Next Article in Journal
Differentiation of Intracerebral Tumor Entities with Quantitative Contrast Attenuation and Iodine Mapping in Dual-Layer Computed Tomography
Previous Article in Journal
Determination of Changes in Tissue Perfusion at Home with Hyperspectral and Thermal Imaging in the First Six Weeks after Endovascular Therapy in Patients with Peripheral Arterial Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Prognostic Role of Cell Blood Count in Chronic Myeloid Neoplasm and Acute Myeloid Leukemia and Its Possible Implications in Hematopoietic Stem Cell Transplantation

1
Hematology Unit, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
2
Department of Medical Sciences and Public Health, University of Cagliari, S554, km 4500, 09042 Monserrato, Italy
3
Department of Gynecologic Oncology, Businco Hospital, ARNAS G. Brotzu, 09124 Cagliari, Italy
4
Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
*
Author to whom correspondence should be addressed.
Diagnostics 2022, 12(10), 2493; https://doi.org/10.3390/diagnostics12102493
Submission received: 6 September 2022 / Revised: 1 October 2022 / Accepted: 12 October 2022 / Published: 14 October 2022
(This article belongs to the Section Pathology and Molecular Diagnostics)

Abstract

:
Numerous prognostic indexes have been developed in hematological diseases based on patient characteristics and genetic or molecular assessment. However, less attention was paid to more accessible parameters, such as neutrophils, lymphocytes, monocytes, and platelet counts. Although many studies have defined the role of neutrophil-to-lymphocyte or platelet-to-lymphocyte in lymphoid malignancies, few applications exist for myeloid neoplasm or hematopoietic stem cell transplantation procedures. In this review, we synthesized literature data on the prognostic value of count blood cells in myeloid malignancies and hematopoietic stem cell transplantation in the context of classical prognostic factors and clinical outcomes.

1. Introduction

A myeloid neoplasm (MN) refers to a heterogeneous group of hematological diseases affecting hematopoietic stem cells, including acute and chronic forms [1]. Different defects can contribute to the genesis of MN diseases, such as mutations in JAK2, CALR, MPL, FLT3 genes, or chromosome translocations. Therefore, different signaling pathways can be activated at the cellular level to promote an uncontrolled expansion of myeloid cells in the bone marrow [1,2]. The diagnosis of myeloid malignancy requires specific tests such as a trephine bone marrow biopsy and a morphology study of a myelogram [3]. Cytogenetic, molecular biology and cytofluorimetric analysis is also required [2]. The first exam required in hematology is a cellular blood count. (CBC). The test is an inexpensive and easy way to diagnose and monitor myeloid malignancies as well as other diseases.
The prognostic role of the distribution of the different cell populations in CBC has been assessed in different diseases, and in particular, the meaning of parameters such as neutrophil to lymphocyte ratio (NLR) and lymphocyte to monocyte ratio (LMR) has been explored [4,5,6]. High NLR has been shown to predict signals of severity in COVID-19 pneumonia [7] and mortality in coronary events [8]. Notably, NLR is an established parameter used for prognostic stratification in patients with solid tumors [9]. In the field of hematology, studies have evaluated the impact of these parameters focusing mainly on lymphomas [10]. However, the apparent alteration of CBC in myeloid diseases did not result in a debate about the proportion of different types of leukocytes. This review aims to assemble several studies that examined the role of NLR, LMR, platelets to lymphocytes ratio (PLR), absolute lymphocyte count (ALC), and absolute monocyte count (AMC) in myeloid malignancies. In addition, we discussed the role of those parameters in defining prognosis or response to therapy in most common myeloid diseases, such as chronic myeloid leukemia, Philadelphia-negative myeloproliferative neoplasm, myelodysplastic syndromes, and acute myeloid leukemia. Finally, we analyzed the effect of NLR, MLR, PLR AMC, and ACL on different aspects of hematopoietic stem cell transplantation (HSCT). Because of the great variability of the available data in HSCT, no distinction was made in terms of donor type, stem cell donor source or conditioning regimes.

2. Bone Marrow Microenvironment

The bone marrow microenvironment is located in the BM cavity, where hematopoietic cells can interact with multiple cellular components like osteoblasts, fibroblasts, mesenchymal cells, macrophages, and different matrix elements such as adhesion molecules, chemokines, cytokines, and soluble or membrane-bound factors. This relative barrier delimiting this microenvironment is also known as the BM niche [11].
The tumor microenvironment is a dynamical and complex milieu closely connected with all steps of carcinogenesis, composed of matrix and stromal cells, neuroendocrine, adipose, immune, and inflammatory cells, and lymphatic and vascular systems. Hematological malignancies present a slightly diverse microenvironment in comparison to solid tumors. The measurable parameters in the peripheral blood cells, such as neutrophils, lymphocytes, and platelets, may reflect the systemic inflammatory response associated with these tumor-induced microenvironment changes [4,6,9,12].
Indeed, the tumor microenvironment is complex and constantly evolving, and both adaptive and innate immune cells play a critical part in tumorigenesis [13]. Tumor progression is largely dependent on inflammation, which acts as a major driver with related oxidative stress, angiogenesis, matrix remodeling, and specific genetic mutations [14,15]. Similarly, in the hematologic field, alterations in the BM microenvironment have been demonstrated to represent a central step in the development of several myeloid malignancies and influence the peripheral CBC composition [16,17,18]. The composition of intra- and peri-tumoral immune cells affects both antitumor immunity and immunodeficiency. The percentage and location of different T cells influence the patient outcome: high intratumoral CD8+ cells are related to better outcomes [19,20], while a low amount of infiltrating CD8+ and CD4+ lymphocytes can promote relapse or metastasis [10]. Cells of the monocyte lineage are fundamental for the innate immune response and play a central role in the tumor microenvironment. In particular, tumor-associated macrophages (TAMs) can favor cancer cell proliferation, migration, and genetic instability and induce angiogenesis and lymphangiogenesis, which promote metastasis [21,22,23]. They can polarize into M1 or M2 phenotype, depending on different stimuli, which have specific pro- or anti-inflammatory profiles and exert different pro- or anti-tumoral actions [24]. A “reprogrammed” inflammatory BM microenvironment is a usual report in myeloid malignancies. Both malignant hematopoietic cells and proinflammatory cytokines are able to stimulate different categories of stromal cells in the bone marrow, induce their secretory activity, and affect their decreased hematopoiesis-supporting ability [10]. Macrophages and monocytes, as players of the innate immune response, are raised in chronic inflammatory conditions in myeloid malignancies. Other crucial cells are neutrophils, which exert anti-tumor activity but are also able to inhibit the cytotoxic action of lymphocytes. Moreover, granulocytes may foster cancer progression by stimulating changes in stromal cells and inducing the expression of specific cytokines, such as hematopoietic growth factor (HGF) and granulocyte colony-stimulating factor (G-CSF) [25,26].
Abnormally proliferating cancer cells in MPN can induce normal hematopoietic and stromal cells in the BM niche to release proinflammatory mediators, which can create chronic inflammation inside the niche [16].
Indeed, it has been described that the MPN BM stem cell niche is a place of chronic inflammation characterized by raised myeloproliferation, altered neutrophil apoptosis, and, probably, unbalanced neutrophil marginal and reserve pool. The specific alterations of these compartments are induced by the inflammatory stimuli and, in turn, can have a major impact on the resolution of inflammation in the MPN, both in the BM and in the peripheral blood, thus influencing the pathogenesis and progression of the disease [27]. Noteworthy, it has been reported that the JAK2 mutation, at the center of the MPN pathogenesis, activates the STAT3 signaling pathway, which is involved in a variety of inflammatory cytokines expressions causing inflammation and dysfunction of the immune system [28]. Hence the production of antibodies in anti-endothelial cells is associated with an increased risk of thrombosis [29]. In turn, inflammation may promote atheromatous damage and, therefore, the occurrence of thrombotic events, which are the major complication of MPN [30].
Then, high NLR can reveal a reduced number of lymphocytes and an increase of neutrophils in the tumor microenvironment. The absolute neutrophil count might act as an indicator of systemic inflammation, which favors tumor progression. Thus, also in myeloid diseases, NLR and MLR can express the connections between the tumor microenvironment and the host’s immune response and be strictly related to patients’ prognosis.

3. Myeloid Malignancies

3.1. Chronic Myeloid Leukemia

Chronic myeloid leukemia (CML) is a clonal myeloproliferative expansion of transformed, primitive hematopoietic progenitor cells. Interestingly, BCR-ABL1, generated by a reciprocal translocation between chromosomes 9 and 22, has been linked to leukemia’s pathogenesis for the first time. In CML, myeloid progenitor cells expand at different stages of maturation, are released prematurely into the peripheral bloodstream, and settle extramedullary [31]. Since the 2000s, specific treatments such as tyrosine kinase inhibitors (TKI) have dramatically improved the disease’s outcome [32].
There is usually a strong correlation between white blood cell (WBC) count and disease prognosis in patients with CML. CML prognostic scores were first established in the CML based on, among other factors, the basophil and platelets count [33,34]. In contrast, low platelets are associated with higher mortality risk in the ELTS score [35,36]. The prognostic implications of ALC in CML were also investigated [37,38,39]. According to Sasaki et al., there were no significant differences in the cumulative incidence of complete cytogenetic response (CCyR) or molecular response (MR) at different time points. However, finding ALC ≥ 4000/µL at 3 or 6 months after TKI therapy was rare but associated with a decreased overall survival (OS) [37]. In patients treated with dasatinib, a significant increase in large granular lymphocytes (LGL) has been observed in peripheral blood. High levels of LGL were associated with a better response to therapy and excellent outcomes [38]. More recently, a report showed that ALC, AMC, and the LMR were not predictive of molecular response status in CML patients [39].

