Next Article in Journal
Protective Role of a Donepezil-Huprine Hybrid against the β-Amyloid (1-42) Effect on Human Erythrocytes
Previous Article in Journal
The Importance of Food for Endotoxemia and an Inflammatory Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Neutrophilic Granulocytes in Philadelphia Chromosome Negative Myeloproliferative Neoplasms

1
Oncologic Center, Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Paracelsus Medical University, 5020 Salzburg, Austria
2
Cancer Cluster Salzburg, 5020 Salzburg, Austria
3
Salzburg Cancer Research Institute-Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), 5020 Salzburg, Austria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9555; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22179555
Submission received: 9 July 2021 / Revised: 26 August 2021 / Accepted: 30 August 2021 / Published: 3 September 2021
(This article belongs to the Section Molecular Immunology)

Abstract

:
Philadelphia chromosome negative myeloproliferative neoplasms (MPN) are composed of polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). The clinical picture is determined by constitutional symptoms and complications, including arterial and venous thromboembolic or hemorrhagic events. MPNs are characterized by mutations in JAK2, MPL, or CALR, with additional mutations leading to an expansion of myeloid cell lineages and, in PMF, to marrow fibrosis and cytopenias. Chronic inflammation impacting the initiation and expansion of disease in a major way has been described. Neutrophilic granulocytes play a major role in the pathogenesis of thromboembolic events via the secretion of inflammatory markers, as well as via interaction with thrombocytes and the endothelium. In this review, we discuss the molecular biology underlying myeloproliferative neoplasms and point out the central role of leukocytosis and, specifically, neutrophilic granulocytes in this group of disorders.

1. Introduction

Myeloproliferative neoplasms (MPN) form a distinct group of hematologic malignancies. According to the current WHO classification, the spectrum of MPNs include: chronic myeloid leukemia (CML); chronic neutrophilic leukemia (CNL); polycythemia vera (PV); primary myelofibrosis (PMF); essential thrombocythemia (ET); chronic eosinophilic leukemia not otherwise specified (CEL, NOS); and otherwise unclassifiable MPNs (MPN-U) [1]. In this review, we will focus strictly on the three most frequent Philadelphia chromosome (Ph) negative disease entities: PV, ET, and PMF, which are mostly associated with mutations in either the Janus kinase 2 (JAK2) gene, the myelproliferative leukemia virus oncogene (MPL), or the calreticulin (CALR) gene [2,3]. These three entities share common features in their biology and pathophysiology and lead to partially overlapping clinical presentations. Since CML arises from a chromosome 9:22 translocation, commonly referred to as the Philadelphia chromosome, which therefore displays a significantly different biology compared to PV, ET, and PMF, we omitted the further discussion of that entity in our review [4].

2. Clinical Picture and Course of Disease

MPNs are characterized by clonal proliferation, which will be described in Section 3. Although the diagnosis of the exact MPN subtype is based on different criteria regarding peripheral blood cell counts and bone marrow biopsy, clinical symptoms may be very similar. These can include constitutional symptoms (most prominently fatigue and weight loss), bone pain, itching, mood alterations, and abdominal discomfort or pain, as splenomegaly is often present at initial diagnosis [5]. Aquagenic pruritus and erythromelalgia are typical symptoms associated with PV and may precede disease onset for years [6,7]. MPNs represent a biological continuum, and transformation between entities is possible. JAK2-positive PV and ET arguably represent the same disease entity with diagnosis determined by allelic frequency and other genetic factors, which will be described in more depth in Section 3 [8]. Although PMF manifests in previously healthy individuals, the transition from PV or ET to post-PV or -ET myelofibrosis (MF) may eventually occur (10-year risk 4.9–6% in PV and 0.8–4.9% in ET) [9,10]. Additionally, at an advanced stage of disease, the risk of transformation into acute myeloid leukemia (AML) increases [11]. Bleeding and thromboembolic events are common complications that have been associated with leukocytosis; treatment generally aims to reduce the rate of thrombo-hemorrhagic events and clinical symptoms [9,12,13,14]. The risk of transformation into AML does not seem to be lowered through the use of currently available treatment options and depends mainly on the subtype of disease: The 10-year transformation risk is less than 1% in ET, around 3% in PV, and 10–20% in PMF [9,15]. In the following, we will further elucidate the roles of neutrophilic granulocytes in MPNs.

3. Molecular Biology of Ph-Negative Myeloproliferative Neoplasms and Predictive/Prognostic Implications

MPNs are known to originate from a single, mutated hematopoietic stem cell (HSC), termed the MPN stem cell [3,16]. A hallmark of MPNs is the activation of the (MPL-)JAK-STAT pathway [17,18]. Apart from thrombopoiesis, the MPL-signaling axis plays a key-role in HSC renewal [19]. Additionally, JAK2 is involved in erythropoiesis downstream of the EPO receptor, as well as in granulopoiesis downstream of granulocyte colony-stimulating factor (G-CSF) receptor (Figure 1). The activating mutation JAK2V617F [20,21,22,23] is found in almost all patients with PV (with a JAK2 exon 12 deletion in a small fraction of patients) [24] and around 60% of patients with ET or PMF [25,26]. In ET and PMF, MPL mutations are found in 3–10%: most prominent are the mutations MPLW515L and MPLW515K [26], while CALR exon 9 frameshift mutations are detected in 20–25% of patients [26,27]. The latter indirectly leads to a constitutive activation of MPL [28]. In around 8% of patients, no alteration in the aforementioned genes can be found and are called “triple-negative” patients [8].
It is important to note that JAK2V617F mutations with a low allelic fraction can be found in a significant number of healthy subjects, therefore, are a common cause for clonal hematopoiesis of indeterminate potential (CHIP) [29] that may precede clinical disease onset by many years [30]. Arguably, however, it confers an elevated risk for portal and mesenteric vein thrombosis in patients not (yet) fulfilling the diagnostic criteria for any MPN [31,32]. The introduction of a JAK2V617F mutation into an HSC resulted in myeloid-lineage biased HSC leading to erythro- and thrombocytosis in a mouse model but was also associated with increased DNA damage and low disease penetration [33]. JAK2V617F mutations have, however, also been detected within lymphoid cell lineages, implying that malignant stem cells maintain their ability for lymphoid differentiation [34,35]. Even though the JAK2V617F mutation is thought to be a driver of disease in affected patients, commonly used mouse models have not shown clonal expansion at the HSC level [36]. Therefore, additional mutations at the HSC level are necessary before MPNs can develop. This may involve regulators of the JAK-STAT pathway or gene expression via epigenetic modifications or transcription factor mutations [37]. As reviewed by several authors, additional mutations might affect cytokine signaling, splicing machinery, transcription factors, and epigenetic modifiers [38,39,40]. Apparently, germline predisposition might also influence disease penetrance as well as phenotype of MPN. Additionally, it is not only the presence of additional somatic mutations, but also the order in which they are acquired that determines the development of disease [41,42]. The disease phenotype is also in part determined by the allelic frequency. For JAK2V617F, the allelic burden seems to be lowest in ET and highest in MF; similarly, for CALR, the allelic burden is higher in MF than ET [43,44,45]. Several genetic factors carry prognostic implications: A worse prognosis in PV is associated with mutations in ASXL1, SRSF2, and IDH2; in ET, with mutations in SH2B3, SF3B1, U2AF1, TP53, IDH2, and EZH2 [46]. Examples for genes associated with poor prognosis in MF include ASXL-1, SRF2, U2AF1-Q157, TP53, or Ras [8,35,47].
Molecular alterations on the HSC level are the underlying cause of all subtypes of MPN and can be carried across all steps of granulopoiesis, i.e., in common myeloid progenitors, granulocyte monocyte progenitors, promyelocytes, myelocytes, metamyelocytes, and, finally, mature neutrophils.

