Next Article in Journal
Expression of HCMV-Encoded miRNA in Subjects Acutely Coinfected with HIV: Correlation with Inflammation and Immune Activation
Next Article in Special Issue
Downregulation of S6 Kinase and Hedgehog–Gli1 by Inhibition of Fatty Acid Synthase in AML with FLT3-ITD Mutation
Previous Article in Journal
Effects of Phenosanic Acid in Rat Seizure Models
Previous Article in Special Issue
Acquired Resistance to Decitabine Associated with the Deoxycytidine Kinase A180P Mutation: Implications for the Order of Hypomethylating Agents in Myeloid Malignancies Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Microenvironment, Inflammation, and Inflammatory Prognostic Indices in Diffuse Large B-Cell Lymphomas: A Narrative Review

by
Zorica Cvetković
1,2,*,
Olivera Marković
2,3,
Gligorije Marinković
1,
Snežana Pejić
4 and
Vesna Vučić
5
1
Department of Hematology, University Hospital Medical Center Zemun, 11080 Belgrade, Serbia
2
Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
3
Department of Hematology, University Hospital Medical Center Bežanijska kosa, 11080 Belgrade, Serbia
4
Department of Molecular Biology and Endocrinology, Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
5
Center of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(12), 5670; https://doi.org/10.3390/ijms26125670
Submission received: 18 May 2025 / Revised: 9 June 2025 / Accepted: 11 June 2025 / Published: 13 June 2025

Abstract

Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin lymphoma, characterized by significant variability in clinical outcomes. Emerging evidence highlights the pivotal role of inflammation in the pathogenesis and prognosis of DLBCL. This narrative review explores the interplay between the tumor microenvironment, inflammatory processes, and prognostic indices used in DLBCL, focusing on biomarkers, immune responses, and systemic inflammation. These indices show promise as predictive and prognostic tools comparable to molecular markers, such as gene expression profiling, which are currently considered gold standards in prognosis but are often costly and technically demanding. By synthesizing findings from the current literature, this article highlights the potential of inflammatory indices as accessible and cost-effective prognostic alternatives to molecular markers in DLBCL, while also underscoring the need for further research to validate their clinical utility.

1. Introduction

Non-Hodgkin lymphomas (NHLs) are the 10th most common malignancy worldwide, according to the most recent GLOBOCAN cancer statistics from the International Agency for Research on Cancer for the year 2020 [1]. Among NHL subtypes, diffuse large B-cell lymphoma (DLBCL) is the most prevalent, accounting for over a third of all NHL cases, with an estimated age-standardized incidence rate of 7.2 per 100,000. In Western countries, the incidence of DLBCL has remained stable since the beginning of the 21st century, likely due to improved control of known host and environmental risk factors, such as obesity, infections, and exposure to toxins [2].
DLBCL, not otherwise specified (NOS), is characterized by an aggressive course and a heterogeneous morphological, molecular, and clinical presentation. Both of the currently coexisting classification systems—the 5th edition of the World Health Organization (WHO) Classification of Hematolymphoid Tumors (WHO-HAEM5, 2022) [3] and the International Consensus Classification (ICC, 2022) [4]—still recommend subtyping according to the cell of origin into DLBCL germinal center B-cell-like (GCB) and DLBCL activated B-cell-like (ABC, non-GCB), with the latter generally associated with a worse prognosis.
The standard first-line treatment for DLBCL is chemoimmunotherapy (CIT), typically combining rituximab with an anthracycline-based chemotherapy regimen such as cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or a CHOP-like alternative. While the majority of patients with DLBCL respond favorably to initial therapy, approximately 40% either fail to achieve remission or experience relapse following treatment [5]. Improved insights into the biology of DLBCL have led to the development of new therapeutic strategies aimed at enhancing patient outcomes and survival rates [6]. However, despite a slight but steady decline in mortality rates in recent years, the 5-year relative survival rate, which remains around 65%, is still considered unsatisfactory [7].
Advancements in modern molecular and genetic techniques have significantly improved our understanding of lymphoma pathogenesis and enabled more accurate prognostic assessments. However, these methods are often costly, time-consuming, and not widely available in many clinical settings. In parallel with the ongoing research into molecular markers as potential therapeutic targets, increasing attention is being given to the development of simple, cost-effective, and accessible clinical prognostic indices. These tools, alongside established models such as the International Prognostic Index (IPI) [8,9], its revised version [10], the age-adjusted IPI [8,11], and the National Comprehensive Cancer Network International Prognostic Index (NCCN-IPI) [12,13], are being investigated to support treatment decisions and follow-up care in patients with DLBCL. The goal of these new indices is to complement existing models by offering additional, clinically practical insights into disease progression, treatment response, and patient outcomes.
Inflammation is a well-established contributor to the development and progression of various cancers, including hematologic malignancies [14]. Emerging therapies, such as immune checkpoint inhibitors (ICPis) targeting cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), programmed cell death-1 (PD-1) and PD ligand 1 (PD-L1), bispecific antibodies (BsAbs), and chimeric antigen receptor (CAR) T-cell therapy, aim to enhance T-cell-mediated antitumor immunity [15]. In parallel, inflammatory markers and indices are increasingly studied as prognostic tools in oncology. In DLBCL, inflammation plays a key role in shaping the tumor microenvironment (TME) and promoting tumorigenesis through immune cell recruitment, cytokine production, angiogenesis, dysregulated signaling pathways, and altered apoptosis. Inflammation not only drives disease progression but also influences treatment response [6,16,17].
Despite the growing evidence, there remain significant gaps in understanding which specific inflammatory markers and indices most accurately predict outcomes in DLBCL, how they interact with treatment responses, and their integration into routine clinical decision making. This narrative review aims to address these gaps by highlighting the role of lipid- and protein-derived inflammatory markers, peripheral blood and TME immune cells, and inflammatory indices as emerging predictive and prognostic biomarkers in DLBCL, with an emphasis on their clinical utility and potential to guide personalized therapy.

2. Lipids and Lipid-Derived Inflammatory Markers in DLBCL

Lipid profile disturbances, commonly associated with inflammatory markers, have been consistently observed in patients with a range of malignancies, including NHL, over the past several decades. Recent studies have highlighted altered lipid metabolism as a major oncogenic factor in aggressive forms of NHL, including DLBCL [18]. Lipids are essential parts of cellular membranes, contributing to their structural integrity and fluidity. They also serve as important secondary messengers in various inflammatory and oncogenic pathways and provide a vital source of energy [19,20,21], as illustrated in Figure 1.

2.1. Cholesterol

A large prospective study, tracking 61,936 patients from the IQVIA (formerly Quintiles and IMS Health, Inc., Durham, NC, USA) database between 2005 and 2019, found that individuals with reduced levels of high-density lipoprotein cholesterol (HDL-C) had a significantly higher risk of developing multiple types of cancer, including hematological malignancies [22]. Notably, a significant and prolonged decrease in HDL-C levels was detected up to 10 years prior to the clinical onset of DLBCL and other NHL subtypes when compared to healthy individuals [23,24,25]. The reduction in cholesterol and HDL-C levels in the plasma of NHL patients correlates with the aggressiveness and clinical stage of the disease, while a sustained increase back to normal levels has been observed in NHL patients who achieved remission [26,27,28]. Furthermore, HDL-C showed remarkably predictive value for progression-free survival (PFS) and overall survival (OS) in patients treated with CIT [29]. Gao et al. combined NCCN-IPI and the cholesterol status to develop a novel prognostic score, Lipo-PI, which demonstrated superior accuracy over NCCN-IPI in predicting 3-year and 5-year OS and PFS [30].
The major structural protein of HDL-C is apolipoprotein A (apoA), which serves as an activator of lecithin: cholesterol acyltransferase (LCAT), an enzyme that converts free cholesterol into cholesteryl ester. In a large retrospective study on 949 newly diagnosed DLBCL patients, lower levels of apoA1 correlated with lymphoma burden symptoms, advanced stage, increased rate of relapse, higher incidence of refractory disease, and shorter OS [31]. Similarly, in a retrospective analysis of 105 DLBCL patients, Yu et al. demonstrated that apoA1 was an independent predictor of OS and PFS and introduced a novel IPI-A score that combines IPI with apoA1 levels [28]. These findings suggest that lipid profiles may serve as non-specific prognostic and predictive biomarkers for DLBCL, reflecting tumor proliferative capacity, tumor burden, and potentially treatment response.
As a crucial component of the cell membrane in both malignant and normal cells, cholesterol is essential for maintaining membrane integrity and fluidity [32]. The cause of cancer-related hypocholesterolemia is more complex than simply the enhanced receptor-mediated influx and consumption of cholesterol by rapidly proliferating tumor cells [19].
It is well established that dyslipidemia, chronic inflammation, and oxidative stress interact synergistically to drive tumorigenesis. The beneficial, multifaceted protective effect of HDL-C in mitigating chronic inflammation states, including in solid and hematological cancers, is still under extensive investigation. Cholesterol regulates numerous processes, including ferroptosis, autophagy, the DNA damage response, the expression of ICP genes, and influences the TME. Specifically, cholesterol modulates the immune response of TME cells through several mechanisms: inhibiting antigen presentation, enriching immunosuppressive cells like neutrophils and tumor-associated macrophages (TAMs), and regulating immune-effector cells such as CD8+ T-cells and natural killer (NK) cells [19,33,34]. These effects occur via both contact-dependent and contact-independent mechanisms, involving soluble molecules like growth factors and lipid-derived cytokines [35,36]. In a recently published prospective observational study by Ou W et al., HDL-C was proposed to be a cardioprotective target, based on the finding that patients with DLBL treated with the standard R-CHOP regimen and higher HDL-C levels exhibited a significantly lower rate of anthracycline-induced subclinical cardiotoxicity [37].

2.2. Plasma Phospolipids

In the context of NHL and plasma phospholipid (PL) fractions, our previous research observed a baseline reduction in all plasma PL species—specifically phosphatidylcholine (PC), lysophosphatidylcholine (LPC), phosphatidylethanolamine (PE), and sphingomyelin (SM). These reductions were more pronounced in aggressive NHL subtypes and remained low in patients who progressed on front-line treatment. In contrast, lipid levels tended to increase in patients who responded well to treatment [38]. Regarding the distribution of plasma PL fatty acids (FAs), we found a reduction in polyunsaturated FA (PUFAs) that correlated with both the aggressiveness of NHL [39] and treatment response [40]. Additionally, our recent pilot study using advanced high-resolution ion mobility mass spectrometry on treatment-naïve DLBCL revealed a decline in 54 plasma PL and sphingolipid (SL) species. Most of these species contained linoleic acid (LA; 18:2(n-6)) or arachidonic acid (AA; 20:4(n-6)), which are key precursors of various cytokines [41].
The observed decline in PL levels, along with the altered FA profile, is thought to result from the complex interplay between the uptake of exogenous fatty acids, de novo lipogenesis, and fatty acid oxidation (FAO or β-oxidation) for ATP production [42,43]. This metabolic reprogramming plays a significant role in the pathogenesis of lymphoma.

2.3. FA

Cancer cells undergo a decisive transformation in their metabolism, compellingly shifting their energy source from glucose to the more potent FA. The overexpression of fatty acid synthase (FASN), a key enzyme involved in the synthesis of 16-carbon FA palmitate and overall lipid biosynthesis, is associated with poor DLBCL prognosis [44], while its selective inhibition disrupts c-Met receptor signaling and induces apoptosis in DLBCL cells [45,46].
The uptake of exogenous fatty acids is regulated by transmembrane proteins, particularly CD36 and CD37.
CD37, a member of the tetraspanin family almost exclusively expressed in lymphoid tissues, modulates lipid metabolism by inhibiting the fatty acid transporter FATP1. This inhibition restricts the uptake and conversion of exogenous palmitate into energy, its incorporation into structural and oncogenic signaling lipids, and its use in posttranslational modifications such as the palmitoylation of PD-L1 [47,48]. CD37 is significantly downregulated in aggressive B-cell lymphoma cell lines resistant to rituximab; nevertheless, this reduction in CD37 expression does not diminish the therapeutic efficacy of CD37-targeted CAR T-cell therapy [49]. Recognizing the prognostic significance of CD37, Xu-Monette et al. proposed two refined risk stratification models for DLBCL: the molecularly adjusted IPI for R-CHOP (M-IPI-R), which incorporates CD37 negativity and the ABC subtype with the IPI; and IPI + IHC, which integrates immunohistochemical (IHC) expression of CD37, Myc, and Bcl-2 with IPI. Both models significantly enhanced prognostic precision compared to IPI alone. These combined indices demonstrated enhanced prognostic accuracy compared to the IPI alone [50].
CD36, a type 2 cell surface scavenger receptor with three distinct domains, binds to proteins containing thrombospondin structural homology repeat (TSR) domains, molecules exhibiting molecular structures consistent with danger-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), and functions as a transmembrane channel for exogenous long-chain FA acid uptake and lipid accumulation. CD36 is expressed on the surface of B cells, as well as various TME cells, including macrophages, monocytes, dendritic cells (DCs), and subsets of T cells [51]. Its upregulation has been linked to several key processes in cancer biology, including tumor cell proliferation, immune evasion, and metastasis [52]. In the context of DLBCL, higher levels of CD36 expression on B cells are strongly correlated with the polarization of TAMs toward the M2 subtype, particularly in CD5+ DLBCL [53]. Additionally, elevated CD36 expression is indicative of poorer prognoses for DLBCL patients [54]. This suggests that CD36 and CD37 may play a critical role in shaping the immune landscape of DLBCL and could serve as potential biomarkers for disease progression and response to treatment and appealing targets for novel therapeutic strategies in lymphoma [20,55,56,57].
Dysregulation of AA metabolism can lead to increased inflammatory responses, tumor progression, and immune suppression within the TME. The dysregulation of AA metabolism, a key n-6 PUFA, can lead to increased inflammatory responses, tumor progression, and immune suppression within the TME. The accumulation of AA in the PLs of cell membranes is associated with the ability of inflammatory cells to produce bioactive lipid mediators. These mediators include prostaglandins (PGs), leukotrienes (LTs), epoxyeicosatetraenoic acids (EETs), and endocannabinoids (ECs), all of which play a role in regulating immune cell function and modulating the TME [58,59,60]. AA is released from PLs in the cell membrane by cytosolic phospholipase A2 (cPLA2) and further catabolized through various pathways, primarily involving cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450 (CYP-450) enzymes [21].
COX is the key and rate-limiting enzyme responsible for the conversion of AA to PGs. There are two distinct isoforms of COX: COX-1, which is constitutively expressed in most tissues, and the inducible COX-2, which is expressed in cells of the TME, endothelial cells, smooth muscle cells, fibroblasts, chondrocytes, and cancer cells [61,62]. Several decades ago, researchers discovered a significant increase in COX-2 protein expression and activity in lymphoma cell lines [63]. In B-NHL, a correlation was identified between COX-2 upregulation and deletions on chromosome 17p, where the tumor suppressor gene TP53 is located [64], which is associated with poor outcomes [65]. COX-2 promotes apoptotic resistance, proliferation, transformation, angiogenesis, inflammation, invasion, and metastasis of malignant cells, mainly through its metabolite PGE2 [66,67,68]. Of interest is that platelets primarily express COX-1, which generates large amounts of thromboxane (TX)A2, a proaggregatory and vasoconstrictor mediator of chronic inflammation associated with several diseases, including tissue fibrosis and cancer [69].
Aspirin (acetylsalicylic acid), a non-selective COX inhibitor, has shown potential therapeutic effect as an adjunct to existing therapies in various cancers, including lymphoma [70,71]. Its anti-inflammatory properties, through the inhibition of COX-1 and COX-2, may reduce the inflammatory response within the TME, which contributes to tumorigenesis and immune evasion [72]. The role of aspirin in cancer prevention is still a matter of debate [73]. A large case-control study including 1703 NHL patients and 2199 frequency-matched controls, found that low-dose aspirin use was associated with a reduced risk of NHL, particularly DLBCL, with stronger protection observed with longer duration of use. This inverse association persisted, though slightly weakened, after adjusting for risk factors, including family history of NHL, body mass index, smoking, alcohol use, and rheumatoid arthritis, though it was slightly attenuated [74].
Selective COX-2 inhibitors like celecoxib have also been investigated in lymphoma therapy. These inhibitors selectively block COX-2 activity, without affecting COX-1, thus reducing unwanted side effects like gastrointestinal toxicity [75,76]. The role of celecoxib in improving chemotherapy efficacy in DLBCL has been explored, including in overcoming doxorubicin-induced multidrug resistance [77] and improving the effectiveness of CAR-T cell immunotherapy [78,79].
On the other hand, n-3 PUFAs, including eicosapentaenoic acid (EPA, 20:5) and docosahexaenoic acid (DHA, 22:6), have the opposite effect to those of n-6 PUFA. A higher ratio of n-6 to n-3 PUFAs in red blood cell membranes prior to diagnosis has been associated with an increased DLBCL risk [80]. The substantial decrease in n-3 PUFAs that corresponded with disease aggressiveness and poor prognosis suggested that n-3 PUFA levels may serve as a potential biomarker for NHL progression [39,40,81]. Supplementation with n-3 PUFAs has demonstrated beneficial effects in cancer patients by enhancing chemotherapy efficacy, mitigating treatment-related side effects, and thereby improving clinical outcomes and quality of life [82,83,84]. A meta-analysis of 20 studies involving 971 cancer patients receiving n-3 PUFA as adjuvant therapy showed a significant reduction in circulating proinflammatory cytokine levels [85]. The results of a recent meta-analysis of 28 clinical trials confirmed the positive anti-inflammatory effects of n-3 PUFA supplementation as an add-on treatment in cancer patients, highlighting that moderate doses (e.g., 850–900 mg/day) can achieve these benefits without causing any significant side effects [86]. In addition to modulating immune responses, n-3 PUFAs exert anti-tumor effects by inducing reactive oxygen species (ROS) production, which leads to oxidative stress and damage to cellular components [87]. Furthermore, n-3 PUFAs play a role in tumor suppression by inducing epigenetic modifications, such as DNA methylation, histone methylation, and acetylation of histones and p53. These changes influence apoptotic signaling pathways, including the RAS/ERK/C/EBPβ, PI3K/AKT/mTOR, ERK/AKT/NF-κB, and BCR/PI3K/NF-κB pathways [88,89,90,91].

3. Protein-Derived Inflammatory Parameters

Protein-derived inflammatory markers like C-reactive protein (CRP), albumin, fibrinogen, lactate dehydrogenase (LDH), beta-2 microglobulin (β2M), and interleukins (ILs) play an important role in assessing the inflammatory state of DLBCL, influencing prognosis, treatment decisions, and disease monitoring. Targeting these inflammatory markers and pathways offers potential therapeutic benefits. For instance, reducing inflammation through therapies targeting cytokines like IL-6 and tumor necrosis factor alpha (TNF-α) could improve clinical outcomes and enhance the effectiveness of chemotherapy and immunotherapy in DLBCL patients.

