Next Article in Journal
Anionic Lipid Catalyzes the Generation of Cytotoxic Insulin Oligomers
Previous Article in Journal
The Unconventional Role of ABHD17A in Increasing the S-Palmitoylation and Antiviral Activity of IFITM1 by Downregulating ABHD16A
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Circulating Biomarker Panorama in HIV-Associated Lymphoma: A Bridge from Early Risk Warning to Prognostic Stratification

1
School of Medicine, Chongqing University, Chongqing 400030, China
2
Chongqing Key Laboratory for the Mechanism and Intervention of Cancer Metastasis, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing 400030, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2025, 15(7), 993; https://doi.org/10.3390/biom15070993
Submission received: 6 May 2025 / Revised: 26 June 2025 / Accepted: 10 July 2025 / Published: 11 July 2025
(This article belongs to the Section Molecular Biomarkers)

Abstract

HIV-associated lymphoma (HAL) is a heterogeneous and highly aggressive group of malignancies. Although antiretroviral therapy (ART) has significantly prolonged the survival of people living with HIV (PLWH), the risk of malignancy secondary to HIV infection remains higher than in HIV-negative individuals, with HAL being among the most frequent. The pathogenesis of HAL is complex, involving multifactorial interactions. In current clinical practice, HAL faces a double challenge: the lack of effective biological risk warning systems and the lack of precise prognostic stratification tools. In recent years, the construction of multidimensional biomarker systems has shown critical value in the comprehensive management of HAL. This review aims to systematically summarize recent advances in circulating biomarkers for HAL, focusing on the potential applications of immune environment indicators, such as inflammatory cytokine profiles and microbial translocation markers, as well as serum protein profiles, lymphocyte subsets, extracellular vesicles (EVs), circulating microRNAs (miRNAs), and viral biomarkers. These biomarkers offer promising avenues for early risk prediction, therapeutic monitoring, and prognostic evaluation. Developing an assessment system based on multidimensional biomarkers will optimize early risk stratification, enable precise prognostic classification, and support personalized therapeutic strategies, thereby providing a novel theoretical basis and practical direction for the clinical management of HAL.

1. Introduction

The disease burden of human immunodeficiency virus (HIV) infection and its oncologic complications remains a major global public health challenge. According to the latest epidemiologic data from the Joint United Nations Programme on HIV/AIDS (UNAIDS), as of 2023, an estimated 39.9 million adults worldwide were living with HIV, and approximately 630,000 deaths were attributed to AIDS-related illnesses [1]. Although the widespread application of antiretroviral therapy (ART) has substantially extended the survival of people living with HIV (PLWH), the incidence of malignancies, particularly AIDS-defining cancers (ADCs) [2,3], continues to rise and has become one of the leading causes of death among PLWH [4,5,6]. Given the highly aggressive nature of HAL, recent research has increasingly focused on building early risk warning systems. Studies have shown that before HAL is clinically diagnosed, PLWH often exhibit characteristic biological alterations in the blood, such as elevated levels of B-cell stimulatory factors (e.g., cytokines and chemokines). This immune activation profile is closely associated with an increased risk of lymphoma development, suggesting that these biomarkers could serve as potential indicators for the early detection of HAL [7,8,9]. These findings also offer a potential window for targeted intervention during the subclinical phase of the disease.
Moreover, due to the considerable variability in HAL prognosis, accurate prognostic evaluation is equally critical [10]. Although prognostic models such as the International Prognostic Index (IPI) and the age-adjusted International Prognostic Index (aaIPI) are widely used in clinical practice [11], they insufficiently account for HIV-specific factors and do not incorporate HIV-related biomarkers, limiting their ability to precisely stratify high-risk patients with poor outcomes [12,13,14]. Thus, there is an urgent need to develop next-generation prognostic models that integrate clinical features with molecular and immunological biomarkers to guide individualized treatment decisions. Against this background, identifying novel biomarkers for early risk prediction, therapeutic monitoring, and prognostic evaluation has become a major focus for optimizing HAL management. Circulating abnormalities in inflammatory mediators, immune activation factors, and microbial translocation-related molecules are now recognized as key features associated with HAL development and poor outcomes.
This review systematically summarizes recent advances in circulating biomarkers in HAL, covering inflammatory molecules, microbial translocation markers, lymphocyte subsets, extracellular vesicles, miRNAs, immunoglobulin (Ig), and viral-related markers, and explores their translational potential for risk stratification, therapeutic monitoring, and prognostic assessment, providing theoretical support for the development of precision intervention strategies aimed at improving outcomes in PLWH.

2. Epidemiological Overview

Non-Hodgkin’s lymphoma (NHL) is one of the most common types of ADCs [15]. It includes diffuse large B-cell lymphoma (DLBCL), Burkitt lymphoma (BL), primary plasmoblastic lymphoma (PBL), and primary effusion lymphoma (PEL), according to the latest World Health Organization (WHO) classification [16]. Epidemiological data indicate that over 90% of HAL cases are classified as NHL [17]. Historical data from the United States show that, before the introduction of ART, the risk of developing NHL among PLWH was 113 times higher compared to HIV-negative individuals [18]. This phenomenon likely contributed to the significant increase in overall NHL cases observed in the United States during the 1980s [19]. Recent meta-analyses have demonstrated that even in the ART era, PLWH continue to have an approximately 23-fold higher risk of developing NHL compared to the general population [20]. As the most common fatal cancer among PLWH, HIV-associated NHL (HIV-NHL) accounts for more than one-third of all HIV-related cancer deaths [4,21,22], a trend confirmed by multiple large cohort studies [23,24,25]. The prognosis for HIV-NHL remains poor. Studies have reported that approximately 34% of patients with HIV-NHL die within the first year after diagnosis [26], with a two-year overall survival rate of around 58% [27] and a five-year survival rate of only 55% [26]. These data underscore the ongoing challenges in the clinical management of HAL and highlight the urgent need for improved early detection, risk stratification, and treatment strategies. Comparatively, Hodgkin lymphoma (HL), as a non-AIDS-defining cancers (NADCs), has a substantially lower overall incidence than NHL. However, studies have shown that PLWH remain at a significantly higher risk of developing HL compared to the general immunocompetent population, with marked variations in incidence observed across different countries and between sexes [28]. In recent years, advancements in treatment have led to a continuous improvement in 5-year survival rates among HL patients [29,30], though outcomes remain suboptimal in resource-limited settings [31]. Given that this review focuses primarily on NHL, the discussion of HL is limited to a brief overview.

3. Pathogenesis

The high morbidity and mortality of HAL are closely linked to its unique pathological mechanisms, which involves multifactorial interactions that have not yet been fully elucidated [32,33]. Current mainstream views suggest that the following mechanisms are involved: (a) HIV infection leads to CD4+ T-cell depletion, resulting in severe immunodeficiency [34]; (b) genomic instability and genetic aberrations, which predispose people to malignant transformation [35]; (c) the direct oncogenic effects of HIV-encoded viral proteins, such as p17, gp120, transactivator of transcription (Tat), and negative factor (Nef) [36]; (d) the synergistic oncogenic effects of opportunistic infections, notably Epstein–Barr virus (EBV) and Kaposi’s sarcoma-associated herpesvirus (KSHV, also known as human herpesvirus 8) [37,38]. In addition, chronic antigenic stimulation and aberrant B-cell activation associated with HIV infection, along with persistent overproduction of cytokines, also play significant roles in the pathogenesis of HAL [39] (Figure 1). Our recent studies have revealed that the highly aggressive nature of HAL may be attributed to malignant B cells’ reliance on oxidative phosphorylation for energy metabolism and to pronounced tumor immune escape mechanisms. Key features include the loss of major histocompatibility complex class I (MHC-I) expression, T-cell exhaustion-like functional impairment, and macrophage-mediated immunosuppressive microenvironment [40].

4. Plasma Metabolite Changes

Due to its accessibility and minimally invasive nature, blood is widely used for biomarker screening and validation in precision oncology medicine [41]. The development of HAL is closely associated with alterations in various plasma metabolites. In recent years, studies have demonstrated that serum cytokines, chemokines, soluble receptors, and serum proteins play crucial roles in the early stages of disease progression, risk prediction, and prognostic evaluation. Moreover, emerging molecular biomarkers such as circulating miRNAs, extracellular vesicles (EVs), and clonally rearranged Ig DNA have further expanded the research landscape of plasma metabolite changes. Immunological dysfunction, persistent HIV viremia, and sustained expression of HIV-encoded viral proteins together contribute to altered plasma metabolic profiles, thereby promoting tumor microenvironment (TME) remodeling and immune evasion. Figure 2 categorizes and presents six classes of circulating biomarkers associated with the occurrence, progression and prognosis of HAL.

5. Immunoinflammation-Related Biomarkers

5.1. Cytokines

Cytokines play important roles in regulating immune responses, development, metabolism and tumorigenesis. Interleukin (IL)-6, IL-10, and tumor necrosis factor-alpha (TNF-α), as immunomodulatory cytokines widely distributed in the human body, are involved in the acute-phase response of the organism and anti-infective immunity by affecting antibody production and cell proliferation and differentiation [42,43,44,45]. Abnormal expression of these cytokines has been closely associated with malignant lymphoid neoplasms, promoting B-cell proliferation, genomic instability, and lymphoma initiation and progression, while also serving as indicators of poor prognosis [46,47,48,49].
In PLWH, genetic variation in IL-10 and its specific genotypes has potential value in HIV-NHL risk identification. Studies have shown significantly increased IL-10 expression and a higher frequency of the 592C/C genotype in the promoter region among HIV-NHL patients [50]. Further genetic analyses have revealed subtype-specific associations of IL-10 single nucleotide polymorphisms (SNPs), where the rs1800896_G allele correlates with increased systemic HIV-NHL risk, while the rs1800871_T and rs1800872_A alleles are associated with a reduced risk of primary central nervous system lymphoma (PCNSL) [51]. These findings suggest that genetic variations in IL-10 regulatory regions influence expression levels, and high-expressing IL-10 genotypes could serve as genetic risk markers for individualized risk stratification based on anatomical lymphoma subtypes. In addition, multicenter cohort studies have also shown a time-dependent increase in the levels of cytokines such as IL-6 and IL-10 before NHL diagnosis in PLWH, independently of CD4+ T-cell counts [52]. Combined testing for other cytokines such as IL-1α, IL-4, IL-5, IL-13, and granulocyte-macrophage colony-stimulating factor (GM-CSF) can also help identify people at high risk for HAL [9]. In animal experiments, HIV-1 transgenic Tg26 mice, a model for studying HIV-associated diseases [53], exhibit progressive elevation of multiple cytokines (e.g., IL-1β, IL-6, IL-10, TNF-α, IL-12p40, and IL-13, and GM-CSF in plasma, closely associated with B-cell lymphoma development [54]. This is consistent with the trend in HAL patients. Some studies have further confirmed that sustained elevations of cytokines including IL-6, IL-10, interferon gamma-induced protein 10 (IP-10)/CXCL10, TNF-α, and neopterin 1–5 years before HIV-NHL diagnosis are positively correlated with disease risk [55,56]. Moreover, combined biomarker models incorporating IL-11, IL-18, IL-29, and CXCL11 have shown superior predictive performance compared to single markers [7,57], highlighting the necessity of multiplex cytokine profiling. These cytokines are also markedly elevated in HIV-associated HL (HIV-HL) [58], suggesting their pivotal roles in HAL pathogenesis and their potential utility for early prediction and diagnosis (Table 1).
In terms of prognosis, high levels of IL-6, IL-10, and B-cell-activating factor of the TNF family/B lymphocyte Stimulator (BAFF/BLyS) have been associated with poorer overall survival (OS) and progression-free survival (PFS) in HIV-NHL patients [59,60]. However, the independent prognostic value of individual cytokines remains somewhat inconsistent across studies. After R+EPOCH therapy (rituximab plus etoposide, vincristine, doxorubicin, cyclophosphamide, and prednisone), although IL-6 and IL-10 levels showed no significant overall change, elevated IL-6 was significantly associated with shortened OS, and high IL-10 with reduced PFS [59]. In HIV-associated DLBCL (HIV-DLBCL) patients, elevated IL-10 and transforming growth factor-β (TGF-β) levels were independently associated with poor OS and PFS, regardless of IPI scores, while IL-6, despite being highly expressed, did not correlate with prognosis [61]. These discrepancies may stem from cohort heterogeneity, lymphoma subtype differences, and variations in treatment regimens (e.g., R+EPOCH versus R±CHOP). Nevertheless, cumulative evidence supports a strong link between cytokine dysregulation and unfavorable HAL outcomes. In addition to the factors mentioned above, IL-7, IL-8, IL-15, and interferon-γ (IFN-γ) were also strongly associated with increased risk of disease-free survival (DFS) and death; particularly, IL-7 and IL-8 were also strong independent predictors of DFS and OS respectively [62].
In summary, cytokines are not only important risk markers for the development of HAL but also hold potential as indicators for prognostic assessment. Jointly monitoring changes in specific cytokine levels prior to disease onset could facilitate early screening of high-risk individuals and the implementation of targeted interventions to improve clinical outcomes. However, given the variability in the predictive value of cytokines across different lymphoma subtypes, future studies should not only expand sample sizes but also consider constructing stratified predictive models based on histological subtypes (e.g., HIV-DLBCL and PCNSL) to enhance the specificity and clinical applicability of cytokine-based biomarkers. In addition, further research is warranted to explore the potential of targeting cytokine-related pathways (such as IL-6R inhibitors) in improving patient prognosis.