4. Inflammation and Neutrophilic Granulocytes in MPN

In recent years, several studies pointed out the importance of inflammation and neutrophils in the initiation and promotion of MPN [41,48,49,50,51,52]. This includes the expansion of JAK2V617F mutated HSCs driven by secretion of proinflammatory cytokines by stromal cells of the bone marrow [52]. Longhitano et al. proposed a role for the inflammasome in MPN [53]. For several inflammatory markers, a positive impact on MPN stem cells was found. Tumor necrosis factor-alpha (TNF-alpha) has been shown to be elevated as a result of JAK2V617F mutation and assisted in the expansion of mutant clones, while having a negative effect on the non-mutant HSC [54]. From a murine model, and subsequently from human bone marrow gathered from patients, IL-33 has been identified to induce colony formation on the HSC level and to lead to prolonged survival of JAK2V617F-positive cell lines [55]. In an in vitro study, the expansion of erythroid precursors seemed to be dependent on IL-11 and hepatocyte growth factor (HGF), even in the presence of a JAK2V617F mutation [56]. Similarly, inflammatory processes may further support the malignant clones even after shutting down JAK2 signaling; JAK2V617F-positive cells have been reported to be protected against JAK2 inhibition by IL-6, CXCL10, and fibroblast growth factor (FGF) [57].
The transcription factor NF-E2 is often found to be overexpressed in patients with MPN [58], independently of JAK2 mutational status. As a mechanism, an increased binding of RUNX-1 to the NF-E2 promotor has been described in neutrophils [59]. Increased levels of NF-E2 in mouse models led to leuko-and thrombocytosis [60]. NF-E2 is also induced by IL-1beta, mainly found in PMF and ET [53], and is shown to induce MF and fibrotic tissue formation in vitro [61], as well as to be secreted by JAK2V617F-positive HSC in animal models [62]. Conversely, NF-E2 induces IL-8 [63], which is often elevated in patients with MPN [64,65]. Apart from altering megakaryopoiesis [65], IL-8 has been described as contributing to stem cell mobilization and to neutrophilia [66], as well as stimulating growth of the erythroid lineage [67]. Additionally, granulocytes in MF have been described as a major source of IL-8 themselves [68]. As summarized by Longhitano et al., elevation of many other cytokines, growth factors, and metalloproteases can be found [53]. Importantly, however, in PMF, prognostic values for IL-8, IL-2R, IL-12, IL-15 [69], and CRP (which is directly related to the JAK2V617F allele burden, [70,71]) levels, have been described. Similarly, a correlation between CRP and JAK2V617F has been described in PV and ET [72]. Although cytokines may be produced by malignant clones, JAK-STAT activation does also occur in non-malignant cells and causes them to further support the inflammatory process [68]. YKL-40, a useful biomarker in many diseases with a proinflammatory state, has also been found to be elevated in MPN. Moreover, it increases with disease progression and transition to post-PV MF and correlates with neutrophils, platelets, CRP, LDH, and the JAK2V617F allele burden [48,73,74]. Neutrophil gelatinase-associated lipocalin (NGAL, also known as lipocalin-2), another useful marker in many inflammatory and malignant diseases (esp. CML), has also been shown to be elevated in patients with PV and ET, and to correlate with neutrophil levels [75,76]. Even higher levels of lipocalin-2 were reported in patients with MF [77]. Experiments by two different groups independently described secretion of NGAL in JAK2V617F-harboring cells, leading to a relative increase in proliferation of malignant hematopoietic stem cells compared to non-mutant cells, as the JAK2V617F mutation conferred resistance against NGAL-induced apoptosis [77,78]. NGAL also contributes to bone marrow fibrosis by stimulating proliferation of stromal cells [77]. Table 1 summarizes different mediators of inflammation involved in MPN.
Leukocytosis, and especially an increase in leukocytosis, is a useful biomarker to assess risk for thrombosis and bleeding in patients’ MPN [13,79,80]. Table 2 summarizes critical areas of neutrophil involvement. Neutrophilic leukocytosis has been described as marking the transition to high-risk post-PV MF and to be a negative predictive marker for OS [81]. In the same study, increased granulocytic proliferation within the bone marrow was seen. As reviewed by Falanga et al. in 2005, constant activation of neutrophils in PV and ET leading to facilitated endothelial adhesion and aggregation with thrombocytes is thought to contribute to thrombosis [82]. This is evidenced by the increased expression of CD11b/CD18 (a pattern recognition receptor important for adhesion as well as phagocytosis) by neutrophils obtained from patients with ET and PV [83]. In a mouse model, neutrophilia, and especially neutrophil activation, contributed to plaque formation and accelerated atherosclerosis by adhesion and entry into the plaque [84]. Like other inflammatory states, premature atherosclerosis is thought to occur [50]. As a side note, respiratory burst was observed to be impaired in these models, implicating neutrophil dysfunction [83,85]. Evidence obtained from in vitro experiments further suggests an activation of beta-1 integrins by JAK2V617F, implying a role of the mutation in abnormal endothelial–neutrophil interaction [86]. Subsequently, it was shown that venous embolism and splenic sequestration of neutrophils in JAK2V617F-positive mice was prevented by application of beta integrin neutralizing antibodies [87]. Additionally, the activation of platelets by leukocytes, including neutrophils, is thought to contribute to platelet dysfunction and activation and may contribute to a prothrombotic state [88,89]. An increase in platelet numbers in the absence of leukocytosis does not appear to elevate the risk for thrombosis [90]. In the peripheral blood of patients across all MPN subtypes, an elevation in leukocyte or neutrophil-platelet aggregates has been observed as an expression of their interaction [91,92]. Thrombo-inflammation initiated by neutrophils has become a generally accepted mechanism for thrombosis, not limited to MPN [93]. On a bone marrow level, a model has been suggested, whereby megakaryocytic sequestration of neutrophilic (and eosinophilic) granulocytes results in bone marrow fibrosis via activation of fibroblasts, thus contributing to the pathogenesis of PMF [94]. This process is known as emperipolesis and is a physiologic phenomenon, the significance of which is still poorly understood, which is augmented in inflammatory states [95]. In the aforementioned study, abnormal P-selectin expression was described to mediate neutrophil–megakaryocyte interaction, which led to a release of alpha granules and growth factors [94]. Another research group described a significantly elevated percentage of neutrophil-containing megakaryocytes within the bone marrow and spleen in a GATA-1 PMF mouse model [96]. Here, emperipolesis-mediated degranulation as a mechanism for the induction of fibrosis was confirmed, and the authors described an additional mechanism for fibrosis, whereby paraptosis and neutrophil degranulation induce inflammation via TGF-beta [96]. Almost thirty years ago, a high percentage of bone marrow specimens from patients with different MPN subtypes were reported to show emperipolesis: 75% of patients with PV and 100% of patients with ET [97]. As 75% of cases with reactive thrombocytosis also showed this, a correlation between emperipolesis and thrombocytosis seems plausible [97]. In vitro experiments published in 2019 reported the reliance on neutrophil beta-integrin (CD18) expression for emperipolesis [98]. As thrombopoietin alone was unable to increase emperipolesis, additional neutrophilic activation, as there is during inflammation, was hypothesized to be necessary. Emperipolesis also seemed to stimulate thrombopoiesis. Apart from this, the same study described the interesting phenomenon of neutrophils transporting parts of their membrane onto megakaryocytes and onto circulating platelets.
In a mouse model, thrombosis was attributed to increased neutrophilic extracellular trap (NET) formation and prevented by JAK-inhibitor treatment [99]. By the same research group, the importance of JAK2V617F was underlined by a statistically significant association between healthy individuals carrying it as a CHIP and the occurrence of venous thromboembolism. NETs have been studied in many inflammatory processes, including infectious diseases, inflammatory states, and autoimmune disorders [100]. Although helpful for combating bacteria, predominantly harmful features have been attributed to NETs in sterile inflammation, including promoting the occurrence of malignancies [101]. A role in both arterial and venous thromboembolism in different conditions has been ascribed to NETs [102]. Generally, they are formed by neutrophils upon the release of chromatin together with components of their membrane and granules, which subsequently leads to sequestration of erythrocytes and thrombocytes and activation of the coagulation cascade. Despite the potential relevance of NET formation in MPN, the current literature is not entirely clear on its role, as reviewed by Ferrer-Marín et al. [103]. Of the three studies conducted on samples obtained from MPN patients, only one demonstrated increased NET production by neutrophils [99,103,104,105]. An explanation for this discrepancy may lie in the fact that JAK2 inhibitor treatment appears to downregulate NET formation, and several patients of the two negative studies received treatment with JAK2 inhibitors [103].
The fact that neutrophilia in MPN is at least partially reflective of ongoing inflammation is underlined by newer gene expression profiles obtained from individuals with MPN [106]. Although similarities between neutrophilic activation in MPN and G-CSF stimulation exist, an increased activation of inflammatory pathways in neutrophils obtained from MPN patients, in comparison to metabolic pathways in G-CSF mobilized neutrophils from healthy controls, was observed [106].

5. Principles of Clinical Management and Conclusions

5.1. Phlebotomy and Cytoreductive Therapy

For ET and PV, the prevention of thromboembolic events is a major treatment goal [9]. Evidence supports the use of low-dose aspirin in all patients [107,108]. Aspirin has been shown to decrease the percentage of circulating neutrophil–platelet complexes, thereby reducing the reciprocal activation of these two cell types [91]. In PV, phlebotomy with a hematocrit goal below 45% significantly reduces the risk for cardiovascular events [109]. Cytoreductive therapy (e.g., hydroxyurea) should be used in patients at higher risk [9]. These agents result in a decline of all cell lines, including neutrophilic granulocytes, and might thereby indirectly reduce inflammation. Hydroxyurea may also be used in low-risk PV or ET for aspirin-refractory patients [9]. Additionally, it may be used as treatment of splenomegaly in MF [47]. Moreover, according to a study from 2010, 82% of patients will experience a relief of constitutional symptoms [110]. Pancytopenia, however, may be aggravated by the use of hydroxyurea. A selective reduction of platelets in patients with ET can be achieved with anagrelide, which has been compared to hydroxyurea in ET [111,112]. One randomized trial reported an increase in arterial but a decrease in venous thromboembolic events, and an increased risk for fibrotic transformation for anagrelide compared to hydroxyurea [111]. This implies the involvement of other cell lines in the development of such complications, possibly leukocytes. A more recent clinical trial, however, did not show significant differences in thrombo-hemorrhagic events between patients treated with anagrelide versus hydroxyurea and met the primary endpoint of anagrelide’s non-inferiority [112].