3.2. Myeloproliferative Neoplasm

Philadelphia-negative myeloproliferative neoplasms (MPNs) include three classical forms of clonal hematological malignancy: polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) [40]. Driver mutations causing MPN include JAK2, CALR, or MPL genes [41]. Clinical features are similar. Myelofibrosis can present either splenomegaly, cytopenias, or overlapping manifestations that lead to a more aggressive disease. Erythrocytosis occurs most often in PV, while thrombocytosis occurs in ET [42].
It is also possible for patients with ET and PV to progress toward myelofibrosis. A thrombotic event and disease progressions are the most severe complications and the leading causes of death in patients with MPNs [43,44,45].
Prognostic scores are based on age and previous thrombotic events [46,47]. The international prognostic score for ET (IPSET) included leukocyte ≥ 11 × 109/L as a parameter associated with a higher risk of thrombotic events [47]. Studies have investigated the role of NLR in preventing ET thrombosis [48,49,50]. According to Hacibekiroglu et al., erythrocyte sedimentation rate, C-reactive protein, RDW, MPV, and NLR in the genesis of thrombosis, are commonly used chronic inflammation indicators [48]. There were significant differences in the main laboratory results between healthy controls and patients. No significant differences were found in the patient cohort between those with and without thrombotic events [48].
An analysis of 70 PV and ET patients found no association among thrombosis, PLR, and NLR [49]. However, in a larger group of 150 ET patients, NLR was the best predictor of thrombosis events. It also reported a high NLR ratio in Jak2-positive patients [50]. Additionally, high NLR was found in patients at high-risk stratification of thrombosis [50]. In PV, leukocytosis played a critical role over time. Indeed, several authors have reported a close relationship between leukocytosis at diagnosis and poor prognosis or high risk of thrombosis [51,52,53]. According to Boiocchi et al., patients with persistent leukocytosis have worse outcomes than those without [54]. In a similar study, Ronner et al. found that persistently elevated leukocytes were associated with increased disease evolution but not thrombotic events [55].
The above suggests that biomarkers like NRL and PLR calculated at disease diagnosis might be a better and easier way of predicting disease outcomes. A recent editorial demonstrated that NRL higher than 3.48 had a shorter time to thrombosis (TTT), and NLR > 2.62 was associated with the lowest OS. Similarly, patients with a high (>148.8) PLR had an inferior OS, and a higher level of PLR > 210.68 was associated with a lower TTT [56]. In addition, the increased value of NLR was an independent predictor of venous thrombosis in a recent revision of the ECLAP trial [57]. Venous thrombosis events were recently associated with a high absolute neutrophil count (ANC) and AMC at diagnosis of MPN [58]. In particular, V617F% ≥ 75% or AMC ≥ 1.5 × 109/L was found to be strongly associated with a higher risk for venous thrombosis in Post-PV MF [59].
MF shows the most aggressive biological behavior among the Ph-MPNs and is associated with the highest mortality rate [60]. Historically, hemogram parameters such as WBC, hemoglobin, or platelets have historically been used to calculate MF scores [61,62,63]. Nevertheless, few studies have examined the correlation between NLR or PLR and MF outcomes [64,65]. There was a significant association between higher NLR and Jak2 mutation, such as ET [64]. A greater NLR and PLR were found in MF patients than in the general population. Among MF patients with higher PLR, a less aggressive disease was revealed with the absence of blast phase disease, constitutional symptoms, smaller spleen size, and lower CRP [64]. Another study showed that low ALC in patients with MF could predict inferior survival [65].

3.3. Myelodysplastic Syndrome

The myelodysplastic syndrome (MDS) is a hematological clonal neoplasm in which ineffective hematopoiesis occurs in one or more blood cell lineages due to dysmorphogenesis. As a result, there were one or more cytopenias [66]. As the disease progressed, prognostic scores were developed on bone marrow blast percentage, karyotype, degree of cytopenia, red blood cell transfusion need, age, and performance status [67,68].
The higher-risk MDS patients are more likely to undergo HSCT because of the risk of leukemic transformation and the very short OS, even without transformation [66]. Adaptive immunity has been linked with an increased interest in MDS prognosis. A different distribution of CBC parameters was observed according to the type of MDS. Notably, high levels of ACL were associated with a specific subgroup of MDS patients with ring sideroblasts, known to have a good prognosis [69]. MDS of multilineage dysplasia, on the other hand, was associated with a lower level of PLR [70]. However, lymphocyte counts were low in therapy-related MDS. In patients with higher-risk MDS (IPSS-R intermediate, high, and very high), there were lower ALC levels compared to lower-risk MDS [69].
Among low-risk MDS patients, an ALC below 1.2 × 109/L was an additional negative prognostic factor. In accordance with the above-mentioned studies, ALC levels < 1.2 × 109/L were associated with poor OS [71,72,73]. Saeed et al. also found that low levels of monocytes were correlated with poor OS in MDS [72]. Following treatment, low baseline neutrophil, monocyte, and lymphocyte levels were associated with an increased risk of infection or bacteremia [74]. Finally, few data are available between CBC and response to treatment. In this context, a low platelet count after therapy may indicate poor response [75].

3.4. Acute Myeloid Leukemia

Acute myeloid leukemia (AML) is a hematological malignancy characterized by the proliferation of abnormal clonal hematopoietic precursors. AML usually occurs de novo in healthy individuals, but it can be triggered by a different hematological disease or from previous chemotherapy or radiotherapy exposure [76]. The clinical condition can manifest as cytopenias or hyperleukocytosis, and it can progress very rapidly. The appropriate therapy depends on the patient’s age, fitness, and prognostic risk score [77,78].
Furthermore, the prognostic importance of CBC has been demonstrated in several studies [79,80,81]. There is a possibility that platelet levels are prognostic in AML. Patients with medium platelet count (50–120 × 109/L) had longer OS and disease-free survival (DFS) than those with a low or high platelet count (<50 × 109/L or >120 × 109/L, respectively) [79]. It appears, however, that treatment response is more variable. Non-response rates were higher in patients with low platelet counts at 21 days after induction treatment [75]. Similarly, a high platelet count at day 14 predicted better outcomes in elderly patients [80]. In contrast, Zhang Y. et al. recently found that for intermediate-risk AML patients treated with chemotherapy, a platelet count < 40 × 109/L was associated with better OS and DFS [81].
Studies focusing on lymphocytes and monocyte cells showed that low AMC and ALC correlate with an increased risk of infectious complications but not death [74]. Monocyte count can predict response and OS in AML patients at different stages of the disease [82].
For instance, Ismail et al. found that an AMC ≥0.8 × 109/L at day 28 after therapy was associated with short OS and DFS [83]. Moreover, a high level of NLR at diagnosis and relapse has been associated with a poor prognosis [84,85]. However, data on the role of ALC are contradictory. According to some authors, higher ALCs assessed before and after the first chemotherapy have poor predictive value [86,87,88,89]. A large study of 1702 AML patients showed that an ALC threshold lower than 1 × 109/L was associated with poor OS and DFS [88]. Other authors have reported better OS and DFS in patients with ALC > 0.35 × 109/L 28 days after starting treatment [83]. Similarly, Keenan et al. found that higher ALC correlates with better OS after each chemotherapy course, even though the difference is more pronounced after induction [90]. As confirmed by additional research, even the HSCT does not seem to improve the prognosis of patients with ALC < 500 × 109/L that had lower OS after chemotherapy [91].
Table 1 and Table 2 summarize the main papers referred to in the article about myeloid malignancies.

4. Hematopoietic Stem Cell Transplantation

Hematopoietic stem cell transplantation (HSCT) has been considered one of the most effective and sometimes the only curative options in the treatment of myeloid hematologic malignancies for decades [92]. HSCT is commonly recommended for CML patients who have failed two or more TKI lineage, but for Ph-negative myeloproliferative neoplasms, it is less indicated [43,93,94]. In contrast, high-risk MDS and AML are the majority of patients to receive this treatment [95,96,97].
The clinical decision to perform an HSCT in these patients is based on balancing the risk of transplant-related mortality, the availability of a suitable donor, and the mortality associated with disease progression and evolution [97,98]. HSCT consists of several phases. The conditioning regime is required to destroy residual hematopoietic activity disease. Following the infusion of the donor, the HSCs engraftment is expected [99,100]. HSCT can be complicated by infections, conditioning-related toxicity, graft failure, and acute or chronic graft versus host disease (GVHD) [98]. Moreover, it can help control minimal residual disease (MRD) as a result of the well-known graft versus leukemia effect (GVL) [101]. Some authors have recommended monitoring the prognosis of HSCT based on CBC. Despite AMC > 0.3–0.5 × 109/L after HSCT being associated with better survival, a more consistent result was seen when lymphocyte subsets were examined [102,103]. Studies have shown that a minimum level of ALC of 300 × 109/L or higher improves OS and is associated with fewer infectious complications after transplantation [104,105,106,107,108,109,110,111]. GVHD is a major cause of morbidity and mortality following allogeneic hematopoietic cell transplantation [112]. ALC and eosinophil count (EC) at the time of chronic GVHD were incorporated into the chronic graft-versus-host disease risk score (CIBMTR). ALC and EC below normal were proven to contribute to adverse OS outcomes [113].
The use of biological drugs to prevent GVHD can affect the outcome of this procedure. Those with high levels of ALC had a worse prognosis when starting the immunomodulating treatment with alemtuzumab [114]. Moreover, higher levels of ALC predict better OS in patients who received high doses of anti-thymocyte globulins (ATG) [115]. Similarly, in a recent report, patients with preconditioning ALC < 500 × 109/L were associated with short OS and higher infectious mortality due to a side effect of excessive doses of ATG and profound T-cell depletion [116]. ALC > 150 × 109/L before ATG infusion correlated with higher rates of acute GVHD requiring steroids and non-relapse mortality [117]. On the contrary, the slow lymphocyte recovery after HSCT (<200 × 109/L) could suggest a change in GVHD treatment based on a higher risk of disease relapse [118,119,120]. Nevertheless, Afzal et al. did not find any correlation between early lymphocyte recovery and the graft versus leukemia effect in pediatric AML patients [121]. In addition, to immune cells, other specific types of immune cells were investigated in HSCT. Natural killers (NK) are among the first type of immune cells to recover after HSCT and are thought to contribute to the graft versus leukemia effect [122]. Indeed, the level of NK was found to be low in patients who relapsed from CML after HSCT [123]. In acute leukemia transplants, higher NK levels were significantly associated with better DFS and TRM than patients with NK below 120/µL [124]. More recently, Minculescu et al. reported that NK >150/mm3 on day +30 predicts better OS, lower TRM, and infections [125]. Table 3 shows the most important articles in this field.