5.2. Chemokines

Chemokines are a family of secreted proteins with molecular weights ranging from 8–12 kDa, classified into CXC, CC, CX3C, and C chemokine subfamilies based on the arrangement of their N-terminal cysteine residues [63]. In the TME, chemokines participate in pathogenesis through dual mechanisms. On the one hand, they guide specific immune cells (such as B cells, T cells, and macrophages) to inflammatory sites via chemotactic gradients to regulate local immune responses. On the other hand, they act directly on tumor cells and endothelial cells to activate proliferative signaling pathways, maintain cancer stem cell phenotypes, and promote epithelial-to-mesenchymal transition (EMT), thereby driving malignant progression [64]. Consequently, the chemokine network serves as a crucial bridge linking tumor immunity to clinical outcomes [64,65,66].
CXCL13 (also known as B-cell-attracting chemokine 1, BCA-1) plays a vital role by interacting with its receptor CXCR5 to direct B-cell trafficking to secondary lymphoid organs such as the spleen and lymph nodes [67,68,69]. Aberrant activation of the CXCL13/CXCR5 axis has been implicated in the pathogenesis of various non-HIV-associated B-cell lymphoma subtypes [70,71,72,73,74,75]. In PLWH, dysregulation of CXCL13 may disrupt B-cell migration and damage lymphoid architecture, thereby promoting HIV-NHL development [76,77]. Several clinical studies have demonstrated the potential value of CXCL13 in the early diagnosis of HIV-NHL [56,78] and have shown significantly elevated serum CXCL13 levels as early as 2.5 years before lymphoma diagnosis [79]. The correlation between CXCL13 and systemic HIV-NHL risk was even more significant [7]. Additionlly, genetic factors may also influence CXCL13 expression and HIV-NHL risk. Specifically, the rs355689 variant in the CXCL13 gene is associated with reduced risk through decreased CXCL13 expression, whereas the rs630923 variant in CXCR5 correlates with increased CXCL13 levels and heightened lymphoma risk [56]. Regarding prognosis, HIV-NHL patients treated with R-EPOCH who had lower baseline CXCL13 levels were more likely to achieve complete remission, while those with higher levels exhibited significantly shortened OS and PFS [59], supporting the role of CXCL13 as a prognostic biomarker and a potential tool for individualized therapy guidance.
Another important chemokine is CXCL10, also known as IP-10, a 10-kDa secreted polypeptide belonging to the CXC family [63]. Previous studies reported that elevated CXCL10 levels are associated with rapid HIV disease progression [80,81,82,83]. In the field of HIV-NHL, early studies found significantly higher circulating CXCL10 levels in HAL patients compared to PLWH without lymphoma [84]. However, this study was limited by the small sample size and the concentration of specimen collection within one year prior to the diagnosis of lymphoma, making it difficult to clarify whether elevated CXCL10 is an early marker of lymphomagenesis or a secondary phenomenon close to the diagnostic period. Vendrame and colleagues found that sustained elevation of CXCL10 in the three-time windows prior to HIV-NHL diagnosis (>3 years, 1–3 years, and 0–1 year) increased the risk of morbidity by more than 3.7-10-fold, and high CXCL10 levels correlated with adverse prognosis significantly [55]. These findings were further validated in independent cohorts and AIDS Malignancy Consortium trials, supporting CXCL10 as both an early biomarker and a predictor of poor outcomes in HIV-NHL [57,60].
Other chemokines such as CCL2 and CCL17 also play significant roles in HIV-NHL. In immunocompetent DLBCL patients, high serum levels of CCL2 are associated with poorer OS and PFS [85]. Under HIV-infected conditions, elevated serum CCL2 and CCL17 levels correlate strongly with increased HIV-NHL risk [7]. Data from animal models also support the predictive value of CCL2 and macrophage inflammatory protein-1β (MIP-1β) for disease progression [54]. Importantly, CCL2 levels partially normalize following therapeutic interventions, suggesting a potential role as a dynamic prognostic marker [60]. CCL8, CCL19, CXCL11, and MIP-1δ have also shown abnormal expression in HIV-NHL [57]. Their potential involvement in immune cell trafficking and immune regulation points to a possible role in HIV-NHL pathogenesis. However, current evidence remains insufficient, and their clinical relevance requires validation in larger cohorts.
In conclusion, chemokines, by modulating the immune microenvironment and tumor behavior, serve multifaceted roles in the early warning, mechanistic understanding, and prognostic evaluation of HIV-NHL (Table 2). Some chemokines have shown promising potential for clinical translation. Nevertheless, for chemokines with less established roles, further large-scale studies are necessary to confirm their clinical utility. Future research should integrate multi-omics approaches and dynamic monitoring strategies to elucidate the temporal evolution of the chemokine network and its clinical implications.

5.3. Soluble Receptors

CD23, a low-affinity Fc receptor for IgE, is primarily expressed on the surface of B cells, and its expression is positively correlated with B-cell activation, serving as an important marker of B-cell activation [86]. The membrane-bound form of CD23 can be proteolytically cleaved to generate soluble CD23 (sCD23), which further promotes B-cell proliferation through a positive feedback mechanism [87,88]. Elevated serum sCD23 levels have been associated with the staging and prognosis of certain B-cell malignancies, such as chronic lymphocytic leukemia [89,90], suggesting that sCD23 may play a role in the pathological regulation of malignant B cells. Within the tumor necrosis factor receptor (TNF-R) superfamily, CD27 and CD30 are key co-stimulatory molecules mainly expressed on memory B cells [91,92]. Their soluble forms, sCD27 and sCD30, are generated by proteolytic cleavage and serve as biomarkers of immune activation [93,94]. Previous studies have demonstrated that sCD27 levels are elevated in HIV infection [95] and other conditions of immune dysregulation [96,97], which is informative for its use in HIV-NHL studies.
The Martínez-Maza’s group has systematically revealed the predictive value of serum sCD23, sCD27, and sCD30 in HIV-NHL [7,52,78,98,99,100]. These markers showed stronger associations with systemic HIV-NHL risk, particularly among individuals who had not received ART [52,78]. It has been observed that sCD23 is more strongly correlated with PCNSL than with systemic HIV-NHL [7], a finding consistent with earlier reports by Bossolasco [101]. This difference may reflect variations in pathological subtypes or blood-brain barrier permeability across study populations.
sCD163, a cleavage product of the macrophage scavenger receptor CD163, has also emerged as a marker of interest [102]. Elevated sCD163 levels are positively associated with HIV-NHL risk in a dose-dependent manner, especially in PCNSL [7], suggesting that it may be involved in virus-driven tumorigenesis, particularly EBV-associated PCNSL [103]. Previous studies in HL have similarly linked sCD163 with EBV-DNA presence [104]. In terms of therapeutic monitoring, recent clinical data showed that sCD25 levels significantly decreased after R-EPOCH treatment, while sCD44 and sCD163 levels remained unchanged [60]. sCD163 may not be useful for monitoring treatment response, dynamic changes in sCD25 could serve as a potential indicator of therapeutic efficacy.
In summary, combined detection of sCD23, sCD27, and sCD30 offers promising potential for ultra-early risk stratification in HIV-NHL, while sCD163 may serve as a specific diagnostic marker for EBV-driven subtypes, particularly PCNSL. Additionally, dynamic monitoring of sCD25 could complement therapeutic response evaluation. These soluble receptors not only deepen our understanding of HIV-NHL pathogenesis but also provide a foundation for biomarker-driven, stratified management strategies (Table 3). Future multicenter prospective studies are warranted to validate their predictive utility and clarify their mechanistic roles in lymphomagenesis.

5.4. Microbial Translocation Biomarkers

The gut microbiota maintains host immune homeostasis through dynamic interactions among epithelial cells, immune cells, and commensal bacteria [105]. Under physiological conditions, host barrier systems effectively prevent microbial products from entering the circulation [106]. However, HIV infection compromises gut barrier integrity, increasing permeability and allowing microbial products such as lipopolysaccharides (LPS) to translocate into the systemic circulation [106,107,108,109]. Even under ART, gut barrier restoration remains incomplete, and persistent microbial translocation becomes a major driver of systemic immune activation and inflammation in PLWH [108,109,110].
sCD14, a specific marker of monocyte/macrophage activation, is released via phospholipase-mediated membrane shedding and vesicle secretion [111]. LPS, a key component of Gram-negative bacterial outer membranes, can translocate into the systemic circulation following gut barrier disruption [112]. LPS binds to lipopolysaccharide-binding protein (LBP) and sCD14 to form a complex that activates the Toll-like receptor 4 (TLR4) signaling pathway, leading to NF-κB–mediated secretion of proinflammatory cytokines such as IL-6 and TNF-α [107,108,111,113]. Serum fatty acid-binding protein 2 (FABP2) levels serve as a marker of gut epithelial damage [114,115]. Endotoxin core antibody (EndoCAb), which neutralizes circulating LPS, reflects mucosal barrier integrity [116]; its reduction indicates microbial overstimulation and is associated with immune overactivation and poor prognosis [107,117]. The bioactivity of LPS is dynamically regulated by a network involving LBP, sCD14, and EndoCAb [118]. Thus, in PLWH, gut barrier dysfunction leads to increased microbial translocation, stimulating monocyte/macrophage activation and sustained systemic inflammation, which promotes lymphomagenesis [110,117] (Figure 3).
Elevated levels of sCD14, LBP, FABP2, combined with decreased EndoCAb, form a “microbial translocation signature” that has been validated as a biomarker for disease monitoring [119]. Given the close association between chronic immune activation and HIV-NHL, microbial translocation markers have attracted considerable attention. Persistent elevation of sCD14, LBP, LPS, FABP2, and haptoglobin, along with decreased EndoCAb, has been linked to increased HIV-NHL risk [7,8]. Notably, sCD14 shows a stronger association with central nervous system (CNS) HIV-NHL, while FABP2 and LBP are more related to systemic HIV-NHL [7]. Recent studies have extended the prognostic relevance of these markers. Baseline sCD14 levels are significantly inversely correlated with OS in HIV-NHL patients, and dynamic reductions in sCD14 and LBP following R-EPOCH therapy may indicate a partial reversal of immune activation, providing additional prognostic information [60].
In summary, microbial translocation biomarkers offer significant potential for risk prediction, prognostic stratification, and treatment response evaluation in HIV-NHL (Table 4). Although the precise molecular mechanisms linking microbial translocation to lymphomagenesis require further elucidation, these markers provide a strong theoretical basis for biomarker-guided stratified management strategies. Future research should aim to clarify the interactions between microbial dysbiosis, host immunity, and tumorigenesis, and explore targeted interventions focused on restoring gut barrier integrity. Additionally, given that HIV infection alters the composition of the intestinal flora, changes in fecal flora in patients with HAL warrant further exploration to help develop targeted intestinal strategies.