5.2. Interferon

Interferon alpha is another cytoreductive agent which has been shown to improve constitutional symptoms and blood counts, as well as to decrease JAK2 allelic burden [113,114]. Although its exact mechanism of action is still subject to research, interferon alpha is thought to preferentially induce cell death in malignant progenitors and has been believed to lead to a depletion of malignant stem cells, as well as to exert an immunomodulatory effect on several inflammatory cells [115]. Moreover, ropeginterferon alfa-2b (R-2b) is approved independently of prior treatment with hydroxyurea [116]. The PROUD-PV and CONTINUATION-PV phase III clinical trials reported the complete hematologic response and the molecular remission to be higher for R-2b than for hydroxyurea after 36 months of treatment [117]. In another clinical trial, significantly improved control of hematocrit with addition of R-2b to phlebotomy was observed [118].

5.3. JAK Inhibitors

Therapeutic approaches to PV (in case of failure of other available therapies) and PMF include JAK2 inhibitor treatment, most notably ruxolitinib, and, more recently, fedratinib [119]. Importantly, they are effective irrespectively of the presence of a JAK2V617F mutation. A significant reduction in constitutional symptoms was reported by the landmark clinical trials for ruxolitinib in PMF and PV, as well as a significant improvement of splenomegaly and prolongation of OS in PMF and control of hematocrit in PV [120,121,122,123]. In the COMFORT-I trial, 41.9% of MF patients treated with ruxolitinib experienced a reduction in spleen volume of at least 35% (versus 0.7% in the placebo group) and a 45.9% reduction in general symptoms (versus 5.3%) [120]. Similarly, the COMFORT-II trial reported a spleen volume reduction of at least 35% in 28% of patients, compared to 0% of patients receiving best available treatment, and the responses were ongoing during a long-term follow up [123,124]. For PV, in the RESPONSE trial, 21% of ruxolitnib-treated patients reached the primary end point (control of hematocrit and a spleen volume reduction of 35%), compared to only 1% of patients with the best available treatment. In the RESPONSE-II trial, hematocrit control was achieved in 62% with ruxolitinib versus 19% with the best available treatment [121,122]. Currently, the role of JAK inhibitors in the treatment of PV is minor compared to its role in treating MF.
Fedratinib has been approved in ruxolitinib-resistant MF after the phase II JAKARTA-2 trial had reported a decrease in spleen volume by one third in 55% of ruxolitinib-pretreated patients [125]. Additionally, a significant improval of symptoms was described [126]. Whereas fedratinib is a very selective JAK2 inhibitor, ruxolitinib inhibits JAK1 and JAK2 [127].
Two novel JAK inhibitors are currently under investigation: Pancratinib (a JAK2 and FLT3 inhibitor) is effective in the reduction in spleen volume and symptoms in MF, and is especially promising in the treatment of patients with baseline cytopenias, which is usually a major limitation for treatment [128]. It has been shown to be more effective in comparison to the best available treatment, including ruxolitinib, and was also effective in cases of a low JAK2V617F allele burden [129,130].
Momelotinib, apart from being an JAK1/2 inhibitor, antagonizes the effects of hepcidin; therefore, it is thought to become useful in MF with transfusion-requiring anemia. Although it decreased transfusion dependency and was non-inferior to ruxolitinib regarding spleen volume reduction, greater symptom control was achieved with ruxolitinib in a phase III trial [131]. For ruxolitinib-pretreated patients, momelotinib significantly decreased transfusion dependency and symptoms, but did not decrease spleen volume when compared to the best available therapy [132].
Although JAK2 inhibitors provide alleviation of symptoms, their ability to induce complete hematologic remission is limited, and complete molecular remission is not observed [133]. Additionally, disease progression (i.e., transformation into MF and AML) can occur despite therapy. As reviewed by Greenfield and colleagues, an explaination for the clinical benefit of ruxolitinib may lie in the reduction in ongoing inflammation rather than selective inhibition of the disease’s driver mutation, and it has even been described as lacking antitumor activity [47,133]. Unsurprisingly, JAK inhibitors have therefore proven to be effective in a variety of other (auto-)inflammatory conditions [47]. These include hemophagocytic lymphohistiocytosis, which is characterized by overproduction of proinflammatory cytokines and hyperactivation of macrophages, as well as graft-versus-host disease (GvHD) [134]. JAK signaling has been demonstrated to be important for T-cell activation and proliferation in GvHD via the production of proinflammatory cytokines and activation of neutrophilic granulocytes [135]. Clinical trials have reported encouraging results for ruxolitinib in steroid-refractory patients with GvHD [135,136].
The clinical presentation of patients with MPN depends, apart from the stage of disease and comorbidities, on the underlying molecular biology of the disease. We have discussed how mutations in JAK2, MPL, and CALR influence the phenotype of disease and how additional mutations influence the course of disease. An increase in leukocytosis, largely determined by the levels of neutrophils, is a risk factor for the development of thrombotic and hemorrhagic complications [13,79,80]. Mechanisms have been described, according to which neutrophils cause thrombotic events via NET formation and via interactions with endothelial cells and thrombocytes [82,86,99]. Chronic inflammation is understood to impact the initiation and progression of disease in a major way, and neutrophils appear to contribute to bone marrow fibrosis [96]. A further dissection of the complex interactions that neutrophils are involved in is necessary in order to establish novel treatment strategies as the currently available options do not specifically influence these processes.

Author Contributions

Conceptualization, T.M. (Thomas Melchardt), writing—original draft preparation, D.K. and T.M. (Thomas Melchardt); writing—review and editing, D.K., T.M. (Teresa Magnes), S.W., T.M. (Thomas Melchardt), A.E., R.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMLacute myleoid leukemia
CALRcalreticulin
CEL, NOSchronic eosinophilic leukemia not otherwise specified
CHIPclonal hematopoiesis of indetermined potential
CMLchronic myeloid leukemia
CNLchronic neutrophilic leukemia
CRPC-reactive protein
GvHDGraft-versus-Host Disease
FGFFibroblast growth factor
HSChematopoietic stem cell
EPOerythropoietin
ERendoplasmic reticulum
ETessential thrombocythemia
HGFhepatocyte growth factor
JAKJanus kinase
LDHlactate dehydrogenase
MFmyelofibrosis
MPNmyeloproliferative neoplasms
MPLmyeloproliferative leukemia virus oncogene/protein
NGneutrophilic granulocyte
NGALneutrophil gelatinase-associated lipocalin
PMFprimary myelofibrosis
PVpolycythemia vera
OSoverall survival
R-2bropeginterferon alfa-2b
TPOthrombopoietin
TNF-alphaTumor necrosis factor-alpha
YKL-40Chitinase-3-like protein 1