5. Discussion

Complete blood counts are crucial to diagnosing, determining prognosis, and monitoring treatments in hematological malignancy [2,40,42,43]. Thus, distinguishing between changes related to the tumor burden and those related to inflammation is complex but crucial. Compared with the general population, myeloid malignancy exhibited a high NLR, PLR, and AMC [48]. In myeloproliferative neoplasm, high leukocyte levels and NLR were found in patients with thrombotic complications [56]. NLR was an independent predictor of venous thrombosis in PV patients [57]. Overall, this results in a lower OS [56]. Further, inflammation can promote thrombosis, and platelets and neutrophils can contribute together [126,127].
Moreover, an inflammatory BM microenvironment with specific changes in the cells of the monocyte-macrophage lineage and the associated release of different proinflammatory cytokines is able to influence not only the development and progression of MPN by inducing fibrosis, thrombosis, tumor angiogenesis, and metastasis, but also the proportion of pro-tumoral/anti-tumoral immune cells both locally and systemically [16,18]. Thus, peripheral CBC parameters may reflect the peculiar changes of BM microenvironment and the status of associated chronic inflammation, which is emerging as a major driver of MPN evolution, and thus represent significant prognostic parameters.
The variation in immune cell expression defines disease manifestation and treatment response. Lymphocytes are the most influential of these cells. Lymphocyte deficiency can allow cancer cells to escape immune system control [128]. In solid tumors, a low amount of lymphocyte infiltration is associated with relapse or metastasis [129,130]. In CML, the level of circulating LGL is correlated with better outcomes in patients receiving dasatinib treatment [38]. MDS and AML patients with lower levels of ALC have been associated with a poorer prognosis of the disease and shorter OS [71,72,73,91,131]. In the transplant setting, ALC prognostic role has been thoroughly investigated and correlated with the immune response reconstitution, a pivotal role after hematopoietic stem cell transplantation [106,107,110,132].
Several studies have concluded that lymphocyte immune reconstitution begins with NK cells in the first month after HSCT, followed by CD8+ and B, and finally by CD4+ cells, improving the patient’s defense against pathogens. The process can take up to two years [105,132]. Thus, ALC at + 21 and +30 can offer a good assessment of the ability of the donor NK cells to control the residual disease [102,104]. Their killing action is well defined, and a low level in CML transplanted patients is associated with relapse of disease [123].
Furthermore, better lymphocyte reconstitution correlates with better infection control, leading to higher DFS and OS rates. The ALC level affects the response to immunomodulating treatment in conditioning regimens and correlates with a better prognosis and a lower TRM [102,104,124]. There is also a delicate balance between immunological recovery and the risk of complications from GVHD in HSCT [132]. In fact, lymphocytes can contribute to GVHD complications, leading to significant morbidity and mortality rates [117]. Dysregulation of the cell pattern may predict the risk for GVHD, especially when a high CD4/CD8 ratio is found [124].
The investigation of the prognostic role of different CBC parameters, such as NLR, ALC, LMR, and PLR, in myeloproliferative neoplasm, represents an intriguing area of research. The definition of their actual clinical significance may help classify the prognosis of patients affected by these hematological malignancies more precisely. Even in HSCT, those parameters could be significant in predicting the risk of complications such as TRM and GVHD. However, there is currently not enough data to define its specific role. Furthermore, many variables, such as conditioning regimen, donor type, and CSE sources, may improperly influence their interpretation.

6. Conclusions

The predictive and protective role of NRL, PLR, or absolute count of lymphocytes and monocytes in myeloid malignancy has been poorly evaluated. Clearly, this is a simple tool to obtain and may have significant effects. Additional studies are needed.