3.1. CRP

CRP is an acute-phase protein produced by the liver in response to inflammatory cytokines, particularly IL-6, which is secreted by macrophages and T cells. Due to its ease of testing through simple blood samples, CRP has become a widely used biomarker for assessing a patient’s inflammatory status. Its levels are often elevated in various malignancies, both solid tumors and hematological cancers, highlighting its potential as a prognostic marker for disease progression and treatment response [92]. In Hodgkin lymphoma, baseline levels of CRP have been shown to correlate directly with advanced stages of the disease and/or the presence of B symptoms. Elevated CRP levels are also considered predictors of treatment response, with persistent high CRP values during treatment indicating resistance to therapy [93].
In DLBCL, CRP was identified as an independent prognostic marker over a decade ago [94]. The patients with low CRP expression receiving R-CHOP chemotherapy exhibit significantly better therapeutic responses compared to those with high CRP levels, emphasizing the negative prognostic impact of elevated CRP [95,96]. The prognostic utility of CRP in DLBCL has been validated by two meta-analyses [97,98]. A recent meta-analysis that included data from 11 studies involving 2314 patients demonstrated that high baseline CRP levels were associated with lower OS in DLBCL patients [98]. Beyond its role as a stand-alone prognostic marker, CRP enhances the predictive accuracy of the R-IPI score in DLBCL patients [99,100].
Elevated CRP levels have been linked to an increased risk of venous thromboembolism (VTE), particularly deep vein thrombosis (DVT), in cancer patients [101]. It has been reported that CRP, along with other inflammatory parameters, can help identify DLBCL patients at risk for VTE, who may benefit from thromboprophylaxis [102].
Furthermore, CRP has shown value as a biomarker in CAR T-cell therapy. In a multicenter clinical study exploring the predictive factors of response to immunotherapy, elevated CRP levels at the time of infusion (e.g., ≥10 mg/L) were found to predict not only treatment response but also the occurrence of potential therapy-related toxicities [103]. Elevated CRP levels prior to CAR T-cell infusion have also been shown to predict the length of intensive care unit stay [104], further underscoring their relevance in both disease monitoring and treatment decision-making.

3.2. Fibrinogen

Fibrinogen is an acute-phase reactant that plays a pivotal role in clot formation and inflammation. Elevated fibrinogen levels have been linked to increased tumor cell proliferation, metastasis, and angiogenesis and have also been associated with poor prognosis in various malignancies [105]. In DLBCL, a significant correlation between fibrinogen levels and the presence of B symptoms and clinical stage has been reported [106]. Moreover, in a retrospective study, Niu et al. revealed that hyperfibrinogenemia was associated with higher NCCN-IPI scores, further highlighting its prognostic value [107].
Several studies have also demonstrated that elevated fibrinogen levels at presentation are linked to poor outcomes, including shorter PFS and OS in DLBCL patients [107,108,109], reinforcing its prognostic relevance. Additionally, Ogura et al. found that elevated fibrinogen levels prior to stem cell transplantation were associated with poorer survival outcomes, emphasizing the importance of this inflammatory marker in the management and prognosis of DLBCL [110].

3.3. Albumin

Albumin is a negative acute-phase protein whose levels decrease in response to systemic inflammation. Low serum albumin levels are associated with poor nutritional status, increased tumor burden, and a worse prognosis in cancer patients [111,112]. In DLBCL, hypoalbuminemia (defined as serum albumin < 35 g/L) has been identified as a significant predictor of poor prognosis in DLBCL [113,114]. Conversely, higher serum albumin levels (≥40 g/L) proved to be an independent prognostic factor of superior outcomes in DLBCL patients aged ≥ 70 years [115]. Hypoalbuminemia is usually considered a marker of malnutrition that is common in elderly population and cancer patients, which can worsen clinical outcome. To assess the nutritional status of patients, two indices that incorporate albumin levels, namely, the Prognostic nutrition index (PNI) and Geriatric nutritional index (GNRI), were originally introduced to identify at-risk medical patients. These indices have been extensively evaluated in patients with hematological malignancies and have proven to be valid predictors of prognosis [116,117,118,119]. In a cohort of 95 older DLBCL patients, low GNRI but not sarcopenia was significantly associated with adverse prognosis [120]. These findings underscore the value of albumin as a critical marker not only for nutritional assessment but also for predicting clinical outcomes, particularly in elderly populations who may experience increased inflammation and nutritional depletion due to both their age and cancer.
Albumin, along with CRP and fibrinogen, are routinely measured in most biochemical laboratories, making them affordable and accessible inflammatory biomarkers.

3.4. IL-6

Numerous cytokines are released during the inflammatory response, some exerting tumor-promoting effects while others play a tumor-suppressive role. A better understanding of the cytokine landscape has provided deeper insights into tumorigenesis. Members of the IL-6 cytokine family, including IL-6 itself, oncostatin M, leukemia inhibitory factor, IL-11, IL-27, IL-31, ciliary neurotrophic factor, cardiotrophin 1, and cardiotrophin-like cytokine factor 1, have been implicated in promoting tumor development by influencing the TME, as illustrated in Figure 2, underscoring their potential as therapeutic targets in oncology [121].
Among these cytokines, IL-6 stands out as a key mediator of the link between inflammation and cancer, and it is the most widely applicable and clinically evaluated cytokine for prognostication in routine clinical practice. IL-6 promotes tumor growth, immune evasion, and resistance to therapy by activating key oncogenic pathways, including JAK/STAT3 and NF-κB [122,123,124]. It also reshapes the tumor microenvironment by modulating immune cell populations and inducing the polarization of TAMs, which further enhances tumor growth and therapy resistance [125,126]. Elevated IL-6 levels are consistently associated with poor clinical outcomes in DLBCL. For instance, Bao et al. found that IL-6 levels ≥ 4.5 pg/mL were predictive of shorter PFS and OS [127]. Furthermore, high IL-6 levels were associated with resistance to treatment [128,129] and with clinical features including advanced disease stage and B symptoms [130]. These findings highlight the importance of IL-6 as a valuable marker in both risk stratification and monitoring therapeutic efficacy in DLBCL patients but also as possible therapeutic target [131]. An IL-6 receptor antagonist, tocilizumab, is used for prevention and treatment of cytokine-release syndrome, a potentially life-threatening complication during T-cell-directed treatment [132,133]. Furthermore, tocilizumab improves cytotoxic activity of antibody–drug conjugate naratuximab emtansine in CD37+ DLBCL cells [134].

3.5. β2M

β2M is an essential component of class I major histocompatibility complex and is present on all nucleated cells. Elevated levels of β2M in blood and bodily fluids are indicative of cell membrane turnover, cellular turnover rates, and kidney function and serve as a useful inflammatory marker in various cancers, particularly B-cell malignancies. The recent research, including a genome-wide meta-analysis (GWMA), bidirectional two-sample Mendelian randomization (TSMR), and pathway enrichment analysis, has established a causal link between increased β2M levels and a higher risk of DLBCL, potentially associated with innate immune system dysfunction [135]. Furthermore, a retrospective analysis involving 3232 newly diagnosed DLBCL patients treated with CIT, as recorded in the Danish Lymphoma Registry, demonstrated that incorporating β2M into the NCCN-IPI enhances the discriminatory performance of this prognostic tool [136].

4. Cells of Innate and Adaptive Immune Responses in the TME of DLBCL

Lymphomagenesis is characterized by complex interactions between tumor cells, innate immune cells, and adaptive immune cells within the TME. Both innate and adaptive immune responses are essential for controlling tumor growth, and the interactions between tumor cells and immune cells in the TME significantly impact clinical outcomes [137]. The polarization of macrophages from M1 to M2 [138]; the dysfunction of B cells [139], NK cells [140], and T-cells [141]; and the upregulation of ICP [142] all contribute to immune evasion and tumor progression, as displayed in Figure 3. Understanding these dynamics among immune cells offers insights into potential therapeutic strategies, such as ICPis, regulatory T-cell (Treg) depletion, and BsAb and CAR T-cell therapies, which could help restore immune surveillance and improve treatment outcomes in DLBCL [143].

4.1. Cells of the Innate Immune Response in DLBCL

4.1.1. Monocytes and TAMs

TAMs, derived from monocytes circulating in the bloodstream, represent the most abundant immune cell populations within the TME and play a crucial role in the progression and prognosis of lymphoma [138]. Upon migration to tissues, monocytes differentiate into macrophages and polarize into two primary phenotypes depending on environmental signals from the TME: M1/classically activated and M2/alternatively activated. M1 macrophages, activated by pro-inflammatory signals like interferon-gamma (IFN-γ), IL-1β, and IL-12, exhibit anti-tumor activity by enhancing cytotoxic T-cell responses and directly killing tumor cells [144]. In contrast, M2 macrophages exert immunosuppressive functions, producing IL-10, transforming growth factor (TGF)-β, PD-L1, and vascular endothelial growth factor (VEGF), which support tumor progression, tissue remodeling, angiogenesis, and metastasis [145]. Additionally, M2 macrophages contribute to immune suppression by inhibiting T-cell function and enhancing tumor resistance to chemotherapy [146]. M2 macrophages also contribute to the degradation of the extracellular matrix through the overexpression of legumain, which further promotes the progression of DLBCL [147]. The polarization of macrophages from M1 to M2 is frequently observed in DLBCL and correlates with poor prognosis [148,149]. TAMs have distinct phenotypes and genetic signatures. M2 macrophages express CD163, while the expression of the pan-macrophage marker CD68 is upregulated in M1 compared to M2 macrophages [150]. A retrospective analysis of clinicopathologic data and survival outcomes from 355 patients diagnosed with DLBCL demonstrated that a CD163+ M2 TAM content of 9.5% or greater negatively affects the prognosis of DLBCL [151]. Guo et al. recently proposed the M2-macrophage-related gene prognostic model for DLBCL constructed on the expression of eight genes (MS4A4A, CCL13, LTB, CCL23, CCL18, XKR4, IL22RA2, and FOLR2). Elevated immune scores in high-risk DLBCL patients were associated with poorer prognosis and greater sensitivity to chemotherapeutic agents and ICPis [152]. Novel treatment strategies targeting TAMs to overcome tumor resistance are intensively being investigated. Both animal models and clinical trials focused on phagocytosis checkpoints, the reversal from M2 to M1 phenotype, and CAR-macrophage (CAR-M) have shown promising results with high efficacy and low toxicity [153].
Monocytes circulate in the peripheral blood for 1 to 3 days before migrating into tissues, where they differentiate into macrophages. The relationship between peripheral absolute monocyte count (AMC) and TAM in DLBCL remains a topic of ongoing research. Monocytosis at the time of DLBCL diagnosis has been identified as an independent prognostic factor associated with poorer outcomes and reduced OS. Wilcox et al., in a cohort of 366 DLBCL patients, demonstrated that an absolute monocyte count (AMC) above 0.63 × 109/L at diagnosis is associated with poorer prognosis, with a 5-year OS rate of 59% compared to 71% in patients with lower AMC [154]. This cutoff was subsequently validated in a large multicenter study of 1017 therapy-naïve DLBCL patients from Israel and Italy, where it retained its negative prognostic value even after adjusting for the IPI (HR 1.54, p = 0.009) [155]. Additionally, patients with negative end-of- treatment (EOT)-PET-CT but AMC > 0.63 × 109/L had significantly lower 3- and 5-year event-free survival (EFS) rates, suggesting that combining AMC with EOT-PET-CT strengthens the prediction of durability of treatment response [156].

4.1.2. Neutrophils and Tumor Associated Neutrophils (TANs)

Neutrophils, the most abundant circulating leukocytes and primary responders to bacterial infections and tissue damage, also play a role in the tumor response in DLBCL. Within TME, pro-inflammatory cytokines such as IL-1, IFN-γ, and TNF-α stimulate the production of chemokines that recruit neutrophils [157]. These TANs promote tumor progression and immune suppression by releasing ROS and proteolytic enzymes that degrade the extracellular matrix, thereby facilitating tumor invasion. Neutrophils contribute to immune evasion by suppressing T-cell-mediated anti-tumor responses through the secretion of inhibitory cytokines like IL-10 and chemokines such as C-X-C motif chemokine ligand 8 (CXCL8), thereby facilitating tumor immune escape [158]. Another key mechanism is the formation of neutrophil extracellular traps (NETs)—web-like structures composed of decondensed chromatin—that can capture circulating tumor cells (CTCs) and enhance metastatic potential by promoting tumor cell survival [159,160].
In clinical settings, an elevated absolute neutrophil count (ANC) has been observed in patients with advanced-stage DLBCL (p = 0.012) and is significantly associated with poorer DFS [161]. While ANC alone has prognostic value, it is more commonly incorporated into composite indices alongside other hematologic and biochemical markers, which will be discussed later.

4.1.3. NK Cells

NK cells are essential effectors of the innate immune system, responsible for eliminating tumor and virus-infected cells through distinct mechanisms, including direct cytotoxicity via the release of perforin and granzymes, as well as induction of apoptosis through death receptor pathways such as Fas ligand and TNF-related apoptosis-inducing ligand. They constitute approximately 10–15% of peripheral blood lymphocytes and are phenotypically defined by the expression of CD3CD56+. NK cells are further categorized into two main subsets based on CD56 expression: CD56bright NK cells, which are relatively immature, account for 5–10% of circulating NK cells and primarily exert immunoregulatory functions through cytokine secretion; and CD56dim NK cells, which are fully mature, comprise 90–95% of peripheral NK cells and are mainly responsible for direct cytotoxicity, including Ab-dependent cell-mediated cytotoxicity (ADCC) [162,163].
In the TME, NK cells often become dysfunctional due to the upregulation of ICP receptors such as PD-1 and T-cell immunoglobulin and mucin-domain-containing protein 3 (TIM-3), which markedly reduce their cytotoxic potential [140,142,164]. Furthermore, macrophage polarization and the accumulation of Tregs contribute to NK cell suppression and immune evasion [165,166]. Clinically, elevated levels of circulating NK cells before treatment have been associated with improved therapeutic outcomes in relapsed/refractory (R/R) DLBCL [167]. Notably, NK cell count < 0.1 × 109/L has been identified as an independent adverse prognostic factor for PFS and OS in the non-GCB subtype of DLBCL [168].

4.1.4. DCs

Myeloid-derived DCs, which present tumor antigens and trigger naïve T-cell activation and differentiation, express the surface marker CD11c, linked to cytotoxic effects against lymphoma cells [169]. Decreased CD11c+ DC in the TME has been linked with MYC and BCL2/BCL6 (double-hit/triple-hit) genotype and survival in DLBCL [170,171]. In contrast, a higher density of DCs in TME [172] and an increased number of CD11c+ DCs in the peripheral blood of DLBCL patients have been linked with better prognosis and OS [173]. Lee et al. developed a prognostic model for patients with EN DLBCL receiving CIT, incorporating the expression of CD11c and Forkhead box P3 (FOXP3), a key transcription factor involved in the regulation of Treg cell function, within the TME [174]. However, the accurate assessment of peripheral NK cell and DCs levels typically requires flow cytometry, a method that is costly and impractical for routine clinical practice.

4.2. Cells of Adaptive Immune Response in DLBCL

Since DLBCL originates from B lymphocytes, dysregulation of B-cell receptor (BCR) signaling plays a pivotal role in its pathogenesis. Aberrant BCR signaling promotes tumor cell survival, enhances proliferation, and confers resistance to apoptosis, primarily through activation of the NF-κB and PI3K/AKT pathways [175]. The antitumor B-cell immune response is further compromised by immunosuppressive elements in the TME, particularly Tregs and TAMs. Tregs, which are essential for maintaining immune homeostasis, can suppress not only NK cell activity but also B-cell and CD4+/CD8+ T-cell responses when enriched in the TME of DLBCL [139].
T cells are central to adaptive immunity: CD8+ cytotoxic T cells directly eliminate target cells, while CD4+ helper T cells assist B cells in antibody production. T-cell activation is initiated when antigens presented on major histocompatibility complex (MHC) molecules are recognized by the T-cell receptor (TCR), leading to downstream signaling cascades. However, in approximately 50% of DLBCL cases, there is a loss of MHC class I expression on tumor cells, which may impair CD8+ T-cell recognition [176]. One of the primary antitumor functions of CD8+ T cells, and a key mechanism targeted by immunotherapies, is the induction of apoptosis via the perforin–granzyme pathway. CD4+ T cells also contribute significantly to antitumor immunity, and a TME enriched with CD4+ T cells (over 20%) has been associated with improved relapse-free survival and OS [177,178]. Similar to NK cells, both CD4+ and CD8+ T cells can become functionally exhausted within the TME. As noted earlier, tumor cells exploit ICP pathways, such as PD-1, PD-L1, CTLA-4, and FOXP3-expressing Tregs, to dampen T-cell activity and escape immune surveillance [141]. Recent studies have shown that elevated expression of OX40, a costimulatory receptor expressed on activated T cells, in DLBCL is associated with co-expression of ICP proteins and enhanced T-cell activation. This contributes to an inflamed TME, favorable clinical characteristics, and improved response to CIT, underscoring the potential of OX40 as both a prognostic biomarker [179] and a therapeutic target [180]. In a separate investigation of the mutational landscape of DLBCL, Zhang et al. identified 227 significantly mutated genes. Notably, tumor protein 53 (TP53) and CD58 mutations displayed a mutually exclusive pattern. The TP53WT and CD58MUT subgroup, especially within the GCB subtype, was associated with poor prognosis, increased immune evasion, and preferential expression of co-inhibitory receptors such as PD-1, TIM-3, and lymphocyte activation gene 3 (LAG-3). This profile suggests potential sensitivity to immunotherapy. Moreover, this subgroup exhibited elevated infiltration of exhausted T cells, macrophages, and NK cells, indicative of a distinctly immunosuppressive TME [181].
Patients with DLBCL exhibit reduced levels of peripheral blood CD4+, CD8+ T cells, and NK cells compared to healthy individuals. High-risk patients also show increased Treg proportions and diminished IFN-γ production by CD4+ and CD8+ T cells, indicating an impaired cellular immune response [182]. The prognostic significance of pretreatment lymphopenia (<1 × 109/L) has been recognized across various cancers, including lymphoma, for decades [183]. A meta-analysis involving 1206 DLBCL patients from six studies further confirmed that low baseline absolute lymphocyte counts (ALC) are associated with poorer DFS and OS [184]. Lymphocyte count in peripheral blood serves as a key surrogate marker of immune reconstitution following autologous stem cell transplantation (ASCT) in NHL. Patients with an ALC on day 15 post-transplant (ALC-15) ≥ 0.5 × 10⁹/L demonstrated improved clinical outcomes [185]. Pre-lymphodepletion ALC has been identified as a predictor of response to CAR T-cell therapy. Patients with higher ALC prior to lymphodepletion showed significantly improved OS (45.17 vs. 9.6 months, p = 0.008) and PFS (45.17 vs. 4.07 months, p = 0.030) compared to those with lower ALC [186].