References

  1. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef]
  2. Barbui, T.; Thiele, J.; Gisslinger, H.; Kvasnicka, H.M.; Vannucchi, A.M.; Guglielmelli, P.; Orazi, A.; Tefferi, A. The 2016 WHO classification and diagnostic criteria for myeloproliferative neoplasms: Document summary and in-depth discussion. Blood Cancer J. 2018, 8, 15. [Google Scholar] [CrossRef] [PubMed]
  3. Mead, A.J.; Mullally, A. Myeloproliferative neoplasm stem cells. Blood 2017, 129, 1607–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Jabbour, E.; Kantarjian, H. Chronic myeloid leukemia: 2020 update on diagnosis, therapy and monitoring. Am. J. Hematol. 2020, 95, 691–709. [Google Scholar] [CrossRef] [PubMed]
  5. Scherber, R.; Dueck, A.C.; Johansson, P.; Barbui, T.; Barosi, G.; Vannucchi, A.M.; Passamonti, F.; Andreasson, B.; Ferarri, M.L.; Rambaldi, A.; et al. The myeloproliferative neoplasm symptom assessment form (MPN-SAF): International prospective validation and reliability trial in 402 patients. Blood 2011, 118, 401–408. [Google Scholar] [CrossRef] [PubMed]
  6. Siegel, F.P.; Tauscher, J.; Petrides, P.E. Aquagenic pruritus in polycythemia vera: Characteristics and influence on quality of life in 441 patients. Am. J. Hematol. 2013, 88, 665–669. [Google Scholar] [CrossRef] [PubMed]
  7. Mann, N.; King, T.; Murphy, R. Review of primary and secondary erythromelalgia. Clin. Exp. Derm. 2019, 44, 477–482. [Google Scholar] [CrossRef] [Green Version]
  8. Tefferi, A.; Lasho, T.L.; Finke, C.M.; Knudson, R.A.; Ketterling, R.; Hanson, C.H.; Maffioli, M.; Caramazza, D.; Passamonti, F.; Pardanani, A. CALR vs. JAK2 vs. MPL-mutated or triple-negative myelofibrosis: Clinical, cytogenetic and molecular comparisons. Leukemia 2014, 28, 1472–1477. [Google Scholar] [CrossRef]
  9. Tefferi, A.; Barbui, T. Polycythemia vera and essential thrombocythemia: 2019 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2019, 94, 133–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Cerquozzi, S.; Tefferi, A. Blast transformation and fibrotic progression in polycythemia vera and essential thrombocythemia: A literature review of incidence and risk factors. Blood Cancer J. 2015, 5, e366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Dunbar, A.J.; Rampal, R.K.; Levine, R. Leukemia secondary to myeloproliferative neoplasms. Blood 2020, 136, 61–70. [Google Scholar] [CrossRef]
  12. Finazzi, G. How to manage essential thrombocythemia. Leukemia 2012, 26, 875–882. [Google Scholar] [CrossRef] [Green Version]
  13. Carobbio, A.; Ferrari, A.; Masciulli, A.; Ghirardi, A.; Barosi, G.; Barbui, T. Leukocytosis and thrombosis in essential thrombocythemia and polycythemia vera: A systematic review and meta-analysis. Blood Adv. 2019, 3, 1729–1737. [Google Scholar] [CrossRef] [PubMed]
  14. Bose, P.; Verstovsek, S. Updates in the management of polycythemia vera and essential thrombocythemia. Adv. Hematol. 2019, 10, 2040620719870052. [Google Scholar] [CrossRef] [Green Version]
  15. Iurlo, A.; Cattaneo, D.; Gianelli, U. Blast transformation in myeloproliferative neoplasms: Risk factors, biological findings, and targeted therapeutic options. Int. J. Mol. Sci. 2019, 20, 1839. [Google Scholar] [CrossRef] [Green Version]
  16. Mullally, A.; Poveromo, L.; Schneider, R.K.; Al-Shahrour, F.; Lane, S.W.; Ebert, B.L. Distinct roles for long-term hematopoietic stem cells and erythroid precursor cells in a murine model of Jak2V617F-mediated polycythemia vera. Blood 2012, 120, 166–172. [Google Scholar] [CrossRef] [PubMed]
  17. Sangkhae, V.; Etheridge, S.L.; Kaushansky, K.; Hitchcock, I.S. The thrombopoietin receptor, MPL, is critical for development of a JAK2V617F-induced myeloproliferative neoplasm. Blood 2014, 124, 3956–3963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Schieber, M.; Crispino, J.D.; Stein, B. Myelofibrosis in 2019: Moving beyond JAK2 inhibition. Blood Cancer J. 2019, 9, 74. [Google Scholar] [CrossRef] [PubMed]
  19. Chou, F.S.; Mulloy, J.C. The thrombopoietin/MPL pathway in hematopoiesis and leukemogenesis. J. Cell. Biochem. 2011, 112, 1491–1498. [Google Scholar] [CrossRef]
  20. Baxter, E.J.; Scott, L.M.; Campbell, P.J.; East, C.; Fourouclas, N.; Swanton, S.; Vassiliou, G.S.; Bench, A.J.; Boyd, E.M.; Curtin, N.; et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005, 365, 1054–1061. [Google Scholar] [CrossRef]
  21. James, C.; Ugo, V.; Le Couédic, J.P.; Staerk, J.; Delhommeau, F.; Lacout, C.; Garçon, L.; Raslova, H.; Berger, R.; Bennaceur-Griscelli, A.; et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005, 434, 1144–1148. [Google Scholar] [CrossRef] [PubMed]
  22. Kralovics, R.; Passamonti, F.; Buser, A.S.; Teo, S.S.; Tiedt, R.; Passweg, J.R.; Tichelli, A.; Cazzola, M.; Skoda, R.C. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med. 2005, 352, 1779–1790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Levine, R.L.; Wadleigh, M.; Cools, J.; Ebert, B.L.; Wernig, G.; Huntly, B.J.; Boggon, T.J.; Wlodarska, I.; Clark, J.J.; Moore, S.; et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005, 7, 387–397. [Google Scholar] [CrossRef] [Green Version]
  24. Scott, L.M.; Tong, W.; Levine, R.L.; Scott, M.A.; Beer, P.A.; Stratton, M.R.; Futreal, P.A.; Erber, W.N.; McMullin, M.F.; Harrison, C.N.; et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N. Engl. J. Med. 2007, 356, 459–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tefferi, A.; Pardanani, A. Myeloproliferative neoplasms: A contemporary review. JAMA Oncol. 2015, 1, 97–105. [Google Scholar] [CrossRef] [PubMed]
  26. Tefferi, A. Primary myelofibrosis: 2019 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2018, 93, 1551–1560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Nangalia, J.; Massie, C.E.; Baxter, E.J.; Nice, F.L.; Gundem, G.; Wedge, D.C.; Avezov, E.; Li, J.; Kollmann, K.; Kent, D.G.; et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N. Engl. J. Med. 2013, 369, 2391–2405. [Google Scholar] [CrossRef] [Green Version]
  28. Edahiro, Y.; Araki, M.; Komatsu, N. Mechanism underlying the development of myeloproliferative neoplasms through mutant calreticulin. Cancer Sci. 2020, 111, 2682–2688. [Google Scholar] [CrossRef]
  29. Perner, F.; Perner, C.; Ernst, T.; Heidel, F.H. Roles of JAK2 in aging, inflammation, hematopoiesis and malignant transformation. Cells 2019, 8, 854. [Google Scholar] [CrossRef] [Green Version]
  30. McKerrell, T.; Park, N.; Chi, J.; Collord, G.; Moreno, T.; Ponstingl, H.; Dias, J.; Gerasimou, P.; Melanthiou, K.; Prokopiou, C.; et al. V617F hematopoietic clones are present several years prior to MPN diagnosis and follow different expansion kinetics. Blood Adv. 2017, 1, 968–971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Rao, R.; Grosel, J. Acute portal vein thrombosis in a 59-year-old male with JAK2 V617F mutation. Radiol. Case Rep. 2018, 13, 1249–1255. [Google Scholar] [CrossRef] [PubMed]
  32. Colaizzo, D.; Amitrano, L.; Tiscia, G.L.; Scenna, G.; Grandone, E.; Guardascione, M.A.; Brancaccio, V.; Margaglione, M. The JAK2 V617F mutation frequently occurs in patients with portal and mesenteric venous thrombosis. J. Thromb. Haemost. 2007, 5, 55–61. [Google Scholar] [CrossRef] [PubMed]
  33. Lundberg, P.; Takizawa, H.; Kubovcakova, L.; Guo, G.; Hao-Shen, H.; Dirnhofer, S.; Orkin, S.H.; Manz, M.G.; Skoda, R.C. Myeloproliferative neoplasms can be initiated from a single hematopoietic stem cell expressing JAK2-V617F. J. Exp. Med. 2014, 211, 2213–2230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Delhommeau, F.; Dupont, S.; Tonetti, C.; Massé, A.; Godin, I.; Le Couedic, J.P.; Debili, N.; Saulnier, P.; Casadevall, N.; Vainchenker, W.; et al. Evidence that the JAK2 G1849T (V617F) mutation occurs in a lymphomyeloid progenitor in polycythemia vera and idiopathic myelofibrosis. Blood 2007, 109, 71–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Mylonas, E.; Yoshida, K.; Frick, M.; Hoyer, K.; Christen, F.; Kaeda, J.; Obenaus, M.; Noerenberg, D.; Hennch, C.; Chan, W.; et al. Single-cell analysis based dissection of clonality in myelofibrosis. Nat. Commun. 2020, 11, 73. [Google Scholar] [CrossRef] [Green Version]