Author Contributions

Conceptualization, O.M. and C.M.; methodology, B.M.; resources, O.M., B.M. and C.M.; writing—original draft preparation, O.M., B.M. and C.M.; writing—review and editing, G.C., A.M. and G.L.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Coltro, G.; Loscocco, G.G.; Vannucchi, A.M. Classical Philadelphia-negative myeloproliferative neoplasms (MPNs): A continuum of different disease entities. Int. Rev. Cell Mol. Biol. 2021, 365, 1–69. [Google Scholar] [CrossRef] [PubMed]
  2. 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] [PubMed]
  3. Torlakovic, E.E.; Brynes, R.K.; Hyjek, E.; Lee, S.-H.; Kreipe, H.; Kremer, M.; McKenna, R.; Sadahira, Y.; Tzankov, A.; Reis, M.; et al. ICSH guidelines for the standardization of bone marrow immunohistochemistry. Int. J. Lab. Hematol. 2015, 37, 431–449. [Google Scholar] [CrossRef] [PubMed]
  4. Russell, C.D.; Parajuli, A.; Gale, H.J.; Bulteel, N.S.; Schuetz, P.; de Jager, C.P.C.; Loonen, A.J.M.; Merekoulias, G.I.; Baillie, J.K. The utility of peripheral blood leucocyte ratios as biomarkers in infectious diseases: A systematic review and meta-analysis. J. Infect. 2019, 78, 339–348. [Google Scholar] [CrossRef] [Green Version]
  5. Liu, W.; Huang, Z.; Tang, S.; Wei, S.; Zhang, Z. An evaluation of homocysteine, C-reactive protein, lipid levels, neutrophils to lymphocyte ratio in postmenopausal osteopenic women. Gynecol. Endocrinol. 2016, 32, 446–448. [Google Scholar] [CrossRef]
  6. Hajibandeh, S.; Hajibandeh, S.; Hobbs, N.; Mansour, M. Neutrophil-to-lymphocyte ratio predicts acute appendicitis and distinguishes between complicated and uncomplicated appendicitis: A systematic review and meta-analysis. Am. J. Surg. 2020, 219, 154–163. [Google Scholar] [CrossRef] [Green Version]
  7. Xia, X.; Wen, M.; Zhan, S.; He, J.; Chen, W. An increased neutrophil/lymphocyte ratio is an early warning signal of severe COVID-19. Nan Fang Yi Ke Da Xue Xue Bao 2020, 40, 333–336. [Google Scholar] [CrossRef]
  8. Dong, C.-H.; Wang, Z.-M.; Chen, S.-Y. Neutrophil to lymphocyte ratio predict mortality and major adverse cardiac events in acute coronary syndrome: A systematic review and meta-analysis. Clin. Biochem. 2018, 52, 131–136. [Google Scholar] [CrossRef]
  9. Templeton, A.J.; McNamara, M.G.; Šeruga, B.; Vera-Badillo, F.E.; Aneja, P.; Ocaña, A.; Leibowitz-Amit, R.; Sonpavde, G.; Knox, J.J.; Tran, B.; et al. Prognostic role of neutrophil-to-lymphocyte ratio in solid tumors: A systematic review and meta-analysis. J. Natl. Cancer Inst. 2014, 106, dju124. [Google Scholar] [CrossRef] [Green Version]
  10. Stefaniuk, P.; Szymczyk, A.; Podhorecka, M. The Neutrophil to Lymphocyte and Lymphocyte to Monocyte Ratios as New Prognostic Factors in Hematological Malignancies—A Narrative Review. Cancer Manag. Res. 2020, 12, 2961–2977. [Google Scholar] [CrossRef]
  11. Schmitt-Graeff, A.H.; Nitschke, R.; Zeiser, R. The Hematopoietic Niche in Myeloproliferative Neoplasms. Mediat. Inflamm. 2015, 2015, 347270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ethier, J.-L.; Desautels, D.; Templeton, A.; Shah, P.S.; Amir, E. Prognostic role of neutrophil-to-lymphocyte ratio in breast cancer: A systematic review and meta-analysis. Breast Cancer Res. 2017, 19, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  15. Fleischman, A.G. Inflammation as a Driver of Clonal Evolution in Myeloproliferative Neoplasm. Mediat. Inflamm. 2015, 2015, 606819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Chen, P.; Wu, B.; Ji, L.; Zhan, Y.; Li, F.; Cheng, L.; Cao, J.; Chen, H.; Ke, Y.; Min, Z.; et al. Cytokine Consistency Between Bone Marrow and Peripheral Blood in Patients with Philadelphia-Negative Myeloproliferative Neoplasms. Front. Med. 2021, 8, 598182. [Google Scholar] [CrossRef]
  17. Seshadri, M.; Qu, C.-K. Microenvironmental regulation of hematopoietic stem cells and its implications in leukemogenesis. Curr. Opin. Hematol. 2016, 23, 339–345. [Google Scholar] [CrossRef] [Green Version]
  18. Mitroulis, I.; Kalafati, L.; Bornhäuser, M.; Hajishengallis, G.; Chavakis, T. Regulation of the Bone Marrow Niche by Inflammation. Front. Immunol. 2020, 11, 1540. [Google Scholar] [CrossRef]
  19. Dave, S.S.; Wright, G.; Tan, B.; Rosenwald, A.; Gascoyne, R.D.; Chan, W.C.; Fisher, R.I.; Braziel, R.M.; Rimsza, L.M.; Grogan, T.M.; et al. Prediction of survival in follicular lymphoma based on molecular features of tumor-infiltrating immune cells. N. Engl. J. Med. 2004, 351, 2159–2169. [Google Scholar] [CrossRef] [Green Version]
  20. Wahlin, B.E.; Sander, B.; Christensson, B.; Ostenstad, B.; Holte, H.; Brown, P.D.; Sundström, C.; Kimby, E. Entourage: The immune microenvironment following follicular lymphoma. Blood Cancer J. 2012, 2, e52. [Google Scholar] [CrossRef]
  21. Ansell, S.M.; Vonderheide, R.H. Cellular composition of the tumor microenvironment. Am. Soc. Clin. Oncol. Educ. Book 2013, 33, e91–e97. [Google Scholar] [CrossRef]
  22. Wang, J.; Gao, K.; Lei, W.; Dong, L.; Xuan, Q.; Feng, M.; Wang, J.; Ye, X.; Jin, T.; Zhang, Z.; et al. Lymphocyte-to-monocyte ratio is associated with prognosis of diffuse large B-cell lymphoma: Correlation with CD163 positive M2 type tumor-associated macrophages, not PD-1 positive tumor-infiltrating lymphocytes. Oncotarget 2017, 8, 5414–5425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gooden, M.J.M.; de Bock, G.H.; Leffers, N.; Daemen, T.; Nijman, H.W. The prognostic influence of tumour-infiltrating lymphocytes in cancer: A systematic review with meta-analysis. Br. J. Cancer 2011, 105, 93–103. [Google Scholar] [CrossRef] [Green Version]
  24. Liu, J.; Geng, X.; Hou, J.; Wu, G. New insights into M1/M2 macrophages: Key modulators in cancer progression. Cancer Cell Int. 2021, 21, 389. [Google Scholar] [CrossRef] [PubMed]
  25. Dobrenis, K.; Gauthier, L.R.; Barroca, V.; Magnon, C. Granulocyte colony-stimulating factor off-target effect on nerve outgrowth promotes prostate cancer development. Int. J. Cancer 2015, 136, 982–988. [Google Scholar] [CrossRef]
  26. He, K.; Liu, X.; Hoffman, R.D.; Shi, R.-Z.; Lv, G.-Y.; Gao, J.-L. G-CSF/GM-CSF-induced hematopoietic dysregulation in the progression of solid tumors. FEBS Open Bio 2022, 12, 1268–1285. [Google Scholar] [CrossRef] [PubMed]
  27. Marković, D.; Maslovarić, I.; Djikić, D.; Čokić, V.P. Neutrophil Death in Myeloproliferative Neoplasms: Shedding More Light on Neutrophils as a Pathogenic Link to Chronic Inflammation. Int. J. Mol. Sci. 2022, 23, 1490. [Google Scholar] [CrossRef] [PubMed]
  28. 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]
  29. Wautier, M.-P.; El Nemer, W.; Gane, P.; Rain, J.-D.; Cartron, J.-P.; Colin, Y.; Le Van Kim, C.; Wautier, J.-L. Increased adhesion to endothelial cells of erythrocytes from patients with polycythemia vera is mediated by laminin alpha5 chain and Lu/BCAM. Blood 2007, 110, 894–901. [Google Scholar] [CrossRef] [Green Version]
  30. Leiva, O.; Hobbs, G.; Ravid, K.; Libby, P. Cardiovascular Disease in Myeloproliferative Neoplasms. JACC CardioOncol. 2022, 4, 166–182. [Google Scholar] [CrossRef]
  31. Faderl, S.; Talpaz, M.; Estrov, Z.; O’Brien, S.; Kurzrock, R.; Kantarjian, H.M. The biology of chronic myeloid leukemia. N. Engl. J. Med. 1999, 341, 164–172. [Google Scholar] [CrossRef] [PubMed]
  32. Hehlmann, R. Chronic Myeloid Leukemia in 2020. Hemasphere 2020, 4, e468. [Google Scholar] [CrossRef] [PubMed]
  33. Sokal, J.E.; Cox, E.B.; Baccarani, M.; Tura, S.; Gomez, G.A.; Robertson, J.E.; Tso, C.Y.; Braun, T.J.; Clarkson, B.D.; Cervantes, F. Prognostic discrimination in “good-risk” chronic granulocytic leukemia. Blood 1984, 63, 789–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Hasford, J.; Baccarani, M.; Hoffmann, V.; Guilhot, J.; Saussele, S.; Rosti, G.; Guilhot, F.; Porkka, K.; Ossenkoppele, G.; Lindoerfer, D.; et al. Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: The EUTOS score. Blood 2011, 118, 686–692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Pfirrmann, M.; Baccarani, M.; Saussele, S.; Guilhot, J.; Cervantes, F.; Ossenkoppele, G.; Hoffmann, V.S.; Castagnetti, F.; Hasford, J.; Hehlmann, R.; et al. Prognosis of long-term survival considering disease-specific death in patients with chronic myeloid leukemia. Leukemia 2016, 30, 48–56. [Google Scholar] [CrossRef]
  36. Specchia, G.; Pregno, P.; Breccia, M.; Castagnetti, F.; Monagheddu, C.; Bonifacio, M.; Tiribelli, M.; Stagno, F.; Caocci, G.; Martino, B.; et al. Prognostic Factors for Overall Survival in Chronic Myeloid Leukemia Patients: A Multicentric Cohort Study by the Italian CML GIMEMA Network. Front. Oncol. 2021, 11, 739171. [Google Scholar] [CrossRef]
  37. Sasaki, K.; Rodriguez-Rivera, I.I.; Kantarjian, H.M.; O’Brien, S.; Jabbour, E.; Borthakur, G.; Ravandi, F.; Burke, M.J.; Zweidler-McKay, P.A.; Cortes, J.E. Correlation of Lymphocyte Count with Treatment Response to Tyrosine Kinase Inhibitors in Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase. Blood 2014, 124, 4538. [Google Scholar] [CrossRef]