6. Protein-Related Biomarkers

6.1. Immunoglobulins

Immunoglobulin consists of two homologous heavy (H) and light (L) chains linked by disulfide bonds. Based on the heavy chain type, they are classified into five major classes: IgG, IgA, IgM, IgD, and IgE. Light chains exist as either kappa (κ) or lambda (λ) isotypes, with each chain containing a variable domain and multiple constant domains [120]. During B-cell development, H and L chains combine to form a complete immunoglobulin, while excess light chains are secreted into the blood as free light chains (FLCs) [121]. The clonal characteristics of FLCs and the κ/λ ratio have become important biomarkers reflecting abnormal B-cell activation, with significant applications in the diagnosis, therapeutic monitoring, and prognosis evaluation of cancers and inflammatory diseases [122,123,124,125]. In recent years, FLCs as a marker of B-cell activation have gained attention for their predictive value in HAL. However, the available findings remain heterogeneous regarding the optimal predictive time window. Several studies have demonstrated that serum κ and λ FLC levels significantly increase within five years before the diagnosis of HIV-NHL [55,126]. Although prediagnostic elevation is consistently observed, the precise temporal window for optimal predictive accuracy remains uncertain. Notably, FLCs appear to retain predictive value throughout this this five-year period in individuals with CD4 counts ≥100 cells/mm3, whereas among those with CD4 counts <100 cells/mm3, the association is primarily evident 2–5 years before diagnosis [126]. Shepherd et al. also reported that FLCs showed better predictive performance when measured more than two years before HIV-NHL diagnosis [127,128]. In contrast, findings from an Italian cohort demonstrated that κ+λ FLC levels exceeding 1.5 times the normal upper limit within two years before HAL diagnosis were associated with a sharply increased risk, with the strongest predictive performance observed during this period [129]. Although FLCs show certain advantages as early risk warning biomarkers, discrepancies in predictive timing have been reported, which may be influenced by multiple factors, including CD4+ T-cell counts, viral load, and lymphoma subtypes. Moreover, the prognostic relevance of FLCs in HAL remains unclear, as current studies have not identified a significant association [61].
Compared with FLCs, the role of total serum immunoglobulin levels in HIV-NHL risk prediction remains controversial. Early studies suggested that elevated serum globulin levels were associated with increased HIV-NHL risk [130], while later larger cohort studies failed to confirm this association [131], possibly due to differences in study design, sample characteristics, detection methods, and the impact of widespread ART use. Additionally, conventional immunoglobulin subtypes such as IgA, IgE, IgG, and IgM have been reported to be not significantly associated with the risk of HIV-NHL [52,126,127].

6.2. PD-1 and PD-L1

Programmed cell death protein 1 (PD-1), an important immunosuppressive molecule, is a member of the immunoglobulin superfamily [132]. PD-1 and its ligand programmed cell death ligand 1 (PD-L1) are key immune checkpoint molecules that regulate immune responses. They maintain immune tolerance by suppressing T-cell function and limiting the overreaction of the immune system [133]. However, this pathway is also exploited by various malignant tumor cells to evade immune surveillance and promote tumor progression [134]. In recent years, immune checkpoint inhibitors targeting the PD-1/PD-L1 pathway have shown significant efficacy in the treatment of several malignancies [133]. In HAL, the overexpression of PD-1/PD-L1 is not only closely related to immune escape mechanisms but may also play a crucial role in tumor initiation and progression. Epeldegui et al. found that the proportion of PD-L1+ B cells was significantly elevated in PLWH, and this phenomenon could be detected several years before the lymphoma diagnosis [135]. These PD-L1+ B cells predominantly exhibit a regulatory B-cell (Breg) phenotype, capable of secreting IL-10 and interacting with T cells through the PD-1/PD-L1 axis to suppress their immune function, thereby providing a favorable microenvironment for lymphoma immune escape. Moreover, in EBV-positive HIV-NHL patients, high expression of PD-1 has also been confirmed to be closely associated with poorer OS and PFS, suggesting that PD-1 is not only a marker of immune escape but also an independent prognostic factor [136].
In the context of HIV infection, chronic immune activation may further induce sustained high expression of PD-1/PD-L1, thus promoting tumor development and progression. Therefore, PD-1/PD-L1 not only holds potential as a biomarker for predicting HAL risk but also provides a theoretical basis for the clinical application of immune checkpoint inhibitors in such patients. Joint monitoring of PD-1 and PD-L1 expression could improve early screening efficiency for high-risk individuals and provide a foundation for personalized immunotherapy strategies.

6.3. Gal-1

Galectin-1 (Gal-1), a β-galactoside-binding protein, plays a critical role in immune regulation [137]. In HL, increased Gal-1 expression has been associated with poor prognosis and is considered a biomarker for disease progression [138]. In HAL, Gal-1 exhibits distinct expression patterns. Serum Gal-1 levels are lower in PLWH, particularly among those who subsequently develop HAL, whereas intratumoral Gal-1 expression is relatively elevated and has been associated with a better prognosis [139]. This discrepancy may be explained by changes in immune status and disease progression. HIV infection alters the host immune system, potentially suppressing the secretion of Gal-1 by immune cells and resulting in lower serum Gal-1 levels. In contrast, intratumoral Gal-1 expression appears to be independently regulated, with tumor cells and cells within the TME capable of autonomously producing Gal-1. High intratumoral Gal-1 expression may represent an adaptive response during tumor development, promoting tumor growth, immune evasion, and angiogenesis. Although current research remains limited, Gal-1 shows promising potential in the HAL field and warrants further in-depth investigation.

6.4. β2-Microglobulin

β2-microglobulin (β2-M) is a classic marker of tumor burden [140,141], and its serum levels are significantly associated with disease staging in HIV-negative DLBCL. Although some studies have suggested that β2-M alone may have limited diagnostic utility in HIV-NHL [142], recent clinical data indicate that elevated serum β2-M is directly associated with increased tumor cell proliferation and tumor burden [143]. These findings support its role as a biomarker for lymphoma progression and poor prognosis. Additionally, elevated β2-M levels have also been reported at the time of HIV-HL diagnosis [144]. These observations suggest that β2-M holds potential for risk prediction and prognostic assessment, although its utility may vary across different HAL subtypes. Further large-scale studies are needed to validate these findings.

6.5. Ferritin

Serum ferritin is recognized as an important prognostic factor in cancer detection [145], with its levels closely associated with tumor burden and disease activity in hematologic malignancies [146]. In immunocompetent patients with DLBCL, elevated ferritin levels are significantly correlated with poor prognosis [147,148], and the combination of ferritin with the IPI further enhances prognostic stratification [149]. Similarly, elevated ferritin levels are significantly associated with reduced survival, independently of IPI scores and CD4+ T-cell counts in HIV-DLBCL [61]. Furthermore, studies have suggested that ferritin levels may be linked to the polarization status of tumor-associated macrophages and could reflect the TME, offering potential guidance for rituximab-based therapy [150]. Overall, these findings underscore the significant prognostic value of ferritin as an independent biomarker in HAL.

6.6. Lactate Dehydrogenase

Serum lactate dehydrogenase (LDH) is one of the most classic biomarkers reflecting tumor burden and metabolic activity, and its elevation is significantly associated with poor prognosis in hematological malignancies [151,152]. In clinical practice for HAL, LDH has become an important independent prognostic indicator due to its accessibility, cost-effectiveness, and strong association with inferior PFS and OS [153,154,155]. However, its independent predictive value, threshold definitions, and clinical significance vary across different subtypes. In HIV-DLBCL, most studies support a strong correlation between LDH levels and OS, particularly when LDH exceeds twice the upper limit of normal (2 × ULN), where the 2-year OS rate significantly declines [156,157,158]. The prognostic value of LDH for PFS remains controversial. Some studies have reported that elevated LDH shortens median PFS by approximately four months but fails to retain independent predictive power [159], while more recent larger cohort studies confirmed LDH as an independent predictor of PFS [158]. These differences across studies may be related to the immune status of the study population or the evolution of treatment regimens. In contrast, in BL, the prognostic value of LDH is even more pronounced. When LDH exceeds 5 × ULN, both 3-year PFS and OS are significantly reduced [160]. In the latest study by Liu et al., LDH levels above 3 × ULN were significantly associated with PFS but required an even higher threshold to impact OS [161]. Such threshold differences suggest underlying biological heterogeneity among study populations, highlighting the need to optimize risk stratification and therapeutic strategies based on disease subtype. Multicenter studies are needed to establish standardized thresholds.
Additionally, LDH holds prognostic value in other special HAL subtypes. In HIV-associated PBL (HIV-PBL), LDH levels above 300 U/L significantly shortened median OS [162]. In HIV-associated PCNSL (HIV-PCNSL), elevated cerebrospinal fluid (CSF) LDH levels were correlated with tumor progression [163], providing a potential tool for disease monitoring in this unique setting. Recently, Liang’s group [164] integrated LDH and other variables to develop a new prognostic model, which outperformed the traditional IPI system in HIV-DLBCL prognostication, offering a feasible stratification approach for resource-limited settings, pending external and multicenter validation. In conclusion, LDH remains a biomarker of significant clinical value in HAL prognosis. Combined detection with other emerging biomarkers may further enhance the precision of prognostic assessment, necessitating ongoing translational research efforts.

6.7. Other Biomarkers

Proteomics provides an efficient tool for exploring HAL biomarkers, and it has demonstrated unique advantages in high-throughput protein screening, disease mechanism analysis and drug target development [165,166]. Detection of serum protein mass spectrometry reveals a number of differentially expressed proteins, such as apolipoprotein C-II (Apo C-II), complement C4-A, and serum amyloid A (SAA), which have important roles in the TME and are closely associated with the risk of HAL development [167]. Notably, elevated SAA levels have also been correlated with prolonged survival in HAL patients [168]. These differentially expressed proteins may assist in the early identification of high-risk PLWH, enabling closer monitoring and early intervention. Regarding prognostic evaluation, levels of the complement C1Q complex are closely associated with disease progression in HIV-NHL patients, and a decrease in C1Q levels after treatment predicts poor prognosis [142].
In summary, the detection of these serum protein biomarkers provides new molecular targets for the early diagnosis and prognostic evaluation of HAL, and holds promise for advancing individualized treatment strategies (Table 5). These molecules, through their dynamic expression patterns, enable the early identification of high-risk PLWH and show significant associations with TME remodeling and treatment response. It is important to note that the applicability of traditional biomarkers, such as β2-microglobulin and LDH, may vary across different HAL subtypes, suggesting that biomarker selection and application should be optimized according to disease-specific characteristics in clinical practice. Future research should build upon multicenter and external validation studies to further integrate proteomic data and develop multi-omics predictive models, ultimately enhancing the accuracy of prognostic assessment and the level of personalized therapy for HAL patients.