  36. Li, J.; Kent, D.G.; Chen, E.; Green, A.R. Mouse models of myeloproliferative neoplasms: JAK of all grades. Dis. Model. Mech. 2011, 4, 311–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Cleary, C.; Kralovics, R. Molecular basis and clonal evolution of myeloproliferative neoplasms. Clin. Chem. Lab. Med. 2013, 51, 1889–1896. [Google Scholar] [CrossRef] [Green Version]
  38. Prick, J.; de Haan, G.; Green, A.R.; Kent, D.G. Clonal heterogeneity as a driver of disease variability in the evolution of myeloproliferative neoplasms. Exp. Hematol. 2014, 42, 841–851. [Google Scholar] [CrossRef]
  39. Viny, A.D.; Levine, R.L. Genetics of myeloproliferative neoplasms. Cancer J. 2014, 20, 61–65. [Google Scholar] [CrossRef] [Green Version]
  40. Them, N.C.; Kralovics, R. Genetic basis of MPN: Beyond JAK2-V617F. Curr. Hematol. Malig. Rep. 2013, 8, 299–306. [Google Scholar] [CrossRef]
  41. Nangalia, J.; Green, A.R. Myeloproliferative neoplasms: From origins to outcomes. Blood 2017, 130, 2475–2483. [Google Scholar] [CrossRef]
  42. Ortmann, C.A.; Kent, D.G.; Nangalia, J.; Silber, Y.; Wedge, D.C.; Grinfeld, J.; Baxter, E.J.; Massie, C.E.; Papaemmanuil, E.; Menon, S.; et al. Effect of mutation order on myeloproliferative neoplasms. N. Engl. J. Med. 2015, 372, 601–612. [Google Scholar] [CrossRef] [Green Version]
  43. Larsen, T.S.; Pallisgaard, N.; Møller, M.B.; Hasselbalch, H.C. The JAK2 V617F allele burden in essential thrombocythemia, polycythemia vera and primary myelofibrosis--impact on disease phenotype. Eur. J. Haematol. 2007, 79, 508–515. [Google Scholar] [CrossRef] [PubMed]
  44. Passamonti, F.; Rumi, E.; Pietra, D.; Della Porta, M.G.; Boveri, E.; Pascutto, C.; Vanelli, L.; Arcaini, L.; Burcheri, S.; Malcovati, L.; et al. Relation between JAK2 (V617F) mutation status, granulocyte activation, and constitutive mobilization of CD34+ cells into peripheral blood in myeloproliferative disorders. Blood 2006, 107, 3676–3682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Rumi, E.; Pietra, D.; Ferretti, V.; Klampfl, T.; Harutyunyan, A.S.; Milosevic, J.D.; Them, N.C.; Berg, T.; Elena, C.; Casetti, I.C.; et al. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood 2014, 123, 1544–1551. [Google Scholar] [CrossRef]
  46. Tefferi, A.; Lasho, T.L.; Guglielmelli, P.; Finke, C.M.; Rotunno, G.; Elala, Y.; Pacilli, A.; Hanson, C.A.; Pancrazzi, A.; Ketterling, R.P.; et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv. 2016, 1, 21–30. [Google Scholar] [CrossRef] [Green Version]
  47. Tefferi, A. Primary myelofibrosis: 2021 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2021, 96, 145–162. [Google Scholar] [CrossRef] [PubMed]
  48. Hasselbalch, H.C.; Bjørn, M.E. MPNs as inflammatory diseases: The evidence, consequences, and perspectives. Mediat. Inflamm. 2015, 2015, 102476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hasselbalch, H.C. Chronic inflammation as a promotor of mutagenesis in essential thrombocythemia, polycythemia vera and myelofibrosis. A human inflammation model for cancer development? Leuk. Res. 2013, 37, 214–220. [Google Scholar] [CrossRef] [PubMed]
  50. Hasselbalch, H.C. Perspectives on chronic inflammation in essential thrombocythemia, polycythemia vera, and myelofibrosis: Is chronic inflammation a trigger and driver of clonal evolution and development of accelerated atherosclerosis and second cancer? Blood 2012, 119, 3219–3225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Lussana, F.; Rambaldi, A. Inflammation and myeloproliferative neoplasms. J. Autoimmun. 2017, 85, 58–63. [Google Scholar] [CrossRef]
  52. Koschmieder, S.; Mughal, T.I.; Hasselbalch, H.C.; Barosi, G.; Valent, P.; Kiladjian, J.J.; Jeryczynski, G.; Gisslinger, H.; Jutzi, J.S.; Pahl, H.L.; et al. Myeloproliferative neoplasms and inflammation: Whether to target the malignant clone or the inflammatory process or both. Leukemia 2016, 30, 1018–1024. [Google Scholar] [CrossRef]
  53. Longhitano, L.; Li Volti, G.; Giallongo, C.; Spampinato, M.; Barbagallo, I.; Di Rosa, M.; Romano, A.; Avola, R.; Tibullo, D.; Palumbo, G.A. The role of inflammation and inflammasome in myeloproliferative disease. J. Clin. Med. 2020, 9, 2334. [Google Scholar] [CrossRef] [PubMed]
  54. Fleischman, A.G.; Aichberger, K.J.; Luty, S.B.; Bumm, T.G.; Petersen, C.L.; Doratotaj, S.; Vasudevan, K.B.; LaTocha, D.H.; Yang, F.; Press, R.D.; et al. TNFα facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood 2011, 118, 6392–6398. [Google Scholar] [CrossRef]
  55. Mager, L.F.; Riether, C.; Schürch, C.M.; Banz, Y.; Wasmer, M.H.; Stuber, R.; Theocharides, A.P.; Li, X.; Xia, Y.; Saito, H.; et al. IL-33 signaling contributes to the pathogenesis of myeloproliferative neoplasms. J. Clin. Investig. 2015, 125, 2579–2591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Boissinot, M.; Cleyrat, C.; Vilaine, M.; Jacques, Y.; Corre, I.; Hermouet, S. Anti-inflammatory cytokines hepatocyte growth factor and interleukin-11 are over-expressed in Polycythemia vera and contribute to the growth of clonal erythroblasts independently of JAK2V617F. Oncogene 2011, 30, 990–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Manshouri, T.; Estrov, Z.; Quintás-Cardama, A.; Burger, J.; Zhang, Y.; Livun, A.; Knez, L.; Harris, D.; Creighton, C.J.; Kantarjian, H.M.; et al. Bone marrow stroma-secreted cytokines protect JAK2(V617F)-mutated cells from the effects of a JAK2 inhibitor. Cancer Res. 2011, 71, 3831–3840. [Google Scholar] [CrossRef] [Green Version]
  58. Goerttler, P.S.; Kreutz, C.; Donauer, J.; Faller, D.; Maiwald, T.; März, E.; Rumberger, B.; Sparna, T.; Schmitt-Gräff, A.; Wilpert, J.; et al. Gene expression profiling in polycythaemia vera: Overexpression of transcription factor NF-E2. Br. J. Haematol. 2005, 129, 138–150. [Google Scholar] [CrossRef]
  59. Wang, W.; Schwemmers, S.; Hexner, E.O.; Pahl, H.L. AML1 is overexpressed in patients with myeloproliferative neoplasms and mediates JAK2V617F-independent overexpression of NF-E2. Blood 2010, 116, 254–266. [Google Scholar] [CrossRef]
  60. Hasselbalch, H.C. A role of NF-E2 in chronic inflammation and clonal evolution in essential thrombocythemia, polycythemia vera and myelofibrosis? Leuk. Res. 2014, 38, 263–266. [Google Scholar] [CrossRef]
  61. Sollazzo, D.; Forte, D.; Polverelli, N.; Romano, M.; Perricone, M.; Rossi, L.; Ottaviani, E.; Luatti, S.; Martinelli, G.; Vianelli, N.; et al. Crucial factors of the inflammatory microenvironment (IL-1β/TNF-α/TIMP-1) promote the maintenance of the malignant hemopoietic clone of myelofibrosis: An in vitro study. Oncotarget 2016, 7, 43974–43988. [Google Scholar] [CrossRef] [Green Version]
  62. Arranz, L.; Sánchez-Aguilera, A.; Martín-Pérez, D.; Isern, J.; Langa, X.; Tzankov, A.; Lundberg, P.; Muntión, S.; Tzeng, Y.S.; Lai, D.M.; et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature 2014, 512, 78–81. [Google Scholar] [CrossRef]
  63. Wehrle, J.; Seeger, T.S.; Schwemmers, S.; Pfeifer, D.; Bulashevska, A.; Pahl, H.L. Transcription factor nuclear factor erythroid-2 mediates expression of the cytokine interleukin 8, a known predictor of inferior outcome in patients with myeloproliferative neoplasms. Haematologica 2013, 98, 1073–1080. [Google Scholar] [CrossRef] [Green Version]
  64. Hermouet, S.; Godard, A.; Pineau, D.; Corre, I.; Raher, S.; Lippert, E.; Jacques, Y. Abnormal production of interleukin (IL)-11 and IL-8 in polycythaemia vera. Cytokine 2002, 20, 178–183. [Google Scholar] [CrossRef]
  65. Emadi, S.; Clay, D.; Desterke, C.; Guerton, B.; Maquarre, E.; Charpentier, A.; Jasmin, C.; Le Bousse-Kerdilès, M.C. French INSERM Research Network on MMM. IL-8 and its CXCR1 and CXCR2 receptors participate in the control of megakaryocytic proliferation, differentiation, and ploidy in myeloid metaplasia with myelofibrosis. Blood 2005, 105, 464–473. [Google Scholar] [CrossRef] [Green Version]