  38. Paydas, S. Dasatinib, large granular lymphocytosis, and pleural effusion: Useful or adverse effect? Crit. Rev. Oncol. Hematol. 2014, 89, 242–247. [Google Scholar] [CrossRef]
  39. Pepedil-Tanrikulu, F.; Buyukkurt, N.; Korur, A.; Sariturk, C.; Aytan, P.; Boga, C.; Ozdogu, H.; Kozanoglu, I. Significance of Lymphocyte Count, Monocyte Count, and Lymphocyte-To-Monocyte Ratio in Predicting Molecular Response in Patients with Chronic Myeloid Leukemia: A Single-Centre Experience. Clin. Lab. 2020, 66, 319–324. [Google Scholar] [CrossRef]
  40. Tremblay, D.; Yacoub, A.; Hoffman, R. Overview of Myeloproliferative Neoplasms: History, Pathogenesis, Diagnostic Criteria, and Complications. Hematol. Oncol. Clin. North Am. 2021, 35, 159–176. [Google Scholar] [CrossRef]
  41. Loscocco, G.G.; Guglielmelli, P.; Vannucchi, A.M. Impact of Mutational Profile on the Management of Myeloproliferative Neoplasms: A Short Review of the Emerging Data. OncoTargets Ther. 2020, 13, 12367–12382. [Google Scholar] [CrossRef] [PubMed]
  42. Barbui, T.; Tefferi, A.; Vannucchi, A.M.; Passamonti, F.; Silver, R.T.; Hoffman, R.; Verstovsek, S.; Mesa, R.; Kiladjian, J.-J.; Hehlmann, R.; et al. Philadelphia chromosome-negative classical myeloproliferative neoplasms: Revised management recommendations from European LeukemiaNet. Leukemia 2018, 32, 1057–1069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Tefferi, A.; Barbui, T. Polycythemia vera and essential thrombocythemia: 2021 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2020, 95, 1599–1613. [Google Scholar] [CrossRef] [PubMed]
  44. Dunbar, A.J.; Rampal, R.K.; Levine, R. Leukemia secondary to myeloproliferative neoplasms. Blood 2020, 136, 61–70. [Google Scholar] [CrossRef] [PubMed]
  45. Schafer, A.I. Thrombotic, Vascular, and Bleeding Complications of the Myeloproliferative Neoplasms. Hematol. Oncol. Clin. North Am. 2021, 35, 305–324. [Google Scholar] [CrossRef] [PubMed]
  46. Marchioli, R.; Finazzi, G.; Landolfi, R.; Kutti, J.; Gisslinger, H.; Patrono, C.; Marilus, R.; Villegas, A.; Tognoni, G.; Barbui, T. Vascular and Neoplastic Risk in a Large Cohort of Patients with Polycythemia Vera. J. Clin. Oncol. 2016, 23, 2224–2232. [Google Scholar] [CrossRef] [PubMed]
  47. Passamonti, F.; Thiele, J.; Girodon, F.; Rumi, E.; Carobbio, A.; Gisslinger, H.; Kvasnicka, H.M.; Ruggeri, M.; Randi, M.L.; Gangat, N.; et al. A prognostic model to predict survival in 867 World Health Organization–defined essential thrombocythemia at diagnosis: A study by the International Working Group on Myelofibrosis Research and Treatment. Blood 2012, 120, 1197–1201. [Google Scholar] [CrossRef]
  48. Hacibekiroglu, T.; Akinci, S.; Basturk, A.; Inal, B.; Guney, T.; Bakanay, S.M.; Dilek, I. Evaluation of Inflammation Parameters in Philadelphia Negative Chronic Myeloproliferative Neoplasia Patients. Asian Pac. J. Cancer Prev. 2015, 16, 5159–5162. [Google Scholar] [CrossRef] [Green Version]
  49. Kocak, M.Z.; Dagli, M.; Ünlü, A. The ratio of platelet/lymphocyte, the ratio of neutrophil/lymphocyte and some haemogram parameters related to thrombosis in essential thrombocytosis and polycythaemia vera. Biomed Res. 2017, 28, 3036–3039. [Google Scholar]
  50. Zhou, D.; Chen, W.; Cheng, H.; Qiao, J.; Zhu, L.; Li, Z.; Xu, K. Clinico-hematological profile and thrombotic/hemorrhagic events in 150 chinese patients with essential thrombocythemia. Leuk. Res. 2018, 69, 1–6. [Google Scholar] [CrossRef]
  51. Landolfi, R.; Di Gennaro, L.; Barbui, T.; De Stefano, V.; Finazzi, G.; Marfisi, R.; Tognoni, G.; Marchioli, R. Leukocytosis as a major thrombotic risk factor in patients with polycythemia vera. Blood 2007, 109, 2446–2452. [Google Scholar] [CrossRef] [PubMed]
  52. Gangat, N.; Strand, J.; Li, C.-Y.; Wu, W.; Pardanani, A.; Tefferi, A. Leucocytosis in polycythaemia vera predicts both inferior survival and leukaemic transformation. Br. J. Haematol. 2007, 138, 354–358. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. 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] [Green Version]
  55. Ronner, L.; Podoltsev, N.; Gotlib, J.; Heaney, M.L.; Kuykendall, A.T.; O’Connell, C.; Shammo, J.; Fleischman, A.G.; Scherber, R.M.; Mesa, R.; et al. Persistent leukocytosis in polycythemia vera is associated with disease evolution but not thrombosis. Blood 2020, 135, 1696–1703. [Google Scholar] [CrossRef] [PubMed]
  56. Krečak, I.; Holik, H.; Morić Perić, M.; Zekanović, I.; Coha, B.; Valovičić Krečak, M.; Gverić-Krečak, V.; Lucijanić, M. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratios as prognostic biomarkers in polycythemia vera. Int. J. Lab. Hematol. 2022, 44, e145–e148. [Google Scholar] [CrossRef]
  57. Carobbio, A.; Vannucchi, A.M.; Guglielmelli, P.; Loscocco, G.G. Neutrophil-to-Lymphocyte Ratio (NLR) Is a Risk Factor for Venous Thrombosis in Polycythemia Vera—ScienceDirect. Available online: https://www.sciencedirect.com/science/article/pii/S0006497121034698 (accessed on 31 July 2022).
  58. Farrukh, F.; Guglielmelli, P.; Loscocco, G.G.; Pardanani, A.; Hanson, C.A.; De Stefano, V.; Barbui, T.; Gangat, N.; Vannucchi, A.M.; Tefferi, A. Deciphering the individual contribution of absolute neutrophil and monocyte counts to thrombosis risk in polycythemia vera and essential thrombocythemia. Am. J. Hematol. 2022, 97, E35–E37. [Google Scholar] [CrossRef] [PubMed]
  59. Teng, G.; Zhou, Y.; Zhang, Y.; Hu, N.; Liu, T.; Han, Y.; Gao, C.; Zhang, L.; Bai, J. Thrombosis in patients with post-polycythemia vera myelofibrosis: Incidence and risk factors. Thromb. Res. 2022, 212, 38–43. [Google Scholar] [CrossRef]
  60. Tefferi, A. Primary myelofibrosis: 2021 update on diagnosis, risk-stratification and management. Am. J. Hematol. 2021, 96, 145–162. [Google Scholar] [CrossRef]
  61. Passamonti, F.; Rumi, E.; Caramella, M.; Elena, C.; Arcaini, L.; Boveri, E.; Del Curto, C.; Pietra, D.; Vanelli, L.; Bernasconi, P.; et al. A dynamic prognostic model to predict survival in post-polycythemia vera myelofibrosis. Blood 2008, 111, 3383–3387. [Google Scholar] [CrossRef] [Green Version]
  62. Gangat, N.; Caramazza, D.; Vaidya, R.; George, G.; Begna, K.; Schwager, S.; Van Dyke, D.; Hanson, C.; Wu, W.; Pardanani, A.; et al. DIPSS plus: A refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J. Clin. Oncol. 2011, 29, 392–397. [Google Scholar] [CrossRef]
  63. Cervantes, F.; Dupriez, B.; Pereira, A.; Passamonti, F.; Reilly, J.T.; Morra, E.; Vannucchi, A.M.; Mesa, R.A.; Demory, J.-L.; Barosi, G.; et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood 2009, 113, 2895–2901. [Google Scholar] [CrossRef] [PubMed]
  64. Lucijanic, M.; Cicic, D.; Stoos-Veic, T.; Pejsa, V.; Lucijanic, J.; Fazlic Dzankic, A.; Vlasac Glasnovic, J.; Soric, E.; Skelin, M.; Kusec, R. Elevated Neutrophil-to-Lymphocyte-ratio and Platelet-to-Lymphocyte Ratio in Myelofibrosis: Inflammatory Biomarkers or Representatives of Myeloproliferation Itself? Anticancer Res. 2018, 38, 3157–3163. [Google Scholar] [CrossRef] [PubMed]
  65. Lucijanic, M.; Veletic, I.; Rahelic, D.; Pejsa, V.; Cicic, D.; Skelin, M.; Livun, A.; Tupek, K.M.; Stoos-Veic, T.; Lucijanic, T.; et al. Assessing serum albumin concentration, lymphocyte count and prognostic nutritional index might improve prognostication in patients with myelofibrosis. Wien. Klin. Wochenschr. 2018, 130, 126–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Garcia-Manero, G.; Chien, K.S.; Montalban-Bravo, G. Myelodysplastic syndromes: 2021 update on diagnosis, risk stratification and management. Am. J. Hematol. 2020, 95, 1399–1420. [Google Scholar] [CrossRef]
  67. Greenberg, P.L.; Tuechler, H.; Schanz, J.; Sanz, G.; Garcia-Manero, G.; Solé, F.; Bennett, J.M.; Bowen, D.; Fenaux, P.; Dreyfus, F.; et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood 2012, 120, 2454–2465. [Google Scholar] [CrossRef] [Green Version]
  68. Greenberg, P.; Cox, C.; LeBeau, M.M.; Fenaux, P.; Morel, P.; Sanz, G.; Sanz, M.; Vallespi, T.; Hamblin, T.; Oscier, D.; et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 1997, 89, 2079–2088. [Google Scholar] [CrossRef]
  69. Silzle, T.; Blum, S.; Schuler, E.; Kaivers, J.; Rudelius, M.; Hildebrande, B.; Gattermann, N.; Haas, R.; Germing, U. Lymphopenia at diagnosis is highly prevalent in myelodysplastic syndromes and has an independent negative prognostic value in IPSS-R-low-risk patients. Blood Cancer J. 2019, 9, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Yikilmaz, A.S.; Akinci, S.; Bakanay, S.M.; Dilek, I. Does the platelet-to-lymphocyte ratio have a prognostic effect in patients with myelodysplastic syndrome? Bratisl. Med. J. Bratisl. Lek. Listy 2020, 121, 422–427. [Google Scholar] [CrossRef]
  71. Jacobs, N.L.; Holtan, S.G.; Porrata, L.F.; Markovic, S.N.; Tefferi, A.; Steensma, D.P. Host immunity affects survival in myelodysplastic syndromes: Independent prognostic value of the absolute lymphocyte count. Am. J. Hematol. 2010, 85, 160–163. [Google Scholar] [CrossRef]
