Central  Google Scholar 

  191. Zalpoor H, Akbari A, Nayerain Jazi N, Liaghat M, Bakhtiyari M (2022) Possible role of autophagy induced by COVID-19 in cancer progression, chemo-resistance, and tumor recurrence. Infect Agents Cancer 17:38

    Article  CAS  Google Scholar 

  192. Klimienė I, Radzevičius M, Matuzevičienė R, Sinkevič-Belliot K, Kučinskienė ZA, Pečeliūnas V (2021) Adhesion molecule immunophenotype of bone marrow multiple myeloma plasma cells impacts the presence of malignant circulating plasma cells in peripheral blood. Int J Lab Hematol 43:403–408

    Article  PubMed  Google Scholar 

  193. Dhodapkar MV (2023) The immune system in multiple myeloma and precursor states: lessons and implications for immunotherapy and interception. Am J Hematol 98:S4–S12

    Article  CAS  PubMed  Google Scholar 

  194. Akhlaghi T, Maclachlan K, Korde N, Mailankody S, Lesokhin AM, Hassoun H, Lu SX, Patel D, Shah UA, Tan C (2022) African American patients with smoldering multiple myeloma may have a lower risk of progression compared to White patients. Am Soc Clin Oncol 40:8045–8045

    Article  Google Scholar 

  195. Yarmohammadi H, Cunningham-Rundles C (2017) Idiopathic CD4 lymphocytopenia: Pathogenesis, etiologies, clinical presentations and treatment strategies. Ann Allergy Asthma Immunol 119:374-8

  196. Sereno M, Gutiérrez-Gutiérrez G, Sandoval C, Falagan S, Jimenez-Gordo AM, Merino M, López-Menchaca R, Martínez-Martin P, Roa S, Casado E (2020) A favorable outcome of pneumonia COVID 19 in an advanced lung cancer patient with severe neutropenia: is immunosuppression a risk factor for SARS-COV2 infection? Lung Cancer 145:213–215

    Article  PubMed  Google Scholar 

  197. Wang B, Van Oekelen O, Mouhieddine TH, Del Valle DM, Richter J, Cho HJ, Richard S, Chari A, Gnjatic S, Merad M (2020) A tertiary center experience of multiple myeloma patients with COVID-19: lessons learned and the path forward. J Hematol Oncol 13:94

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Ribas A, Dhodapkar MV, Campbell KM, Davies FE, Gore SD, Levy R, Greenberger LM (2021) How to provide the needed protection from COVID-19 to patients with hematologic malignancies. Blood Cancer Discov 2:562–567

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Hasselbalch HC, Elvers M, Schafer AI (2021) The pathobiology of thrombosis, microvascular disease, and hemorrhage in the myeloproliferative neoplasms. Blood 137:2152–2160

    Article  CAS  PubMed  Google Scholar 

  200. Landgren O, Goldin LR, Kristinsson SY, Helgadottir EA, Samuelsson J, Björkholm M (2008) Increased risks of polycythemia vera, essential thrombocythemia, and myelofibrosis among 24 577 first-degree relatives of 11 039 patients with myeloproliferative neoplasms in Sweden. Blood 112:2199–2204

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Veninga A, De Simone I, Heemskerk JW, Ten Cate H, van der Meijden PE (2020) Clonal hematopoietic mutations linked to platelet traits and the risk of thrombosis or bleeding. Haematologica 105:2020–2031

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Ejaz H, Alsrhani A, Zafar A, Javed H, Junaid K, Abdalla AE, Abosalif KO, Ahmed Z, Younas S (2020) COVID-19 and comorbidities: deleterious impact on infected patients. J Infect Public Health 13:1833–1839

    Article  PubMed  PubMed Central  Google Scholar 

  203. Wolach O, Shacham Abulafia A (2021) Can Novel Insights into the Pathogenesis of Myeloproliferative Neoplasm-Related Thrombosis Inform Novel Treatment Approaches?. Hemato 2:305-28.