  66. Laterveer, L.; Lindley, I.J.; Hamilton, M.S.; Willemze, R.; Fibbe, W.E. Interleukin-8 induces rapid mobilization of hematopoietic stem cells with radioprotective capacity and long-term myelolymphoid repopulating ability. Blood 1995, 85, 2269–2275. [Google Scholar] [CrossRef] [Green Version]
  67. Corre-Buscail, I.; Pineau, D.; Boissinot, M.; Hermouet, S. Erythropoietin-independent erythroid colony formation by bone marrow progenitors exposed to interleukin-11 and interleukin-8. Exp. Hematol. 2005, 33, 1299–1308. [Google Scholar] [CrossRef] [PubMed]
  68. Kleppe, M.; Kwak, M.; Koppikar, P.; Riester, M.; Keller, M.; Bastian, L.; Hricik, T.; Bhagwat, N.; McKenney, A.S.; Papalexi, E.; et al. JAK-STAT pathway activation in malignant and nonmalignant cells contributes to MPN pathogenesis and therapeutic response. Cancer Discov. 2015, 5, 316–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Tefferi, A.; Vaidya, R.; Caramazza, D.; Finke, C.; Lasho, T.; Pardanani, A. Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: A comprehensive cytokine profiling study. J. Clin. Oncol. 2011, 29, 1356–1363. [Google Scholar] [CrossRef] [PubMed]
  70. Barosi, G.; Massa, M.; Campanelli, R.; Fois, G.; Catarsi, P.; Viarengo, G.; Villani, L.; Poletto, V.; Bosoni, T.; Magrini, U.; et al. Primary myelofibrosis: Older age and high JAK2V617F allele burden are associated with elevated plasma high-sensitivity C-reactive protein levels and a phenotype of progressive disease. Leuk. Res. 2017, 60, 18–23. [Google Scholar] [CrossRef]
  71. Barbui, T.; Carobbio, A.; Finazzi, G.; Guglielmelli, P.; Salmoiraghi, S.; Rosti, V.; Rambaldi, A.; Vannucchi, A.M.; Barosi, G. Elevated C-reactive protein is associated with shortened leukemia-free survival in patients with myelofibrosis. Leukemia 2013, 27, 2084–2086. [Google Scholar] [CrossRef]
  72. Barbui, T.; Carobbio, A.; Finazzi, G.; Vannucchi, A.M.; Barosi, G.; Antonioli, E.; Guglielmelli, P.; Pancrazzi, A.; Salmoiraghi, S.; Zilio, P.; et al. Inflammation and thrombosis in essential thrombocythemia and polycythemia vera: Different role of C-reactive protein and pentraxin 3. Haematologica 2011, 96, 315–318. [Google Scholar] [CrossRef]
  73. Bjørn, M.E.; Andersen, C.L.; Jensen, M.K.; Hasselbalch, H.C. Circulating YKL-40 in myelofibrosis a potential novel biomarker of disease activity and the inflammatory state. Eur. J. Haematol. 2014, 93, 224–228. [Google Scholar] [CrossRef]
  74. Andersen, C.L.; Bjørn, M.E.; McMullin, M.F.; Harrison, C.; Samuelsson, J.; Ejerblad, E.; Zweegman, S.; Fernandes, S.; Bareford, D.; Knapper, S.; et al. Circulating YKL-40 in patients with essential thrombocythemia and polycythemia vera treated with the novel histone deacetylase inhibitor vorinostat. Leuk. Res. 2014, 38, 816–821. [Google Scholar] [CrossRef]
  75. Allegra, A.; Alonci, A.; Bellomo, G.; Campo, S.; Cannavò, A.; Penna, G.; Russo, S.; Centorrino, R.; Gerace, D.; Petrungaro, A.; et al. Increased serum levels of neutrophil gelatinase-associated lipocalin in patients with essential thrombocythemia and polycythemia vera. Leuk. Lymphoma 2011, 52, 101–107. [Google Scholar] [CrossRef]
  76. Bauvois, B.; Susin, S.A. Revisiting neutrophil gelatinase-associated lipocalin (NGAL) in cancer: Saint or sinner? Cancers 2018, 10, 336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lu, M.; Xia, L.; Liu, Y.C.; Hochman, T.; Bizzari, L.; Aruch, D.; Lew, J.; Weinberg, R.; Goldberg, J.D.; Hoffman, R. Lipocalin produced by myelofibrosis cells affects the fate of both hematopoietic and marrow microenvironmental cells. Blood 2015, 126, 972–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Kagoya, Y.; Yoshimi, A.; Tsuruta-Kishino, T.; Arai, S.; Satoh, T.; Akira, S.; Kurokawa, M. JAK2V617F+ myeloproliferative neoplasm clones evoke paracrine DNA damage to adjacent normal cells through secretion of lipocalin-2. Blood 2014, 124, 2996–3006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Bonicelli, G.; Abdulkarim, K.; Mounier, M.; Johansson, P.; Rossi, C.; Jooste, V.; Andreasson, B.; Maynadié, M.; Girodon, F. Leucocytosis and thrombosis at diagnosis are associated with poor survival in polycythaemia vera: A population-based study of 327 patients. Br. J. Haematol. 2013, 160, 251–254. [Google Scholar] [CrossRef] [Green Version]
  80. Tefferi, A.; Rumi, E.; Finazzi, G.; Gisslinger, H.; Vannucchi, A.M.; Rodeghiero, F.; Randi, M.L.; Vaidya, R.; Cazzola, M.; Rambaldi, A.; et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera: An international study. Leukemia 2013, 27, 1874–1881. [Google Scholar] [CrossRef] [Green Version]
  81. Boiocchi, L.; Gianelli, U.; Iurlo, A.; Fend, F.; Bonzheim, I.; Cattaneo, D.; Knowles, D.M.; Orazi, A. Neutrophilic leukocytosis in advanced stage polycythemia vera: Hematopathologic features and prognostic implications. Mod. Pathol. 2015, 28, 1448–1457. [Google Scholar] [CrossRef]
  82. Falanga, A.; Marchetti, M.; Barbui, T.; Smith, C.W. Pathogenesis of thrombosis in essential thrombocythemia and polycythemia vera: The role of neutrophils. Semin. Hematol. 2005, 42, 239–247. [Google Scholar] [CrossRef]
  83. Burgaleta, C.; González, N.; César, J. Increased CD11/CD18 expression and altered metabolic activity on polymorphonuclear leukocytes from patients with polycythemia vera and essential thrombocythemia. Acta Haematol. 2002, 108, 23–28. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, W.; Liu, W.; Fidler, T.; Wang, Y.; Tang, Y.; Woods, B.; Welch, C.; Cai, B.; Silvestre-Roig, C.; Ai, D.; et al. Macrophage inflammation, erythrophagocytosis, and accelerated atherosclerosis in Jak2. Circ. Res. 2018, 123, e35–e47. [Google Scholar] [CrossRef] [PubMed]
  85. Carulli, G.; Minnucci, S.; Azzara, A.; Gianfaldoni, M.L.; Angiolini, C.; Sagripanti, A.; Ferretti, A.; Ambrogi, F. Neutrophil functions in essential thrombocythemia. Hematol. Pathol. 1995, 9, 37–47. [Google Scholar] [CrossRef]
  86. Gupta, N.; Edelmann, B.; Schnoeder, T.M.; Saalfeld, F.C.; Wolleschak, D.; Kliche, S.; Schraven, B.; Heidel, F.H.; Fischer, T. JAK2-V617F activates β1-integrin-mediated adhesion of granulocytes to vascular cell adhesion molecule 1. Leukemia 2017, 31, 1223–1226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Edelmann, B.; Gupta, N.; Schnoeder, T.M.; Oelschlegel, A.M.; Shahzad, K.; Goldschmidt, J.; Philipsen, L.; Weinert, S.; Ghosh, A.; Saalfeld, F.C.; et al. JAK2-V617F promotes venous thrombosis through β1/β2 integrin activation. J. Clin. Investig. 2018, 128, 4359–4371. [Google Scholar] [CrossRef]
  88. Arellano-Rodrigo, E.; Alvarez-Larrán, A.; Reverter, J.C.; Villamor, N.; Colomer, D.; Cervantes, F. Increased platelet and leukocyte activation as contributing mechanisms for thrombosis in essential thrombocythemia and correlation with the JAK2 mutational status. Haematologica 2006, 91, 169–175. [Google Scholar]
  89. Marin Oyarzún, C.P.; Heller, P.G. Platelets as mediators of thromboinflammation in chronic myeloproliferative neoplasms. Front. Immunol. 2019, 10, 1373. [Google Scholar] [CrossRef] [Green Version]
  90. Campbell, P.J.; MacLean, C.; Beer, P.A.; Buck, G.; Wheatley, K.; Kiladjian, J.J.; Forsyth, C.; Harrison, C.N.; Green, A.R. Correlation of blood counts with vascular complications in essential thrombocythemia: Analysis of the prospective PT1 cohort. Blood 2012, 120, 1409–1411. [Google Scholar] [CrossRef] [Green Version]
  91. Falanga, A.; Marchetti, M.; Vignoli, A.; Balducci, D.; Barbui, T. Leukocyte-platelet interaction in patients with essential thrombocythemia and polycythemia vera. Exp. Hematol. 2005, 33, 523–530. [Google Scholar] [CrossRef]
  92. Alvarez-Larrán, A.; Arellano-Rodrigo, E.; Reverter, J.C.; Domingo, A.; Villamor, N.; Colomer, D.; Cervantes, F. Increased platelet, leukocyte, and coagulation activation in primary myelofibrosis. Ann. Hematol. 2008, 87, 269–276. [Google Scholar] [CrossRef]
  93. Pircher, J.; Engelmann, B.; Massberg, S.; Schulz, C. Platelet-neutrophil crosstalk in atherothrombosis. Thromb. Haemost. 2019, 119, 1274–1282. [Google Scholar] [CrossRef]