5. Role of Platelets and Anemia in Inflammation and DLBCL Prognostication

Beyond their role in hemostasis, platelets contribute to immune modulation by releasing cytokines and chemokines that attract neutrophils and monocytes and activate DCs, thereby influencing both innate and adaptive immunity [187]. In addition to immune cells, platelets activate fibroblasts and vascular cells through the release of various bioactive molecules, including lipid-derived eicosanoids, growth and angiogenic factors, and RNA-rich extracellular vesicles. During early tumorigenesis, platelet activation induces COX-2 overexpression in stromal cells, leading to increased PGE2 production, which, in turn, promotes cancer progression by inhibiting apoptosis, enhancing tumor cell proliferation and migration, and enabling immune evasion [188]. In DLBCL, pretreatment platelet count has emerged as a potential prognostic marker. In a study of 1007 patients treated with frontline R-CHOP or R-CHOP-like regimens, a platelet count ≤ 157 × 109/L was independently associated with poorer OS (HR 1.960, 95% CI 1.418–2.709, p < 0.001) and PFS (HR 1.443, 95% CI 1.080–1.927, p = 0.013) [189].
Cancer-related anemia has a multifactorial origin, with its prevalence varying from 30% to 90% depending on the cancer type and site [190]. In follicular NHL, anemia is a well-established prognostic factor and is incorporated into standard risk models when hemoglobin (Hb) falls below 120 g/L [191,192]. However, in DLBCL, specific Hb thresholds with prognostic or predictive significance have yet to be clearly defined. According to Hong et al., a baseline Hb level below 100 g/L (determined as the lowest value within seven days before initiating R-CHOP, without prior red blood cell transfusion) was significantly associated with shorter DFS and OS in a cohort of 157 DLBCL patients [193]. Furthermore, Matsumoto et al., proposed a simple prognostic index based on grade 2 anemia (defined as Hb < 100–80 g/L) and advanced Ann Arbor clinical stages (CS) to predict outcome in DLBCL patients [194].

6. Inflammatory Indices Evaluated in DLBCL

Although developed in the pre-rituximab era, the IPI remains the most widely used tool for risk stratification in newly diagnosed DLBCL patients across all age groups. Other proposed prognostic indices, such as R-IPI, age-adjusted IPI (aa-IPI), and NCCN-IPI are all derived from the same clinical variables as the original IPI and none can accurately identify patients at ultra-high risk [195]. Efforts to enhance prognostic precision, such as the IPI-IPM, which incorporates molecular markers including CMBL, TLCD3B, SYNDIG1, ESM1, EPHA3, HUNK, PTX3, and IL12A, have shown promise [196], but their complexity limits their routine clinical application.

6.1. Prognostic Indices That Include Inflammatory Biomarkers

The success of immune-based therapies, coupled with growing insights into the impact of systemic inflammation and the TME on DLBCL progression, treatment response, and survival, has driven efforts to enhance the traditional IPI. Due to their established prognostic relevance, routinely available lipid-derived (e.g., HDL-C, apoA) and protein-based (e.g., CRP, albumin, hemoglobin, fibrinogen, and β2M) inflammatory markers have been used either as standalone biomarkers, as previously discussed, or more commonly integrated into existing prognostic models—or combined to create novel indices that are easily applicable in everyday clinical practice.
Most of the developed indices include albumin as a key component. Both the pretreatment albumin-to-globulin ratio and the CRP-to-albumin ratio have been associated with poorer prognosis in patients with DLBCL [197,198]. Additionally, the CRP-to-albumin ratio and albumin-to-fibrinogen have been suggested as a potential complement to the IPI for improved risk stratification in DLBCL [199,200]. In addition, more complex indices have also been developed. For instance, Melchardt et al. refined the NCCN-IPI for elderly DLBCL patients by incorporating low serum albumin and β2M, enhancing its prognostic accuracy [201]. Similarly, Gang et al. integrated hypoalbuminemia with traditional IPI variables to develop age-adjusted prognostic indices with a 70-year cutoff, improving risk stratification [202]. The Kyoto Prognostic Index (KPI), which combines LDH, albumin, ECOG-PS, and EN involvement, has demonstrated superior predictive power in the rituximab era [203]. Additionally, GNRI, calculated from serum albumin and body weight ratios, has been identified as an independent prognostic marker reflecting patients’ nutritional status [204].
The key components and principal prognostic and predictive findings of several proposed models containing lipid- and protein-derived markers are summarized in Table 1.

6.2. Prognostic Indices That Include Peripheral Blood Cells

Peripheral blood cells (PBCs)—including lymphocytes, monocytes, neutrophils, and platelets—not only reflect systemic immune and inflammatory responses but also mirror the biological activity of the TME, as we have described earlier in this review. Their prognostic significance in DLBCL is well established, and they have become central components of several novel prognostic models that we will further analyze. These indices, whether based on ratios of absolute PBC counts or integrated with other inflammatory markers, substantially improve risk stratification and offer a more accurate and comprehensive prediction of clinical outcomes in newly diagnosed DLBCL.
Among these, platelet count and ALC are most frequently utilized in the composite prognostic indices. Some indices are based on two variables, such as LAR (LDH to ALC ratio) [200], PNI (albumin and ALC) [117], PA (platelet and albumin) [205], and HP (Hb and platelet) [206]. Others, like the model proposed by Chen H. et al., incorporate three components—β2M, platelet count, and red cell distribution width (RDW) [207]. In an effort to better identify high-risk patients, some indices integrate multiple blood cell types. For example, Chen et al. developed a composite model incorporating baseline ANC, AMC, β2M, Eastern Cooperative Oncology Group Performance Scale (ECOG PS), and number of extranodal (EN) sites. This model demonstrated superior risk discrimination compared to the standard IPI [208]. Jelicic et al. recently proposed the Elderly-IPI (E-IPI), which includes all NCCN-IPI parameters (excluding EN involvement), along with serum albumin and platelet counts to enhance prognostic precision in elderly patients [209].
Indices based on ratio of PBCs, such as lymphocyte-to-monocyte ratio (LMR), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) have demonstrated strong predictive value in both solid tumors and hematological malignancies [210,211]. These ratios are also proven to be effective in predicting treatment response and survival outcomes in DLBCL [212,213,214]. More complex models combining three or more blood cell types have also been developed [215,216], while others integrate blood cell ratios with additional inflammatory markers [213], as detailed in Table 2.
Some of these indices have been validated through systematic reviews and meta-analyses. A meta-analysis of studies involving 1311 patients demonstrated that a low Prognostic Nutritional Index (PNI) is an independent indicator of poor prognosis in DLBCL, associated with dismal prognosis and poor DFS and OS [217]. Further supporting the clinical relevance of nutritional and inflammatory markers, two separate systematic reviews assessed the GNRI. One analysis of 2448 elderly patients with DLBCL showed that lower GNRI values were significantly linked to reduced DFS and OS [218]. Another review, based on data from 2353 patients, confirmed that low GNRI not only predicts inferior survival outcomes but is also correlated with poor PS, advanced stage, and the presence of B symptoms [219].
Regarding indices incorporating PBC ratios, a meta-analysis of 1931 DLBCL patients found that elevated PLR was significantly associated with inferior OS and adverse clinical features such as B symptoms, elevated LDH, advanced stage, and ECOG PS ≥ 2, though it showed no association with PFS, age, gender, or cell of origin [220]. Likewise, a systematic review of 11 studies involving 4578 patients consistently identified low LMR as a strong predictor of reduced OS and DFS, reinforcing its prognostic relevance in DLBCL [221].
Beyond meta-analyses, direct comparisons of various prognostic indices within the same cohort have also been conducted. For example, Wang et al. demonstrated that the systemic immune–inflammation index (SII) outperformed both PLR and NLR in predicting outcomes in 224 newly diagnosed DLBCL patients [216]. In R/R DLBCL, Kim et al. evaluated multiple inflammation-based models: NLR, derived NLR (dNLR), LMR, PLR, Glasgow Prognostic Score (GPS), PNI, systemic inflammation response index (SIRI), and SII, alongside NCCN-IPI. Their findings identified high NCCN-IPI, GPS 2, and elevated dNLR as independent predictors of poor prognosis, leading to the development of a new risk model for PFS and OS [222].
Most recently, Ide D. et al. introduced the Kyoto Prognostic Index for relapsed/refractory DLBCL, which integrates low LMR, elevated LDH, and high CRP. This model showed promising predictive power for treatment response and survival in transplant-ineligible patients [223].

7. Conclusions and Further Directions

This narrative review highlights the critical role of inflammation in the pathogenesis and prognosis of DLBCL, with a particular emphasis on its incorporation into prognostic indices. A range of inflammation-based markers has been proposed as accessible, cost-effective tools designed to bridge the gap between conventional clinical scoring systems and complex, resource-intensive molecular models, thereby enhancing the accuracy of DLBCL risk stratification. While many of these models appear promising, most have been derived from retrospective, single-center cohorts with limited sample sizes and lack rigorous external validation. To incorporate inflammatory markers into routine clinical practice, several challenges must be addressed, including standardization of measurement methods, establishment of uniform cut-off values, and controlling for potential confounding factors.
Notably, a comparative analysis of 13 inflammation-based prognostic models in over 5000 patients from the Danish lymphoma registry found no advantage over the established NCCN IPI [224]. This highlights the gap between theoretical promise and clinical applicability. To advance personalized care in DLBCL, these inflammation-driven indices require rigorous validation through large-scale, prospective, multicenter prospective studies, and randomized clinical trials. Only with such high-quality evidence can these markers be confidently integrated into routine prognostic assessments and therapeutic decision making, firmly establishing their role as reliable and useful tools in clinical practice.

Author Contributions

Conceptualization, Z.C.; writing—original draft preparation, Z.C., O.M. and G.M.; writing—review and editing, Z.C. and V.V.; visualization, Z.C. and G.M.; supervision, Z.C., S.P. and V.V.; 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.