  204. Murray PG, Young LS (2019) An etiological role for the Epstein-Barr virus in the pathogenesis of classical Hodgkin lymphoma. Blood 134:591–596

    Article  CAS  PubMed  Google Scholar 

  205. Pasin F, Calveri MM, Pizzarelli G, Calabrese A, Andreoli M, Bongiovanni I, Cattaneo C, Rignanese G (2020) Oncolytic effect of SARS-CoV2 in a patient with NK lymphoma. Acta Biomed 91:e2020047

  206. Yousif NG, Oton A, Chetterje K, Gupta M, Shankar M (2022) Complete remission of Hodgkin Lymphoma after a concurrent infection with COVID-19: systematic review and meta-analysis. Muthanna Med J 6:57-67

  207. Osman M, Faridi RM, Sligl W, Shabani-Rad MT, Dharmani-Khan P, Parker A, Kalra A, Tripathi MB, Storek J, Cohen Tervaert JW, Khan FM (2020) Impaired natural killer cell counts and cytolytic activity in patients with severe COVID-19. Blood Adv 4:5035-9

  208. Huang I, Pranata R (2020) Lymphopenia in severe coronavirus disease-2019 (COVID-19): systematic review and meta-analysis. J Intensive Care 8:1-0

  209. van Eeden C, Khan L, Osman MS, Cohen Tervaert JW (2020) Natural killer cell dysfunction and its role in COVID-19. Int J Mol Sci 21:6351

    Article  PubMed  PubMed Central  Google Scholar 

  210. Sparano JA, Lee JY, Kaplan LD, Levine AM, Ramos JC, Ambinder RF, Wachsman W, Aboulafia D, Noy A, Henry DH (2010) Rituximab plus concurrent infusional EPOCH chemotherapy is highly effective in HIV-associated B-cell non-Hodgkin lymphoma. Blood 115:3008–3016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Perry C, Luttwak E, Balaban R, Shefer G, Morales MM, Aharon A, Tabib Y, Cohen YC, Benyamini N, Beyar-Katz O, Neaman M (2021) Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with B-cell non-Hodgkin lymphoma. Blood advances 5:3053–61

  212. Terpos E, Gavriatopoulou M, Fotiou D, Giatra C, Asimakopoulos I, Dimou M, Sklirou AD, Ntanasis-Stathopoulos I, Darmani I, Briasoulis A, Kastritis E (2021) Poor neutralizing antibody responses in 132 patients with CLL, NHL and HL after vaccination against SARS-CoV-2: a prospective study. Cancers 13:4480

  213. Riise J, Meyer S, Blaas I, Chopra A, Tran TT, Delic-Sarac M, Hestdalen ML, Brodin E, Rustad EH, Dai KZ (2022) Rituximab-treated patients with lymphoma develop strong CD8 T-cell responses following COVID-19 vaccination. Br J Haematol 197:697–708

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Passamonti F, Cattaneo C, Arcaini L, Bruna R, Cavo M, Merli F, Angelucci E, Krampera M, Cairoli R, Della Porta MG (2020) Clinical characteristics and risk factors associated with COVID-19 severity in patients with haematological malignancies in Italy: a retrospective, multicentre, cohort study. Lancet Haematol 7:e737–e745

    Article  PubMed  PubMed Central  Google Scholar 

  215. Yigenoglu TN, Ata N, Altuntas F, Bascı S, Dal MS, Korkmaz S, Namdaroglu S, Basturk A, Hacıbekiroglu T, Dogu MH (2021) The outcome of COVID-19 in patients with hematological malignancy. J Med Virol 93:1099–1104

    Article  CAS  PubMed  Google Scholar 

  216. Yang K, Sheng Y, Huang C, Jin Y, Xiong N, Jiang K, Lu H, Liu J, Yang J, Dong Y (2020) Clinical characteristics, outcomes, and risk factors for mortality in patients with cancer and COVID-19 in Hubei, China: a multicentre, retrospective, cohort study. Lancet Oncol 21:904–913

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Sanchez-Pina JM, Rodríguez Rodriguez M, Castro Quismondo N, Gil Manso R, Colmenares R, Gil Alos D, Paciello ML, Zafra D, Garcia-Sanchez C, Villegas C (2020) Clinical course and risk factors for mortality from COVID-19 in patients with haematological malignancies. Eur J Haematol 105:597–607

    Article  CAS  PubMed  Google Scholar 

  218. Bergamaschi G, de BorrelliAndreis F, Aronico N, Lenti MV, Barteselli C, Merli S, Pellegrino I, Coppola L, Cremonte EM, Croce G (2021) Anemia in patients with Covid-19: pathogenesis and clinical significance. Clin Exp Med 21:239–246