7. Genetic-Related Biomarkers

7.1. MiRNAs

MiRNAs are a class of endogenous, non-coding single-stranded RNAs, typically 18–24 nucleotides in length. Each miRNA can target multiple messenger RNAs (mRNAs), negatively regulating gene expression by promoting mRNA degradation or inhibiting translation [169,170,171]. Due to their tissue-specific expression patterns and high stability, miRNAs can be easily extracted from blood and other body fluids, making them promising candidates as diagnostic biomarkers and prognostic tools for cancer [172,173,174]. In recent years, the critical role of miRNAs in tumor biology has attracted increasing attention. Aberrant miRNA expression profiles have been recognized as common features across various malignancies, where they may function either as oncogenes or tumor suppressors [175,176]. In solid tumors, the regulatory networks involving miRNAs have been gradually elucidated [177,178,179]. Similarly, in hematological malignancies, dysregulation of miRNAs has been shown to be closely associated with the pathogenesis of lymphomas [180,181] and leukemias [182,183].The development of normal B cells is tightly regulated by miRNAs, and disruption of this regulation is considered a major driving factor in lymphomagenesis [184,185].
In HAL, miR-21 as an oncogenic miRNA shows significant elevation several years before diagnosis and can be used as a marker for early risk prediction [186,187,188]. In contrast, the high expression of miR-122 and miR-223 was more significantly associated with the risk of developing HIV-DLBCL and PCNSL [187]. The miR-17-92 cluster and its related miRNAs (such as miR-17, miR-106a, and miR-106b) are also markedly overexpressed in HIV-NHL, likely promoting tumor progression through negative regulation of the cell cycle inhibitor p21 [189].
While most studies have focused on the overexpression of miRNAs, recent investigations have also addressed miRNA downregulation. In animal models, miR-200c-3p was found to be significantly downregulated in HIV-1-treated BL cells [190], leading to the upregulation of its target proteins ZEB1 and ZEB2, which subsequently promote cancer cell migration and invasion [191,192]. These findings suggest that HIV-1 infection may accelerate lymphoma progression and metastasis by reshaping miRNA regulatory networks, highlighting the importance of early miRNA-based screening in high-risk HAL populations. However, whether downregulation patterns provide comparable predictive utility remains to be systematically validated. In addition, genetic studies have identified specific SNPs, such as DDX20 rs197412, miR-196a2 rs11614913, and HIF1A rs2057482, that are associated with an increased risk of HIV-NHL [193], which provides a genetic rationale for individualized risk stratification for screening.
In a word, current evidence strongly supports the role of miRNAs in HAL and potential targets for risk stratification, and the value of circulating miRNAs as an important biomarker for the early detection of HAL should not be ignored. However, issues such as disease specificity of miRNA expression and the impact of ART on miRNA profiles require further exploration. Future studies should aim to develop more precise miRNA detection methods, elucidate the specific regulatory networks involved, and refine individualized risk prediction and therapeutic strategies.

7.2. Circulating DNA

In recent years, studies have revealed that HIV-NHL possess distinct molecular and immunogenomic characteristics compared to those in HIV-negative individuals. M.I. Tiirikainen et al. found that in immunocompromised HIV-positive hosts, the progression of DLBCL requires fewer somatic genomic alterations. Tumor samples from HIV-positive patients exhibit fewer regional DNA copy number alterations (CNAs), and the frequency of CNAs is significantly lower than that observed in HIV-negative individuals [194]. In HAL, Trine Engelbrecht Hybel et al. identified multiple gene mutations, most commonly in KMT2D, TNFAIP3, and TP53. However, no subtype-specific gene variants were identified, indicating substantial genetic heterogeneity among different histological subtypes of HAL [195]. Moreover, the immunogenomic landscape of HIV-NHL is profoundly influenced by EBV status. In EBV-negative patients, T cells retain their ability to recognize tumor-specific neoantigens, particularly those related to IgH rearrangements, offering potential avenues for T-cell-based immunotherapy [196].
At the molecular level, under physiological conditions, B lymphocytes generate diverse antibody repertoires through random recombination of Ig gene segments [120]. In contrast, malignant transformation of B cells typically results in clonal Ig gene rearrangement, a hallmark of B-cell lymphomas [197,198]. In recent years, various high-throughput sequencing technologies have been applied to the study of circulating free DNA (cfDNA), particularly tumor-derived circulating tumor DNA (ctDNA). Clonal Ig gene rearrangements can be detected in the cfDNA of patients with NHL or HL [199,200,201,202,203], and such rearrangements are frequently observed in PLWH diagnosed with lymphoma [204]. The presence of Ig DNA rearrangement in plasma serves as a lymphoma-specific tumor biomarker (Table 6); patients who fail to clear clonal Ig DNA after treatment may be at high risk of failure with standard therapy [204]. The correlation between plasma cIg levels and both lymphoma disease burden and treatment response suggests that clonal Ig could serve as a sensitive and specific biomarker [202,205]. A more recent study [206], used next-generation sequencing (NGS) to detect plasma cfDNA in patients with HIV-associated aggressive B-cell lymphoma, finding clonal Ig heavy chain sequences in 55% of cases. In contrast, no such sequences were detected in HIV-infected patients with concomitant tuberculosis, further supporting the disease specificity of this biomarker.
Table 6. Genetic-related biomarkers.
Table 6. Genetic-related biomarkers.
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
miRNAsmiRNA SNPs
miR-17
miR-21
miR-122
miR-106a
miR-106b
miR-223
miR-200c-3p
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-BL cell line
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Decreased
Predictive
Predictive
Predictive
Predictive
Predictive
Predictive
Predictive
Predictive
[193]
[189]
[186,187,188]
[187]
[189]
[189]
[187]
[190]
Circulating DNADNA-CNAs
Clonal Ig DNA
cfDNA
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
Decreased
Increased
Increased
Predictive
Predictive/Treatment Monitoring
Predictive
[194]
[204]
[206]
Abbreviations: miRNAs, microRNAs; HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; HIV-BL, HIV-associated Burkitt lymphoma; SNPs, single nucleotide polymorphisms; DNA-CNAs, copy number alterations; Ig, Immunoglobulin; circulating free DNA.

8. Extracellular Vesicles

EVs are membrane-bound structures secreted by cells, capable of carrying nucleic acids, proteins, and metabolites [207]. EVs can specifically express immune regulatory molecules such as PD-L1 and CD40 ligands on their surface, facilitating intercellular communication and modulation of the TME through ligand–receptor interactions, thereby promoting tumor progression [208,209,210,211]. EVs have attracted significant attention in the field of cancer liquid biopsy [208,212]. In immunocompetent NHL patients, circulating EVs have been validated as biomarkers for diagnosis and prognostic assessment [213,214]. In the context of HAL, EVs carrying immune regulatory molecules such as PD-L1, CD40, CD40L, and TNF receptors-II (TNF-RII) are significantly elevated and correlate with levels of inflammatory cytokines [215]. Baseline levels of EVs expressing PD-L1 or IL-6R can independently predict the risk of HIV-NHL, with TNF-RII+ EVs being particularly associated with the DLBCL subtype [216].
Overall, EVs represent a promising emerging class of biomarkers for early risk prediction and therapeutic monitoring in HIV-NHL (Table 7). The specific molecules they carry may also directly regulate disease progression. Furthermore, EV-associated miRNAs, proteins, and DNA offer considerable potential for liquid biopsy applications. However, studies investigating EVs in HAL remain limited, and several challenges persist, including small sample sizes, unclear mechanistic insights (particularly in the HIV setting), and technical barriers in EV isolation. Future research should focus on expanding cohort sizes, clarifying the molecular mechanisms of EVs in HAL, and optimizing isolation and detection techniques to facilitate their clinical translation.

9. Lymphocyte Subpopulation

9.1. CD4+ T Cells and CD4/CD8 Ratio

CD4+ T cells are the primary targets of HIV infection. Their decline, along with a reduced CD4/CD8 ratio, reflects immune dysfunction and has been widely used to assess immune status and lymphoma risk in PLWH. Studies have confirmed that lower CD4+ T-cell counts and CD4/CD8 ratios not only sensitively reflect immune reconstitution but also serve as independent predictors for the development of ADCs [217]. Regarding predictive value, earlier studies suggested that the historical nadir of CD4+ T-cell counts was closely associated with HAL risk [218,219]. In mid to late ART, more studies have highlighted that recent CD4+ T-cell counts are stronger independent predictors [131,220,221,222,223]. A study from the UK National HIV Oncology Centre demonstrated that while CD4+ T-cell counts were generally lower at HAL diagnosis in the pre-ART era, significant immune recovery was observed following the widespread introduction of ART, with higher CD4+ T-cell counts at lymphoma diagnosis thereafter [224]. This suggests that once CD4+ T-cell counts are restored under ART, the impact of prior immunodeficiency on NHL risk diminishes [225], and greater emphasis should be placed on current immune status. Recent advancements in dynamic immune monitoring have enabled comparisons across baseline, nadir, and current CD4+ T-cell counts, as well as CD4/CD8 ratios. It has been found that low current CD4+ T-cell counts and a CD4/CD8 ratio <0.4 are independent predictors of HIV-NHL [226,227]. In the United States, a CD4/CD8 ratio <0.3 significantly increased NHL risk and demonstrated long-term predictive value [228], consistent with findings from Spanish cohorts [229]. Moreover, an international study across Europe and Australia indicated that a CD4/CD8 ratio <0.5 independently predicted HIV-NHL [230]. However, Caby et al. based on a French multicenter cohort proposed different conclusions, as they found that the CD4/CD8 ratio did not show a statistical association with the risk of HIV-NHL in PLWH with ≥500/mm3 CD4+ T cells [231]. Such discrepancies may be attributable to variations in baseline immune status, timing of measurements, methodologies, or lymphoma subtype distributions.
In terms of prognosis, low CD4+ T cells are associated with inferior OS and PFS among HAL patients [232,233]. A CD4/CD8 ratio <0.5 is also an independent predictor of non-AIDS-related and all-cause mortality among PLWH, including those with NHL [234]. Our center’s data further demonstrate that newly diagnosed HAL patients exhibit significantly reduced CD4+ T cells and CD4/CD8 ratios, accompanied by elevated CD8+ T-cell counts, IL-6, and IL-2R levels—all factors associated with poor prognosis [235]. Additionally, S.K. Barta’s team integrated CD4+ T-cell counts, viral load, and AIDS history into a HAL-specific prognostic model, which outperformed the traditional aaIPI for OS prediction and risk stratification, although its performance for predicting CR and PFS remained limited [236]. Combining the CD4/CD8 ratio with IPI further improves prognostic accuracy and offers practical guidance for therapeutic decision-making in HAL [237].

9.2. Lymphocyte-to-Monocyte Ratio

The lymphocyte-to-monocyte ratio (LMR) reflects the extent of systemic inflammatory responses in cancer patients and has demonstrated prognostic value in both solid tumors and HIV-negative diffuse large B-cell lymphoma [238,239]. Zeng et al. were the first to validate its applicability in HIV-DLBCL [240]. Higher LMR (≥2) was significantly associated with longer survival in HAL [241]. Our center has recently developed an ARDPI model that includes LMR and CD4/CD8 ratio. Compared with IPI, the ARDPI model has higher predictive accuracy and accurately predicts survival in newly diagnosed HIV-DLBCL patients [14]. However, the prognostic value of LMR in other HIV-associated aggressive lymphoma subtypes remains to be further validated.
Overall, CD4+ T-cell counts, CD4/CD8 ratio, and LMR have become important biomarkers for risk assessment and prognostic prediction in HAL (Table 8). A persistently low CD4/CD8 ratio suggests a high risk of HAL development, and its combination with traditional indices such as the IPI can further enhance the accuracy of risk prediction and prognostic stratification. Future studies should focus on large-scale, multicenter prospective cohorts to determine the optimal timing and cutoff values for these biomarkers. In clinical practice, it is recommended to strengthen lymphoma screening for PLWH with a persistently inverted CD4/CD8 ratio and to incorporate emerging prognostic models into therapeutic decision-making and follow-up monitoring.