Data Availability Statement

The data that are discussed in this article are presented in cited studies.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, S.S. Epidemiology and etiology of diffuse large B-cell lymphoma. Semin. Hematol. 2023, 60, 255–266. [Google Scholar] [CrossRef] [PubMed]
  3. Kurz, K.S.; Ott, M.; Kalmbach, S.; Steinlein, S.; Kalla, C.; Horn, H.; Ott, G.; Staiger, A.M. Large B-Cell Lymphomas in the 5th Edition of the WHO-Classification of Haematolymphoid Neoplasms—Updated Classification and New Concepts. Cancers 2023, 15, 2285. [Google Scholar] [CrossRef] [PubMed]
  4. Campo, E.; Jaffe, E.S.; Cook, J.R.; Quintanilla-Martinez, L.; Swerdlow, S.H.; Anderson, K.C.; Brousset, P.; Cerroni, L.; de Leval, L.; Dirnhofer, S.; et al. The International Consensus Classification of Mature Lymphoid Neoplasms: A report from the Clinical Advisory Committee. Blood 2022, 140, 1229–1253. [Google Scholar] [CrossRef]
  5. Melchardt, T.; Egle, A.; Greil, R. How I treat diffuse large B-cell lymphoma. ESMO Open 2023, 8, 100750. [Google Scholar] [CrossRef]
  6. Masnikosa, R.; Cvetković, Z.; Pirić, D. Tumor Biology Hides Novel Therapeutic Approaches to Diffuse Large B-Cell Lymphoma: A Narrative Review. Int. J. Mol. Sci. 2024, 25, 11384. [Google Scholar] [CrossRef]
  7. National Cancer Institute Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Non-Hodgkin Lymphoma—Diffuse Large B-Cell Lymphoma (DLBCL). Available online: https://seer.cancer.gov/statfacts/html/dlbcl.html (accessed on 20 February 2025).
  8. International Non-Hodgkin’s Lymphoma Prognostic Factors Project. A predictive model for aggressive non-Hodgkin’s lymphoma. N. Engl. J. Med. 1993, 329, 987–994. [Google Scholar] [CrossRef]
  9. Ziepert, M.; Hasenclever, D.; Kuhnt, E.; Glass, B.; Schmitz, N.; Pfreundschuh, M.; Loeffler, M. Standard International Prognostic Index remains a valid predictor of outcome for patients with aggressive CD20+ B-cell lymphoma in the rituximab era. J. Clin. Oncol. 2010, 28, 2373–2380. [Google Scholar] [CrossRef]
  10. Sehn, L.H.; Berry, B.; Chhanabhai, M.; Fitzgerald, C.; Gill, K.; Hoskins, P.; Klasa, R.; Savage, K.J.; Shenkier, T.; Sutherland, J.; et al. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood 2007, 109, 1857–1861. [Google Scholar] [CrossRef]
  11. Hamlin, P.A.; Zelenetz, A.D.; Kewalramani, T.; Qin, J.; Satagopan, J.M.; Verbel, D.; Noy, A.; Portlock, C.S.; Straus, D.J.; Yahalom, J.; et al. Age-adjusted International Prognostic Index predicts autologous stem cell transplantation outcome for patients with relapsed or primary refractory diffuse large B-cell lymphoma. Blood 2003, 102, 1989–1996. [Google Scholar] [CrossRef]
  12. Zhou, Z.; Sehn, L.H.; Rademaker, A.W.; Gordon, L.I.; Lacasce, A.S.; Crosby-Thompson, A.; Vanderplas, A.; Zelenetz, A.D.; Abel, G.A.; Rodriguez, M.A.; et al. An enhanced International Prognostic Index (NCCN-IPI) for patients with diffuse large B-cell lymphoma treated in the rituximab era. Blood 2014, 123, 837–842. [Google Scholar] [CrossRef]
  13. Ruppert, A.S.; Dixon, J.G.; Salles, G.; Wall, A.; Cunningham, D.; Poeschel, V.; Haioun, C.; Tilly, H.; Ghesquieres, H.; Ziepert, M.; et al. International prognostic indices in diffuse large B-cell lymphoma: A comparison of IPI, R-IPI, and NCCN-IPI. Blood 2020, 135, 2041–2048. [Google Scholar] [CrossRef] [PubMed]
  14. Saluja, S.; Bansal, I.; Bhardwaj, R.; Beg, M.S.; Palanichamy, J.K. Inflammation as a driver of hematological malignancies. Front. Oncol. 2024, 14, 1347402. [Google Scholar] [CrossRef]
  15. Davoodi-Moghaddam, Z.; Jafari-Raddani, F.; Noori, M.; Bashash, D. A systematic review and meta-analysis of immune checkpoint therapy in relapsed or refractory non-Hodgkin lymphoma; a friend or foe? Transl. Oncol. 2023, 30, 101636. [Google Scholar] [CrossRef] [PubMed]
  16. Tamma, R.; Ranieri, G.; Ingravallo, G.; Annese, T.; Oranger, A.; Gaudio, F.; Musto, P.; Specchia, G.; Ribatti, D. Inflammatory Cells in Diffuse Large B Cell Lymphoma. J. Clin. Med. 2020, 9, 2418. [Google Scholar] [CrossRef] [PubMed]
  17. Cerchietti, L. Genetic mechanisms underlying tumor microenvironment composition and function in diffuse large B-cell lymphoma. Blood 2024, 143, 1101–1111. [Google Scholar] [CrossRef]
  18. Zhang, Z.; Zhao, C.; Yang, S.; Lu, W.; Shi, J. A novel lipid metabolism-based risk model associated with immunosuppressive mechanisms in diffuse large B-cell lymphoma. Lipids Health Dis. 2024, 23, 20. [Google Scholar] [CrossRef]
  19. Xiao, M.; Xu, J.; Wang, W.; Zhang, B.; Liu, J.; Li, J.; Xu, H.; Zhao, Y.; Yu, X.; Shi, S. Functional significance of cholesterol metabolism in cancer: From threat to treatment. Exp. Mol. Med. 2023, 55, 1982–1995. [Google Scholar] [CrossRef]
  20. Guerrero-Rodríguez, S.L.; Mata-Cruz, C.; Pérez-Tapia, S.M.; Velasco-Velázquez, M.A. Role of CD36 in cancer progression, stemness, and targeting. Front. Cell Dev. Biol. 2022, 10, 1079076. [Google Scholar] [CrossRef]
  21. Zhang, Y.; Liu, Y.; Sun, J.; Zhang, W.; Guo, Z.; Ma, Q. Arachidonic acid metabolism in health and disease. MedComm 2023, 4, e363. [Google Scholar] [CrossRef]
  22. Loosen, S.H.; Kostev, K.; Luedde, M.; Luedde, T.; Roderburg, C. Low blood levels of high-density lipoprotein (HDL) cholesterol are positively associated with cancer. J. Cancer Res. Clin. Oncol. 2022, 148, 3039–3046. [Google Scholar] [CrossRef] [PubMed]
  23. Lim, U.; Gayles, T.; Katki, H.A.; Stolzenberg-Solomon, R.; Weinstein, S.J.; Pietinen, P.; Taylor, P.R.; Virtamo, J.; Albanes, D. Serum high-density lipoprotein cholesterol and risk of non-Hodgkin lymphoma. Cancer Res. 2007, 67, 5569–5574. [Google Scholar] [CrossRef]
  24. Alford, S.H.; Divine, G.; Chao, C.; Habel, L.A.; Janakiraman, N.; Wang, Y.; Feigelson, H.S.; Scholes, D.; Roblin, D.; Epstein, M.M.; et al. Serum cholesterol trajectories in the 10 years prior to lymphoma diagnosis. Cancer Causes Control 2018, 29, 143–156. [Google Scholar] [CrossRef]
  25. Jeong, S.M.; Choi, T.; Kim, D.; Han, K.; Kim, S.J.; Rhee, S.Y.; Giovannucci, E.L.; Shin, D.W. Association between high-density lipoprotein cholesterol level and risk of hematologic malignancy. Leukemia 2021, 35, 1356–1364. [Google Scholar] [CrossRef] [PubMed]
  26. Cvetković, Z.; Cvetković, B.; Petrović, M.; Ranić, M.; Debeljak-Martarčić, J.; Vučić, V.; Glibetić, M. Lipid profile as a prognostic factor in cancer patients. J. BUON 2009, 14, 501–506. [Google Scholar]
  27. Kuliszkiewicz-Janus, M.; Małecki, R.; Mohamed, A.S. Lipid changes occurring in the course of hematological cancers. Cell Mol. Biol. Lett. 2008, 13, 465–474. [Google Scholar] [CrossRef]
  28. Yu, T.; Luo, D.; Luo, C.; Xu-Monette, Z.Y.; Yu, L. Prognostic and therapeutic value of serum lipids and a new IPI score system based on apolipoprotein A-I in diffuse large B-cell lymphoma. Am. J. Cancer Res. 2023, 13, 475–484. [Google Scholar] [PubMed]
  29. Wang, F.; Lu, L.; Chen, H.; Yue, Y.; Sun, Y.; Yan, F.; He, B.; Lin, R.; Gu, W. Altered serum lipid levels are associated with prognosis of diffuse large B cell lymphoma and influenced by utility of rituximab. Ann. Hematol. 2023, 102, 393–402. [Google Scholar] [CrossRef]
  30. Gao, R.; Liang, J.H.; Wang, L.; Zhu, H.Y.; Wu, W.; Cao, L.; Fan, L.; Li, J.Y.; Yang, T.; Xu, W. Low serum cholesterol levels predict inferior prognosis and improve NCCN-IPI scoring in diffuse large B cell lymphoma. Int. J. Cancer 2018, 143, 1884–1895. [Google Scholar] [CrossRef]
  31. Huang, X.; Wang, Y.; Huang, Z.; Chen, X.; Lin, Q.; Huang, H.; Fan, L. Low serum apolipoprotein A1 level predicts poor prognosis of patients with diffuse large B-cell lymphoma in the real world: A retrospective study. BMC Cancer 2024, 24, 62. [Google Scholar] [CrossRef]
  32. Bielecka-Dąbrowa, A.; Hannam, S.; Rysz, J.; Banach, M. Malignancy-associated dyslipidemia. Open Cardiovasc. Med. J. 2011, 5, 35–40. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, K.; Zhang, Y.; Si, C.; Cao, Y.; Shao, P.; Zhang, P.; Wang, N.; Su, G.; Qian, J.; Yang, L. Cholesterol: The driving force behind the remodeling of tumor microenvironment in colorectal cancer. Heliyon 2024, 10, e39425. [Google Scholar] [CrossRef] [PubMed]
  34. Allegra, A.; Murdaca, G.; Mirabile, G.; Gangemi, S. Protective Effects of High-Density Lipoprotein on Cancer Risk: Focus on Multiple Myeloma. Biomedicines 2024, 12, 514. [Google Scholar] [CrossRef] [PubMed]
  35. Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef]
  36. Cockerill, G.W.; Huehns, T.Y.; Weerasinghe, A.; Stocker, C.; Lerch, P.G.; Miller, N.E.; Haskard, D.O. Elevation of plasma high-density lipoprotein concentration reduces interleukin-1-induced expression of E-selectin in an in vivo model of acute inflammation. Circulation 2001, 103, 108–112. [Google Scholar] [CrossRef]
  37. Ou, W.; Jiang, T.; Zhang, N.; Lu, K.; Weng, Y.; Zhou, X.; Wang, D.; Dong, Q.; Tang, X. Role of HDL cholesterol in anthracycline-induced subclinical cardiotoxicity: A prospective observational study in patients with diffuse large B-cell lymphoma treated with R-CHOP. BMJ Open 2024, 14, e074541. [Google Scholar] [CrossRef]
  38. Cvetković, Z.; Milošević, M.; Cvetković, B.; Masnikosa, R.; Arsić, A.; Petrović, S.; Vučić, V. Plasma phospholipid changes are associated with response to chemotherapy in non-Hodgkin lymphoma patients. Leuk. Res. 2017, 54, 39–46. [Google Scholar] [CrossRef]
  39. Cvetković, Z.; Vučić, V.; Cvetković, B.; Petrović, M.; Ristić-Medić, D.; Tepsić, J.; Glibetić, M. Abnormal fatty acid distribution of the serum phospholipids of patients with non-Hodgkin lymphoma. Ann. Hematol. 2010, 89, 775–782. [Google Scholar] [CrossRef]
  40. Cvetković, Z.; Vučić, V.; Cvetković, B.; Karadžić, I.; Ranić, M.; Glibetić, M. Distribution of plasma fatty acids is associated with response to chemotherapy in non-Hodgkin’s lymphoma patients. Med. Oncol. 2013, 30, 741. [Google Scholar] [CrossRef]
  41. Masnikosa, R.; Pirić, D.; Post, J.M.; Cvetković, Z.; Petrović, S.; Paunović, M.; Vučić, V.; Bindila, L. Disturbed Plasma Lipidomic Profiles in Females with Diffuse Large B-Cell Lymphoma: A Pilot Study. Cancers 2023, 15, 3653. [Google Scholar] [CrossRef]
  42. Koundouros, N.; Poulogiannis, G. Reprogramming of fatty acid metabolism in cancer. Br. J. Cancer 2020, 122, 4–22. [Google Scholar] [CrossRef]
  43. Montagne, A.; Kotta, K.; Kielbassa-Elkadi, K.; Martins, I.; Martinez-Climent, J.Á.; Kroemer, G.; Thieblemont, C.; Baud, V. Fatty Acid Metabolism Provides an Essential Survival Signal in OxPhos and BCR DLBCL Cells. Biomedicines 2025, 13, 707. [Google Scholar] [CrossRef]
  44. Pang, Y.; Lu, T.; Xu-Monette, Z.Y.; Young, K.H. Metabolic Reprogramming and Potential Therapeutic Targets in Lymphoma. Int. J. Mol. Sci. 2023, 24, 5493. [Google Scholar] [CrossRef]
  45. Wu, J.; Meng, F.; Ran, D.; Song, Y.; Dang, Y.; Lai, F.; Yang, L.; Deng, M.; Song, Y.; Zhu, J. The Metabolism and Immune Environment in Diffuse Large B-Cell Lymphoma. Metabolites 2023, 13, 734. [Google Scholar] [CrossRef]
  46. Liu, B.; Zhao, X.; Zhang, S.; Li, Q.; Li, X.; Huang, D.; Xia, J.; Ma, N.; Duan, Y.; Zhang, X.; et al. Targeting ZDHHC21/FASN axis for the treatment of diffuse large B-cell lymphoma. Leukemia 2024, 38, 351–364. [Google Scholar] [CrossRef] [PubMed]
  47. Peeters, R.; Cuenca-Escalona, J.; Zaal, E.A.; Hoekstra, A.T.; Balvert, A.C.G.; Vidal-Manrique, M.; Blomberg, N.; van Deventer, S.J.; Stienstra, R.; Jellusova, J.; et al. Fatty acid metabolism in aggressive B-cell lymphoma is inhibited by tetraspanin CD37. Nat. Commun. 2022, 13, 5371. [Google Scholar] [CrossRef]
  48. Zheng, S.; Song, Q.; Zhang, P. Metabolic Modifications, Inflammation, and Cancer Immunotherapy. Front. Oncol. 2021, 11, 703681. [Google Scholar] [CrossRef] [PubMed]
  49. Bobrowicz, M.; Kusowska, A.; Krawczyk, M.; Zhylko, A.; Forcados, C.; Slusarczyk, A.; Barankiewicz, J.; Domagala, J.; Kubacz, M.; Šmída, M.; et al. CD20 expression regulates CD37 levels in B-cell lymphoma—Implications for immunotherapies. Oncoimmunology 2024, 13, 2362454. [Google Scholar] [CrossRef] [PubMed]
  50. Xu-Monette, Z.Y.; Li, L.; Byrd, J.C.; Jabbar, K.J.; Manyam, G.C.; Maria de Winde, C.; van den Brand, M.; Tzankov, A.; Visco, C.; Wang, J.; et al. Assessment of CD37 B-cell antigen and cell of origin significantly improves risk prediction in diffuse large B-cell lymphoma. Blood 2016, 128, 3083–3100. [Google Scholar] [CrossRef]
  51. Chen, Y.; Zhang, J.; Cui, W.; Silverstein, R.L. CD36, a signaling receptor and fatty acid transporter that regulates immune cell metabolism and fate. J. Exp. Med. 2022, 219, e20211314. [Google Scholar] [CrossRef]
  52. Zhou, X.; Su, M.; Lu, J.; Li, D.; Niu, X.; Wang, Y. CD36: The Bridge between Lipids and Tumors. Molecules 2024, 29, 531. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, M.K.; Cheng, L.L.; Yi, H.M.; He, Y.; Li, X.; Fu, D.; Dai, Y.T.; Fang, H.; Cheng, S.; Xu, P.P.; et al. Enhanced lipid metabolism confers the immunosuppressive tumor microenvironment in CD5-positive non-MYC/BCL2 double expressor lymphoma. Front. Oncol. 2022, 12, 885011. [Google Scholar] [CrossRef] [PubMed]
  54. Cheng, W.; Zhou, L.; Luo, Q.; Luo, Y.; Yu, L. The Molecular Mechanism of CD36 in Regulating Cholesterol Metabolism Reprogramming to Drive the Pathogenesis of Diffuse Large B-Cell Lymphoma Cells. Blood 2024, 144 (Suppl. S1), 5769. [Google Scholar] [CrossRef]
  55. Golubovskaya, V.; Zhou, H.; Li, F.; Valentine, M.; Sun, J.; Berahovich, R.; Xu, S.; Quintanilla, M.; Ma, M.C.; Sienkiewicz, J.; et al. Novel CD37, Humanized CD37 and Bi-Specific Humanized CD37-CD19 CAR-T Cells Specifically Target Lymphoma. Cancers 2021, 13, 981. [Google Scholar] [CrossRef] [PubMed]
  56. Chu, Y.; Zhou, X.; Wang, X. Antibody-drug conjugates for the treatment of lymphoma: Clinical advances and latest progress. J. Hematol. Oncol. 2021, 14, 88. [Google Scholar] [CrossRef]
  57. Mougiakakos, D. Anti-CD37 CAR T cells: Another arrow in the quiver. Blood 2024, 144, 1133–1134. [Google Scholar] [CrossRef]
  58. Fernandes Messias, M.C.; Mecatti, G.C.; Figueiredo Angolini, C.F.; Eberlin, M.N.; Credidio, L.; Real Martinez, C.A.; Rodrigues Coy, C.S.; de Oliveira Carvalho, P. Plasma Lipidomic Signature of Rectal Adenocarcinoma Reveals Potential Biomarkers. Front. Oncol. 2018, 7, 325. [Google Scholar] [CrossRef]
  59. Wang, B.; Wu, L.; Chen, J.; Dong, L.; Chen, C.; Wen, Z.; Hu, J.; Fleming, I.; Wang, D.W. Metabolism pathways of arachidonic acids: Mechanisms and potential therapeutic targets. Signal Transduct. Target. Ther. 2021, 6, 94. [Google Scholar] [CrossRef]
  60. Tredicine, M.; Mucci, M.; Recchiuti, A.; Mattoscio, D. Immunoregulatory mechanisms of the arachidonic acid pathway in cancer. FEBS Lett. 2025, 599, 927–951. [Google Scholar] [CrossRef]
  61. Pannunzio, A.; Coluccia, M. Cyclooxygenase-1 (COX-1) and COX-1 Inhibitors in Cancer: A Review of Oncology and Medicinal Chemistry Literature. Pharmaceuticals 2018, 11, 101. [Google Scholar] [CrossRef]
  62. Zhang, Y.; Tighe, S.; Zhu, Y.T. COX-2 Signaling in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1277, 87–104. [Google Scholar] [CrossRef]
  63. Wun, T.; McKnight, H.; Tuscano, J.M. Increased cyclooxygenase-2 (COX-2): A potential role in the pathogenesis of lymphoma. Leuk. Res. 2004, 28, 179–190. [Google Scholar] [CrossRef] [PubMed]
  64. Qi, L.; Pan, X.; Chen, X.; Liu, P.; Chen, M.; Zhang, Q.; Hang, X.; Tang, M.; Wen, D.; Dai, L.; et al. COX-2/PGE2 upregulation contributes to the chromosome 17p-deleted lymphoma. Oncogenesis 2023, 12, 5. [Google Scholar] [CrossRef] [PubMed]
  65. Zaky, A.H.; Elsers, D.; Bakry, R.; Abdelwanis, M.; Nabih, O.; Hafez, R.; Rezk, M. Prognostic Value of Accumulative Expression of COX-2 and p53 in Small and Diffuse Large B Cell Lymphoma. Pathol. Oncol. Res. 2020, 26, 1183–1190. [Google Scholar] [CrossRef] [PubMed]
  66. Hashemi Goradel, N.; Najafi, M.; Salehi, E.; Farhood, B.; Mortezaee, K. Cyclooxygenase-2 in cancer: A review. J. Cell Physiol. 2019, 234, 5683–5699. [Google Scholar] [CrossRef]
  67. Pu, D.; Yin, L.; Huang, L.; Qin, C.; Zhou, Y.; Wu, Q.; Li, Y.; Zhou, Q.; Li, L. Cyclooxygenase-2 Inhibitor: A Potential Combination Strategy With Immunotherapy in Cancer. Front. Oncol. 2021, 11, 637504. [Google Scholar] [CrossRef]
  68. Jin, K.; Qian, C.; Lin, J.; Liu, B. Cyclooxygenase-2-Prostaglandin E2 pathway: A key player in tumor-associated immune cells. Front. Oncol. 2023, 13, 1099811. [Google Scholar] [CrossRef]
  69. Bruno, A.; Tacconelli, S.; Contursi, A.; Ballerini, P.; Patrignani, P. Cyclooxygenases and platelet functions. Adv. Pharmacol. 2023, 97, 133–165. [Google Scholar] [CrossRef]
  70. Zhang, H.; Lu, J.; Jiao, Y.; Chen, Q.; Li, M.; Wang, Z.; Yu, Z.; Huang, X.; Yao, A.; Gao, Q.; et al. Aspirin Inhibits Natural Killer/T-Cell Lymphoma by Modulation of VEGF Expression and Mitochondrial Function. Front. Oncol. 2019, 8, 679. [Google Scholar] [CrossRef]
  71. Elwood, P.; Morgan, G.; Watkins, J.; Protty, M.; Mason, M.; Adams, R.; Dolwani, S.; Pickering, J.; Delon, C.; Longley, M. Aspirin and cancer treatment: Systematic reviews and meta-analyses of evidence: For and against. Br. J. Cancer 2024, 130, 3–8. [Google Scholar] [CrossRef]
  72. Zhang, X.; Feng, Y.; Liu, X.; Ma, J.; Li, Y.; Wang, T.; Li, X. Beyond a chemopreventive reagent, aspirin is a master regulator of the hallmarks of cancer. J. Cancer Res. Clin. Oncol. 2019, 145, 1387–1403. [Google Scholar] [CrossRef]
  73. Sun, M.; Yu, J.; Wan, J.; Dou, X.; Chen, X.; Ye, F. Role of aspirin in cancer prevention. Cancer Treat. Res. Commun. 2025, 43, 100884. [Google Scholar] [CrossRef] [PubMed]
  74. Liebow, M.; Larson, M.C.; Thompson, C.A.; Nowakowski, G.S.; Call, T.G.; Macon, W.R.; Kay, N.E.; Habermann, T.M.; Slager, S.L.; Cerhan, J.R. Aspirin and other nonsteroidal anti-inflammatory drugs, statins and risk of non-Hodgkin lymphoma. Int. J. Cancer 2021, 149, 535–545. [Google Scholar] [CrossRef] [PubMed]