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  219. Tao Z, Xu J, Chen W, Yang Z, Xu X, Liu L, Chen R, Xie J, Liu M, Wu J (2021) Anemia is associated with severe illness in COVID-19: a retrospective cohort study. J Med Virol 93:1478–1488

    Article  CAS  PubMed  Google Scholar 

  220. Candoni A, Petruzzellis G, Sperotto A, Andreotti V, Giavarra M, Corvaja C, Minisini A, Comuzzi C, Tascini C, Fanin R (2022) Detection of SARS-CoV-2 infection prevalence in 860 cancer patients with a combined screening procedure including triage, molecular nasopharyngeal swabs and rapid serological test. A report from the first epidemic wave. PloS one 17:e0262784

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Perry C, Luttwak E, Balaban R, Shefer G, Morales MM, Aharon A, Tabib Y, Cohen YC, Benyamini N, Beyar-Katz O, Neaman M (2021) Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with B-cell non-Hodgkin lymphoma. Blood Adv 5:3053-61

  222. Langerbeins P, Hallek M (2022) COVID-19 in patients with hematologic malignancy. Blood 140:236-52

  223. Baden LR, El Sahly HM, Essink B, Kotloff K, Frey S, Novak R, Diemert D, Spector SA, Rouphael N, Creech CB (2021) Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. N Engl J Med 384:403–416

    Article  CAS  PubMed  Google Scholar 

  224. Racine É, Gilca V, Amini R, Tunis M, Ismail S, Sauvageau C (2020) A systematic literature review of the recombinant subunit herpes zoster vaccine use in immunocompromised 18–49 year old patients. Vaccine 38:6205-14

  225. Dooling KL, Guo A, Patel M, Lee GM, Moore K, Belongia EA, Harpaz R (2018) Recommendations of the advisory committee on immunization practices for use of herpes zoster vaccines. Am J Transpl 18:756–762

    Article  Google Scholar 

  226. Dagnew AF, Ilhan O, Lee WS, Woszczyk D, Kwak JY, Bowcock S, Sohn SK, Macías GR, Chiou TJ, Quiel D, Aoun M (2019) Immunogenicity and safety of the adjuvanted recombinant zoster vaccine in adults with haematological malignancies: a phase 3, randomised, clinical trial and post-hoc efficacy analysis. Lancet Infect Dis 19:988-1000

  227. Natarajan K, Prasad N, Dascomb K, Irving SA, Yang DH, Gaglani M, Klein NP, DeSilva MB, Ong TC, Grannis SJ, Stenehjem E (2022) Effectiveness of homologous and heterologous COVID-19 booster doses following 1 Ad. 26. COV2. S (Janssen [Johnson & Johnson]) vaccine dose against COVID-19–associated emergency department and urgent care encounters and hospitalizations among adults—VISION Network, 10 states, December 2021–March 2022. Morbidity and Mortality Weekly Report. 71:495

  228. Livingston EH, Malani PN, Creech CB (2021) The Johnson & Johnson vaccine for COVID-19. Jama 325:1575–1575

    Article  CAS  PubMed  Google Scholar 

  229. Rossignol J, Michallet A, Oberic L, Picard M, Garon A, Willekens C, Dulery R, Leleu X, Cazin B, Ysebaert L (2011) Rituximab–cyclophosphamide–dexamethasone combination in the management of autoimmune cytopenias associated with chronic lymphocytic leukemia. Leukemia 25:473–478

    Article  CAS  PubMed  Google Scholar 

  230. Scheinberg M, Machado LA, Castro LGM, Ferreira SB, Michalany N (2021) Successful treatment of ulcerated pyoderma gangrenosum with baricitinib, a novel JAK inhibitor. J Transl Autoimmun 4:100099

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Campbell R, Chong G, Hawkes EA (2018) Novel indications for Bruton’s tyrosine kinase inhibitors, beyond hematological malignancies. J Clin Med 7(4):62

    Article  PubMed  PubMed Central  Google Scholar 

  232. McGann PT, Ware RE (2015) Hydroxyurea therapy for sickle cell anemia. Exp Opin Drug Saf 14:1749–1758

    Article  CAS  Google Scholar 

  233. Zhang R, Mylonakis E (2021) In inpatients with COVID-19, none of remdesivir, hydroxychloroquine, lopinavir, or interferon β-1a differed from standard care for in-hospital mortality. Ann Intern Med 174:JC17