10. Virus-Related Biomarkers

10.1. HIV Viraemia

HIV viremia contributes to the increased risk of HAL not only by inducing CD4+ T-cell depletion and chronic immune activation but also through the direct oncogenic mechanisms of HIV-1 itself [242,243,244]. Studies have shown that cumulative HIV viraemia is an independent predictor of HAL development [223]. PLWH with HIV RNA levels exceeding 105 copies/mL exhibit a significantly higher risk of developing HIV-NHL [131,221,245]. However, the quantification of cumulative viremia effects varies across regions. In the German study, it was found that for every 2000 days log10 copies/mL increase in cumulative viremia was associated with a 67% higher overall lymphoma risk, with the greatest risk observed in the BL subtype [246]. In contrast, studies from the United States focused on the impact of low-level viremia, showing that HIV RNA levels between 51–500 copies/mL increased the risk of HIV-NHL by 66–110%, with DLBCL being the most strongly associated subtype [247]. Multivariate analyses further indicated that for each 1 log10 copies/mL increase in HIV RNA, the risk of NHL increased by 24–42% [247,248]. These findings suggest a clear dose-dependent relationship between HIV viremia levels and HIV-NHL risk.
By comparison, some studies have shown that although high HIV viral loads prior to ART initiation increase the risk of lymphoma and other cancers, early initiation of ART can substantially mitigate this risk. Consequently, the association between HIV viremia and lymphoma risk becomes less pronounced during effective ART treatment [249,250]. Notably, persistent viremia even after HIV-NHL diagnosis has been associated with increased mortality [251], underscoring the importance of maintaining strict viral suppression during chemotherapy.
Animal model studies have revealed that although ART effectively reduces plasma viral load, it is powerless to remove viral reservoirs in the lymph nodes, and the virus can rebound rapidly after stopping the drug [252]. Future HIV cure strategies must therefore focus on targeting these hidden reservoirs within deep tissues such as lymph nodes. Additionally, in humanized mouse models, the use of caspase-1 inhibitor VX-765 significantly reduced immune activation, CD4+ T-cell depletion, plasma viremia, and the establishment of HIV-1 reservoirs [253]. These findings suggest that early ART initiation combined with anti-inflammatory therapy could effectively limit reservoir formation. Furthermore, in patients initiating ART during the AIDS phase, the proviral integration sites showed monoclonal expansion, which progressively intensified over time. This clonal expansion reduces immune system heterogeneity, potentially increasing susceptibility to infections and malignancies, including lymphoma. These observations offer mechanistic insights into the elevated risk of opportunistic infections and cancers among long-term survivors [254].

10.2. HIV-Encoded Proteins

The prevailing view is that the development of HAL is closely related to the sustained action of HIV-encoded proteins. These viral proteins can drive oncogenic processes through multiple mechanisms, with their sustained presence forming a molecular foundation for cumulative risk [255]. Among them, the Tat and p17 matrix proteins are considered pivotal contributors to HAL development. Tat promotes lymphomagenesis by inducing immune evasion [256], oxidative stress and DNA damage [257], chromosomal translocations and genomic instability [258,259,260,261], dysregulation of signaling pathways and transcriptional networks [260,262], as well as compensatory responses and adaptive evolution [263]. These processes establish a vicious cycle of “immune escape–genomic instability–aberrant proliferation” that ultimately drives lymphoma development. Similarly, p17 promotes malignant transformation by activating key signaling cascades such as PI3K/Akt and MEK/ERK, inducing abnormal B-cell proliferation, genomic instability, and remodeling of the TME [54,264,265,266]. Studies have shown that p17 is highly detectable in plasma and tissues [267,268], even in patients with a good response to ART therapy. Compared to other HIV proteins (such as gp120, p24, nef, and tat), p17 is already highly expressed during the early stages of lymphoma, whereas the latter proteins tend to rise during the later stages [54,264,269]. Moreover, natural variation in the p17 gene during HIV-1 infection has been shown to differentially regulate intracellular signaling pathways and promote clonal expansion of B cells, thereby accelerating lymphomagenesis [270]. Specific p17 genevariants further enhance B-cell clonal growth and increase the risk of HAL development [271,272].
To conclude, persistent HIV viremia and the continuous expression of HIV-encoded proteins jointly contribute to the development and progression of HAL. Cumulative viral load shows a dose-dependent association with lymphoma risk, emphasizing the need for dynamic monitoring and sustained viral suppression to minimize oncogenic stimulation and improve outcomes. In diagnosed patients, early initiation of intensified antiretroviral and anti-inflammatory therapies is critical. Among viral proteins, p17 exhibits superior potential over other HIV-encoded proteins as a biomarker for early HAL risk prediction by promoting B-cell proliferation and remodeling the TME. Future directions should prioritize the establishment of p17-based dynamic risk assessment models and strategies targeting viral reservoirs to enhance early detection and optimize the management of HAL in high-risk populations.

10.3. EBV Viraemia

EBV has long been closely associated with HIV-PCNSL. The detection of EBV DNA in CSF demonstrates a sensitivity of 100% and a specificity of 98.5%, making it a potentially reliable diagnostic biomarker [273]. All PCNSL samples show strong positive expression of EBV-encoded small RNA (EBER1), indicating a 100% association between EBV and HIV-PCNSL [274]. Although some studies have found that EBV viremia does not serve as an independent prognostic marker and shows no significant predictive value in HIV-NHL [9,275,276,277], other research has reported that a high pretreatment plasma EBV DNA level is a poor prognostic marker in HIV-DLBCL [12,278]. Taken together, while the prognostic value of plasma EBV DNA in HIV-NHL remains controversial, its diagnostic utility in HIV-PCNSL—particularly via CSF detection—has been well established and validated.

10.4. KSHV Viraemia

KSHV is one of the key pathogenic factors in HIV-related malignant lymphoproliferative disorders, particularly in PEL and multicentric Castleman disease (MCD), where it has a well-established oncogenic role. PEL is almost invariably associated with KSHV infection, rendering KSHV serostatus highly specific for PEL and providing some predictive value [279,280]. Although recent advances in antiviral and immunomodulatory therapies have improved outcomes for KSHV-related MCD, patients still face a significantly elevated risk of NHL [281]. Untreated KSHV-related MCD typically follows a progressive and often fatal course, potentially evolving into PEL or KSHV-related DLBCL [282]. Such DLBCL cases may represent a clonal plasmablastic expansion driven by KSHV infection, suggesting that KSHV can mediate B-cell malignant transformation under certain pathological conditions. Moreover, in KSHV-related MCD and malignancies, KSHV frequently coexists with EBV, and the two viruses may synergistically promote tumorigenesis by modulating each other’s latent and lytic cycles, disrupting host immune responses, and altering cell fate pathways [283]. HIV infection itself contributes to this process through immunosuppression and activation of viral oncogenic mechanisms, creating a favorable microenvironment for KSHV and EBV-driven oncogenesis [283]. However, the pathogenic role of KSHV in HALs is subtype-specific. For instance, in more common HAL subtypes such as BL, DLBCL, and HL, KSHV has not been consistently detected or linked to tumor development [279,280]. Instead, these lymphomas are typically EBV-associated. Thus, while KSHV holds significant etiological and predictive relevance in PEL and certain MCD-related lymphomas, it is not suitable as a universal risk-predictive biomarker for all HAL (Table 9).
Table 9. Virus-related biomarkers.
Table 9. Virus-related biomarkers.
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
HIV-related markersHIV viraemia
p17
p17 gene variation
Tat
Nef
gp120
HIV-NHL patients
HIV-NHL patients/mice
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients/mice
Mice
Increased
Increased
Increased
Increased
Increased
Increased
Predictive/Prognostic
Predictive
Predictive
Predictive
Predictive
Predictive
[131,221,223,245,246,247,248,249,250,251]
[54,264,269]
[270,271,272]
[54,264,269]
[54,264,269]
[54,264,269]
Other virusesEBV viraemia
KSHV viraemia
HAL patients
HIV-PEL patients
Increased
Increased
Predictive/Prognostic
Predictive
[9,12,275,276,277,278]
[279,280,281,282,283]
Abbreviations: HAL, HIV-associated lymphoma; HIV-PEL, HIV-associated primary effusion lymphoma; Tat, transactivator of transcription; Nef, negative factor; EBV, Epstein–Barr virus; KSHV, Kaposi’s sarcoma-associated herpesvirus.

11. Conclusions and Perspectives

HAL remains one of the leading causes of mortality among PLWH, characterized by aggressive behavior and rapid progression. Over recent years, substantial progress has been made in the discovery of circulating biomarkers related to immune-inflammatory mediators, chemokines, soluble receptors, serum proteins, lymphocyte subsets, HIV viral parameters, and virus-encoded proteins. These biomarkers offer valuable information for early risk warning, therapeutic response monitoring, and prognostic stratification (Table 10). Dysregulated cytokine and chemokine profiles reflect immune activation and B-cell dysregulation, providing early indicators of HAL risk. Markers such as FLCs, clonal Ig DNA, β2-M, ferritin, and LDH have demonstrated potential for monitoring tumor burden and disease activity. Meanwhile, emerging biomarkers including circulating miRNAs and EVs have expanded the horizons for minimally invasive liquid biopsy applications. Lymphocyte subpopulation analysis and HIV viral load monitoring further underscore the critical role of immune function and viral persistence in HAL pathogenesis and outcomes.
Nevertheless, several challenges remain in translating these findings into clinical practice. Case number limitations, subtype heterogeneity, technical barriers, and retrospective study biases may affect biomarker validation and generalizability. Future research should prioritize large-scale, multi-center, prospective cohort studies integrating multi-omics approaches to map biomarker dynamics across HAL subtypes comprehensively. The validation of predictive models and the optimization of threshold definitions will be crucial steps toward clinical implementation. Ultimately, integrating dynamic biomarker monitoring into routine screening could enable early identification and precision intervention for high-risk PLWH. Progress in biomarker-driven strategies holds promise for transitioning HAL management toward a paradigm of dynamic surveillance and real-time intervention to significantly improve patient outcomes.