  94. Schmitt, A.; Jouault, H.; Guichard, J.; Wendling, F.; Drouin, A.; Cramer, E.M. Pathologic interaction between megakaryocytes and polymorphonuclear leukocytes in myelofibrosis. Blood 2000, 96, 1342–1347. [Google Scholar] [CrossRef]
  95. Cunin, P.; Nigrovic, P.A. Megakaryocyte emperipolesis: A new frontier in cell-in-cell interaction. Platelets 2020, 31, 700–706. [Google Scholar] [CrossRef] [PubMed]
  96. Centurione, L.; Di Baldassarre, A.; Zingariello, M.; Bosco, D.; Gatta, V.; Rana, R.A.; Langella, V.; Di Virgilio, A.; Vannucchi, A.M.; Migliaccio, A.R. Increased and pathologic emperipolesis of neutrophils within megakaryocytes associated with marrow fibrosis in GATA-1(low) mice. Blood 2004, 104, 3573–3580. [Google Scholar] [CrossRef] [Green Version]
  97. Cashell, A.W.; Buss, D.H. The frequency and significance of megakaryocytic emperipolesis in myeloproliferative and reactive states. Ann. Hematol. 1992, 64, 273–276. [Google Scholar] [CrossRef]
  98. Cunin, P.; Bouslama, R.; Machlus, K.R.; Martínez-Bonet, M.; Lee, P.Y.; Wactor, A.; Nelson-Maney, N.; Morris, A.; Guo, L.; Weyrich, A.; et al. Megakaryocyte emperipolesis mediates membrane transfer from intracytoplasmic neutrophils to platelets. Elife 2019, 8. [Google Scholar] [CrossRef] [PubMed]
  99. Wolach, O.; Sellar, R.S.; Martinod, K.; Cherpokova, D.; McConkey, M.; Chappell, R.J.; Silver, A.J.; Adams, D.; Castellano, C.A.; Schneider, R.K.; et al. Increased neutrophil extracellular trap formation promotes thrombosis in myeloproliferative neoplasms. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef] [PubMed]
  101. Jorch, S.K.; Kubes, P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat. Med. 2017, 23, 279–287. [Google Scholar] [CrossRef] [PubMed]
  102. Laridan, E.; Martinod, K.; De Meyer, S.F. Neutrophil extracellular traps in arterial and venous thrombosis. Semin. Thromb. Hemost. 2019, 45, 86–93. [Google Scholar] [CrossRef]
  103. Ferrer-Marín, F.; Cuenca-Zamora, E.J.; Guijarro-Carrillo, P.J.; Teruel-Montoya, R. Emerging role of neutrophils in the thrombosis of chronic myeloproliferative neoplasms. Int. J. Mol. Sci. 2021, 22, 1143. [Google Scholar] [CrossRef]
  104. Marin Oyarzún, C.P.; Carestia, A.; Lev, P.R.; Glembotsky, A.C.; Castro Ríos, M.A.; Moiraghi, B.; Molinas, F.C.; Marta, R.F.; Schattner, M.; Heller, P.G. Neutrophil extracellular trap formation and circulating nucleosomes in patients with chronic myeloproliferative neoplasms. Sci. Rep. 2016, 6, 38738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Guy, A.; Favre, S.; Labrouche-Colomer, S.; Deloison, L.; Gourdou-Latyszenok, V.; Renault, M.A.; Riviere, E.; James, C. High circulating levels of MPO-DNA are associated with thrombosis in patients with MPN. Leukemia 2019, 33, 2544–2548. [Google Scholar] [CrossRef] [PubMed]
  106. Slezak, S.; Jin, P.; Caruccio, L.; Ren, J.; Bennett, M.; Zia, N.; Adams, S.; Wang, E.; Ascensao, J.; Schechter, G.; et al. Gene and microRNA analysis of neutrophils from patients with polycythemia vera and essential thrombocytosis: Down-regulation of micro RNA-1 and -133a. J. Transl. Med. 2009, 7, 39. [Google Scholar] [CrossRef] [Green Version]
  107. Landolfi, R.; Marchioli, R.; Kutti, J.; Gisslinger, H.; Tognoni, G.; Patrono, C.; Barbui, T.; European Collaboration on Low-Dose Aspirin in Polycythemia Vera Investigators. Efficacy and safety of low-dose aspirin in polycythemia vera. N. Engl. J. Med. 2004, 350, 114–124. [Google Scholar] [CrossRef] [Green Version]
  108. Alvarez-Larrán, A.; Cervantes, F.; Pereira, A.; Arellano-Rodrigo, E.; Pérez-Andreu, V.; Hernández-Boluda, J.C.; Ayats, R.; Salvador, C.; Muntañola, A.; Bellosillo, B.; et al. Observation versus antiplatelet therapy as primary prophylaxis for thrombosis in low-risk essential thrombocythemia. Blood 2010, 116, 1205–1210. [Google Scholar] [CrossRef] [Green Version]
  109. Marchioli, R.; Finazzi, G.; Specchia, G.; Cacciola, R.; Cavazzina, R.; Cilloni, D.; De Stefano, V.; Elli, E.; Iurlo, A.; Latagliata, R.; et al. Cardiovascular events and intensity of treatment in polycythemia vera. N. Engl. J. Med. 2013, 368, 22–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Martínez-Trillos, A.; Gaya, A.; Maffioli, M.; Arellano-Rodrigo, E.; Calvo, X.; Díaz-Beyá, M.; Cervantes, F. Efficacy and tolerability of hydroxyurea in the treatment of the hyperproliferative manifestations of myelofibrosis: Results in 40 patients. Ann. Hematol. 2010, 89, 1233–1237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Harrison, C.N.; Campbell, P.J.; Buck, G.; Wheatley, K.; East, C.L.; Bareford, D.; Wilkins, B.S.; van der Walt, J.D.; Reilly, J.T.; Grigg, A.P.; et al. Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N. Engl. J. Med. 2005, 353, 33–45. [Google Scholar] [CrossRef] [Green Version]
  112. Gisslinger, H.; Gotic, M.; Holowiecki, J.; Penka, M.; Thiele, J.; Kvasnicka, H.M.; Kralovics, R.; Petrides, P.E.; Group, A.S. Anagrelide compared with hydroxyurea in WHO-classified essential thrombocythemia: The ANAHYDRET Study, a randomized controlled trial. Blood 2013, 121, 1720–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kiladjian, J.J.; Cassinat, B.; Chevret, S.; Turlure, P.; Cambier, N.; Roussel, M.; Bellucci, S.; Grandchamp, B.; Chomienne, C.; Fenaux, P. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood 2008, 112, 3065–3072. [Google Scholar] [CrossRef]
  114. Quintás-Cardama, A.; Kantarjian, H.; Manshouri, T.; Luthra, R.; Estrov, Z.; Pierce, S.; Richie, M.A.; Borthakur, G.; Konopleva, M.; Cortes, J.; et al. Pegylated interferon alfa-2a yields high rates of hematologic and molecular response in patients with advanced essential thrombocythemia and polycythemia vera. J. Clin. Oncol. 2009, 27, 5418–5424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. How, J.; Hobbs, G. Use of interferon alfa in the treatment of myeloproliferative neoplasms: Perspectives and review of the literature. Cancers 2020, 12, 1954. [Google Scholar] [CrossRef]
  116. Wagner, S.M.; Melchardt, T.; Greil, R. Ropeginterferon alfa-2b for the treatment of patients with polycythemia vera. Drugs Today 2020, 56, 195–202. [Google Scholar] [CrossRef] [PubMed]
  117. Gisslinger, H.; Klade, C.; Georgiev, P.; Krochmalczyk, D.; Gercheva-Kyuchukova, L.; Egyed, M.; Rossiev, V.; Dulicek, P.; Illes, A.; Pylypenko, H.; et al. Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): A randomised, non-inferiority, phase 3 trial and its extension study. Lancet Haematol. 2020, 7, e196–e208. [Google Scholar] [CrossRef]
  118. Barbui, T.; Vannucchi, A.M.; De Stefano, V.; Masciulli, A.; Carobbio, A.; Ferrari, A.; Ghirardi, A.; Rossi, E.; Ciceri, F.; Bonifacio, M.; et al. Ropeginterferon alfa-2b versus phlebotomy in low-risk patients with polycythaemia vera (Low-PV study): A multicentre, randomised phase 2 trial. Lancet Haematol. 2021, 8, e175–e184. [Google Scholar] [CrossRef]
  119. Bose, P.; Verstovsek, S. JAK2 inhibitors for myeloproliferative neoplasms: What is next? Blood 2017, 130, 115–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Verstovsek, S.; Mesa, R.A.; Gotlib, J.; Levy, R.S.; Gupta, V.; DiPersio, J.F.; Catalano, J.V.; Deininger, M.; Miller, C.; Silver, R.T.; et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N. Engl. J. Med. 2012, 366, 799–807. [Google Scholar] [CrossRef] [Green Version]
  121. Vannucchi, A.M.; Kiladjian, J.J.; Griesshammer, M.; Masszi, T.; Durrant, S.; Passamonti, F.; Harrison, C.N.; Pane, F.; Zachee, P.; Mesa, R.; et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N. Engl. J. Med. 2015, 372, 426–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Passamonti, F.; Griesshammer, M.; Palandri, F.; Egyed, M.; Benevolo, G.; Devos, T.; Callum, J.; Vannucchi, A.M.; Sivgin, S.; Bensasson, C.; et al. Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): A randomised, open-label, phase 3b study. Lancet Oncol. 2017, 18, 88–99. [Google Scholar] [CrossRef]
  123. Cervantes, F.; Vannucchi, A.M.; Kiladjian, J.J.; Al-Ali, H.K.; Sirulnik, A.; Stalbovskaya, V.; McQuitty, M.; Hunter, D.S.; Levy, R.S.; Passamonti, F.; et al. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood 2013, 122, 4047–4053. [Google Scholar] [CrossRef] [Green Version]