    Article  PubMed  Google Scholar 

  234. Hendriks RW, Yuvaraj S, Kil LP (2014) Targeting Bruton’s tyrosine kinase in B cell malignancies. Nat Rev Cancer 14:219–232

    Article  CAS  PubMed  Google Scholar 

  235. Pardanani A, Tefferi A (2014) Definition and management of ruxolitinib treatment failure in myelofibrosis. Blood Cancer J 4:e268–e268

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Juárez-Salcedo LM, Desai V, Dalia S (2019) Venetoclax: evidence to date and clinical potential. Drugs Context 8:212574

    Article  PubMed  PubMed Central  Google Scholar 

  237. Ng CM, Bruno R, Combs D, Davies B (2005) Population pharmacokinetics of rituximab (anti-CD20 monoclonal antibody) in rheumatoid arthritis patients during a phase II clinical trial. J Clin Pharmacol 45:792–801

    Article  CAS  PubMed  Google Scholar 

  238. Owen C, Robinson S, Christofides A, Sehn LH (2022) A Canadian perspective: Monoclonal antibodies for pre-and post-exposure protection from COVID-19 in vulnerable patients with hematological malignancies. Curr Oncol 29:3940–3949

    Article  PubMed  PubMed Central  Google Scholar 

  239. Cicchitto G, Cardillo L, de Martinis C, Sabatini P, Marchitiello R, Abate G, Rovetti A, Cavallera A, Apuzzo C, Ferrigno F (2022) Effects of casirivimab/imdevimab monoclonal antibody treatment among vaccinated patients infected by SARS-CoV-2 delta variant. Viruses 14:650

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Xu C, Rafique A, Potocky T, Paccaly A, Nolain P, Lu Q, Iglesias-Rodriguez M, St John G, Nivens MC, Kanamaluru V (2021) Differential binding of Sarilumab and Tocilizumab to IL-6Rα and effects of receptor occupancy on clinical parameters. J Clin Pharmacol 61:714–724

    Article  CAS  PubMed  Google Scholar 

  241. Mouliou DS, Dardiotis E (2022) Temelimab for MS and SARS-CoV-2: could it be a double-edged blessing? Mult Scler Relat Disord 64:103938

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Stricker E, Peckham-Gregory EC, Scheurer ME (2023) HERVs and cancer—A comprehensive review of the relationship of human endogenous retroviruses and human cancers. Biomedicines 11:936

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Mikhael J, Belhadj-Merzoug K, Hulin C, Vincent L, Moreau P, Gasparetto C, Pour L, Spicka I, Vij R, Zonder J (2021) A phase 2 study of isatuximab monotherapy in patients with multiple myeloma who are refractory to daratumumab. Blood cancer journal. 11:89

    Article  PubMed  PubMed Central  Google Scholar 

  244. Scheinberg P, Nunez O, Weinstein B, Scheinberg P, Biancotto A, Wu CO, Young NS (2011) Horse versus rabbit antithymocyte globulin in acquired aplastic anemia. N Engl J Med 365:430–438

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  245. Passweg JR, Baldomero H, Basak GW, Chabannon C, Corbacioglu S, Duarte R, Kuball J, Lankester A, Montoto S, de Latour RP (2019) The EBMT activity survey report 2017: a focus on allogeneic HCT for nonmalignant indications and on the use of non-HCT cell therapies. Bone Marrow Transpl 54:1575–1585

    Article  CAS  Google Scholar 

  246. Shankar R, Radhakrishnan N, Dua S, Arora S, Rana M, Sahu DK, Rai S, Gupta DK (2021) Convalescent plasma to aid in recovery of COVID-19 pneumonia in a child with acute lymphoblastic leukemia. Transfus Apher Sci 60:102956

    Article  PubMed  Google Scholar 

  247. Hashino K, Ishii M, Iemura M, Akagi T, Kato H (2001) Re-treatment for immune globulin-resistant Kawasaki disease: a comparative study of additional immune globulin and steroid pulse therapy. Pediatr Int 43:211–217