Author Contributions

X.S. and Q.X., Writing-original draft, Methodology, Visualization; Y.L. (Yi Liu), Methodology, Visualization; Y.L. (Ya Li), X.X. and S.H., Data curation, Validation; J.L., Software, Validation; X.Z., Writing-review & editing; Y.L. (Yao Liu), Conceptualization, Resources, Funding acquisition, Writing-review & editing, Project administration. X.Z. and Yao Liu are co-corresponding authors of this article. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Chongqing Science and Health Joint Medical Research Sprint, Key Projects, and Major Projects (2025DBXM002), Chongqing Innovative Medical Device Application Demonstration Project (CQEIC2024MDAD-050), the Key Project of Chongqing Technology Innovation and Application Development Special Project (CSTB2024TIAD-KPX0031), Natural Science Foundation of Chongqing, China (CSTB2024NSCQ-KJFZMSX0077) and the Fundamental Research Funds for the Central Universities (2022CDJYGRH-001).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Thanks to all authors for their contribution to this manuscript. The authors are thankful to Huihui Fu, Liuyue Zhai and Yifeng Tang for their help in reviewing quality improvement.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. UNAIDS. Global HIV & AIDS Statistics—Fact Sheet. Available online: https://www.unaids.org/en/resources/fact-sheet (accessed on 2 May 2025).
  2. Hernández-Ramírez, R.U.; Shiels, M.S.; Dubrow, R.; Engels, E.A. Cancer risk in HIV-infected people in the USA from 1996 to 2012: A population-based, registry-linkage study. Lancet HIV 2017, 4, e495–e504. [Google Scholar] [CrossRef] [PubMed]
  3. Gibson, T.M.; Morton, L.M.; Shiels, M.S.; Clarke, C.A.; Engels, E.A. Risk of non-Hodgkin lymphoma subtypes in HIV-infected people during the HAART era: A population-based study. AIDS 2014, 28, 2313–2318. [Google Scholar] [CrossRef]
  4. Simard, E.P.; Engels, E.A. Cancer as a cause of death among people with AIDS in the United States. Clin. Infect. Dis. 2010, 51, 957–962. [Google Scholar] [CrossRef]
  5. Yarchoan, R.; Uldrick, T.S. HIV-Associated Cancers and Related Diseases. N. Engl. J. Med. 2018, 378, 1029–1041. [Google Scholar] [CrossRef] [PubMed]
  6. Xiao, Q.; He, S.; Wang, C.; Liu, Y.; Zhang, X.; Liu, Y. Urgent Call for Attention to HIV-Associated Lymphoma: After AIDS 2024, the 25th International AIDS Conference. J. Med. Virol. 2025, 97, e70159. [Google Scholar] [CrossRef]
  7. Epeldegui, M.; Magpantay, L.; Guo, Y.; Halec, G.; Cumberland, W.G.; Yen, P.K.; Macatangay, B.; Margolick, J.B.; Rositch, A.F.; Wolinsky, S.; et al. A prospective study of serum microbial translocation biomarkers and risk of AIDS-related non-Hodgkin lymphoma. AIDS 2018, 32, 945–954. [Google Scholar] [CrossRef]
  8. Marks, M.A.; Rabkin, C.S.; Engels, E.A.; Busch, E.; Kopp, W.; Rager, H.; Goedert, J.J.; Chaturvedi, A.K. Markers of microbial translocation and risk of AIDS-related lymphoma. AIDS 2013, 27, 469–474. [Google Scholar] [CrossRef]
  9. Rabkin, C.S.; Engels, E.A.; Landgren, O.; Schuurman, R.; Camargo, M.C.; Pfeiffer, R.; Goedert, J.J. Circulating cytokine levels, Epstein-Barr viremia, and risk of acquired immunodeficiency syndrome-related non-Hodgkin lymphoma. Am. J. Hematol. 2011, 86, 875–878. [Google Scholar] [CrossRef]
  10. Wang, C.; Xiao, Q.; Zhang, X.; Liu, Y. HIV associated lymphoma: Latest updates from 2023 ASH annual meeting. Exp. Hematol. Oncol. 2024, 13, 65. [Google Scholar] [CrossRef]
  11. Barta, S.K.; Samuel, M.S.; Xue, X.; Wang, D.; Lee, J.Y.; Mounier, N.; Ribera, J.M.; Spina, M.; Tirelli, U.; Weiss, R.; et al. Changes in the influence of lymphoma- and HIV-specific factors on outcomes in AIDS-related non-Hodgkin lymphoma. Ann. Oncol. 2015, 26, 958–966. [Google Scholar] [CrossRef]
  12. Dunleavy, K.; Little, R.F.; Pittaluga, S.; Grant, N.; Wayne, A.S.; Carrasquillo, J.A.; Steinberg, S.M.; Yarchoan, R.; Jaffe, E.S.; Wilson, W.H. The role of tumor histogenesis, FDG-PET, and short-course EPOCH with dose-dense rituximab (SC-EPOCH-RR) in HIV-associated diffuse large B-cell lymphoma. Blood 2010, 115, 3017–3024. [Google Scholar] [CrossRef]
  13. Chen, J.; Wu, Y.; Kang, Z.; Qin, S.; Ruan, G.; Zhao, H.; Tao, X.; Xie, Z.; Peng, J. A promising prognostic model for predicting survival of patients with HIV-related diffuse large B-cell lymphoma in the cART era. Cancer Med. 2023, 12, 12470–12481. [Google Scholar] [CrossRef] [PubMed]
  14. Yang, T.; Lei, H.; Li, J.; Liang, Y.; Wang, C.; Liu, J.; Wu, Y.; Liu, J.; Zhou, Q.; Min, H.; et al. A Visual Nomogram Survival Prediction Model in Acquired Immune Deficiency Syndrome (AIDS)-Related Diffuse Large B-Cell Lymphoma. J. Med. Virol. 2025, 97, e70359. [Google Scholar] [CrossRef]
  15. HIV.pdf. Available online: https://www.nccn.org/professionals/physician_gls/pdf/hiv.pdf (accessed on 3 May 2025).
  16. Vaccher, E.; Chadburn, A.; Gloghini, A.; Antinori, A.; Bower, M.; Carbone, A. Lymphomas included in the AIDS case definition: An update 30 years later. Lancet HIV 2023, 10, e635. [Google Scholar] [CrossRef] [PubMed]
  17. Al-Naeeb, A.B.; Ajithkumar, T.; Behan, S.; Hodson, D.J. Non-Hodgkin lymphoma. BMJ 2018, 362, k3204. [Google Scholar] [CrossRef]
  18. Goedert, J.J.; Coté, T.R.; Virgo, P.; Scoppa, S.M.; Kingma, D.W.; Gail, M.H.; Jaffe, E.S.; Biggar, R.J. Spectrum of AIDS-associated malignant disorders. Lancet 1998, 351, 1833–1839. [Google Scholar] [CrossRef]
  19. Coté, T.R.; Biggar, R.J.; Rosenberg, P.S.; Devesa, S.S.; Percy, C.; Yellin, F.J.; Lemp, G.; Hardy, C.; Geodert, J.J.; Blattner, W.A. Non-Hodgkin’s lymphoma among people with AIDS: Incidence, presentation and public health burden. AIDS/Cancer Study Group. Int. J. Cancer 1997, 73, 645–650. [Google Scholar] [CrossRef]
  20. Chen, Y.; Zhao, J.; Sun, P.; Cheng, M.; Xiong, Y.; Sun, Z.; Zhang, Y.; Li, K.; Ye, Y.; Shuai, P.; et al. Estimates of the global burden of non-Hodgkin lymphoma attributable to HIV: A population attributable modeling study. EClinicalMedicine 2024, 67, 102370. [Google Scholar] [CrossRef]
  21. Kimani, S.M.; Painschab, M.S.; Horner, M.J.; Muchengeti, M.; Fedoriw, Y.; Shiels, M.S.; Gopal, S. Epidemiology of haematological malignancies in people living with HIV. Lancet HIV 2020, 7, e641–e651. [Google Scholar] [CrossRef]
  22. Nishijima, T.; Inaba, Y.; Kawasaki, Y.; Tsukada, K.; Teruya, K.; Kikuchi, Y.; Gatanaga, H.; Oka, S. Mortality and causes of death in people living with HIV in the era of combination antiretroviral therapy compared with the general population in Japan. AIDS 2020, 34, 913–921. [Google Scholar] [CrossRef]
  23. Poizot-Martin, I.; Lions, C.; Allavena, C.; Huleux, T.; Bani-Sadr, F.; Cheret, A.; Rey, D.; Duvivier, C.; Jacomet, C.; Ferry, T.; et al. Spectrum and Incidence Trends of AIDS- and Non-AIDS-Defining Cancers between 2010 and 2015 in the French Dat’AIDS Cohort. Cancer Epidemiol. Biomark. Prev. 2021, 30, 554–563. [Google Scholar] [CrossRef]
  24. Shiels, M.S.; Pfeiffer, R.M.; Gail, M.H.; Hall, H.I.; Li, J.; Chaturvedi, A.K.; Bhatia, K.; Uldrick, T.S.; Yarchoan, R.; Goedert, J.J.; et al. Cancer burden in the HIV-infected population in the United States. J. Natl. Cancer Inst. 2011, 103, 753–762. [Google Scholar] [CrossRef]
  25. Simard, E.P.; Pfeiffer, R.M.; Engels, E.A. Cumulative incidence of cancer among individuals with acquired immunodeficiency syndrome in the United States. Cancer 2011, 117, 1089–1096. [Google Scholar] [CrossRef]
  26. Collaboration of Observational HIV Epidemiological Research Europe (COHERE) Study Group; Bohlius, J.; Schmidlin, K.; Costagliola, D.; Fätkenheuer, G.; May, M.; Caro Murillo, A.M.; Mocroft, A.; Bonnet, F.; Clifford, G.; et al. Prognosis of HIV-associated non-Hodgkin lymphoma in patients starting combination antiretroviral therapy. AIDS 2009, 23, 2029–2037. [Google Scholar]
  27. Schommers, P.; Hentrich, M.; Hoffmann, C.; Gillor, D.; Zoufaly, A.; Jensen, B.; Bogner, J.R.; Thoden, J.; Wasmuth, J.-C.; Wolf, T.; et al. Survival of AIDS-related diffuse large B-cell lymphoma, Burkitt lymphoma, and plasmablastic lymphoma in the German HIV Lymphoma Cohort. Br. J. Haematol. 2015, 168, 806–810. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, C.; Liu, J.; Liu, Y. Progress in the Treatment of HIV-Associated Lymphoma When Combined With the Antiretroviral Therapies. Front. Oncol. 2021, 11, 798008. [Google Scholar] [CrossRef]
  29. Hentrich, M.; Müller, M.; Wyen, C.; Pferschy, A.; Jurinovic, V.; Siehl, J.; Rockstroh, J.K.; Schürmann, D.; Hoffmann, C.; German HIV-Related Lymphoma Study Group. Stage-adapted treatment of HIV-associated Hodgkin lymphoma: Long-term results of a prospective, multicenter study. Hemasphere 2024, 8, e68. [Google Scholar] [CrossRef]
  30. Carbone, A.; Gloghini, A.; Serraino, D.; Spina, M.; Tirelli, U.; Vaccher, E. Immunodeficiency-associated Hodgkin lymphoma. Expert. Rev. Hematol. 2021, 14, 547–559. [Google Scholar] [CrossRef] [PubMed]
  31. Opie, J.; Mohamed, Z.; Chetty, D.; Bailey, J.; Brown, K.; Verburgh, E.; Hardie, D. Hodgkin lymphoma: The role of EBV plasma viral load testing in an HIV-endemic setting. Clin. Exp. Med. 2024, 25, 10. [Google Scholar] [CrossRef]
  32. Huguet, M.; Navarro, J.-T.; Moltó, J.; Ribera, J.-M.; Tapia, G. Diffuse Large B-Cell Lymphoma in the HIV Setting. Cancers 2023, 15, 3191. [Google Scholar] [CrossRef]
  33. Carroll, V.; Garzino-Demo, A. HIV-associated lymphoma in the era of combination antiretroviral therapy: Shifting the immunological landscape. Pathog. Dis. 2015, 73, ftv044. [Google Scholar] [CrossRef] [PubMed]
  34. Dal Maso, L.; Franceschi, S. Epidemiology of non-Hodgkin lymphomas and other haemolymphopoietic neoplasms in people with AIDS. Lancet Oncol. 2003, 4, 110–119. [Google Scholar] [CrossRef]