  124. Harrison, C.; Kiladjian, J.J.; Al-Ali, H.K.; Gisslinger, H.; Waltzman, R.; Stalbovskaya, V.; McQuitty, M.; Hunter, D.S.; Levy, R.; Knoops, L.; et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N. Engl. J. Med. 2012, 366, 787–798. [Google Scholar] [CrossRef] [Green Version]
  125. Harrison, C.N.; Schaap, N.; Vannucchi, A.M.; Kiladjian, J.J.; Tiu, R.V.; Zachee, P.; Jourdan, E.; Winton, E.; Silver, R.T.; Schouten, H.C.; et al. Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): A single-arm, open-label, non-randomised, phase 2, multicentre study. Lancet Haematol. 2017, 4, e317–e324. [Google Scholar] [CrossRef]
  126. Harrison, C.N.; Schaap, N.; Vannucchi, A.M.; Kiladjian, J.J.; Jourdan, E.; Silver, R.T.; Schouten, H.C.; Passamonti, F.; Zweegman, S.; Talpaz, M.; et al. Fedratinib in patients with myelofibrosis previously treated with ruxolitinib: An updated analysis of the JAKARTA2 study using stringent criteria for ruxolitinib failure. Am. J. Hematol. 2020, 95, 594–603. [Google Scholar] [CrossRef]
  127. Zhou, T.; Georgeon, S.; Moser, R.; Moore, D.J.; Caflisch, A.; Hantschel, O. Specificity and mechanism-of-action of the JAK2 tyrosine kinase inhibitors ruxolitinib and SAR302503 (TG101348). Leukemia 2014, 28, 404–407. [Google Scholar] [CrossRef]
  128. Mesa, R.A.; Vannucchi, A.M.; Mead, A.; Egyed, M.; Szoke, A.; Suvorov, A.; Jakucs, J.; Perkins, A.; Prasad, R.; Mayer, J.; et al. Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): An international, randomised, phase 3 trial. Lancet Haematol. 2017, 4, e225–e236. [Google Scholar] [CrossRef]
  129. Tremblay, D.; Mesa, R.; Scott, B.; Buckley, S.; Roman-Torres, K.; Verstovsek, S.; Mascarenhas, J. Pacritinib demonstrates spleen volume reduction in patients with myelofibrosis independent of JAK2V617F allele burden. Blood Adv. 2020, 4, 5929–5935. [Google Scholar] [CrossRef]
  130. Mascarenhas, J.; Hoffman, R.; Talpaz, M.; Gerds, A.T.; Stein, B.; Gupta, V.; Szoke, A.; Drummond, M.; Pristupa, A.; Granston, T.; et al. Pacritinib vs. best available therapy, including ruxolitinib, in patients with myelofibrosis: A randomized clinical trial. JAMA Oncol. 2018, 4, 652–659. [Google Scholar] [CrossRef]
  131. Mesa, R.A.; Kiladjian, J.J.; Catalano, J.V.; Devos, T.; Egyed, M.; Hellmann, A.; McLornan, D.; Shimoda, K.; Winton, E.F.; Deng, W.; et al. SIMPLIFY-1: A phase III randomized trial of momelotinib versus ruxolitinib in janus kinase inhibitor-naïve patients with myelofibrosis. J. Clin. Oncol. 2017, 35, 3844–3850. [Google Scholar] [CrossRef] [PubMed]
  132. Harrison, C.N.; Vannucchi, A.M.; Platzbecker, U.; Cervantes, F.; Gupta, V.; Lavie, D.; Passamonti, F.; Winton, E.F.; Dong, H.; Kawashima, J.; et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): A randomised, open-label, phase 3 trial. Lancet Haematol. 2018, 5, e73–e81. [Google Scholar] [CrossRef]
  133. Greenfield, G.; McPherson, S.; Mills, K.; McMullin, M.F. The ruxolitinib effect: Understanding how molecular pathogenesis and epigenetic dysregulation impact therapeutic efficacy in myeloproliferative neoplasms. J. Transl. Med. 2018, 16, 360. [Google Scholar] [CrossRef]
  134. Wang, J.; Wang, Y.; Wu, L.; Wang, X.; Jin, Z.; Gao, Z.; Wang, Z. Ruxolitinib for refractory/relapsed hemophagocytic lymphohistiocytosis. Haematologica 2020, 105, e210–e212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Zeiser, R.; von Bubnoff, N.; Butler, J.; Mohty, M.; Niederwieser, D.; Or, R.; Szer, J.; Wagner, E.M.; Zuckerman, T.; Mahuzier, B.; et al. Ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease. N. Engl. J. Med. 2020, 382, 1800–1810. [Google Scholar] [CrossRef] [PubMed]
  136. Jagasia, M.; Perales, M.A.; Schroeder, M.A.; Ali, H.; Shah, N.N.; Chen, Y.B.; Fazal, S.; Dawkins, F.W.; Arbushites, M.C.; Tian, C.; et al. Ruxolitinib for the treatment of steroid-refractory acute GVHD (REACH1): A multicenter, open-label phase 2 trial. Blood 2020, 135, 1739–1749. [Google Scholar] [CrossRef]
Figure 1. Schematic overview of hallmark mutations in MPN. JAK2 lies downstream of the EPO receptor, the thrombopoietin receptor (MPL), and the G-CSF receptor. The activation of JAK2 by a JAK2V617F or a JAK2 exon 12 mutations, therefore, enhances signaling downstream of pathways that would normally be activated by the growth factors for erythropoiesis, thrombopoiesis, or granulopoiesis. An activating mutation in MPL leads to increased signaling through the thrombopoietin (TPO)-thrombopoietin receptor-(MPL)-axis. Similarly, a mutation in the chaperone protein CALR leads to a constitutive activation of MPL upon binding of CALR to MPL in the endoplasmic reticulum (ER).
Figure 1. Schematic overview of hallmark mutations in MPN. JAK2 lies downstream of the EPO receptor, the thrombopoietin receptor (MPL), and the G-CSF receptor. The activation of JAK2 by a JAK2V617F or a JAK2 exon 12 mutations, therefore, enhances signaling downstream of pathways that would normally be activated by the growth factors for erythropoiesis, thrombopoiesis, or granulopoiesis. An activating mutation in MPL leads to increased signaling through the thrombopoietin (TPO)-thrombopoietin receptor-(MPL)-axis. Similarly, a mutation in the chaperone protein CALR leads to a constitutive activation of MPL upon binding of CALR to MPL in the endoplasmic reticulum (ER).
Ijms 22 09555 g001
Table 1. Overview of the roles of important inflammatory markers in MPN.
Table 1. Overview of the roles of important inflammatory markers in MPN.
Inflammatory Cell/Organelle/ProteinRole
Bone marrow stromal cells [52]Expansion of JAK2V617F mutant HSC
Inflammasome [53]Inflammation aiding disease progression
TNF-alpha [54]Expansion of JAK2V617F mutant HSC and suppression of non-mutant HSC
IL-33 [55]Prolonged survival of JAK2V617F-positive cell lines
IL-11, HGF [56]Necessary for expansion of erythroid precursors harboring JAK2V617F
IL-6, CXCL10, FGF [57]Protection of malignant clones against JAK2 inhibition
NF-E2 [60,63]Leuko- and thrombocytosis, increases IL-8
IL-8 [65,66,67] 0.75Alteration of megakaryopoiesis, neutrophilia, erythrocytosis
CRP [70,71,72]Correlation with JAK2V617F allele burden in PV, ET, PMFPrognostic marker in PMF
IL-8, IL-2R, IL-12, IL-15 [69]Prognostic markers in PMF
YKL-40 [48,73,74]Correlated with disease progression, transition to post-PV MF, neutrophilia, thrombocytosis, CRP, LDH, JAK2V617 allele burden
Neutrophil gelatinase-associated lipocalin (NGAL) [75,76,77]Correlation with neutrophilia in PV, ET, MF
Table 2. Roles of neutrophilic granulocytes and leukocytes in MPN.
Table 2. Roles of neutrophilic granulocytes and leukocytes in MPN.
Major MechanismArea of Involvement
Increased endothelial adhesion and accelerated atherosclerosis [82,86]Arterial thrombosis
Increased NET formation [99]Thrombosis
Aggregation of thromboyctes with neutrophils [82]Thrombosis
Emperipolesis [94,96]Bone marrow fibrosis, thrombocytosis
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kiem, D.; Wagner, S.; Magnes, T.; Egle, A.; Greil, R.; Melchardt, T. The Role of Neutrophilic Granulocytes in Philadelphia Chromosome Negative Myeloproliferative Neoplasms. Int. J. Mol. Sci. 2021, 22, 9555. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22179555

AMA Style

Kiem D, Wagner S, Magnes T, Egle A, Greil R, Melchardt T. The Role of Neutrophilic Granulocytes in Philadelphia Chromosome Negative Myeloproliferative Neoplasms. International Journal of Molecular Sciences. 2021; 22(17):9555. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22179555

Chicago/Turabian Style

Kiem, Dominik, Sandro Wagner, Teresa Magnes, Alexander Egle, Richard Greil, and Thomas Melchardt. 2021. "The Role of Neutrophilic Granulocytes in Philadelphia Chromosome Negative Myeloproliferative Neoplasms" International Journal of Molecular Sciences 22, no. 17: 9555. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22179555

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

Article Metrics

Back to TopTop