    Article  CAS  PubMed  Google Scholar 

  248. Swanson PA, Padilla M, Hoyland W, McGlinchey K, Fields PA, Bibi S, Faust SN, McDermott AB, Lambe T, Pollard AJ (2021) AZD1222/ChAdOx1 nCoV-19 vaccination induces a polyfunctional spike protein–specific TH1 response with a diverse TCR repertoire. Sci Transl Med 13:eabj7211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Ewer KJ, Barrett JR, Belij-Rammerstorfer S, Sharpe H, Makinson R, Morter R, Flaxman A, Wright D, Bellamy D, Bittaye M (2021) T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat Med 27:270–278

    Article  CAS  PubMed  Google Scholar 

  250. Welniak LA, Blazar BR, Murphy WJ (2007) Immunobiology of allogeneic hematopoietic stem cell transplantation. Annu Rev Immunol 25:139-70

  251. Stuver R, Shah GL, Korde NS, Roeker LE, Mato AR, Batlevi CL, Chung DJ, Doddi S, Falchi L, Gyurkocza B (2022) Activity of AZD7442 (tixagevimab-cilgavimab) against Omicron SARS-CoV-2 in patients with hematologic malignancies. Cancer Cell 40:590–591

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Chang A, Koff JL, Lai L, Orellana-Noia VM, Surati M, Leal AM, Ellis ML, Wali B, Moreno A, Linderman SL (2023) Low neutralizing activity of AZD7442 against current SARS-CoV-2 Omicron variants in patients with B cell malignancies. Blood Adv 7:2459–2462

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  253. Otiniano A, van de Wyngaert Z, Brissot E, Dulery R, Gozlan J, Daguenel A, Abi Aad Y, Ricard L, Stocker N, Banet A (2023) Tixagevimab/cilgavimab for Omicron SARS-CoV-2 infection in patients with haematologic diseases. Bone Marrow Transpl 58:340–342

    Article  CAS  Google Scholar 

  254. Scarfò L, Herishanu Y (2022) CLL and COVID-19: light at the end of the tunnel? Blood 140:407–409

    Article  PubMed  PubMed Central  Google Scholar 

  255. Djebbari F, Rampotas A, Vallance G, Panitsas F, Basker N, Sangha G, Salhan B, Karim F, Firas A-K, Gudger A (2022) Infections in relapsed myeloma patients treated with isatuximab plus pomalidomide and dexamethasone during the COVID-19 pandemic: Initial results of a UK-wide real-world study. Hematology 27:691–699

    Article  CAS  PubMed  Google Scholar 

  256. Group RC (2021) Dexamethasone in hospitalized patients with Covid-19. N Engl J Med 384:693–704

    Article  Google Scholar 

  257. Martinez MA (2021) Plitidepsin: a repurposed drug for the treatment of COVID-19. Antimicrob Agents Chemother 65:e00200–00221

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  258. He W, Gao Y, Zhou J, Shi Y, Xia D, Shen H-M (2022) Friend or Foe? Implication of the autophagy-lysosome pathway in SARS-CoV-2 infection and COVID-19. Int J Biol Sci 18:4690

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  259. Papapanou M, Papoutsi E, Giannakas T, Katsaounou P (2021) Plitidepsin: Mechanisms and clinical profile of a promising antiviral agent against COVID-19. J Personal Med 11:668

    Article  Google Scholar 

  260. Dell’Isola GB, Felicioni M, Ferraro L, Capolsini I, Cerri C, Gurdo G, Mastrodicasa E, Massei MS, Perruccio K, Brogna M (2021) Case report: remdesivir and convalescent plasma in a newly acute B lymphoblastic leukemia diagnosis with concomitant sars-CoV-2 infection. Front Pediatr 9:712603

    Article  PubMed  PubMed Central  Google Scholar 

  261. Guo W, Zheng Y, Feng S (2023) Omicron related COVID-19 prevention and treatment measures for patients with hematological malignancy and strategies for modifying hematologic treatment regimes. Frontiers in Cellular and Infection Microbiology 13: 1207225

  262. Pommeret F, Colomba J, Bigenwald C, Laparra A, Bockel S, Bayle A, Michot J-M, Hueso T, Albiges L, Tiberghien P (2021) Bamlanivimab+ etesevimab therapy induces SARS-CoV-2 immune escape mutations and secondary clinical deterioration in COVID-19 patients with B-cell malignancies. Ann Oncol 32:1445–1447