  75. Bernard, M.P.; Bancos, S.; Sime, P.J.; Phipps, R.P. Targeting cyclooxygenase-2 in hematological malignancies: Rationale and promise. Curr. Pharm. Des. 2008, 14, 2051–2060. [Google Scholar] [CrossRef]
  76. Li, S.; Jiang, M.; Wang, L.; Yu, S. Combined chemotherapy with cyclooxygenase-2 (COX-2) inhibitors in treating human cancers: Recent advancement. Biomed. Pharmacother. 2020, 129, 110389. [Google Scholar] [CrossRef]
  77. Karai, E.; Szebényi, K.; Windt, T.; Fehér, S.; Szendi, E.; Dékay, V.; Vajdovich, P.; Szakács, G.; Füredi, A. Celecoxib Prevents Doxorubicin-Induced Multidrug Resistance in Canine and Mouse Lymphoma Cell Lines. Cancers 2020, 12, 1117. [Google Scholar] [CrossRef] [PubMed]
  78. Torres-Collado, A.X.; Jazirehi, A.R. Overcoming Resistance of Human Non-Hodgkin’s Lymphoma to CD19-CAR CTL Therapy by Celecoxib and Histone Deacetylase Inhibitors. Cancers 2018, 10, 200. [Google Scholar] [CrossRef]
  79. Khvorost, D.; Kendall, B.; Jazirehi, A.R. Immunotherapy of Hematological Malignancies of Human B-Cell Origin with CD19 CAR T Lymphocytes. Cells 2024, 13, 662. [Google Scholar] [CrossRef]
  80. Ardisson Korat, A.V.; Chiu, Y.H.; Bertrand, K.A.; Zhang, S.; Epstein, M.M.; Rosner, B.A.; Chiuve, S.; Campos, H.; Giovannucci, E.L.; Chavarro, J.E.; et al. A prospective analysis of red blood cell membrane polyunsaturated fatty acid levels and risk of non-Hodgkin lymphoma. Leuk. Lymphoma 2022, 63, 3351–3361. [Google Scholar] [CrossRef]
  81. Thanarajasingam, G.; Maurer, M.J.; Habermann, T.M.; Nowakowski, G.S.; Bennani, N.N.; Thompson, C.A.; Cerhan, J.R.; Witzig, T.E. Low Plasma Omega-3 Fatty Acid Levels May Predict Inferior Prognosis in Untreated Diffuse Large B-Cell Lymphoma: A New Modifiable Dietary Biomarker? Nutr. Cancer 2018, 70, 1088–1090. [Google Scholar] [CrossRef]
  82. Azrad, M.; Turgeon, C.; Demark-Wahnefried, W. Current evidence linking polyunsaturated Fatty acids with cancer risk and progression. Front. Oncol. 2013, 3, 224. [Google Scholar] [CrossRef] [PubMed]
  83. Gu, Z.; Shan, K.; Chen, H.; Chen, Y.Q. n-3 Polyunsaturated Fatty Acids and their Role in Cancer Chemoprevention. Curr. Pharmacol. Rep. 2015, 1, 283–294. [Google Scholar] [CrossRef] [PubMed]
  84. Lam, C.N.; Watt, A.E.; Isenring, E.A.; de van der Schueren, M.A.E.; van der Meij, B.S. The effect of oral omega-3 polyunsaturated fatty acid supplementation on muscle maintenance and quality of life in patients with cancer: A systematic review and meta-analysis. Clin. Nutr. 2021, 40, 3815–3826. [Google Scholar] [CrossRef]
  85. Guo, Y.; Ma, B.; Li, X.; Hui, H.; Zhou, Y.; Li, N.; Xie, X. n-3 PUFA can reduce IL-6 and TNF levels in patients with cancer. Br. J. Nutr. 2023, 129, 54–65. [Google Scholar] [CrossRef]
  86. Jamali, M.; Zarezadeh, M.; Jamilian, P.; Ghoreishi, Z. The effect of n-3 polyunsaturated fatty acids on inflammation status in cancer patients: Updated systematic review and dose- and time-response meta-analysis. PharmaNutrition 2024, 27, 100372. [Google Scholar] [CrossRef]
  87. Moloudizargari, M.; Mortaz, E.; Asghari, M.H.; Adcock, I.M.; Redegeld, F.A.; Garssen, J. Effects of the polyunsaturated fatty acids, EPA and DHA, on hematological malignancies: A systematic review. Oncotarget 2018, 9, 11858–11875. [Google Scholar] [CrossRef]
  88. Ceccarelli, V.; Nocentini, G.; Billi, M.; Racanicchi, S.; Riccardi, C.; Roberti, R.; Grignani, F.; Binaglia, L.; Vecchini, A. Eicosapentaenoic acid activates RAS/ERK/C/EBPβ pathway through H-ras intron 1 CpG island demethylation in U937 leukemia cells. PLoS ONE 2014, 9, e85025. [Google Scholar] [CrossRef] [PubMed]
  89. Kapadia, B.; Nanaji, N.M.; Bhalla, K.; Bhandary, B.; Lapidus, R.; Beheshti, A.; Evens, A.M.; Gartenhaus, R.B. Fatty Acid Synthase induced S6Kinase facilitates USP11-eIF4B complex formation for sustained oncogenic translation in DLBCL. Nat. Commun. 2018, 9, 829. [Google Scholar] [CrossRef]
  90. Bakhshi, T.J.; Way, T.; Muncy, B.; Georgel, P.T. Effects of the omega-3 fatty acid DHA on histone and p53 acetylation in diffuse large B cell lymphoma. Biochem. Cell Biol. 2023, 101, 172–191. [Google Scholar] [CrossRef]
  91. Nishida, A.; Andoh, A. The Role of Inflammation in Cancer: Mechanisms of Tumor Initiation, Progression, and Metastasis. Cells 2025, 14, 488. [Google Scholar] [CrossRef]
  92. Hart, P.C.; Rajab, I.M.; Alebraheem, M.; Potempa, L.A. C-Reactive Protein and Cancer-Diagnostic and Therapeutic Insights. Front. Immunol. 2020, 11, 595835. [Google Scholar] [CrossRef]
  93. Negreiros, E.A.D.S.; da Silveira, T.M.B.; Fortier, S.C.; Chiattone, C.S. Evaluation of C-reactive protein and its prognostic relationship in patients with Hodgkin’s Lymphoma. Hematol. Transfus. Cell Ther. 2024, 46, S53–S58. [Google Scholar] [CrossRef] [PubMed]
  94. Cao, Y.; Shi, Y.X.; Chen, J.O.; Tan, Y.T.; Cai, Y.C.; Luo, H.Y.; Qiu, M.Z.; Cai, X.Y.; Jin, Y.; Sun, Y.L.; et al. Serum C-reactive protein as an important prognostic variable in patients with diffuse large B cell lymphoma. Tumour Biol. 2012, 33, 1039–1044. [Google Scholar] [CrossRef] [PubMed]
  95. Adams, H.J.; de Klerk, J.M.; Fijnheer, R.; Heggelman, B.G.; Dubois, S.V.; Nievelstein, R.A.; Kwee, T.C. Prognostic Value of Anemia and C-Reactive Protein Levels in Diffuse Large B-Cell Lymphoma. Clin. Lymphoma Myeloma Leuk. 2015, 15, 671–679. [Google Scholar] [CrossRef] [PubMed]
  96. Wang, J.; Zhou, M.; Wang, X.; Xu, J.; Chen, B.; Ouyang, J. Pretreatment C-reactive protein was an independent prognostic factor for patients with diffuse large B-cell lymphoma treated with RCHOP. Clin. Chim. Acta 2016, 459, 150–154. [Google Scholar] [CrossRef]
  97. Qin, W.; Yuan, Q.; Wu, J.; Yu, H.; Wang, Y.; Chen, Q. Prognostic value of pre-therapy C-reactive protein level in diffuse large B-cell lymphoma: A meta-analysis. Leuk. Lymphoma 2019, 60, 358–366. [Google Scholar] [CrossRef]
  98. Huang, Z.; Wang, K.; Huang, S.; Lu, Q. Prognostic value of baseline C-reactive protein in diffuse large B-cell lymphoma: A systematic review and meta-analysis. Transl. Cancer Res. 2023, 12, 2169–2180. [Google Scholar] [CrossRef]
  99. Troppan, K.T.; Schlick, K.; Deutsch, A.; Melchardt, T.; Egle, A.; Stojakovic, T.; Beham-Schmid, C.; Weiss, L.; Neureiter, D.; Wenzl, K.; et al. C-reactive protein level is a prognostic indicator for survival and improves the predictive ability of the R-IPI score in diffuse large B-cell lymphoma patients. Br. J. Cancer 2014, 111, 55–60. [Google Scholar] [CrossRef]
  100. Gradel, K.O.; Larsen, T.S.; Frederiksen, H.; Vinholt, P.J.; Iachina, M.; Póvoa, P.; Zampieri, F.G.; Nielsen, S.L.; Dessau, R.B.; Møller, J.K.; et al. Impact of C-reactive protein and albumin levels on short, medium, and long term mortality in patients with diffuse large B-cell lymphoma. Ann. Med. 2022, 54, 713–722. [Google Scholar] [CrossRef]
  101. Dix, C.; Zeller, J.; Stevens, H.; Eisenhardt, S.U.; Shing, K.S.C.T.; Nero, T.L.; Morton, C.J.; Parker, M.W.; Peter, K.; McFadyen, J.D. C-reactive protein, immunothrombosis and venous thromboembolism. Front. Immunol. 2022, 13, 1002652. [Google Scholar] [CrossRef]
  102. Otasevic, V.; Mihaljevic, B.; Milic, N.; Stanisavljevic, D.; Vukovic, V.; Tomic, K.; Fareed, J.; Antic, D. Immune activation and inflammatory biomarkers as predictors of venous thromboembolism in lymphoma patients. Thromb. J. 2022, 20, 20. [Google Scholar] [CrossRef] [PubMed]
  103. Detroit, M.; Collier, M.; Beeker, N.; Willems, L.; Decroocq, J.; Deau-Fischer, B.; Vignon, M.; Birsen, R.; Moufle, F.; Leclaire, C.; et al. Predictive Factors of Response to Immunotherapy in Lymphomas: A Multicentre Clinical Data Warehouse Study (PRONOSTIM). Cancers 2023, 15, 4028. [Google Scholar] [CrossRef] [PubMed]
  104. Nierengarten, M.B. Simple blood test predicts patients most likely to have poor CAR T-cell outcomes. Cancer 2024, 130, 2084. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, X.; Yu, X.; Chen, C.; Chen, C.; Wang, Y.; Su, D.; Zhu, L. Fibrinogen and tumors. Front. Oncol. 2024, 14, 1393599. [Google Scholar] [CrossRef]
  106. Shehata, A.M.F.; Aldesoky, A.I.; Gohar, S.F. Plasma fibrinogen level as possible prognostic biomarker in diffuse large B-cell lymphoma. Hematology 2019, 24, 103–107. [Google Scholar] [CrossRef]
  107. Zhang, B.; Lin, L.; Ji, J.M.; Wu, Y.; Shen, Q. Prognostic Value of Plasma Fibrinogen Levels in Patients with Diffuse Large B-Cell Lymphoma. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2025, 33, 114–120. [Google Scholar] [CrossRef]
  108. Troppan, K.T.; Melchardt, T.; Wenzl, K.; Schlick, K.; Deutsch, A.; Bullock, M.D.; Reitz, D.; Beham-Schmid, C.; Weiss, L.; Neureiter, D.; et al. The clinical significance of fibrinogen plasma levels in patients with diffuse large B cell lymphoma. J. Clin. Pathol. 2016, 69, 326–330. [Google Scholar] [CrossRef]
  109. Çiftçiler, R.; Ciftciler, A.E.; Selim, C. Evaluation of the association between serum danger molecules at diagnosis and survival outcomes in patients with diffuse large B cell lymphoma. Med. Int. 2025, 5, 25. [Google Scholar] [CrossRef]
  110. Ogura, S.; Nakazato, T. The Prognostic Significance of Pretransplant Plasma Fibrinogen Levels in Autologous Hematopoietic Stem Cell Transplantation. Transpl. Proc. 2021, 53, 405–407. [Google Scholar] [CrossRef]
  111. Guven, D.C.; Sahin, T.K.; Erul, E.; Rizzo, A.; Ricci, A.D.; Aksoy, S.; Yalcin, S. The association between albumin levels and survival in patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Front. Mol. Biosci. 2022, 9, 1039121. [Google Scholar] [CrossRef]
  112. Tang, Q.; Li, X.; Sun, C.R. Predictive value of serum albumin levels on cancer survival: A prospective cohort study. Front. Oncol. 2024, 14, 1323192. [Google Scholar] [CrossRef] [PubMed]
  113. Bairey, O.; Shacham-Abulafia, A.; Shpilberg, O.; Gurion, R. Serum albumin level at diagnosis of diffuse large B-cell lymphoma: An important simple prognostic factor. Hematol. Oncol. 2016, 34, 184–192. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, L.; Pan, L.; Liu, T. Prognostic value of serum albumin level in patients with diffuse large B cell lymphoma. Acta Biochim. Pol. 2023, 70, 767–776. [Google Scholar] [CrossRef]
  115. Hu, X.; Feng, X.; Wang, H.; Miao, Y.; Lian, X.; Gao, Q.; Gao, Y.; Zhai, X.; Zhang, D.; Niu, B.; et al. Association between serum albumin levels and survival in elderly patients with diffuse large B-cell lymphoma: A single-center retrospective study. Transl. Cancer Res. 2023, 12, 1577–1587. [Google Scholar] [CrossRef]
  116. Bouillanne, O.; Morineau, G.; Dupont, C.; Coulombel, I.; Vincent, J.P.; Nicolis, I.; Benazeth, S.; Cynober, L.; Aussel, C. Geriatric Nutritional Risk Index: A new index for evaluating at-risk elderly medical patients. Am. J. Clin. Nutr. 2005, 82, 777–783. [Google Scholar] [CrossRef]
  117. Zhou, Q.; Wei, Y.; Huang, F.; Wei, X.; Wei, Q.; Hao, X.; Zhang, Y.; Feng, R. Low prognostic nutritional index predicts poor outcome in diffuse large B-cell lymphoma treated with R-CHOP. Int. J. Hematol. 2016, 104, 485–490. [Google Scholar] [CrossRef]
  118. Yan, D.; Shen, Z.; Zhang, S.; Hu, L.; Sun, Q.; Xu, K.; Jin, Y.; Sang, W. Prognostic values of geriatric nutritional risk index (GNRI) and prognostic nutritional index (PNI) in elderly patients with Diffuse Large B-Cell Lymphoma. J. Cancer 2021, 12, 7010–7017. [Google Scholar] [CrossRef] [PubMed]
  119. Yu, Q.; Tian, M.; Pi, G.; Jia, Y.; Jin, X. Geriatric nutritional risk index as a predictor of prognosis in hematologic malignancies: A systematic review and meta-analysis. Front. Nutr. 2023, 10, 1274592. [Google Scholar] [CrossRef]
  120. Pénichoux, J.; Lanic, H.; Thill, C.; Ménard, A.L.; Camus, V.; Stamatoullas, A.; Lemasle, E.; Leprêtre, S.; Lenain, P.; Contentin, N.; et al. Prognostic relevance of sarcopenia, geriatric, and nutritional assessments in older patients with diffuse large B-cell lymphoma: Results of a multicentric prospective cohort study. Ann. Hematol. 2023, 102, 1811–1823. [Google Scholar] [CrossRef]
  121. Soler, M.F.; Abaurrea, A.; Azcoaga, P.; Araujo, A.M.; Caffarel, M.M. New perspectives in cancer immunotherapy: Targeting IL-6 cytokine family. J. Immunother. Cancer 2023, 11, e007530. [Google Scholar] [CrossRef]
  122. Guo, Q.; Jin, Y.; Chen, X.; Ye, X.; Shen, X.; Lin, M.; Zeng, C.; Zhou, T.; Zhang, J. NF-κB in biology and targeted therapy: New insights and translational implications. Signal Transduct. Target. Ther. 2024, 9, 53. [Google Scholar] [CrossRef]
  123. Huang, B.; Lang, X.; Li, X. The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front. Oncol. 2022, 12, 1023177. [Google Scholar] [CrossRef]
  124. Thuya, W.L.; Cao, Y.; Ho, P.C.; Wong, A.L.; Wang, L.; Zhou, J.; Nicot, C.; Goh, B.C. Insights into IL-6/JAK/STAT3 signaling in the tumor microenvironment: Implications for cancer therapy. Cytokine Growth Factor Rev. 2025; in press. [Google Scholar] [CrossRef]
  125. Gholiha, A.R.; Hollander, P.; Glimelius, I.; Hedstrom, G.; Molin, D.; Hjalgrim, H.; Smedby, K.E.; Hashemi, J.; Amini, R.M.; Enblad, G. Revisiting IL-6 expression in the tumor microenvironment of classical Hodgkin lymphoma. Blood Adv. 2021, 5, 1671–1681. [Google Scholar] [CrossRef]
  126. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef]
  127. Bao, C.; Gu, J.; Huang, X.; You, L.; Zhou, Z.; Jin, J. Cytokine profiles in patients with newly diagnosed diffuse large B-cell lymphoma: IL-6 and IL-10 levels are associated with adverse clinical features and poor outcomes. Cytokine 2023, 169, 156289. [Google Scholar] [CrossRef]
  128. Burger, R. Impact of interleukin-6 in hematological malignancies. Transfus. Med. Hemother. 2013, 40, 336–343. [Google Scholar] [CrossRef]
  129. Kim, J.H.; Kim, W.S.; Park, C. Interleukin-6 mediates resistance to PI3K-pathway-targeted therapy in lymphoma. BMC Cancer 2019, 19, 936. [Google Scholar] [CrossRef]
  130. Guney, N.; Soydinc, H.O.; Basaran, M.; Bavbek, S.; Derin, D.; Camlica, H.; Yasasever, V.; Topuz, E. Serum levels of interleukin-6 and interleukin-10 in Turkish patients with aggressive non-Hodgkin’s lymphoma. Asian Pac. J. Cancer Prev. 2009, 10, 669–674. [Google Scholar]
  131. Hashwah, H.; Bertram, K.; Stirm, K.; Stelling, A.; Wu, C.T.; Kasser, S.; Manz, M.G.; Theocharides, A.P.; Tzankov, A.; Müller, A. The IL-6 signaling complex is a critical driver, negative prognostic factor, and therapeutic target in diffuse large B-cell lymphoma. EMBO Mol. Med. 2019, 11, e10576. [Google Scholar] [CrossRef]
  132. Santomasso, B.D.; Nastoupil, L.J.; Adkins, S.; Lacchetti, C.; Schneider, B.J.; Anadkat, M.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; et al. Management of Immune-Related Adverse Events in Patients Treated With Chimeric Antigen Receptor T-Cell Therapy: ASCO Guideline. J. Clin. Oncol. 2021, 39, 3978–3992. [Google Scholar] [CrossRef]
  133. Crombie, J.L.; Graff, T.; Falchi, L.; Karimi, Y.H.; Bannerji, R.; Nastoupil, L.; Thieblemont, C.; Ursu, R.; Bartlett, N.; Nachar, V.; et al. Consensus recommendations on the management of toxicity associated with CD3×CD20 bispecific antibody therapy. Blood 2024, 143, 1565–1575. [Google Scholar] [CrossRef] [PubMed]
  134. Arribas, A.J.; Napoli, S.; Gaudio, E.; Herbaux, C.; Cannas, E.; Tarantelli, C.; Bordone-Pittau, R.; Cascione, L.; Munz, N.; Aresu, L.; et al. PI3Kδ activation, IL-6 overexpression, and CD37 loss cause resistance to naratuximab emtansine in lymphomas. Blood Adv. 2024, 8, 6268–6281. [Google Scholar] [CrossRef]
  135. Li, J.; Wu, Y.; Zhang, X.; Wang, X. Causal relationship between beta-2 microglobulin and B-cell malignancies: Genome-wide meta-analysis and a bidirectional two-sample Mendelian randomization study. Front. Immunol. 2024, 15, 1448476. [Google Scholar] [CrossRef] [PubMed]
  136. Jelicic, J.; Juul-Jensen, K.; Bukumiric, Z.; Runason Simonsen, M.; Roost Clausen, M.; Ludvigsen Al-Mashhadi, A.; Schou Pedersen, R.; Bjørn Poulsen, C.; Ortved Gang, A.; Brown, P.; et al. Revisiting beta-2 microglobulin as a prognostic marker in diffuse large B-cell lymphoma. Cancer Med. 2024, 13, e7239. [Google Scholar] [CrossRef]
  137. Takahara, T.; Nakamura, S.; Tsuzuki, T.; Satou, A. The Immunology of DLBCL. Cancers 2023, 15, 835. [Google Scholar] [CrossRef]
  138. Xiong, X.; Xie, X.; Wang, Z.; Zhang, Y.; Wang, L. Tumor-associated macrophages in lymphoma: From mechanisms to therapy. Int. Immunopharmacol. 2022, 112, 109235. [Google Scholar] [CrossRef] [PubMed]
  139. Zhou, Z.; Zhu, P.; Ge, J.; Li, Q.; Li, H.; Zhe, N.; Liu, Z.; Chen, D. Gene regulatory mechanisms of T cell exhaustion in diffuse large B cell lymphoma based on single-cell transcriptome data. Leuk. Res. 2024, 146, 107588. [Google Scholar] [CrossRef]
  140. Jia, H.; Yang, H.; Xiong, H.; Luo, K.Q. NK cell exhaustion in the tumor microenvironment. Front. Immunol. 2023, 14, 1303605. [Google Scholar] [CrossRef]
  141. Zhang, A.; Fan, T.; Liu, Y.; Yu, G.; Li, C.; Jiang, Z. Regulatory T cells in immune checkpoint blockade antitumor therapy. Mol. Cancer 2024, 23, 251. [Google Scholar] [CrossRef]
  142. Dong, W.; Wu, X.; Ma, S.; Wang, Y.; Nalin, A.P.; Zhu, Z.; Zhang, J.; Benson, D.M.; He, K.; Caliguri, M.A.; et al. The Mechanism of Anti-PD-L1 Antibody Efficacy against PD-L1-Negative Tumors Identifies NK Cells Expressing PD-L1 as a Cytolytic Effector. Cancer Discov. 2019, 9, 1422–1437. [Google Scholar] [CrossRef]
  143. Liu, Y.; Zhou, X.; Wang, X. Targeting the tumor microenvironment in B-cell lymphoma: Challenges and opportunities. J. Hematol. Oncol. 2021, 14, 125. [Google Scholar] [CrossRef] [PubMed]
  144. Sica, A.; Bronte, V. Altered macrophage differentiation and immune dysfunction in tumor development. J. Clin. Investig. 2007, 117, 1155–1166. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, H.; Kim, C.; Zou, W. Metabolism and macrophages in the tumor microenvironment. Curr. Opin. Immunol. 2024, 91, 102491. [Google Scholar] [CrossRef]