  72. Saeed, L.; Patnaik, M.M.; Begna, K.H.; Al-Kali, A.; Litzow, M.R.; Hanson, C.A.; Ketterling, R.P.; Porrata, L.F.; Pardanani, A.; Gangat, N.; et al. Prognostic relevance of lymphocytopenia, monocytopenia and lymphocyte-to-monocyte ratio in primary myelodysplastic syndromes: A single center experience in 889 patients. Blood Cancer J. 2017, 7, e550. [Google Scholar] [CrossRef]
  73. Saeed, L.; Patnaik, M.M.; Begna, K.H.; Al-Kali, A.; Hanson, C.A.; Litzow, M.R.; Ketterling, R.P.; Porrata, L.; Pardanani, A.; Gangat, N.; et al. Subnormal Lymphocyte Count Predicts Inferior Survival in Myelodysplastic Syndromes: A Single Center Experience in 889 Patients. Blood 2016, 128, 5534. [Google Scholar] [CrossRef]
  74. Buckley, S.A.; Othus, M.; Vainstein, V.; Abkowitz, J.L.; Estey, E.H.; Walter, R.B. Prediction of adverse events during intensive induction chemotherapy for acute myeloid leukemia or high-grade myelodysplastic syndromes. Am. J. Hematol. 2014, 89, 423–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Chen, X.; Newell, L.F.; Xie, H.; Walter, R.B.; Pagel, J.M.; Sandhu, V.K.; Becker, P.S.; Hendrie, P.C.; Abkowitz, J.L.; Appelbaum, F.R.; et al. Low platelet count reduces subsequent complete remission rate despite marrow with <5% blasts after AML induction therapy. Leukemia 2015, 29, 1779–1780. [Google Scholar] [CrossRef] [PubMed]
  76. De Kouchkovsky, I.; Abdul-Hay, M. Acute myeloid leukemia: A comprehensive review and 2016 update. Blood Cancer J. 2016, 6, e441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Estey, E.H. Acute myeloid leukemia: 2019 update on risk-stratification and management. Am. J. Hematol. 2018, 93, 1267–1291. [Google Scholar] [CrossRef] [Green Version]
  78. Mrózek, K.; Marcucci, G.; Nicolet, D.; Maharry, K.S.; Becker, H.; Whitman, S.P.; Metzeler, K.H.; Schwind, S.; Wu, Y.-Z.; Kohlschmidt, J.; et al. Prognostic significance of the European LeukemiaNet standardized system for reporting cytogenetic and molecular alterations in adults with acute myeloid leukemia. J. Clin. Oncol. 2012, 30, 4515–4523. [Google Scholar] [CrossRef]
  79. Zhang, Q.; Dai, K.; Bi, L.; Jiang, S.; Han, Y.; Yu, K.; Zhang, S. Pretreatment platelet count predicts survival outcome of patients with de novo non-M3 acute myeloid leukemia. PeerJ 2017, 5, e4139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Huang, J.; Zhao, H.; Hong, M.; Zhu, H.; Zhu, Y.; Lian, Y.; Li, S.; Li, J.; Qian, S. Early recovery of the platelet count after decitabine-based induction chemotherapy is a prognostic marker of superior response in elderly patients with newly diagnosed acute myeloid leukaemia. Bmc Cancer 2018, 18, 1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Zhang, Y.; Gu, H.; Chen, Q.; Zhang, Y.; Cheng, H.; Yang, J.; Wang, J.; Hu, X. Low Platelet Counts at Diagnosis Predict Better Survival for Patients with Intermediate-Risk Acute Myeloid Leukemia. Acta Haematol. 2020, 143, 9–18. [Google Scholar] [CrossRef]
  82. Feng, J.; Zhang, W.; Wu, J.; Gao, S.; Ye, H.; Sun, L.; Chen, Y.; Yu, K.; Xing, C.-Y. Effect of initial absolute monocyte count on survival outcome of patients with de novo non-M3 acute myeloid leukemia. Leuk. Lymphoma 2016, 57, 2548–2554. [Google Scholar] [CrossRef] [PubMed]
  83. Ismail, M.M.; Abdulateef, N.A.B. Absolute Monocyte Count is Superior than Absolute Lymphocyte Count at Day 28 as an Independent Prognostic Factor in Acute Myeloid Leukemia. Indian J. Hematol. Blood Transfus. 2019, 35, 100–108. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Q.; Yang, Q.; Weng, Y.; Huang, Z.; Chen, R.; Zhu, Y.; Dai, K.; Zhang, S.; Jiang, S.; Yu, K. Neutrophil-to-lymphocyte ratio correlates with prognosis and response to chemotherapy in patients with non-M3 de novo acute myeloid leukemia. Transl. Cancer Res. 2021, 10, 1013–1024. [Google Scholar] [CrossRef] [PubMed]
  85. Mushtaq, M.U.; Chaudhary, S.G.; Murthy, G.S.G.; Hall, A.C.; Atallah, E.L.; Mattison, R.J. Prognostic Significance of Neutrophil-to-Lymphocyte Ratio in Relapsed/Refractory Acute Myeloid Leukemia. Blood 2018, 132, 5246. [Google Scholar] [CrossRef]
  86. Lobanova, T.I.; Parovichnikova, E.N.; Galtseva, I.V.; Davydova, J.O.; Kapranov, N.M.; Fidarova, Z.T.; Gavrilina, O.A.; Troitskaya, V.V.; Savchenko, V. Absolute Lymphocyte Count after First Induction Is Associated with Relapse Free Survival in Acute Myeloid Leukemia. Blood 2017, 130, 5087. [Google Scholar]
  87. Jang, J.; Kim, Y.R.; Cho, H.; Chung, H.; Kim, S.-J.; Kim, J.S.; Cheong, J.-W.; Min, Y.H. Peripheral Blood Absolute Lymphocyte Count at Diagnosis As a New Prognostic Factor in Acute Myeloid Leukemia with t(8;21) Patients. Blood 2019, 134, 5178. [Google Scholar] [CrossRef]
  88. Le Jeune, C.; Bertoli, S.; Elhamri, M.; Vergez, F.; Borel, C.; Huguet, F.; Michallet, M.; Dumontet, C.; Recher, C.; Thomas, X. Initial absolute lymphocyte count as a prognostic factor for outcome in acute myeloid leukemia. Leuk. Lymphoma 2014, 55, 855–862. [Google Scholar] [CrossRef] [PubMed]
  89. Bar, M.; Othus, M.; Park, H.M.; Sandhu, V.; Chen, X.; Wood, B.L.; Estey, E. Elevated lymphocyte count at time of acute myeloid leukemia diagnosis is associated with shorter remission. Leuk. Lymphoma 2015, 56, 3109–3115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Keenan, R.; Williamson, R.; Caswell, M.; Salim, R.; Campbell, H.J. Early Absolute Lymphocyte Count Is a Strong Predictor of Long-Term Improved Survival in Childhood Acute Myeloid Leukaemia. Blood 2012, 120, 3553. [Google Scholar] [CrossRef]
  91. Bumma, N.; Ai, J.; Jia, X.; Hobson, S.; Abounader, D.; Sekeres, M.A.; Kalaycio, M.E.; Sobecks, R.; Gerds, A.; Hamilton, B.K.; et al. Impact of Day 28 Absolute Lymphocyte Count on Outcome of Adult Patients with Acute Myeloid Leukemia. Blood 2014, 124, 1472. [Google Scholar] [CrossRef]
  92. Bazinet, A.; Popradi, G. A general practitioner’s guide to hematopoietic stem-cell transplantation. Curr. Oncol. Tor. Ont 2019, 26, 187–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Hochhaus, A.; Baccarani, M.; Silver, R.T.; Schiffer, C.; Apperley, J.F.; Cervantes, F.; Clark, R.E.; Cortes, J.E.; Deininger, M.W.; Guilhot, F.; et al. European LeukemiaNet 2020 recommendations for treating chronic myeloid leukemia. Leukemia 2020, 34, 966–984. [Google Scholar] [CrossRef] [PubMed]
  94. Ali, H.; Bacigalupo, A. 2021 Update on Allogeneic Hematopoietic Stem Cell Transplant for Myelofibrosis: A Review of Current Data and Applications on risk Stratification and Management. Am. J. Hematol. 2021, 96, 1532–1538. [Google Scholar] [CrossRef] [PubMed]
  95. Passamonti, F. Stem cell transplant in MF: It’s time to personalize. Blood 2019, 133, 2118–2120. [Google Scholar] [CrossRef] [Green Version]
  96. Masouridi-Levrat, S.; Olavarria, E.; Iacobelli, S.; Aljurf, M.; Morozova, E.; Niittyvuopio, R.; Sengeloev, H.; Reményi, P.; Helbig, G.; Browne, P.; et al. Outcomes and toxicity of allogeneic hematopoietic cell transplantation in chronic myeloid leukemia patients previously treated with second-generation tyrosine kinase inhibitors: A prospective non-interventional study from the Chronic Malignancy Working Party of the EBMT. Bone Marrow Transplant. 2022, 57, 23–30. [Google Scholar] [CrossRef] [PubMed]
  97. Killick, S.B.; Ingram, W.; Culligan, D.; Enright, H.; Kell, J.; Payne, E.M.; Krishnamurthy, P.; Kulasekararaj, A.; Raghavan, M.; Stanworth, S.J.; et al. British Society for Haematology guidelines for the management of adult myelodysplastic syndromes. Br. J. Haematol. 2021, 194, 267–281. [Google Scholar] [CrossRef] [PubMed]
  98. Pelcovits, A.; Niroula, R. Acute Myeloid Leukemia: A Review. Rhode Isl. Med. J. 2020, 103, 38–40. [Google Scholar]
  99. Barrett, J.; Craddock, C. Bone marrow transplantation in the United Kingdom—Past, present and future. Br. J. Haematol. 2020, 191, 612–616. [Google Scholar] [CrossRef] [PubMed]
  100. Cluzeau, T.; Lambert, J.; Raus, N.; Dessaux, K.; Absi, L.; Delbos, F.; Devys, A.; De Matteis, M.; Dubois, V.; Filloux, M.; et al. Risk factors and outcome of graft failure after HLA matched and mismatched unrelated donor hematopoietic stem cell transplantation: A study on behalf of SFGM-TC and SFHI. Bone Marrow Transplant. 2016, 51, 687–691. [Google Scholar] [CrossRef] [PubMed]
  101. Sweeney, C.; Vyas, P. The Graft-Versus-Leukemia Effect in AML. Front. Oncol. 2019, 9, 1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Thoma, M.D.; Huneke, T.J.; DeCook, L.J.; Johnson, N.D.; Wiegand, R.A.; Litzow, M.R.; Hogan, W.J.; Porrata, L.F.; Holton, S.G. Peripheral Blood Lymphocyte and Monocyte Recovery and Survival in Acute Leukemia Postmyeloablative Allogeneic Hematopoietic Stem Cell Transplant. Biol. Blood Marrow Transplant. 2012, 18, 600–607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Tang, L.; Wang, N.; Xing, C.; Zhuang, Q.; Liang, B.; Sun, L.; Chen, Y.; Qian, Y.; Shen, Z.; Jiang, S.; et al. Effect of absolute monocyte count post-transplant on the outcome of patients with acute myeloid leukemia undergoing myeloablative allogeneic hematopoietic stem cell transplant with busulfan and cyclophosphamide conditioning. Leuk. Res. 2018, 69, 60–65. [Google Scholar] [CrossRef]