  35. Gaidano, G.; Pasqualucci, L.; Capello, D.; Berra, E.; Deambrogi, C.; Rossi, D.; Maria Larocca, L.; Gloghini, A.; Carbone, A.; Dalla-Favera, R. Aberrant somatic hypermutation in multiple subtypes of AIDS-associated non-Hodgkin lymphoma. Blood 2003, 102, 1833–1841. [Google Scholar] [CrossRef] [PubMed]
  36. Dolcetti, R.; Gloghini, A.; Caruso, A.; Carbone, A. A lymphomagenic role for HIV beyond immune suppression? Blood 2016, 127, 1403–1409. [Google Scholar] [CrossRef]
  37. Lurain, K.; Ramaswami, R.; Yarchoan, R. The role of viruses in HIV-associated lymphomas. Semin. Hematol. 2022, 59, 183–191. [Google Scholar] [CrossRef]
  38. Carbone, A.; Volpi, C.C.; Gualeni, A.V.; Gloghini, A. Epstein-Barr virus associated lymphomas in people with HIV. Curr. Opin. HIV AIDS 2017, 12, 39–46. [Google Scholar] [CrossRef]
  39. Berhan, A.; Bayleyegn, B.; Getaneh, Z. HIV/AIDS Associated Lymphoma: Review. Blood Lymphat. Cancer 2022, 12, 31–45. [Google Scholar] [CrossRef]
  40. Zhang, X.; Yang, Z.; Xie, X.; Li, J.; Xiao, Q.; Xu, G.; Ma, B.; Xie, X.; Liu, Y.; Zhai, L.; et al. The single-cell immune landscape of HIV-associated aggressive B-cell lymphoma. J. Natl. Cancer Cent. 2025, 5, 221–235. [Google Scholar] [CrossRef]
  41. Hampel, H.; O’Bryant, S.E.; Molinuevo, J.L.; Zetterberg, H.; Masters, C.L.; Lista, S.; Kiddle, S.J.; Batrla, R.; Blennow, K. Blood-based biomarkers for Alzheimer disease: Mapping the road to the clinic. Nat. Rev. Neurol. 2018, 14, 639–652. [Google Scholar] [CrossRef]
  42. Kang, S.; Narazaki, M.; Metwally, H.; Kishimoto, T. Historical overview of the interleukin-6 family cytokine. J. Exp. Med. 2020, 217, e20190347. [Google Scholar] [CrossRef]
  43. Fu, C.; Zhang, X.; Zhang, X.; Wang, D.; Han, S.; Ma, Z. Advances in IL-7 Research on Tumour Therapy. Pharmaceuticals 2024, 17, 415. [Google Scholar] [CrossRef]
  44. Ouyang, W.; Rutz, S.; Crellin, N.K.; Valdez, P.A.; Hymowitz, S.G. Regulation and functions of the IL-10 family of cytokines in inflammation and disease. Annu. Rev. Immunol. 2011, 29, 71–109. [Google Scholar] [CrossRef] [PubMed]
  45. D’mello, K.P.; Zhao, L.; Kaser, E.C.; Zhu, Z.; Xiao, H.; Wakefield, M.R.; Bai, Q.; Fang, Y. The role of interleukins and the widely studied TNF-α in non-Hodgkin’s lymphoma. Med. Oncol. 2021, 38, 56. [Google Scholar] [CrossRef] [PubMed]
  46. 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]
  47. Nacinović-Duletić, A.; Stifter, S.; Dvornik, S.; Skunca, Z.; Jonjić, N. Correlation of serum IL-6, IL-8 and IL-10 levels with clinicopathological features and prognosis in patients with diffuse large B-cell lymphoma. Int. J. Lab. Hematol. 2008, 30, 230–239. [Google Scholar] [CrossRef]
  48. Edlefsen, K.L.; Martínez-Maza, O.; Madeleine, M.M.; Magpantay, L.; Mirick, D.K.; Kopecky, K.J.; LaCroix, A.Z.; De Roos, A.J. Cytokines in serum in relation to future non-Hodgkin lymphoma risk: Evidence for associations by histologic subtype. Int. J. Cancer 2014, 135, 913–922. [Google Scholar] [CrossRef]
  49. Hosgood, H.D.; Au, W.-Y.; Kim, H.N.; Liu, J.; Hu, W.; Tse, J.; Song, B.; Wong, K.-F.; Lee, J.-J.; Chanock, S.J.; et al. IL10 and TNF variants and risk of non-Hodgkin lymphoma among three Asian populations. Int. J. Hematol. 2013, 97, 793–799. [Google Scholar] [CrossRef]
  50. Breen, E.C.; Boscardin, W.J.; Detels, R.; Jacobson, L.P.; Smith, M.W.; O’Brien, S.J.; Chmiel, J.S.; Rinaldo, C.R.; Lai, S.; Martínez-Maza, O. Non-Hodgkin’s B cell lymphoma in persons with acquired immunodeficiency syndrome is associated with increased serum levels of IL10, or the IL10 promoter-592 C/C genotype. Clin. Immunol. 2003, 109, 119–129. [Google Scholar] [CrossRef]
  51. Wong, H.-L.; Breen, E.C.; Pfeiffer, R.M.; Aissani, B.; Martinson, J.J.; Margolick, J.B.; Kaslow, R.A.; Jacobson, L.P.; Ambinder, R.F.; Chanock, S.; et al. Cytokine signaling pathway polymorphisms and AIDS-related non-Hodgkin lymphoma risk in the multicenter AIDS cohort study. AIDS 2010, 24, 1025–1033. [Google Scholar] [CrossRef]
  52. Breen, E.C.; Hussain, S.K.; Magpantay, L.; Jacobson, L.P.; Detels, R.; Rabkin, C.S.; Kaslow, R.A.; Variakojis, D.; Bream, J.H.; Rinaldo, C.R.; et al. B-cell stimulatory cytokines and markers of immune activation are elevated several years prior to the diagnosis of systemic AIDS-associated non-Hodgkin B-cell lymphoma. Cancer Epidemiol. Biomark. Prev. 2011, 20, 1303–1314. [Google Scholar] [CrossRef]
  53. Hanna, Z.; Kay, D.G.; Cool, M.; Jothy, S.; Rebai, N.; Jolicoeur, P. Transgenic mice expressing human immunodeficiency virus type 1 in immune cells develop a severe AIDS-like disease. J. Virol. 1998, 72, 121–132. [Google Scholar] [CrossRef] [PubMed]
  54. Curreli, S.; Krishnan, S.; Reitz, M.; Lunardi-Iskandar, Y.; Lafferty, M.K.; Garzino-Demo, A.; Zella, D.; Gallo, R.C.; Bryant, J. B cell lymphoma in HIV transgenic mice. Retrovirology 2013, 10, 92. [Google Scholar] [CrossRef] [PubMed]
  55. Vendrame, E.; Hussain, S.K.; Breen, E.C.; Magpantay, L.I.; Widney, D.P.; Jacobson, L.P.; Variakojis, D.; Knowlton, E.R.; Bream, J.H.; Ambinder, R.F.; et al. Serum levels of cytokines and biomarkers for inflammation and immune activation, and HIV-associated non-Hodgkin B-cell lymphoma risk. Cancer Epidemiol. Biomark. Prev. 2014, 23, 343–349. [Google Scholar] [CrossRef]
  56. Hussain, S.K.; Zhu, W.; Chang, S.-C.; Breen, E.C.; Vendrame, E.; Magpantay, L.; Widney, D.; Conn, D.; Sehl, M.; Jacobson, L.P.; et al. Serum levels of the chemokine CXCL13, genetic variation in CXCL13 and its receptor CXCR5, and HIV-associated non-hodgkin B-cell lymphoma risk. Cancer Epidemiol. Biomark. Prev. 2013, 22, 295–307. [Google Scholar] [CrossRef]
  57. Nolen, B.M.; Breen, E.C.; Bream, J.H.; Jenkins, F.J.; Kingsley, L.A.; Rinaldo, C.R.; Lokshin, A.E. Circulating mediators of inflammation and immune activation in AIDS-related non-hodgkin lymphoma. PLoS ONE 2014, 9, e99144. [Google Scholar] [CrossRef]
  58. Lievin, R.; Hendel-Chavez, H.; Baldé, A.; Lancar, R.; Algarte-Génin, M.; Krzysiek, R.; Costagliola, D.; As-soumou, L.; Taoufik, Y.; Besson, C. Increased Production of B-Cell Activating Cytokines and Altered Peripheral B-Cell Subset Distribution during HIV-Related Classical Hodgkin Lymphoma. Cancers 2021, 14, 128. [Google Scholar] [CrossRef]
  59. Epeldegui, M.; Lee, J.Y.; Martínez, A.C.; Widney, D.P.; Magpantay, L.I.; Regidor, D.; Mitsuyasu, R.; Sparano, J.A.; Ambinder, R.F.; Martínez-Maza, O. Predictive Value of Cytokines and Immune Activation Biomarkers in AIDS-Related Non-Hodgkin Lymphoma Treated with Rituximab plus Infusional EPOCH (AMC-034 trial). Clin. Cancer Res. 2016, 22, 328–336. [Google Scholar] [CrossRef]
  60. Martínez, L.E.; Lensing, S.; Chang, D.; Magpantay, L.I.; Mitsuyasu, R.; Ambinder, R.F.; Sparano, J.A.; Mar-tínez-Maza, O.; Epeldegui, M. Immune Activation and Microbial Translocation as Prognostic Biomarkers for AIDS-Related Non-Hodgkin Lymphoma in the AMC-034 Study. Clin. Cancer Res. 2021, 27, 4642–4651. [Google Scholar] [CrossRef]
  61. Vaughan, J.; Patel, M.; Suchard, M.; Gededzha, M.; Ranchod, H.; Howard, W.; Snyman, T.; Wiggill, T. Derangements of immunological proteins in HIV-associated diffuse large B-cell lymphoma: The frequency and prognostic impact. Front. Cell Infect. Microbiol. 2024, 14, 1340096. [Google Scholar] [CrossRef]
  62. Bortolin, M.T.; Tedeschi, R.; Bidoli, E.; Zanussi, S.; Pratesi, C.; Vaccher, E.; Tirelli, U.; De Paoli, P. Multiplex analysis of blood cytokines as a prognostic tool in HIV related non-Hodgkin lymphoma patients: A potential role of interleukin-7. Cytokine 2012, 60, 90–95. [Google Scholar] [CrossRef]
  63. Zlotnik, A.; Yoshie, O. Chemokines: A new classification system and their role in immunity. Immunity 2000, 12, 121–127. [Google Scholar] [CrossRef]
  64. Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef]
  65. Luster, A.D. Chemokines—Chemotactic cytokines that mediate inflammation. N. Engl. J. Med. 1998, 338, 436–445. [Google Scholar] [CrossRef]
  66. Balkwill, F. Cancer and the chemokine network. Nat. Rev. Cancer. 2004, 4, 540–550. [Google Scholar] [CrossRef]
  67. Hussain, M.; Adah, D.; Tariq, M.; Lu, Y.; Zhang, J.; Liu, J. CXCL13/CXCR5 signaling axis in cancer. Life Sci. 2019, 227, 175–186. [Google Scholar] [CrossRef]
  68. Gunn, M.D.; Ngo, V.N.; Ansel, K.M.; Ekland, E.H.; Cyster, J.G.; Williams, L.T. A B-cell-homing chemokine made in lymphoid follicles activates Burkitt’s lymphoma receptor-1. Nature 1998, 391, 799–803. [Google Scholar] [CrossRef]
  69. Gao, S.-H.; Liu, S.-Z.; Wang, G.-Z.; Zhou, G.-B. CXCL13 in Cancer and Other Diseases: Biological Functions, Clinical Significance, and Therapeutic Opportunities. Life 2021, 11, 1282. [Google Scholar] [CrossRef]
  70. Purdue, M.P.; Hofmann, J.N.; Kemp, T.J.; Chaturvedi, A.K.; Lan, Q.; Park, J.-H.; Pfeiffer, R.M.; Hildesheim, A.; Pinto, L.A.; Rothman, N. A prospective study of 67 serum immune and inflammation markers and risk of non-Hodgkin lymphoma. Blood 2013, 122, 951–957. [Google Scholar] [CrossRef]
  71. Trentin, L.; Cabrelle, A.; Facco, M.; Carollo, D.; Miorin, M.; Tosoni, A.; Pizzo, P.; Binotto, G.; Nicolardi, L.; Zambello, R.; et al. Homeostatic chemokines drive migration of malignant B cells in patients with non-Hodgkin lymphomas. Blood 2004, 104, 502–508. [Google Scholar] [CrossRef]
  72. Rubenstein, J.L.; Wong, V.S.; Kadoch, C.; Gao, H.-X.; Barajas, R.; Chen, L.; Josephson, S.A.; Scott, B.; Douglas, V.; Maiti, M.; et al. CXCL13 plus interleukin 10 is highly specific for the diagnosis of CNS lymphoma. Blood 2013, 121, 4740–4748. [Google Scholar] [CrossRef]
  73. Kurtova, A.V.; Tamayo, A.T.; Ford, R.J.; Burger, J.A. Mantle cell lymphoma cells express high levels of CXCR4, CXCR5, and VLA-4 (CD49d): Importance for interactions with the stromal microenvironment and specific targeting. Blood 2009, 113, 4604–4613. [Google Scholar] [CrossRef]