  146. Wang, S.; Wang, J.; Chen, Z.; Luo, J.; Guo, W.; Sun, L.; Lin, L. Targeting M2-like tumor-associated macrophages is a potential therapeutic approach to overcome antitumor drug resistance. NPJ Precis. Oncol. 2024, 8, 31. [Google Scholar] [CrossRef] [PubMed]
  147. Shen, L.; Li, H.; Shi, Y.; Wang, D.; Gong, J.; Xun, J.; Zhou, S.; Xiang, R.; Tan, X. M2 tumour-associated macrophages contribute to tumour progression via legumain remodelling the extracellular matrix in diffuse large B cell lymphoma. Sci. Rep. 2016, 6, 30347. [Google Scholar] [CrossRef]
  148. Nam, S.J.; Go, H.; Paik, J.H.; Kim, T.M.; Heo, D.S.; Kim, C.W.; Jeon, Y.K. An increase of M2 macrophages predicts poor prognosis in patients with diffuse large B-cell lymphoma treated with rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone. Leuk. Lymphoma 2014, 55, 2466–2476. [Google Scholar] [CrossRef]
  149. Marchesi, F.; Cirillo, M.; Bianchi, A.; Gately, M.; Olimpieri, O.M.; Cerchiara, E.; Renzi, D.; Micera, A.; Balzamino, B.O.; Bonini, S.; et al. High density of CD68+/CD163+ tumour-associated macrophages (M2-TAM) at diagnosis is significantly correlated to unfavorable prognostic factors and to poor clinical outcomes in patients with diffuse large B-cell lymphoma. Hematol. Oncol. 2015, 33, 110–112. [Google Scholar] [CrossRef]
  150. Ribatti, D.; Tamma, R.; Annese, T.; Ingravallo, G.; Specchia, G. Macrophages and angiogenesis in human lymphomas. Clin. Exp. Med. 2024, 24, 26. [Google Scholar] [CrossRef]
  151. Wang, J.; Gao, K.; Lei, W.; Dong, L.; Xuan, Q.; Feng, M.; Wang, J.; Ye, X.; Jin, T.; Zhanh, 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]
  152. Guo, B.; Duan, Y.; Cen, H. Prognostic model based on M2 macrophage-related signatures for predicting outcomes, enhancing risk stratification, and providing therapeutic insights in diffuse large B-cell lymphoma. Heliyon 2024, 10, e41007. [Google Scholar] [CrossRef]
  153. Liu, Y.; Tan, H.; Dai, J.; Lin, J.; Zhao, K.; Hu, H.; Zhong, C. Targeting macrophages in cancer immunotherapy: Frontiers and challenges. J. Adv. Res. 2025; in press. [Google Scholar] [CrossRef]
  154. Wilcox, R.A.; Ristow, K.; Habermann, T.M.; Inwards, D.J.; Micallef, I.N.; Johnston, P.B.; Colgan, J.P.; Nowakowski, G.S.; Ansell, S.M.; Witzig, T.E.; et al. The absolute monocyte and lymphocyte prognostic score predicts survival and identifies high-risk patients in diffuse large-B-cell lymphoma. Leukemia 2011, 25, 1502–1509. [Google Scholar] [CrossRef] [PubMed]
  155. Tadmor, T.; Bari, A.; Sacchi, S.; Marcheselli, L.; Liardo, E.V.; Avivi, I.; Benyamini, N.; Attias, D.; Pozzi, S.; Cox, M.C.; et al. Monocyte count at diagnosis is a prognostic parameter in diffuse large B-cell lymphoma: Results from a large multicenter study involving 1191 patients in the pre- and post-rituximab era. Haematologica 2014, 99, 125–130. [Google Scholar] [CrossRef] [PubMed]
  156. Marcheselli, R.; Franceschetto, A.; Sacchi, S.; Bari, A.; Levy, I.; Pizzichini, P.; Prosper, D.; D’Apollo, R.; Massi, L.; Casolo, A.; et al. The prognostic role of end of treatment FDG-PET-CT in patients with diffuse large B cell lymphoma can be improved by considering it with absolute monocyte count at diagnosis. Leuk. Lymphoma 2019, 60, 1958–1964. [Google Scholar] [CrossRef]
  157. Liu, C.; Chu, D.; Kalantar-Zadeh, K.; George, J.; Young, H.A.; Liu, G. Cytokines: From Clinical Significance to Quantification. Adv. Sci. 2021, 8, 2004433. [Google Scholar] [CrossRef] [PubMed]
  158. Wang, K.; Wang, X.; Song, L. Unraveling the complex role of neutrophils in lymphoma: From pathogenesis to therapeutic approaches (Review). Mol. Clin. Oncol. 2024, 21, 85. [Google Scholar] [CrossRef]
  159. Nie, M.; Yang, L.; Bi, X.; Wang, Y.; Sun, P.; Yang, H.; Liu, P.; Li, Z.; Xia, Y.; Jiang, Z. Neutrophil Extracellular Traps Induced by IL8 Promote Diffuse Large B-cell Lymphoma Progression via the TLR9 Signaling. Clin. Cancer Res. 2019, 25, 1867–1879. [Google Scholar] [CrossRef]
  160. Hu, W.; Lee, S.M.L.; Bazhin, A.V.; Guba, M.; Werner, J.; Nieß, H. Neutrophil extracellular traps facilitate cancer metastasis: Cellular mechanisms and therapeutic strategies. J. Cancer Res. Clin. Oncol. 2023, 149, 2191–2210. [Google Scholar] [CrossRef]
  161. Yang, Z.; Yu, W. Clinical significance of circulating neutrophils and lymphocyte subsets in newly diagnosed patients with diffuse large B-cell lymphoma. Clin. Exp. Med. 2023, 23, 815–822. [Google Scholar] [CrossRef]
  162. Freud, A.G.; Mundy-Bosse, B.L.; Yu, J.; Caligiuri, M.A. The Broad Spectrum of Human Natural Killer Cell Diversity. Immunity 2017, 47, 820–833. [Google Scholar] [CrossRef]
  163. Phan, M.T.; Chun, S.; Kim, S.H.; Ali, A.K.; Lee, S.H.; Kim, S.; Kim, S.H.; Cho, D. Natural killer cell subsets and receptor expression in peripheral blood mononuclear cells of a healthy Korean population: Reference range, influence of age and sex, and correlation between NK cell receptors and cytotoxicity. Hum. Immunol. 2017, 78, 103–112. [Google Scholar] [CrossRef]
  164. Wang, J.; Li, H.; Kulkarni, A.; Anderson, J.L.; Upadhyay, P.; Onyekachi, O.V.; Arantes, L.M.R.B.; Banerjee, H.; Kane, L.P.; Zhang, X.; et al. Differential impact of TIM-3 ligands on NK cell function. J. Immunother. Cancer 2025, 13, e010618. [Google Scholar] [CrossRef] [PubMed]
  165. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef] [PubMed]
  166. Huang, R.; Kang, T.; Chen, S. The role of tumor-associated macrophages in tumor immune evasion. J. Cancer Res. Clin. Oncol. 2024, 150, 238. [Google Scholar] [CrossRef]
  167. Hontecillas-Prieto, L.; García-Domínguez, D.J.; Palazón-Carrión, N.; Martín García-Sancho, A.; Nogales-Fernández, E.; Jimenez-Cortegana, C.; Sanchez-Leon, M.L.; Silva-Romeiro, S.; Flores-Campos, R.; Carnicero-Gonzalez, F.; et al. CD8+ NKs as a potential biomarker of complete response and survival with lenalidomide plus R-GDP in the R2-GDP-GOTEL trial in recurrent/refractory diffuse large B cell lymphoma. Front. Immunol. 2024, 15, 1293931. [Google Scholar] [CrossRef] [PubMed]
  168. Kim, J.K.; Chung, J.S.; Shin, H.J.; Song, M.K.; Yi, J.W.; Shin, D.H.; Lee, D.S.; Baek, S.M. Influence of NK cell count on the survival of patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood Res. 2014, 49, 162–169. [Google Scholar] [CrossRef]
  169. Patente, T.A.; Pinho, M.P.; Oliveira, A.A.; Evangelista, G.C.M.; Bergami-Santos, P.C.; Barbuto, J.A.M. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front. Immunol. 2019, 9, 3176. [Google Scholar] [CrossRef]
  170. Yuan, C.T.; Chuang, S.S.; Cheng, P.Y.; Chang, K.; Wang, H.; Tsai, J.H.; Liau, J.Y.; Chou, W.C. Decreased CD11c-positive dendritic cells in the tumor microenvironment predict double-hit/triple-hit genotype and survival in diffuse large B-cell lymphoma. J. Pathol. Clin. Res. 2022, 8, 436–447. [Google Scholar] [CrossRef]
  171. Koumpis, E.; Papoudou-Bai, A.; Papathanasiou, K.; Kolettas, E.; Kanavaros, P.; Hatzimichael, E. Unraveling the Immune Microenvironment in Diffuse Large B-Cell Lymphoma: Prognostic and Potential Therapeutic Implications. Curr. Issues Mol. Biol. 2024, 46, 7048–7064. [Google Scholar] [CrossRef]
  172. Ciavarella, S.; Vegliante, M.C.; Fabbri, M.; De Summa, S.; Melle, F.; Motta, G.; De Iuliis, V.; Opinto, G.; Enjuanes, A.; Rega, S.; et al. Dissection of DLBCL microenvironment provides a gene expression-based predictor of survival applicable to formalin-fixed paraffin-embedded tissue. Ann. Oncol. 2018, 29, 2363–2370. [Google Scholar] [CrossRef]
  173. Elhelbawy, N.G.; Nassar, A.A.H.; Eltorgoman, A.E.A.; Saber, S.M.; Badr, E.A. Immunological microenvironment gene expression in patients with diffuse large B cell non Hodgkin lymphoma. Biochem. Biophys. Rep. 2020, 21, 100731. [Google Scholar] [CrossRef]
  174. Lee, S.; Kim, D.H.; Oh, S.Y.; Kim, S.Y.; Koh, M.S.; Lee, J.H.; Lee, S.; Kim, S.H.; Kwak, J.Y.; Pak, M.G.; et al. Clinicopathologic significance of tumor microenvironment CD11c, and FOXP3 expression in diffuse large B-cell lymphoma patients receiving rituximab, cyclophosphamide, anthracycline, vincristine, and prednisone (R-CHOP) combination chemotherapy. Korean J. Intern. Med. 2017, 32, 335–344. [Google Scholar] [CrossRef] [PubMed]
  175. Schmitz, R.; Wright, G.W.; Huang, D.W.; Johnson, C.A.; Phelan, J.D.; Wang, J.Q.; Roulland, S.; Kasbekar, M.; Young, R.M.; Shaffer, A.L.; et al. Genetics and Pathogenesis of Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2018, 378, 1396–1407. [Google Scholar] [CrossRef] [PubMed]
  176. Fangazio, M.; Ladewig, E.; Gomez, K.; Garcia-Ibanez, L.; Kumar, R.; Teruya-Feldstein, J.; Rossi, D.; Filip, I.; Pan-Hammarström, Q.; Inghirami, G.; et al. Genetic mechanisms of HLA-I loss and immune escape in diffuse large B cell lymphoma. Proc. Natl. Acad. Sci. USA 2021, 118, e2104504118. [Google Scholar] [CrossRef]
  177. Ansell, S.M.; Stenson, M.; Habermann, T.M.; Jelinek, D.F.; Witzig, T.E. Cd4+ T-1cell immune response to large B-cell non-Hodgkin’s lymphoma predicts patient outcome. J. Clin. Oncol. 2001, 19, 720–726. [Google Scholar] [CrossRef]
  178. Duell, J.; Westin, J. The future of immunotherapy for diffuse large B-cell lymphoma. Int. J. Cancer 2025, 156, 251–261. [Google Scholar] [CrossRef] [PubMed]
  179. Lu, Y.; Li, Y.; Yu, J.; Meng, S.; Bi, C.; Guan, Q.; Li, L.; Qiu, L.; Qian, Z.; Zhou, S.; et al. OX40 shapes an inflamed tumor immune microenvironment and predicts response to immunochemotherapy in diffuse large B-cell lymphoma. Clin. Immunol. 2023, 251, 109637. [Google Scholar] [CrossRef]
  180. Thapa, B.; Kato, S.; Nishizaki, D.; Miyashita, H.; Lee, S.; Nesline, M.K.; Previs, R.A.; Conroy, J.M.; DePietro, P.; Pabla, S.; et al. OX40/OX40 ligand and its role in precision immune oncology. Cancer Metastasis Rev. 2024, 43, 1001–1013. [Google Scholar] [CrossRef]
  181. Zhang, T.; Lu, Y.; Liu, X.; Zhao, M.; He, J.; Liu, X.; Li, L.; Qiu, L.; Qian, Z.; Zhou, S.; et al. Comprehensive analysis of TP53 mutation characteristics and identification of patients with inferior prognosis and enhanced immune escape in diffuse large B-cell lymphoma. Am. J. Hematol. 2022, 97, E14–E17. [Google Scholar] [CrossRef]
  182. Hou, H.; Luo, Y.; Tang, G.; Zhang, B.; Ouyang, R.; Wang, T.; Huang, M.; Wu, S.; Li, D.; Wang, F. Dynamic changes in peripheral blood lymphocyte subset counts and functions in patients with diffuse large B cell lymphoma during chemotherapy. Cancer Cell Int. 2021, 21, 282. [Google Scholar] [CrossRef]
  183. Ray-Coquard, I.; Cropet, C.; Van Glabbeke, M.; Sebban, C.; Le Cesne, A.; Judson, I.; Tredan, O.; Verweij, J.; Biron, P.; Labidi, I.; et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009, 69, 5383–5391. [Google Scholar] [CrossRef]
  184. Feng, J.; Wang, Z.; Guo, X.; Chen, Y.; Cheng, Y.; Tang, Y. Prognostic significance of absolute lymphocyte count at diagnosis of diffuse large B-cell lymphoma: A meta-analysis. Int. J. Hematol. 2012, 95, 143–148. [Google Scholar] [CrossRef] [PubMed]
  185. Partanen, A.; Turunen, A.; Kuittinen, O.; Kuitunen, H.; Mäntymaa, P.; Varmavuo, V.; Jantunen, E. Predictive Factors for Early Immune Recovery in NHL Patients after Autologous Transplantation: A Multicenter Prospective Study. Cancers 2024, 16, 2550. [Google Scholar] [CrossRef] [PubMed]
  186. Lu, Y.; Zhu, H.; Liu, Y.; Wang, Y.; Sun, Y.; Cheng, H.; Yan, Z.; Cao, J.; Sang, W.; Zhu, F.; et al. Prognostic value of prelymphodepletion absolute lymphocyte counts in relapsed/refractory diffuse large B-cell lymphoma patients treated with chimeric antigen receptor T cells. Front. Immunol. 2023, 14, 1155216. [Google Scholar] [CrossRef] [PubMed]
  187. Sonmez, O.; Sonmez, M. Role of platelets in immune system and inflammation. Porto Biomed. J. 2017, 2, 311–314. [Google Scholar] [CrossRef]
  188. Contursi, A.; Tacconelli, S.; Di Berardino, S.; De Michele, A.; Patrignani, P. Platelets as crucial players in the dynamic interplay of inflammation, immunity, and cancer: Unveiling new strategies for cancer prevention. Front. Pharmacol. 2024, 15, 1520488. [Google Scholar] [CrossRef]
  189. Chen, H.; Qin, Y.; Yang, J.; Liu, P.; He, X.; Zhou, S.; Zhang, C.; Gui, L.; Yang, S.; Shi, Y. The pretreatment platelet count predicts survival outcomes of diffuse large B-cell lymphoma: An analysis of 1007 patients in the rituximab era. Leuk. Res. 2021, 110, 106715. [Google Scholar] [CrossRef]
  190. Knight, K.; Wade, S.; Balducci, L. Prevalence and outcomes of anemia in cancer: A systematic review of the literature. Am. J. Med. 2004, 116, 11–26. [Google Scholar] [CrossRef]
  191. Solal-Céligny, P.; Roy, P.; Colombat, P.; White, J.; Armitage, J.O.; Arranz-Saez, R.; Au, W.Y.; Bellei, M.; Brice, P.; Caballero, D.; et al. Follicular lymphoma international prognostic index. Blood 2004, 104, 1258–1265. [Google Scholar] [CrossRef]
  192. Federico, M.; Bellei, M.; Marcheselli, L.; Luminari, S.; Lopez-Guillermo, A.; Vitolo, U.; Pro, B.; Pileri, S.; Pulsoni, A.; Soubeyran, P.; et al. Follicular lymphoma international prognostic index 2: A new prognostic index for follicular lymphoma developed by the international follicular lymphoma prognostic factor project. J. Clin. Oncol. 2009, 27, 4555–4562. [Google Scholar] [CrossRef]
  193. Hong, J.; Woo, H.S.; Kim, H.; Ahn, H.K.; Sym, S.J.; Park, J.; Ahn, J.Y.; Cho, E.K.; Shin, D.B.; Lee, J.H. Anemia as a useful biomarker in patients with diffuse large B-cell lymphoma treated with R-CHOP immunochemotherapy. Cancer Sci. 2014, 105, 1569–1575. [Google Scholar] [CrossRef]
  194. Matsumoto, K.; Fujisawa, S.; Ando, T.; Koyama, M.; Koyama, S.; Ishii, Y.; Numata, A.; Yamamoto, W.; Motohashi, K.; Hagihara, M.; et al. Anemia associated with worse outcome in diffuse large B-cell lymphoma patients: A single-center retrospective study. Turk. J. Haematol. 2018, 35, 181–184. [Google Scholar] [CrossRef] [PubMed]
  195. Wight, J.C.; Chong, G.; Grigg, A.P.; Hawkes, E.A. Prognostication of diffuse large B-cell lymphoma in the molecular era: Moving beyond the IPI. Blood Rev. 2018, 32, 400–415. [Google Scholar] [CrossRef] [PubMed]
  196. Mu, S.; Shi, D.; Ai, L.; Fan, F.; Peng, F.; Sun, C.; Hu, Y. International Prognostic Index-Based Immune Prognostic Model for Diffuse Large B-Cell Lymphoma. Front. Immunol. 2021, 12, 732006. [Google Scholar] [CrossRef] [PubMed]
  197. Yue, W.; Liu, B.; Gao, L.; He, M.; Wang, J.; Zhang, W.; Chen, L.; Hu, X.; Xu, L.; Yang, J. The pretreatment albumin to globulin ratio as a significant predictor in patients with diffuse large B cell lymphoma. Clin. Chim. Acta 2018, 485, 316–322. [Google Scholar] [CrossRef]
  198. Li, X.; Zhang, Y.; Zhao, W.; Liu, Z.; Shen, Y.; Li, J.; Shen, Z. The Glasgow Prognostic Score as a significant predictor of diffuse large B cell lymphoma treated with R-CHOP in China. Ann. Hematol. 2015, 94, 57–63. [Google Scholar] [CrossRef]
  199. Jung, J.; Lee, H.; Heo, J.Y.; Chang, M.H.; Lee, E.; Park, W.S.; Park, J.H.; Eom, H.S. High level of pre-treatment C-reactive protein to albumin ratio predicts inferior prognosis in diffuse large B-cell lymphoma. Sci. Rep. 2021, 11, 2674. [Google Scholar] [CrossRef]
  200. Shi, X.; Wu, C.; Deng, W.; Wu, J. Prognostic value of lactate dehydrogenase to absolute lymphocyte count ratio and albumin to fibrinogen ratio in diffuse large B-cell lymphoma. Medicine 2024, 103, e39097. [Google Scholar] [CrossRef]
  201. Melchardt, T.; Troppan, K.; Weiss, L.; Hufnagl, C.; Neureiter, D.; Tränkenschuh, W.; Hopfinger, G.; Magnes, T.; Deutsch, A.; Neumeister, P.; et al. A modified scoring of the NCCN-IPI is more accurate in the elderly and is improved by albumin and β2-microglobulin. Br. J. Haematol. 2015, 168, 239–245. [Google Scholar] [CrossRef]
  202. Gang, A.O.; Pedersen, M.; d’Amore, F.; Pedersen, L.M.; Jensen, B.A.; Jensen, P.; Møller, M.B.; Mourits-Andersen, H.T.; Pedersen, R.S.; Klausen, T.W.; et al. A clinically based prognostic index for diffuse large B-cell lymphoma with a cut-off at 70 years of age significantly improves prognostic stratification: Population-based analysis from the Danish Lymphoma Registry. Leuk. Lymphoma 2015, 56, 2556–2562. [Google Scholar] [CrossRef]
  203. Kobayashi, T.; Kuroda, J.; Yokota, I.; Tanba, K.; Fujino, T.; Kuwahara, S.; Isa, R.; Yamaguchi, J.; Kawata, E.; Akaogi, T.; et al. The Kyoto Prognostic Index for patients with diffuse large B-cell lymphoma in the rituximab era. Blood Cancer J. 2016, 6, e383. [Google Scholar] [CrossRef]
  204. Atas, U.; Sozel, H.; Iltar, U.; Yucel, O.K.; Salim, O.; Undar, L. The Prognostic Impact of Pretreatment Geriatric Nutritional Risk Index in Patients with Diffuse Large B-Cell Lymphoma. Nutr. Cancer 2023, 75, 591–598. [Google Scholar] [CrossRef]
  205. Ochi, Y.; Kazuma, Y.; Hiramoto, N.; Ono, Y.; Yoshioka, S.; Yonetani, N.; Matsushita, A.; Imai, Y.; Hashimoto, H.; Ishikawa, T. Utility of a simple prognostic stratification based on platelet counts and serum albumin levels in elderly patients with diffuse large B cell lymphoma. Ann. Hematol. 2017, 96, 1–8. [Google Scholar] [CrossRef]
  206. Nakayama, S.; Matsuda, M.; Adachi, T.; Sueda, S.; Ohashi, Y.; Awaji, S.; Hashimoto, S.; Matsumura, I. Novel prognostic index based on hemoglobin level and platelet count for diffuse large B-cell lymphoma, not otherwise specified in the R-CHOP era. Platelets 2019, 30, 637–645. [Google Scholar] [CrossRef]
  207. Chen, H.; Zhong, Q.; Zhou, Y.; Qin, Y.; Yang, J.; Liu, P.; He, X.; Zhou, S.; Zhang, C.; Gui, L.; et al. Enhancement of the International prognostic index with β2-microglobulin, platelet count and red blood cell distribution width: A new prognostic model for diffuse large B-cell lymphoma in the rituximab era. BMC Cancer 2022, 22, 583. [Google Scholar] [CrossRef] [PubMed]
  208. Chen, Y.; Neelapu, S.; Feng, L.; Bi, W.; Yang, T.H.; Wang, M.; Fanale, M.A.; Westin, J.R.; Hagemeister, F.B.; Fayad, L.E.; et al. Prognostic significance of baseline peripheral absolute neutrophil, monocyte and serum β2-microglobulin level in patients with diffuse large B-cell lymphoma: A new prognostic model. Br. J. Haematol. 2016, 175, 290–299. [Google Scholar] [CrossRef] [PubMed]