  104. Chang, Y.-J.; Zhao, X.-Y.; Xu, L.-P.; Liu, D.-H.; Liu, K.-Y.; Chen, Y.-H.; Wang, Y.; Zhang, X.-H.; Zhao, X.-S.; Han, W.; et al. Early lymphocyte recovery predicts superior overall survival after unmanipulated haploidentical blood and marrow transplant for myelodysplastic syndrome and acute myeloid leukemia evolving from myelodysplastic syndrome. Leuk. Lymphoma 2013, 54, 2671–2677. [Google Scholar] [CrossRef] [PubMed]
  105. Bayraktar, U.D.; Milton, D.R.; Guindani, M.; Rondon, G.; Chen, J.; Al-Atrash, G.; Rezvani, K.; Champlin, R.; Ciurea, S.O. Optimal Threshold and Time of Absolute Lymphocyte Count Assessment for Outcome Prediction after Bone Marrow Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 505–513. [Google Scholar] [CrossRef] [Green Version]
  106. Chakrabarti, S.; Brown, J.; Guttridge, M.; Pamphilon, D.H.; Lankester, A.; Marks, D.I. Early lymphocyte recovery is an important determinant of outcome following allogeneic transplantation with CD34+selected graft and limited T-cell addback. Bone Marrow Transplant. 2003, 32, 23–30. [Google Scholar] [CrossRef] [Green Version]
  107. Fu, Q.; Xu, L.-P.; Zhang, X.-H.; Wang, Y.; Chang, Y.-J.; Huang, X.-J. Early lymphocyte recovery predicts superior outcomes after unmanipulated haploidentical blood and marrow transplant for acute myeloid leukemia. Clin. Transplant. 2016, 30, 954–958. [Google Scholar] [CrossRef] [PubMed]
  108. Gul, Z.; Van Meter, E.; Abidi, M.; Ditah, I.; Abdul-Hussein, M.; Deol, A.; Ayash, L.; Lum, L.G.; Waller, E.K.; Ratanatharathorn, V.; et al. Low blood lymphocyte count at 30 days post transplant predicts worse acute GVHD and survival but not relapse in a large retrospective cohort. Bone Marrow Transplant. 2015, 50, 432–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Han, D.K.; Baek, H.J.; Kim, S.Y.; Hwang, T.J.; Kook, H. Implication of Early Lymphocyte Recovery after Allogeneic Hematopoietic Stem Cell Transplantation in Children with Leukemia. Yonsei Med. J. 2013, 54, 62–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Porrata, L.F.; Litzow, M.R.; Tefferi, A.; Letendre, L.; Kumar, S.; Geyer, S.M.; Markovic, S.N. Early lymphocyte recovery is a predictive factor for prolonged survival after autologous hematopoietic stem cell transplantation for acute myelogenous leukemia. Leukemia 2002, 16, 1311–1318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Le Bourgeois, A.; Peterlin, P.; Guillaume, T.; Delaunay, J.; Duquesne, A.; Le Gouill, S.; Moreau, P.; Mohty, M.; Campion, L.; Chevallier, P. Higher Early Monocyte and Total Lymphocyte Counts Are Associated with Better Overall Survival after Standard Total Body Irradiation, Cyclophosphamide, and Fludarabine Reduced-Intensity Conditioning Double Umbilical Cord Blood Allogeneic Stem Cell Transplantation in Adults. Biol. Blood Marrow Transplant. 2016, 22, 1473–1479. [Google Scholar] [CrossRef] [Green Version]
  112. Hill, G.R.; Betts, B.C.; Tkachev, V.; Kean, L.S.; Blazar, B.R. Current Concepts and Advances in Graft-Versus-Host Disease Immunology. Annu. Rev. Immunol. 2021, 39, 19–49. [Google Scholar] [CrossRef] [PubMed]
  113. Moon, J.H.; Hamad, N.; Sohn, S.K.; Uhm, J.; Alam, N.; Gupta, V.; Lipton, J.H.; Messner, H.A.; Seftel, M.; Kuruvilla, J.; et al. Improved prognostic stratification power of CIBMTR risk score with the addition of absolute lymphocyte and eosinophil counts at the onset of chronic GVHD. Ann. Hematol. 2017, 96, 805–815. [Google Scholar] [CrossRef]
  114. Sheth, V.; Kennedy, V.; de Llavallade, H.; Mclornan, D.; Potter, V.; Engelhardt, B.G.; Savani, B.; Chinratanalab, W.; Goodman, S.; Greer, J.; et al. Differential Interaction of Peripheral Blood Lymphocyte Counts (ALC) With Different in vivo Depletion Strategies in Predicting Outcomes of Allogeneic Transplant: An International 2 Center Experience. Front. Oncol. 2019, 9, 623. [Google Scholar] [CrossRef]
  115. Kennedy, V.E.; Chen, H.; Savani, B.N.; Greer, J.; Kassim, A.A.; Engelhardt, B.G.; Goodman, S.; Sengsayadeth, S.; Chinratanalab, W.; Jagasia, M. Optimizing Antithymocyte Globulin Dosing for Unrelated Donor Allogeneic Hematopoietic Cell Transplantation Based on Recipient Absolute Lymphocyte Count. Biol. Blood Marrow Transplant. 2018, 24, 150–155. [Google Scholar] [CrossRef] [Green Version]
  116. Seo, J.; Shin, D.-Y.; Koh, Y.; Kim, I.; Yoon, S.-S.; Byun, J.M.; Hong, J. Association between preconditioning absolute lymphocyte count and transplant outcomes in patients undergoing matched unrelated donor allogeneic hematopoietic stem cell transplantation with reduced-intensity conditioning and anti-thymocyte globulin. Ther. Adv. Hematol. 2021, 12, 20406207211063784. [Google Scholar] [CrossRef] [PubMed]
  117. Shiratori, S.; Ohigashi, H.; Ara, T.; Yasumoto, A.; Goto, H.; Nakagawa, M.; Sugita, J.; Onozawa, M.; Kahata, K.; Endo, T.; et al. High lymphocyte counts before antithymocyte globulin administration predict acute graft-versus-host disease. Ann. Hematol. 2021, 100, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
  118. Kumar, S.; Chen, M.G.; Gastineau, D.A.; Gertz, M.A.; Inwards, D.J.; Lacy, M.Q.; Tefferi, A.; Litzow, M.R. Effect of slow lymphocyte recovery and type of graft-versus-host disease prophylaxis on relapse after allogeneic bone marrow transplantation for acute myelogenous leukemia. Bone Marrow Transplant. 2001, 28, 951–956. [Google Scholar] [CrossRef] [Green Version]
  119. Powles, R.; Singhal, S.; Treleaven, J.; Kulkarni, S.; Horton, C.; Mehta, J. Identification of patients who may benefit from prophylactic immunotherapy after bone marrow transplantation for acute myeloid leukemia on the basis of lymphocyte recovery early after transplantation. Blood 1998, 91, 3481–3486. [Google Scholar] [CrossRef]
  120. Michelis, F.V.; Messner, H.A.; Loach, D.; Uhm, J.; Gupta, V.; Lipton, J.H.; Seftel, M.D.; Kuruvilla, J.; Kim, D.D. Early lymphocyte recovery at 28 d post-transplant is predictive of reduced risk of relapse in patients with acute myeloid leukemia transplanted with peripheral blood stem cell grafts. Eur. J. Haematol. 2014, 93, 273–280. [Google Scholar] [CrossRef]
  121. Afzal, S.; Ishaqi, M.K.; Dupuis, A.; Doyle, J.; Gassas, A. Early lymphocyte recovery after allogeneic hematopoietic SCT is associated with significant GVL effect in pediatric ALL but not acute myelogenous leukemia-Update study. Bone Marrow Transplant. 2009, 44, 799–804. [Google Scholar] [CrossRef] [Green Version]
  122. Niederwieser, D.; Gastl, G.; Rumpold, H.; Marth, C.; Kraft, D.; Huber, C. Rapid reappearance of large granular lymphocytes (LGL) with concomitant reconstitution of natural killer (NK) activity after human bone marrow transplantation (BMT). Br. J. Haematol. 1987, 65, 301–305. [Google Scholar] [CrossRef]
  123. Jiang, Y.Z.; Barrett, A.J.; Goldman, J.M.; Mavroudis, D.A. Association of natural killer cell immune recovery with a graft-versus-leukemia effect independent of graft-versus-host disease following allogeneic bone marrow transplantation. Ann. Hematol. 1997, 74, 1–6. [Google Scholar] [CrossRef]
  124. Huttunen, P.; Taskinen, M.; Siitonen, S.; Saarinen-Pihkala, U.M. Impact of Very Early CD4(+)/CD8(+) T Cell Counts on the Occurrence of Acute Graft-Versus-Host Disease and NK Cell Counts on Outcome After Pediatric Allogeneic Hematopoietic Stem Cell Transplantation. Pediatr. Blood Cancer 2015, 62, 522–528. [Google Scholar] [CrossRef]
  125. Minculescu, L.; Marquart, H.V.; Friis, L.S.; Petersen, S.L.; Schiodt, I.; Ryder, L.P.; Andersen, N.S.; Sengeloev, H. Early Natural Killer Cell Reconstitution Predicts Overall Survival in T Cell-Replete Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 2187–2193. [Google Scholar] [CrossRef]
  126. Aksu, K.; Donmez, A.; Keser, G. Inflammation-induced thrombosis: Mechanisms, disease associations and management. Curr. Pharm. Des. 2012, 18, 1478–1493. [Google Scholar] [CrossRef]
  127. Iba, T.; Levy, J.H. Inflammation and thrombosis: Roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis. J. Thromb. Haemost. JTH 2018, 16, 231–241. [Google Scholar] [CrossRef] [Green Version]
  128. Vinay, D.S.; Ryan, E.P.; Pawelec, G.; Talib, W.H.; Stagg, J.; Elkord, E.; Lichtor, T.; Decker, W.K.; Whelan, R.L.; Kumara, H.M.C.S.; et al. Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin. Cancer Biol. 2015, 35, S185–S198. [Google Scholar] [CrossRef]
  129. Ménétrier-Caux, C.; Ray-Coquard, I.; Blay, J.-Y.; Caux, C. Lymphopenia in Cancer Patients and its Effects on Response to Immunotherapy: An opportunity for combination with Cytokines? J. Immunother. Cancer 2019, 7, 85. [Google Scholar] [CrossRef] [Green Version]
  130. Fridman, W.H.; Pagès, F.; Sautès-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef]