    Article  CAS  PubMed  Google Scholar 

  263. Mao B, Le-Trilling VTK, Wang K, Mennerich D, Hu J, Zhao Z, Zheng J, Deng Y, Katschinski B, Xu S (2022) Obatoclax inhibits SARS-CoV-2 entry by altered endosomal acidification and impaired cathepsin and furin activity in vitro. Emerg Microbes Infect 11:483–497

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Chan M, Holland EC, Gujral TS (2022) Olverembatinib inhibits SARS-CoV-2-Omicron variant-mediated cytokine release in human peripheral blood mononuclear cells. EMBO Mol Med 14:e15919

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  265. Gurung AB, Ali MA, Elshikh MS, Aref I, Amina M, Lee J (2022) An in silico approach unveils the potential of antiviral compounds in preclinical and clinical trials as SARS-CoV-2 omicron inhibitors. Saudi J Biol Sci 29:103297

    Article  Google Scholar 

  266. Lin X, Ke X, Jian X, Xia L, Yang Y, Zhang T, Xiong H, Zhao B, Liu W, Chen Q (2022) Azacytidine targeting SARS-CoV-2 viral RNA as a potential treatment for COVID-19. Sci Bullet 67:1022

    Article  CAS  ADS  Google Scholar 

  267. Carter-Timofte ME, Arulanandam R, Kurmasheva N, Fu K, Laroche G, Taha Z, van Der Horst D, Cassin L, van der Sluis RM, Palermo E (2021) Antiviral potential of the antimicrobial drug atovaquone against SARS-CoV-2 and emerging variants of concern. ACS Infect Dis 7:3034–3051

    Article  CAS  PubMed  Google Scholar 

  268. Parry H, McIlroy G, Bruton R, Damery S, Tyson G, Logan N, Davis C, Willett B, Zuo J, Ali M (2022) Impaired neutralisation of SARS-CoV-2 delta variant in vaccinated patients with B cell chronic lymphocytic leukaemia. J Hematol Oncol 15:1–12

    Article  Google Scholar 

  269. Brullo C, Villa C, Tasso B, Russo E, Spallarossa A (2021) Btk inhibitors: a medicinal chemistry and drug delivery perspective. Int J Mol Sci 22:7641

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Henriquez S, Zerbit J, Bruel T, Ouedrani A, Planas D, Deschamps P, Staropoli I, Hadjadj J, Varet B, Suarez F, Ermark N (2021) Anti-CD38 therapy impairs SARS-CoV-2 vaccine response in multiple myeloma patients. MedRxiv

  271. Nooka AK, Shanmugasundaram U, Cheedarla N, Verkerke H, Edara VV, Valanparambil R, Kaufman JL, Hofmeister CC, Joseph NS, Lonial S, Azeem M (2022) Determinants of neutralizing antibody response after SARS CoV-2 vaccination in patients with myeloma. J Clin Oncol 40:3057

  272. Blixt L, Gao Y, Wullimann D, Murén Ingelman-Sundberg H, Muschiol S, Healy K, Bogdanovic G, Pin E, Nilsson P, Kjellander C (2022) Hybrid immunity in immunocompromised patients with CLL after SARS-CoV-2 infection followed by booster mRNA vaccination. Blood 140:2403–2407

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Xu A, Hong B, Lou F, Wang S, Li W, Shafqat A, An X, Zhao Y, Song L, Tong Y (2022) Sub-lineages of the SARS-CoV-2 Omicron variants: characteristics and prevention. MedComm 3:e172

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  274. Bellusci L, Grubbs G, Srivastava P, Nemeth MJ, Griffiths EA, Golding H, Khurana S (2022) Neutralization of SARS-CoV-2 Omicron after vaccination of patients with myelodysplastic syndromes or acute myeloid leukemia. Blood 139:2842–2846

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  275. Henriquez S, Zerbit J, Bruel T, Ouedrani A, Planas D, Deschamps P, Staropoli I, Hadjadj J, Varet B, Ermak N (2022) Anti-CD38 therapy impairs SARS-CoV-2 vaccine response against alpha and delta variants in patients with multiple myeloma. Blood 139:942–946

    Article  CAS  PubMed  Google Scholar 

  276. Greenberger LM, Saltzman LA, Senefeld JW, Johnson PW, DeGennaro LJ, Nichols GL (2021) Anti-spike antibody response to SARS-CoV-2 booster vaccination in patients with B cell-derived hematologic malignancies. Cancer Cell 39:1297–1299