  209. Jelicic, J.; Juul-Jensen, K.; Bukumiric, Z.; Runason Simonsen, M.; Kragh Jørgensen, R.R.; Roost Clausen, M.; Ludvigsen Al-Mashhadi, A.; Schou Pedersen, R.; Bjørn Poulsen, C.; Ortved Gang, A.; et al. Validation of prognostic models in elderly patients with diffuse large B-cell lymphoma in a real-world nationwide population-based study—Development of a clinical nomogram. Ann. Hematol. 2025, 104, 433–444. [Google Scholar] [CrossRef] [PubMed]
  210. Tan, D.; Fu, Y.; Tong, W.; Li, F. Prognostic significance of lymphocyte to monocyte ratio in colorectal cancer: A meta-analysis. Int. J. Surg. 2018, 55, 128–138. [Google Scholar] [CrossRef]
  211. Cvetković, M.; Arsenović, I.; Smiljanić, M.; Sobas, M.; Bogdanović, A.; Leković, D. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio as novel prognostic biomarkers in BCR-ABL negative myeloproliferative neoplasms. Ann. Hematol. 2024, 103, 4545–4556. [Google Scholar] [CrossRef]
  212. Markovic, O.; Popovic, L.; Marisavljevic, D.; Jovanovic, D.; Filipovic, B.; Stanisavljevic, D.; Matovina-Brko, G.; Hajder, J.; Matkovic, T.; Živkovic, R.; et al. Comparison of prognostic impact of absolute lymphocyte count, absolute monocyte count, absolute lymphocyte count/absolute monocyte count prognostic score and ratio in patients with diffuse large B cell lymphoma. Eur. J. Intern. Med. 2014, 25, 296–302. [Google Scholar] [CrossRef]
  213. Zhao, P.; Zang, L.; Zhang, X.; Chen, Y.; Yue, Z.; Yang, H.; Zhao, H.; Yu, Y.; Wang, Y.; Zhao, Z.; et al. Novel prognostic scoring system for diffuse large B-cell lymphoma. Oncol. Lett. 2018, 15, 5325–5332. [Google Scholar] [CrossRef]
  214. Wang, J.; Zhou, M.; Xu, J.Y.; Yang, Y.G.; Zhang, Q.G.; Zhou, R.F.; Chen, B.; Ouyang, J. Prognostic role of pretreatment neutrophil-lymphocyte ratio in patients with diffuse large B-cell lymphoma treated with RCHOP. Medicine 2016, 95, e4893. [Google Scholar] [CrossRef] [PubMed]
  215. Chu, Y.; Liu, Y.; Jiang, Y.; Ge, X.; Yuan, D.; Ding, M.; Qu, H.; Liu, F.; Zhou, X.; Wang, X. Prognosis and complications of patients with primary gastrointestinal diffuse large B-cell lymphoma: Development and validation of the systemic inflammation response index-covered score. Cancer Med. 2023, 12, 9570–9582. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, Z.; Zhang, J.; Luo, S.; Zhao, X. Prognostic Significance of Systemic Immune-Inflammation Index in Patients With Diffuse Large B-Cell Lymphoma. Front. Oncol. 2021, 11, 655259. [Google Scholar] [CrossRef] [PubMed]
  217. Luan, C.; Wang, F.; Wei, N.; Chen, B. Prognostic nutritional index and the prognosis of diffuse large B-cell lymphoma: A meta-analysis. Cancer Cell Int. 2020, 20, 455. [Google Scholar] [CrossRef]
  218. Yan, C.; Xie, Y.; Hua, Y.; Li, S.; Fu, H.; Cheng, Z.; Wu, J. Prognostic value of geriatric nutritional risk index in patients with diffuse large B-cell lymphoma: A meta-analysis. Clin. Transl. Oncol. 2024, 26, 515–523. [Google Scholar] [CrossRef]
  219. Cao, D.; Zhang, Z. Prognostic and clinicopathological role of geriatric nutritional risk index in patients with diffuse large B-cell lymphoma: A meta-analysis. Front. Oncol. 2023, 13, 1169749. [Google Scholar] [CrossRef]
  220. Chen, Y.; Zhang, Z.; Fang, Q.; Jian, H. Prognostic impact of platelet-to-lymphocyte ratio on diffuse large B-cell lymphoma: A meta-analysis. Cancer Cell Int. 2019, 19, 245. [Google Scholar] [CrossRef]
  221. Sun, H.L.; Pan, Y.Q.; He, B.S.; Nie, Z.L.; Lin, K.; Peng, H.X.; Cho, W.C.; Wang, S.K. Prognostic performance of lymphocyte-to-monocyte ratio in diffuse large B-cell lymphoma: An updated meta-analysis of eleven reports. Onco Targets Ther. 2016, 9, 3017–3023. [Google Scholar] [CrossRef]
  222. Kim, D.Y.; Song, M.K.; Chung, J.S.; Shin, H.J.; Yang, D.H.; Lim, S.N.; Oh, S.Y. Clinical impacts of inflammatory markers and clinical factors in patients with relapsed or refractory diffuse large B-cell lymphoma. Blood Res. 2019, 54, 244–252. [Google Scholar] [CrossRef]
  223. Ide, D.; Fujino, T.; Kobayashi, T.; Egashira, A.; Miyashita, A.; Mizuhara, K.; Isa, R.; Tsukamoto, T.; Mizutani, S.; Uchiyama, H.; et al. Prognostic model for relapsed/refractory transplant-ineligible diffuse large B-cell lymphoma utilizing the lymphocyte-to-monocyte ratio. Int. J. Hematol. 2024, 119, 697–706. [Google Scholar] [CrossRef]
  224. Jelicic, J.; Juul-Jensen, K.; Bukumiric, Z.; Roost Clausen, M.; Ludvigsen Al-Mashhadi, A.; Pedersen, R.S.; Poulsen, C.B.; Brown, P.; El-Galaly, T.C.; Stauffer Larsen, T. Prognostic indices in diffuse large B-cell lymphoma: A population-based comparison and validation study of multiple models. Blood Cancer J. 2023, 13, 157. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Altered lipid metabolism and arachidonic acid (AA)-derived signaling pathways in diffuse large B-cell lymphoma (DLBCL). DLBCL is associated with decreased HDL and apoA1, impaired cholesterol efflux, and altered phospholipid composition, including reductions in PC, SM, LPC, and PE. This leads to accumulation of FFAs and decreased n-3 PUFAs. CD36-mediated uptake of oxLDL, LPS, and LCFAs activates MAPK, AKT, and STAT3 signaling, promoting glycolysis, proliferation, and EMT. Elevated intracellular AA, released by cPLA2, is metabolized via three key enzymatic routes: the COX pathway (producing PGs and TXs), the LOX pathway (yielding HETEs, LTs, and lipoxins), and the CYP450 pathway (producing EETs and HETEs). These pathways collectively contribute to inflammation, angiogenesis, and tumor progression. Explanation: ↑, increased; ↓, decreased. Abbreviations: HDL, high-density lipoprotein; LCAT, lecithin–cholesterol acyltransferase; apoA1, apolipoprotein A1; FFAs, free fatty acids; PUFAs, polyunsaturated fatty acids; oxLDL, oxidized low-density lipoprotein; LCFAs, long-chain fatty acids; LPS, lipopolysaccharide; CD36, cluster of differentiation 36; MAPK, mitogen-activated protein kinase; AKT, protein kinase B; STAT3, signal transducer and activator of transcription 3; EMT, epithelial–mesenchymal transition; LOX, lipoxygenase; 5-LOX, 5-lipoxygenase; 12,15-LOX, 12/15-lipoxygenase; 8,12,15-LOX, 8/12/15-lipoxygenase; 5-HPETE, 5-hydroperoxyeicosatetraenoic acid; 5-HETE, 5-hydroxyeicosatetraenoic acid; 8,12,15-HETE, 8,12,15-hydroxyeicosatetraenoic acid; 5-oxo ETE; 5-oxo-eicosatetraenoic acid; LTA-4, leukotriene A4; LTB-4, leukotriene B4; LTC-4, leukotriene C4; LTD-4, leukotriene D4; LTE-4, leukotriene E4; LXA-4, lipoxin A4; LXB-4, lipoxin B4; GPX-1, glutathione peroxidase 1; COX-1, cyclooxygenase 1; COX-2, cyclooxygenase 2; cPLA2, cytosolic phospholipase A2; PGH-2, prostaglandin H2; PGD-2, prostaglandin D2; PGE-2, prostaglandin E2; PGF-2, prostaglandin F2; PGI-2, prostaglandin I2; TXA-2, thromboxane A2; TXB-2, thromboxane B2; CYP-450, cytochrome P450; EETs, epoxyeicosatrienoic acids; DHETs, dihydroxyeicosatrienoic acids; 19,20-HETE, 19,20-hydroxyeicosatetraenoic acid.
Figure 1. Altered lipid metabolism and arachidonic acid (AA)-derived signaling pathways in diffuse large B-cell lymphoma (DLBCL). DLBCL is associated with decreased HDL and apoA1, impaired cholesterol efflux, and altered phospholipid composition, including reductions in PC, SM, LPC, and PE. This leads to accumulation of FFAs and decreased n-3 PUFAs. CD36-mediated uptake of oxLDL, LPS, and LCFAs activates MAPK, AKT, and STAT3 signaling, promoting glycolysis, proliferation, and EMT. Elevated intracellular AA, released by cPLA2, is metabolized via three key enzymatic routes: the COX pathway (producing PGs and TXs), the LOX pathway (yielding HETEs, LTs, and lipoxins), and the CYP450 pathway (producing EETs and HETEs). These pathways collectively contribute to inflammation, angiogenesis, and tumor progression. Explanation: ↑, increased; ↓, decreased. Abbreviations: HDL, high-density lipoprotein; LCAT, lecithin–cholesterol acyltransferase; apoA1, apolipoprotein A1; FFAs, free fatty acids; PUFAs, polyunsaturated fatty acids; oxLDL, oxidized low-density lipoprotein; LCFAs, long-chain fatty acids; LPS, lipopolysaccharide; CD36, cluster of differentiation 36; MAPK, mitogen-activated protein kinase; AKT, protein kinase B; STAT3, signal transducer and activator of transcription 3; EMT, epithelial–mesenchymal transition; LOX, lipoxygenase; 5-LOX, 5-lipoxygenase; 12,15-LOX, 12/15-lipoxygenase; 8,12,15-LOX, 8/12/15-lipoxygenase; 5-HPETE, 5-hydroperoxyeicosatetraenoic acid; 5-HETE, 5-hydroxyeicosatetraenoic acid; 8,12,15-HETE, 8,12,15-hydroxyeicosatetraenoic acid; 5-oxo ETE; 5-oxo-eicosatetraenoic acid; LTA-4, leukotriene A4; LTB-4, leukotriene B4; LTC-4, leukotriene C4; LTD-4, leukotriene D4; LTE-4, leukotriene E4; LXA-4, lipoxin A4; LXB-4, lipoxin B4; GPX-1, glutathione peroxidase 1; COX-1, cyclooxygenase 1; COX-2, cyclooxygenase 2; cPLA2, cytosolic phospholipase A2; PGH-2, prostaglandin H2; PGD-2, prostaglandin D2; PGE-2, prostaglandin E2; PGF-2, prostaglandin F2; PGI-2, prostaglandin I2; TXA-2, thromboxane A2; TXB-2, thromboxane B2; CYP-450, cytochrome P450; EETs, epoxyeicosatrienoic acids; DHETs, dihydroxyeicosatrienoic acids; 19,20-HETE, 19,20-hydroxyeicosatetraenoic acid.
Ijms 26 05670 g001
Figure 2. IL-6 cytokine family signaling in the tumor microenvironment (TME). Various components of the TME, including dendritic cells, T cells, macrophages, endothelial cells, tumor-associated neutrophils (TANs), fibroblasts, and tumor cells, secrete members of the IL-6 cytokine family, such as IL-6, IL-11, IL-27, oncostatin M (OSM), leukemia inhibitory factor (LIF), IL-31, and ciliary neurotrophic factor (CNTF). These cytokines signal through specific receptor complexes that typically include glycoprotein 130 (gp130) in combination with ligand-specific receptor subunits. Ligand binding triggers the activation of canonical intracellular signaling pathways, which collectively mediate immunosuppressive functions within the TME and contribute to tumor cell survival, proliferation, and immune evasion. Explanation: ↑, increased; ↓, decreased. Abbreviations: IL-6, interleukin-6; IL-11, interleukin-11; IL-27, interleukin-27; IL-31, interleukin-31; CNTF, ciliary neurotrophic factor; OSM, oncostatin M; LIF, leukemia inhibitory factor; IL-6R, interleukin-6 receptor; IL-11R, interleukin-11 receptor; IL-27R, interleukin-27 receptor; IL-31R, interleukin-31 receptor; LIFR, leukemia inhibitory factor receptor; OSMR, oncostatin M receptor; gp130, glycoprotein 130; JAK, Janus kinase; STAT, signal transducer and activator of transcription; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; mTORC1, mammalian target of rapamycin complex 1; p70S6K, p70 ribosomal protein S6 kinase; Treg, regulatory T cell; Th, T helper cell; Tc, cytotoxic T cell; TAM, tumor-associated macrophage; TAN, tumor-associated neutrophil; MDSC, myeloid-derived suppressor cell; CAF, cancer-associated fibroblast; TME, tumor microenvironment; EMT, epithelial-to-mesenchymal transition.
Figure 2. IL-6 cytokine family signaling in the tumor microenvironment (TME). Various components of the TME, including dendritic cells, T cells, macrophages, endothelial cells, tumor-associated neutrophils (TANs), fibroblasts, and tumor cells, secrete members of the IL-6 cytokine family, such as IL-6, IL-11, IL-27, oncostatin M (OSM), leukemia inhibitory factor (LIF), IL-31, and ciliary neurotrophic factor (CNTF). These cytokines signal through specific receptor complexes that typically include glycoprotein 130 (gp130) in combination with ligand-specific receptor subunits. Ligand binding triggers the activation of canonical intracellular signaling pathways, which collectively mediate immunosuppressive functions within the TME and contribute to tumor cell survival, proliferation, and immune evasion. Explanation: ↑, increased; ↓, decreased. Abbreviations: IL-6, interleukin-6; IL-11, interleukin-11; IL-27, interleukin-27; IL-31, interleukin-31; CNTF, ciliary neurotrophic factor; OSM, oncostatin M; LIF, leukemia inhibitory factor; IL-6R, interleukin-6 receptor; IL-11R, interleukin-11 receptor; IL-27R, interleukin-27 receptor; IL-31R, interleukin-31 receptor; LIFR, leukemia inhibitory factor receptor; OSMR, oncostatin M receptor; gp130, glycoprotein 130; JAK, Janus kinase; STAT, signal transducer and activator of transcription; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; mTORC1, mammalian target of rapamycin complex 1; p70S6K, p70 ribosomal protein S6 kinase; Treg, regulatory T cell; Th, T helper cell; Tc, cytotoxic T cell; TAM, tumor-associated macrophage; TAN, tumor-associated neutrophil; MDSC, myeloid-derived suppressor cell; CAF, cancer-associated fibroblast; TME, tumor microenvironment; EMT, epithelial-to-mesenchymal transition.
Ijms 26 05670 g002
Figure 3. Tumor microenvironment (TME) in diffuse large B-cell lymphoma (DLBCL). The tumor microenvironment in DLBCL is characterized by immunosuppressive signaling, enhanced angiogenesis, and impaired anti-tumor immunity. DLBCL cells express programmed cell death protein PD 1 promoting T cell exhaustion through PD-1/PD-L1 interaction. Regulatory T cells and M2 macrophages secrete IL-10, TGF-β, and VEGF, which inhibit CD4+, CD8+, and NK cells and stimulate neovascularization. TANs promote tumor invasion by releasing ROS, proteolytic enzymes, and NETs. M2 macrophages further support tumor progression, with their polarization induced by IL-4 and IL-13. Dendritic cell function is suppressed, leading to reduced T cell activation. Additionally, BCR signaling in B cells is dysregulated through activation of the NF-κB and phosphoinositide 3-PI3K/AKT pathways, contributing to immune evasion and lymphoma cell survival. Explanation: ↑, increased; ↓, decreased. Abbreviations: Abbreviations: TME, tumor microenvironment; CD36, cluster of differentiation 36; CD4, cluster of differentiation 4; CD8, cluster of differentiation 8; CD16, cluster of differentiation 16; CD32, cluster of differentiation 32; CD64, cluster of differentiation 64; CD80, cluster of differentiation 80; CD86, cluster of differentiation 86; CD163, cluster of differentiation 163; CD200R, cluster of differentiation 200 receptor; CD206, cluster of differentiation 206; CD209, cluster of differentiation 209; CD301, cluster of differentiation 301; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; TIM-3, T-cell immunoglobulin and mucin-domain containing molecule 3; CTLA-4, cytotoxic T lymphocyte-associated protein 4; Treg, regulatory T cell; CD11c⁺DC, CD11c-positive dendritic cell; NK cell, natural killer cell; NETs, neutrophil extracellular traps; TAN, tumor-associated neutrophil; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor; TGF-β, transforming growth factor beta; IL, interleukin; TNF-α, tumor necrosis factor alpha; CXCL12, C-X-C motif chemokine ligand 12; PI3K/AKT, Phosphoinositide 3-kinase/protein kinase B; NF-κB, Nuclear Factor kappa B; STAT3, signal transducer and activator of transcription 3; STAT6, signal transducer and activator of transcription 6; PPARγ, Peroxisome Proliferator-Activated Receptor Gamma; M-CSF, macrophage colony-stimulating factor.
Figure 3. Tumor microenvironment (TME) in diffuse large B-cell lymphoma (DLBCL). The tumor microenvironment in DLBCL is characterized by immunosuppressive signaling, enhanced angiogenesis, and impaired anti-tumor immunity. DLBCL cells express programmed cell death protein PD 1 promoting T cell exhaustion through PD-1/PD-L1 interaction. Regulatory T cells and M2 macrophages secrete IL-10, TGF-β, and VEGF, which inhibit CD4+, CD8+, and NK cells and stimulate neovascularization. TANs promote tumor invasion by releasing ROS, proteolytic enzymes, and NETs. M2 macrophages further support tumor progression, with their polarization induced by IL-4 and IL-13. Dendritic cell function is suppressed, leading to reduced T cell activation. Additionally, BCR signaling in B cells is dysregulated through activation of the NF-κB and phosphoinositide 3-PI3K/AKT pathways, contributing to immune evasion and lymphoma cell survival. Explanation: ↑, increased; ↓, decreased. Abbreviations: Abbreviations: TME, tumor microenvironment; CD36, cluster of differentiation 36; CD4, cluster of differentiation 4; CD8, cluster of differentiation 8; CD16, cluster of differentiation 16; CD32, cluster of differentiation 32; CD64, cluster of differentiation 64; CD80, cluster of differentiation 80; CD86, cluster of differentiation 86; CD163, cluster of differentiation 163; CD200R, cluster of differentiation 200 receptor; CD206, cluster of differentiation 206; CD209, cluster of differentiation 209; CD301, cluster of differentiation 301; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; TIM-3, T-cell immunoglobulin and mucin-domain containing molecule 3; CTLA-4, cytotoxic T lymphocyte-associated protein 4; Treg, regulatory T cell; CD11c⁺DC, CD11c-positive dendritic cell; NK cell, natural killer cell; NETs, neutrophil extracellular traps; TAN, tumor-associated neutrophil; ROS, reactive oxygen species; VEGF, vascular endothelial growth factor; TGF-β, transforming growth factor beta; IL, interleukin; TNF-α, tumor necrosis factor alpha; CXCL12, C-X-C motif chemokine ligand 12; PI3K/AKT, Phosphoinositide 3-kinase/protein kinase B; NF-κB, Nuclear Factor kappa B; STAT3, signal transducer and activator of transcription 3; STAT6, signal transducer and activator of transcription 6; PPARγ, Peroxisome Proliferator-Activated Receptor Gamma; M-CSF, macrophage colony-stimulating factor.
Ijms 26 05670 g003
Table 1. Prognostic indices in DLBCL incorporating lipid- and protein-derived inflammatory markers.
Table 1. Prognostic indices in DLBCL incorporating lipid- and protein-derived inflammatory markers.
Index
Author, Year
Parameters IncludedNCut Off/Prognostic GroupsKey Findings
Matsumoto
Model