  131. Jacobs, N.L.; Holtan, S.G.; Porrata, L.F.; Markovic, S.N.; Tefferi, A.; Steensma, D.P. Low Absolute Lymphocyte Count (ALC) at Diagnosis Is An IPSS-Independent Predictor of Poorer Survival in Myelodysplastic Syndromes (MDS). Blood 2008, 112, 3633. [Google Scholar] [CrossRef]
  132. Mehta, R.S.; Rezvani, K. Immune reconstitution post allogeneic transplant and the impact of immune recovery on the risk of infection. Virulence 2016, 7, 901–916. [Google Scholar] [CrossRef]
Table 1. Prognostic role of peripheral count blood cells parameters in chronic myeloproliferative neoplasms.
Table 1. Prognostic role of peripheral count blood cells parameters in chronic myeloproliferative neoplasms.
AuthorsYearsDiseasesNo. of PtsOutcomesComments
Sasaki [37]2014CML483OSALC ≥ 4 × 109/L at 3 or 6 months of TKI start was associated with lower OS
Pepedil-Tanrikulu [39]2020CML95ResponseALC, AMC, and LMR did not predict the molecular response
Hacibekiroglu [48]2015ET99ThrombosisNo differences in CRP, NLR, RDW, MPV, and sedimentation levels
Kocak [49]2017ET70ThrombosisNo differences in NLR and PLR in patients with or without thrombosis
Zhou [50]2018ET150ThrombosisHigher NLR in Jak2 positive and in patients at high-risk stratification of thrombosis
Boiocchi [54]2015PV10EvolutionPersistent hyperleukocytosis is associated with poor prognosis in MF-post-PV pts
Ronner [55]2020PV520 Evolution, thrombosis Persistently elevated leukocyte was associated with an increased hazard of disease evolution but not of thrombotic events
Krečak [56]2021PV109OS, thrombosisHigher NRL and PLR are associated with a high risk of disease, shorter TTT
Carobbio [57]2021PV1508ThrombosisHigh NLR is an independent predictor of venous thrombosis
Farruk [58]2022PV, TE487ThrombosisANC and AMC associated with venous thrombosis
Teng [59]2022Post-PV MF163ThrombosisPatients with V617F% ≥ 75% or AMC ≥ 1.5 × 109/L had a higher risk for venous thrombosis
Lucijanic [64]2018MF102OSHigher NLR and Jak2 mutation; High NLR and low PLR poor prognosis
Lucijanic [65]2018MF83OSLow ALC associated with poor prognosis
Abbreviations: OS, overall survival; TTT, time to thrombosis; CML, chronic myeloid leukemia; ET, essential thrombocythemia; PV, polycythemia vera; MF, myelofibrosis; ALC, absolute lymphocyte count; ANC, absolute neutrophil count; TKI, tyrosine kinase inhibitors; AMC, absolute monocyte count; NLR, neutrophil-to-lymphocyte ratio; LMR, lymphocyte-to-monocyte ratio; CRP, C-reactive protein; RDW, Red blood cell distribution width; MPV, mean platelet volume; PLR, platelets-to-lymphocytes ratio.
Table 2. Prognostic role of peripheral count blood cell parameters in acute myeloid leukemia and myelodysplastic syndromes.
Table 2. Prognostic role of peripheral count blood cell parameters in acute myeloid leukemia and myelodysplastic syndromes.
AuthorsYearsDiseasesNo. of PtsOutcomesComments
Silzle [58]2019MDS1023OSLow ALC and lower OS, most apparent in lower-risk patients
Yikilmaz [70]2020MDS63ClassificationLow PLR and multilinear dysplasia
Jacobs [71]2010MDS503OSALC > 1.2 × 109/L better OS
Saeed [72]2017MDS889OSLow ALC, low AMC, inferior OS. ALC, AMC, and LMR are not influenced by LFS
Saeed [73]2016MDS889OS, LFSLow ALC lower OS but not lower LFS, most apparent in lower-risk patients
Buckley [74]2014MDS/AML205Complication Low AMC and ALC at induction treatment high risk of infection or bacteremia
Chen [75]2015AML/MDS343ResponseLow PLT counts at 21 after induction was associated with no response
Zhang [79]2017AML209OS, DFSPLT between 50 and 120 × 109/L better OS and DFS
Huang [80]2018AML117Prognosis PLT count recovery on day 14 after D-CAG IC is associated with the response
Zhang [81]2020AML291OS and DFSLow platelets levels at diagnosis predict better OS and DFS
Feng [82]2016AML193OSHigh AMC appeared as a poor prognostic factor for OS
Ismail [83]2019AML83OS, DFSAMC ≥ 0.8 × 109/L + 28 shorter OS and LFS, ALC > 0.35 × 109/L higher OS and LFS
Zhang [84]2021AML181OS, DFSNLR < 2 at diagnosis better OS and DFS
Mushtaq [85]2018AML63OSHigh NLR independently predicts poor OS in RR-AML patients.
Lobanova [86]2017AML35DFSALC more than 0.8 × 109/L poor DFS
Jang [87]2019AML65LFS, OSHigher ALC poor LFS and OS
Le Jeune [88]2013AML1702OS, DFS Initial ALC < 1 × 109/L poor DFS and OS, ALC > 4.5 × 109/L lower response rate IC
Bar [89]2015AML259OSHigher ALC lower remission and poor RFS and OS
Keenan [90]2012AML59OSAt +28 days post IC ALC > 1.35 × 109/L better OS
Bumma [91]2014AML180OSALC < 0.5 × 109/L poor outcome in IC
Abbreviations: MDS, myelodysplastic syndrome; AML, acute myeloid leukemia; OS, overall survival; LFS, leukemia-free survival; DFS, disease-free survival; IC, induction chemotherapy; D-CAG, decitabine, cytarabine, aclarubicin, and granulocyte colony-stimulating factor; ALC, absolute lymphocyte count; AMC, absolute monocyte count; PLR, platelet-to-lymphocyte ratio; NLR, neutrophil-to-lymphocyte ratio; PLT, platelets; RFS, relapse-free survival.
Table 3. Prognostic role of different peripheral count blood cell parameters in bone marrow transplantation.
Table 3. Prognostic role of different peripheral count blood cell parameters in bone marrow transplantation.
AuthorsYearsNo. of PtsOutcomesComments
Thoma [102]2012135OSALC and AMC > 0.3 × 109/L from +30 +60 and +100 from HSCT had better OS
Tang [103]201859OSAMC > 0.57 × 109/L +15 from HSCT had better OS
Chang [104]201378OS, TRMALC > 0.3 × 109/L lower relapse rates and lower infections. Better OS, LFS, and low TRM
Bayraktar [105]2015518OS, NRMALC > 0.3 × 109/L at +60 better OS and NRM after HSCT
Chakrbarti [106]200329OS, NRMALC > 0.3 × 109/L at +30 was the strongest predictor of NRM and OS
Fu [107]2016134OS, LFSALC > 0.294 × 109/L at +30 better OS and LFS, but was not related to relapse
Gul [108]2015381OS, NRMALC < 0.4 × 109/L lower OS and increased NRM. No association with relapse
Han [109]201369 OS, EFSALC > 0.5 × 109/L at +21 and +30 better engraftment. High ALC at 30 days had better OS and EFS. There were no differences in the GVHD or relapse rates.
Porrata [110]200245OSALC > 0.5 × 109/L at day +15 had better OS
Le Bourgeois [111]201647 OS+30 ALC > 2.76 × 109/L and +42 ALC > 4.25 × 109/L had better OS.
Moon [113]2017307cGVHDALC < 1 × 109/L and eosinophil count < 0.5 × 109/L relate to lower OS and helped improve the risk stratification power of CIBMTR
Sheth [114]2019364OS, DFS ALC > 0.08 × 109/L in 2 days of alemtuzumab infusion had poor DFS and OS
Kennedy [115]2018135OSHigh ALC in higher recipient ATG dose had a lower risk of death
Seo [116]202164OSPC ALC < 0.5 × 109/L shorter OS and higher infectious mortality in patients receiving ATG
Shiratori [117]202153GVHDALC > 0.15 × 109/L before ATG predicts GVHD requiring systemic steroids
Kumar [118]200187OS, relapseALC < 0.15 × 109/L at +30 lower OS and higher relapse rates.
Powles [119]1998201RelapseALC > 0.2 × 109/L associated with lower relapse rates
Michelis [120]2014191OS, relapseALC > 0.5 × 109/L +28 is associated with lower relapse
Afzal [121]200971EFS relapseALC does not correlate to GVL and was not predictive of relapse in AML children
Jiang [123]199715GVHDNo correlation between CD4c, CD8c, or NK cells and the development of GVHD
Huttunen [124]201583GVHD, EFSCD4/CD8 higher in patients with GVHD, NK > 0.12 × 109/L at +32 had better TRM and EFS
Minculescu [125]2016298 OS, relapseNK > 0.15 × 109/L on +30 had better OS less TRM and infections. No link to relapse.
Abbreviations: OS, overall survival; EFS, event-free survival; DFS, disease-free survival; NRM, non-relapse mortality; TRM, transplant-related mortality; GVHD, graft versus host disease; ALC, absolute lymphocyte count; AMC, absolute monocyte count; CIBMTR, chronic graft-versus-host disease risk score; GVL, graft versus leukemia; NK, natural killer; HSCT, hematopoietic stem cell transplantation.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mulas, O.; Mola, B.; Madeddu, C.; Caocci, G.; Macciò, A.; Nasa, G.L. Prognostic Role of Cell Blood Count in Chronic Myeloid Neoplasm and Acute Myeloid Leukemia and Its Possible Implications in Hematopoietic Stem Cell Transplantation. Diagnostics 2022, 12, 2493. https://doi.org/10.3390/diagnostics12102493

AMA Style

Mulas O, Mola B, Madeddu C, Caocci G, Macciò A, Nasa GL. Prognostic Role of Cell Blood Count in Chronic Myeloid Neoplasm and Acute Myeloid Leukemia and Its Possible Implications in Hematopoietic Stem Cell Transplantation. Diagnostics. 2022; 12(10):2493. https://doi.org/10.3390/diagnostics12102493

Chicago/Turabian Style

Mulas, Olga, Brunella Mola, Clelia Madeddu, Giovanni Caocci, Antonio Macciò, and Giorgio La Nasa. 2022. "Prognostic Role of Cell Blood Count in Chronic Myeloid Neoplasm and Acute Myeloid Leukemia and Its Possible Implications in Hematopoietic Stem Cell Transplantation" Diagnostics 12, no. 10: 2493. https://doi.org/10.3390/diagnostics12102493

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