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Lim SH, Stuart B, Joseph-Pietras D, Johnson M, Campbell N, Kelly A, Jeffrey D, Turaj AH, Rolfvondenbaumen K, Galloway C (2022) Immune responses against SARS-CoV-2 variants after two and three doses of vaccine in B-cell malignancies: UK PROSECO study. Nat Cancer 3:552–564

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  278. Kornek B, Leutmezer F, Rommer PS, Koblischke M, Schneider L, Haslacher H, Thalhammer R, Zimprich F, Zulehner G, Bsteh G (2022) B cell depletion and SARS-CoV-2 vaccine responses in neuroimmunologic patients. Ann Neurol 91:342–352

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  279. Gressens SB, Wiedemann A, Déchenaud M, Dupuis J, Gallien S, Melica G, Haioun C, Lemonnier F, Levy Y (2023) Anti-SARS-CoV-2 cellular response after 2 and 3 doses of BNT162b2 mRNA vaccine in lymphoma patients receiving anti-CD20 antibodies. Vaccine 41:1550–1553

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  280. Terpos E, Rosati M, Bear J, Burns R, Ntanasis-Stathopoulos I, Devasundaram S, Gavriatopoulou M, Kastritis E, Dimopoulos M-A, Pavlakis GN (2022) Antibody Response to COVID-19 mRNA vaccine in patients with multiple myeloma and waldenstrom’s macroglobulinemia after primary immunization and booster: reactivity to the Sars-Cov-2 WT virus, delta and omicron variants. Blood. 140:4334–4335

    Article  Google Scholar 

  281. Fredericks AM, East KW, Shi Y, Liu J, Maschietto F, Ayala A, Cioffi WG, Cohen M, Fairbrother WG, Lefort CT (2022) Identification and mechanistic basis of non-ACE2 blocking neutralizing antibodies from COVID-19 patients with deep RNA sequencing and molecular dynamics simulations. Front Mol Biosci 9:1362

    Article  Google Scholar 

  282. Du W, Hurdiss DL, Drabek D, Mykytyn AZ, Kaiser FK, González-Hernández M, Muñoz-Santos D, Lamers MM, van Haperen R, Li W (2022) An ACE2-blocking antibody confers broad neutralization and protection against Omicron and other SARS-CoV-2 variants of concern. Sci Immunol 7:eabp9312

    Article  CAS  PubMed  Google Scholar 

  283. Denkinger CM, Janssen M, Schaekel U, Gall J, Leo A, Stelmach P, Weber SF, Krisam J, Baumann L, Stermann J (2023) Anti-SARS-CoV-2 antibody-containing plasma improves outcome in patients with hematologic or solid cancer and severe COVID-19: a randomized clinical trial. Nat Cancer 4:96–107

    CAS  PubMed  Google Scholar 

  284. Atanackovic D, Luetkens T, Omili D, Iraguha T, Lutfi F, Hardy NM, Fan X, Avila SV, Saharia KK, Husson JS (2022) Vaccine-induced T-cell responses against SARS-CoV-2 and its Omicron variant in patients with B cell–depleted lymphoma after CART therapy. Blood 140:152–156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  285. Zerbit J, Detroit M, Meyer A, Decroocq J, Deau-Fischer B, Deschamps P, Birsen R, Mondesir J, Franchi P, Miekoutima E (2022) Patients with hematological malignancies treated with T-cell or B-cell immunotherapy remain at high risk of severe forms of COVID-19 in the omicron era. Viruses 14:2377

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  286. Haggenburg S, Hofsink Q, Rutten CE, Nijhof IS, Hazenberg MD, Goorhuis A (2022) SARS-CoV-2 vaccine-induced humoral and cellular immunity in patients with hematologic malignancies. Semin Hematol 59:192–197

    Article  PubMed  PubMed Central  Google Scholar 

  287. Conway SR, Keller MD, Bollard CM (2022) Cellular therapies for the treatment and prevention of SARS-CoV-2 infection. Blood 140:208–221

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Li Z, Yong H, Wang W, Gao Y, Wang P, Chen X, Lu J, Zheng J, Bai J (2023) GSK3326595 is a promising drug to prevent SARS-CoV-2 Omicron and other variants infection by inhibiting ACE2-R671 di-methylation. J Med Virol 95:e28158