Matsumoto et al., 2018 [194]
CS ≥ III
Hb < 100 g/L
185Scores 0, 1, 23-year PFS—score 0: 89.1%, score 1: 73.9%, score 2: 35.5%, (p < 0.001)
3-year OS—score 0: 94.6%, score 1: 82.0%, score 2: 61.4%, (p < 0.001)
Lipo-PI

Gao et al., 2018 [30]
Concurrently:
HDL-C
<1.03 mmol/L
LDL-C
<2.60 mmol/L

and NCCN IPI variables *
550
(T: 367,
V: 183)
L: score 0–2
LI: score 3–4
HI: score 5–6
H: ≥7
5-year PFS—L: 88.6%,
LI: 65%, HI: 29.5%, H: 10%
5-year OS—L: 96.9%,
LI: 79.6%, HI: 45%, H: 22.5%
Lipo-PI widened the definition of high-risk patients for 5-year OS and improved. the risk stratification of NCCN-IPI.
HDL-C or LDL-C elevations after treatment correlated with better survival
IPI-A

Yu et al., 2023 [28]
ApoA-I < 0.81 g/L

and IPI variables **:
105Comparison of IPI-A with IPI using a ROC curvePFS (1 year: AUC = 0.745,
3 years: AUC = 0.827,
5 years: AUC = 0.763)
A significant increase in TG, LDL-C, HDL-C, ApoA-I, and ApoB levels after
Chemotherapy
AGR

Yue et al., 2018 [197]
Albumin-to-globulin3351.3Low AGR is an independent adverse predictor for OS (HR = 0.613; 95% CI = 0.412–0.910, p = 0.015)
AGR distinguished patients with different prognosis in stage III–IV and the ABC DLBCL groups
GPS
(Glasgow
Prognostic score)

Li et al., 2015 [198]
CRP > 10 mg/L
Albumin < 35 g/L
160GPS-0
GPS-1
GPS-2
Lower GPS predict better outcome, including PFS (p < 0.001) and OS (p < 0.001)
CAR

Jung et al., 2021 [199]
CRP-to-albumin ratio1860.158Low CAR is an independent adverse predictor for
CRR (64.4% vs. 92.6%),
3-year PFS (53.5% vs. 88.0%). 3-year OS (68.3% vs. 96.2%)
AFR

Shi et al., 2024 [200]
Albumin-to-fibrinogen7412.642-year PFS: high AFR-85.7%, low AFR-22.2%
AFR can be used as good supplements for IPI to predict the prognosis of DLBCL
Modified
NCCN-IPI

Melchardt et al., 2015 [201]
Albumin < 35 g/L
β2M > 3.0 mg/L
NCCN-IPI variables *
499L: score 0–2
LI: score 3
HI: score 4–7
H: score 8–10
3-year OS—L: 97.8%,
LI: 82.7%, HI: 65.9%, H: 4.2%

5-year OS—L:93%, LI:78%,
HI: 55.7%, H: 36.8%
DLBCL-PI
(Modified DLBCL prognostic index)

aaDLBCL-PI (for age ≤70 years)

Gang et al., 2015 [202]
Albumin < 35 g/L
and IPI variables ** (except EN)

Albumin <35 g/L and IPI variables ** (except CS)
1990L: score0–1
LI: score 2
HI: score 3
H: score 4–5
5-year OS in four risk groups—IPI: 83%, 64%, 57%, and 38% DLBCL-PI: 87%, 69%, 53%, and 37%
aaDLBCL-PI: 92%, 84%, 74%, and 47%.
KPI
(Kyoto Prognostic Index)

Kobayashi et al., 2016 [203]
LDH ratio
>1–3/>3,
ECOG-PS ≥ 2,
Albumin < 35 g/L
EN (BM, bone, skin and/or lung/pleura)
323L: score 0,
LI: score 1–2, HI: score 3,
H: score 4–5
3-year OS—L: 96.4%,
LI: 84.7%, HI: 63.8%, H: 33.3%

3-year PFS—L: 84.4%,
LI: 70.2%, HI: 53.4%, H: 24.1%
GNRI
(Geriatric nutritional index)

Atas et al., 2023 [204]
Lorentz formula ***206L and H groups according to the ROC curve
(cut off 104.238)
Low GNRI is an independent adverse predictor DFS and OS
Abbreviations: HDL-C, high density cholesterol; LDL-C, low density cholesterol, TG, triglycerides, Apo, apolipoprotein; CRP, C-reactive protein; LDH, lactate dehydrogenase, EN, extranodal sites; IPI, International Prognostic Score; CS, Ann Arbor Clinical Stage; ECOG-PS, Eastern Cooperative Oncology Group Performance Scale; T, training cohort; V, validation cohort; L, low risk; LI, low–intermediate risk; HI, high–intermediate risk; H, high risk; ROC, receiver operating characteristic; AUC, area under the curve; CRR, complete remission rate; PFS, progression free survival, DFS, disease free survival; OS, overall survival. * NCCN-IPI variables: Age 1: ≤40–1, 2: 41–60, 3: 61–75; 4: >75, CS III/IV, ECOG PS > 1, EN, LDH 1: 1–3UNL, 2: >3UNL; ** IPI variables: Age > 60 years, CS III/IV, ECOG PS > 1, EN > 1, LDH > ULN; *** Lorenz formula [1.489 × serum albumin (g/L)] + [41.7 × (current body weight/ideal body weight)].
Table 2. Prognostic indices in newly diagnosed DLBCL incorporating peripheral blood cells.
Table 2. Prognostic indices in newly diagnosed DLBCL incorporating peripheral blood cells.
Index
Author, Year
Parameters IncludedNCut Off/Prognostic GroupsKey Findings
LAR

Shi et al., 2024 [200]
LDH to ALC ratio74244.952-year PFS—low LAR: 78.6%, high LAR:13.8%
LAR can be used as good supplements for IPI to predict the prognosis of DLBCL.
PNI
(Prognostic nutritional index)

Zhou et al., 2016) [117]
albumin (g/L) +
5 × ALC × 109/L
253L and H groups
according to the ROC curve
Low PNI was an independent adverse predictor EFS and OS
PA
(Platelet–albumin)

Ochi et al., 2017 [205]
platelet count
<100 × 109/L,
albumin ≥ 35 g/L
391L/I/H5-year OS—L: 77.6%,
I: 47.9%, H: 19.0%
HP
(Hb–platelet)

Nakayama et al., 2019 [206]
Hb < 120 g/L
platelet counts
<135 × 109/L
89L: score 0
I: score 1
H: score 2
Higher HP index was associated with worse OS and predicted OS rate
independently of the IPI
Chen et al., 2022 [207]β2M UNL
platelets
<157 × 109/L
RDW ≥ 14.5%
998
(T:701,
V: 297)
L: score 0
LI: score 1
HI: score 2–3
H: score 4
VH: score ≥ 5
The AUC of the new model for OS prediction at specific time points (6 months to 10 years) was consistently higher than that of conventional prognostic models in both T and V
Chen et al., 2016 [208]ANC
AMC
ECOG PS
β2M
EN
817L1: score 0
L2: score 1
I: score 2
H1: score 3–5
H2: score 6–7
Better stratifies patients into various risk categories than the IPI for newly diagnosed DLBCL.
E-IPI
(Elderly-IPI)

Jelicic et al., 2025 [209]
NCCN-IPI variables *
(excluding EN),
albumin
platelet
2835L/LI/HI/H groups3-year OS—L: 88.6%,
H: 45%
5-year OS—L: 80.6%,
H: 38.3%
Δ c-index OS: 3–0.024
(0.021; 0.026)
LMR

Markovic et al., 2014. [212]
Lymphocyte-to-monocyte ratio ≤ 2.8222L/HL group had significantly shorter EFS and OS
PLR

Zhao et al., 2018 [213]
Platelet-to-lymphocyte ratio
≥ 170
309L/HPLR was a significant prognostic factor for OS (p < 0.001) and PFS (p = 0.003)
NLR

Wang et al., 2016 [214]
Neutrophil-to-lymphocyte ratio1563.0
L and H
groups according to the ROC curve
5-year OS—L: −82.5%,
H: 57.5%
5-year PFS—L: 64.5%,
H: 30.0%
SIRI-PI
(Systemic inflammation response index)

Chu et al., 2023 [215]
Neutrophil ×
monocyte/
lymphocyte
153
PGl-DLBCL
(T: 102,
V: 51)
1.34
(L/H)
more precise high-risk assessment compared to NCCN-IPI with a higher AUC (0.916 vs. 0.835) and C-index (0.912 vs. 0.836) for OS
SII
(Systemic immune–inflammation index)

Wang et al., 2021 [216]
Neutrophil,
Platelet, and
lymphocyte counts
2241046.1
L and H
groups according to the ROC curve
3-year OS—L: 88.3%,
H: 58.6%
3-year PFS—L: 74.4%,
H: 3.7%
SII superior to NLR and PLR
Zhao et al., 2018 [213]PLR < 170 vs. ≥170
IPI < 2 vs. ≥2 or
aaIPI 0 vs. ≥1
β2M normal vs. UNL
309L/I/H5-year OS—L: 86.4%,
I: 54.1% H: 21.1%
Abbreviations: LDH, lactate dehydrogenase, ALC, absolute lymphocyte count; Hb, hemoglobin β2M, beta-2 microglobulin; RDW, red blood cell distribution width; ANC; absolute neutrophil count; AMC, absolute monocyte count; ECOG PS, Eastern Cooperative Oncology Group Performance Scale; EN, extranodal sites; PLR, platelet-to-lymphocyte ratio; IPI, International prognostic index, PGl, primary gastrointestinal: UNL, upper normal limit, T, training cohort; V, validation cohort; L, low risk; LI, low–intermediate risk; HI, high–intermediate risk; H, high risk, VH, very high risk; OS, overall survival; EFS, event free survival; * NCCN-IPI variables: Age 1: ≤40–1, 2: 41–60, 3: 61–75; 4: >75, CS III/IV, ECOG PS > 1, EN, LDH 1: 1–3UNL, 2: >3UNL.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cvetković, Z.; Marković, O.; Marinković, G.; Pejić, S.; Vučić, V. Tumor Microenvironment, Inflammation, and Inflammatory Prognostic Indices in Diffuse Large B-Cell Lymphomas: A Narrative Review. Int. J. Mol. Sci. 2025, 26, 5670. https://doi.org/10.3390/ijms26125670

AMA Style

Cvetković Z, Marković O, Marinković G, Pejić S, Vučić V. Tumor Microenvironment, Inflammation, and Inflammatory Prognostic Indices in Diffuse Large B-Cell Lymphomas: A Narrative Review. International Journal of Molecular Sciences. 2025; 26(12):5670. https://doi.org/10.3390/ijms26125670

Chicago/Turabian Style

Cvetković, Zorica, Olivera Marković, Gligorije Marinković, Snežana Pejić, and Vesna Vučić. 2025. "Tumor Microenvironment, Inflammation, and Inflammatory Prognostic Indices in Diffuse Large B-Cell Lymphomas: A Narrative Review" International Journal of Molecular Sciences 26, no. 12: 5670. https://doi.org/10.3390/ijms26125670

APA Style

Cvetković, Z., Marković, O., Marinković, G., Pejić, S., & Vučić, V. (2025). Tumor Microenvironment, Inflammation, and Inflammatory Prognostic Indices in Diffuse Large B-Cell Lymphomas: A Narrative Review. International Journal of Molecular Sciences, 26(12), 5670. https://doi.org/10.3390/ijms26125670

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