    Article  CAS  PubMed  Google Scholar 

  289. Keppler-Hafkemeyer A, Greil C, Wratil PR, Shoumariyeh K, Stern M, Hafkemeyer A, Ashok D, Hollaus A, Lupoli G, Priller A (2023) Potent high-avidity neutralizing antibodies and T cell responses after COVID-19 vaccination in individuals with B cell lymphoma and multiple myeloma. Nat Cancer 4:81–95

    CAS  PubMed  Google Scholar 

  290. Martits-Chalangari K, Spak CW, Askar M, Killian A, Fisher TL, Atillasoy E, Marshall WL, McNeel D, Miller MD, Mathai SK (2022) ALVR109, an off-the-shelf partially HLA matched SARS-CoV-2–specific T cell therapy, to treat refractory severe COVID-19 pneumonia in a heart transplant patient: case report. Am Jo Transpl 22:1261–1265

    Article  CAS  Google Scholar 

  291. He X, Zeng XX (2022) Immunotherapy and CRISPR cas systems: potential cure of COVID-19? Drug Des Devel Ther 16:951–972

    Article  PubMed  PubMed Central  Google Scholar 

  292. Elliott W Jr, Guda MR, Asuthkar S, Teluguakula N, Prasad DV, Tsung AJ, Velpula KK (2021) PAD inhibitors as a potential treatment for SARS-CoV-2 immunothrombosis. Biomedicines 9:1867

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  293. Buske C, Dreyling M, Alvarez-Larrán A, Apperley J, Arcaini L, Besson C, Bullinger L, Corradini P, Della Porta MG, Dimopoulos M (2022) Managing hematological cancer patients during the COVID-19 pandemic: an ESMO-EHA Interdisciplinary Expert Consensus. ESMO open 7:100403

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  294. Yu Y-Q, Herrmann A, Thonn V, Cordsmeier A, Neurath MF, Ensser A, Becker C (2022) SMYD2 inhibition downregulates TMPRSS2 and decreases SARS-CoV-2 infection in human intestinal and airway epithelial cells. Cells 11:1262

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. Hueso T, Godron A-S, Lanoy E, Pacanowski J, Levi LI, Gras E, Surgers L, Guemriche A, Meynard J-L, Pirenne F (2022) Convalescent plasma improves overall survival in patients with B-cell lymphoid malignancy and COVID-19: a longitudinal cohort and propensity score analysis. Leukemia 36:1025–1034

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  296. Palanques-Pastor T, Megías-Vericat JE, Martínez P, López Lorenzo JL, Cornago Navascués J, Rodriguez Macias G, Cano I, Arnan Sangerman M, Vidriales Vicente MB, Algarra Algarra JL (2021) Characteristics, clinical outcomes, and risk factors of SARS-COV-2 infection in adult acute myeloid leukemia patients: experience of the PETHEMA group. Leukemia & Lymphoma. 62:2928–2938

    Article  CAS  Google Scholar 

  297. Weinstein JB, Bates TA, Leier HC, McBride SK, Barklis E, Tafesse FG (2022) A potent alpaca-derived nanobody that neutralizes SARS-CoV-2 variants. Iscience 25:103960

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

Download references

Acknowledgements

We are deeply grateful to BioChem Studio for designing some of the very attractive schematic illustrations in this article. We also are thankful to BioRender for providing such attractive templates.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

S.K., B.H., P.A., F.Z., M.N.A., M.R.F., H.Z., M.A., M.I.S., B.G.F., E.T., and I.Z.: Conceptualization, Data curation, Formal analysis, and Investigation; S.K., F.Z., and I.Z.: Visualization; S.K., B.H., P.A., M.N.A.: Writing—original draft; S.K. and I.Z.: Project administration; Supervision: I.Z. and M.M.; S.K., M.R.F., H.Z., M.A., M.I.S., B.G.F., E.T., I.Z., and MM: Editing; S.K., I.Z., and M.M.: Editing Validation.

Corresponding authors

Correspondence to Iman Zare or Mohammad Motamedifar.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kakavandi, S., Hajikhani, B., Azizi, P. et al. COVID-19 in patients with anemia and haematological malignancies: risk factors, clinical guidelines, and emerging therapeutic approaches. Cell Commun Signal 22, 126 (2024). https://doi.org/10.1186/s12964-023-01316-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12964-023-01316-9

Keywords