  74. Kim, S.J.; Ryu, K.J.; Hong, M.; Ko, Y.H.; Kim, W.S. The serum CXCL13 level is associated with the Glasgow Prognostic Score in extranodal NK/T-cell lymphoma patients. J. Hematol. Oncol. 2015, 8, 49. [Google Scholar] [CrossRef]
  75. Charbonneau, B.; Wang, A.H.; Maurer, M.J.; Asmann, Y.W.; Zent, C.S.; Link, B.K.; Ansell, S.M.; Weiner, G.J.; Ozsan, N.; Feldman, A.L.; et al. CXCR5 polymorphisms in non-Hodgkin lymphoma risk and prognosis. Cancer Immunol. Immunother. 2013, 62, 1475–1484. [Google Scholar] [CrossRef]
  76. Förster, R.; Schweigard, G.; Johann, S.; Emrich, T.; Kremmer, E.; Nerl, C.; Lipp, M. Abnormal expression of the B-cell homing chemokine receptor BLR1 during the progression of acquired immunodeficiency syndrome. Blood 1997, 90, 520–525. [Google Scholar] [CrossRef]
  77. Cagigi, A.; Mowafi, F.; Phuong Dang, L.V.; Tenner-Racz, K.; Atlas, A.; Grutzmeier, S.; Racz, P.; Chiodi, F.; Nilsson, A. Altered expression of the receptor-ligand pair CXCR5/CXCL13 in B cells during chronic HIV-1 infection. Blood 2008, 112, 4401–4410. [Google Scholar] [CrossRef]
  78. Hussain, S.K.; Hessol, N.A.; Levine, A.M.; Breen, E.C.; Anastos, K.; Cohen, M.; D’Souza, G.; Gustafson, D.R.; Silver, S.; Martínez-Maza, O. Serum biomarkers of immune activation and subsequent risk of non-hodgkin B-cell lymphoma among HIV-infected women. Cancer Epidemiol. Biomark. Prev. 2013, 22, 2084–2093. [Google Scholar] [CrossRef]
  79. Widney, D.P.; Gui, D.; Popoviciu, L.M.; Said, J.W.; Breen, E.C.; Huang, X.; Kitchen, C.M.R.; Alcantar, J.M.; Smith, J.B.; Detels, R.; et al. Expression and Function of the Chemokine, CXCL13, and Its Receptor, CXCR5, in Aids-Associated Non-Hodgkin’s Lymphoma. AIDS Res. Treat. 2010, 2010, 164586. [Google Scholar] [CrossRef]
  80. Valverde-Villegas, J.M.; de Medeiros, R.M.; Ellwanger, J.H.; Santos, B.R.; de Melo, M.G.; de Matos Almeida, S.E.; Chies, J.A.B. High CXCL10/IP-10 levels are a hallmark in the clinical evolution of the HIV infection. Infect. Genet. Evol. 2018, 57, 51–58. [Google Scholar] [CrossRef]
  81. Liovat, A.-S.; Rey-Cuillé, M.-A.; Lécuroux, C.; Jacquelin, B.; Girault, I.; Petitjean, G.; Zitoun, Y.; Venet, A.; Bar-ré-Sinoussi, F.; Lebon, P.; et al. Acute plasma biomarkers of T cell activation set-point levels and of disease progression in HIV-1 infection. PLoS ONE 2012, 7, e46143. [Google Scholar] [CrossRef]
  82. Ploquin, M.J.; Madec, Y.; Casrouge, A.; Huot, N.; Passaes, C.; Lécuroux, C.; Essat, A.; Boufassa, F.; Jacquelin, B.; Jochems, S.P.; et al. Elevated Basal Pre-infection CXCL10 in Plasma and in the Small Intestine after Infection Are Associated with More Rapid HIV/SIV Disease Onset. PLoS Pathog. 2016, 12, e1005774. [Google Scholar] [CrossRef]
  83. Lee, S.; Chung, Y.-S.; Yoon, C.-H.; Shin, Y.; Kim, S.; Choi, B.-S.; Kim, S.S. Interferon-inducible protein 10 (IP-10) is associated with viremia of early HIV-1 infection in Korean patients. J. Med. Virol. 2015, 87, 782–789. [Google Scholar] [CrossRef]
  84. Ouedraogo, D.E.; Makinson, A.; Kuster, N.; Nagot, N.; Rubbo, P.-A.; Bollore, K.; Foulongne, V.; Cartron, G.; Olive, D.; Reynes, J.; et al. Increased T-cell activation and Th1 cytokine concentrations prior to the diagnosis of B-cell lymphoma in HIV infected patients. J. Clin. Immunol. 2013, 33, 22–29. [Google Scholar] [CrossRef]
  85. Li, Y.-L.; Shi, Z.-H.; Wang, X.; Gu, K.-S.; Zhai, Z.-M. Prognostic significance of monocyte chemoattractant protein-1 and CC chemokine receptor 2 in diffuse large B cell lymphoma. Ann. Hematol. 2019, 98, 413–422. [Google Scholar] [CrossRef]
  86. Delespesse, G.; Suter, U.; Mossalayi, D.; Bettler, B.; Sarfati, M.; Hofstetter, H.; Kilcherr, E.; Debre, P.; Dalloul, A. Expression, structure, and function of the CD23 antigen. Adv. Immunol. 1991, 49, 149–191. [Google Scholar]
  87. Bonnefoy, J.Y.; Lecoanet-Henchoz, S.; Aubry, J.P.; Gauchat, J.F.; Graber, P. CD23 and B-cell activation. Curr. Opin. Immunol. 1995, 7, 355–359. [Google Scholar] [CrossRef]
  88. De Roos, A.J.; Mirick, D.K.; Edlefsen, K.L.; LaCroix, A.Z.; Kopecky, K.J.; Madeleine, M.M.; Magpantay, L.; Martínez-Maza, O. Markers of B-cell activation in relation to risk of non-Hodgkin lymphoma. Cancer Res. 2012, 72, 4733–4743. [Google Scholar] [CrossRef] [PubMed]
  89. Reinisch, W.; Willheim, M.; Hilgarth, M.; Gasché, C.; Mader, R.; Szepfalusi, S.; Steger, G.; Berger, R.; Lechner, K.; Boltz-Nitulescu, G. Soluble CD23 reliably reflects disease activity in B-cell chronic lymphocytic leukemia. J. Clin. Oncol. 1994, 12, 2146–2152. [Google Scholar] [CrossRef] [PubMed]
  90. Beguin, Y.; Lampertz, S.; De Groote, D.; Igot, D.; Malaise, M.; Fillet, G. Soluble CD23 and other receptors (CD4, CD8, CD25, CD71) in serum of patients with chronic lymphocytic leukemia. Leukemia 1993, 7, 2019–2025. [Google Scholar]
  91. Camerini, D.; Walz, G.; Loenen, W.A.; Borst, J.; Seed, B. The T cell activation antigen CD27 is a member of the nerve growth factor/tumor necrosis factor receptor gene family. J. Immunol. 1991, 147, 3165–3169. [Google Scholar] [CrossRef] [PubMed]
  92. Jacquot, S. CD27/CD70 interactions regulate T dependent B cell differentiation. Immunol. Res. 2000, 21, 23–30. [Google Scholar] [CrossRef]
  93. Del Prete, G.; De Carli, M.; Almerigogna, F.; Daniel, C.K.; D’Elios, M.M.; Zancuoghi, G.; Vinante, F.; Pizzolo, G.; Romagnani, S. Preferential expression of CD30 by human CD4+ T cells producing Th2-type cytokines. FASEB J. 1995, 9, 81–86. [Google Scholar] [CrossRef]
  94. Dang, L.V.P.; Nilsson, A.; Ingelman-Sundberg, H.; Cagigi, A.; Gelinck, L.B.S.; Titanji, K.; De Milito, A.; Grutzmeier, S.; Hedlund, J.; Kroon, F.P.; et al. Soluble CD27 induces IgG production through activation of antigen-primed B cells. J. Intern. Med. 2012, 271, 282–293. [Google Scholar] [CrossRef] [PubMed]
  95. De Milito, A.; Aleman, S.; Marenzi, R.; Sonnerborg, A.; Fuchs, D.; Zazzi, M.; Chiodi, F. Plasma levels of soluble CD27: A simple marker to monitor immune activation during potent antiretroviral therapy in HIV-1-infected subjects. Clin. Exp. Immunol. 2002, 127, 486–494. [Google Scholar] [CrossRef]
  96. Caligaris-Cappio, F.; Bertero, M.T.; Converso, M.; Stacchini, A.; Vinante, F.; Romagnani, S.; Pizzolo, G. Circulating levels of soluble CD30, a marker of cells producing Th2-type cytokines, are increased in patients with systemic lupus erythematosus and correlate with disease activity. Clin. Exp. Rheumatol. 1995, 13, 339–343. [Google Scholar]
  97. Bohnhorst, J.Ø.; Bjørgan, M.B.; Thoen, J.E.; Jonsson, R.; Natvig, J.B.; Thompson, K.M. Abnormal B cell differentiation in primary Sjögren’s syndrome results in a depressed percentage of circulating memory B cells and elevated levels of soluble CD27 that correlate with Serum IgG concentration. Clin. Immunol. 2002, 103, 79–88. [Google Scholar] [CrossRef]
  98. Yawetz, S.; Cumberland, W.G.; van der Meyden, M.; Martínez-Maza, O. Elevated serum levels of soluble CD23 (sCD23) precede the appearance ofacquired immunodeficiency syndrome--associated non-Hodgkin’s lymphoma. Blood 1995, 85, 1843–1849. [Google Scholar] [CrossRef]
  99. Widney, D.; Gundapp, G.; Said, J.W.; van der Meijden, M.; Bonavida, B.; Demidem, A.; Trevisan, C.; Taylor, J.; Detels, R.; Martínez-Maza, O. Aberrant expression of CD27 and soluble CD27 (sCD27) in HIV infection and in AIDS-associated lymphoma. Clin. Immunol. 1999, 93, 114–123. [Google Scholar] [CrossRef]
  100. Breen, E.C.; Fatahi, S.; Epeldegui, M.; Boscardin, W.J.; Detels, R.; Martínez-Maza, O. Elevated serum soluble CD30 precedes the development of AIDS-associated non-Hodgkin’s B cell lymphoma. Tumour Biol. 2006, 27, 187–194. [Google Scholar] [CrossRef]
  101. Bossolasco, S.; Nilsson, A.; de Milito, A.; Lazzarin, A.; Linde, A.; Cinque, P.; Chiodi, F. Soluble CD23 in cerebrospinal fluid: A marker of AIDS-related non-Hodgkin’s lymphoma in the brain. AIDS 2001, 15, 1109–1113. [Google Scholar] [CrossRef]
  102. Plevriti, A.; Lamprou, M.; Mourkogianni, E.; Skoulas, N.; Giannakopoulou, M.; Sajib, M.S.; Wang, Z.; Matthe-olabakis, G.; Chatzigeorgiou, A.; Marazioti, A.; et al. The Role of Soluble CD163 (sCD163) in Human Physiology and Pathophysiology. Cells 2024, 13, 1679. [Google Scholar] [CrossRef]
  103. Wang, Y.; Yang, J.; Wen, Y. Lessons from Epstein-Barr virus DNA detection in cerebrospinal fluid as a diagnostic tool for EBV-induced central nervous system dysfunction among HIV-positive patients. Biomed. Pharmacother. 2022, 145, 112392. [Google Scholar] [CrossRef]
  104. Jones, K.; Vari, F.; Keane, C.; Crooks, P.; Nourse, J.P.; Seymour, L.A.; Gottlieb, D.; Ritchie, D.; Gill, D.; Gandhi, M.K. Serum CD163 and TARC as disease response biomarkers in classical Hodgkin lymphoma. Clin. Cancer Res. 2013, 19, 731–742. [Google Scholar] [CrossRef]
  105. Scott, N.A.; Andrusaite, A.; Andersen, P.; Lawson, M.; Alcon-Giner, C.; Leclaire, C.; Caim, S.; Le Gall, G.; Shaw, T.; Connolly, J.P.R.; et al. Antibiotics induce sustained dysregulation of intestinal T cell immunity by perturbing macrophage homeostasis. Sci. Transl. Med. 2018, 10, eaao4755. [Google Scholar] [CrossRef]
  106. Mudd, J.C.; Brenchley, J.M. Gut Mucosal Barrier Dysfunction, Microbial Dysbiosis, and Their Role in HIV-1 Disease Progression. J. Infect. Dis. 2016, 214 (Suppl. 2), S58–S66. [Google Scholar] [CrossRef]
  107. Marchetti, G.; Tincati, C.; Silvestri, G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin. Microbiol. Rev. 2013, 26, 2–18. [Google Scholar] [CrossRef]
  108. Sandler, N.G.; Douek, D.C. Microbial translocation in HIV infection: Causes, consequences and treatment opportunities. Nat. Rev. Microbiol. 2012, 10, 655–666. [Google Scholar] [CrossRef]
  109. Klatt, N.R.; Funderburg, N.T.; Brenchley, J.M. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013, 21, 6–13. [Google Scholar] [CrossRef]
  110. Brenchley, J.M.; Price, D.A.; Schacker, T.W.; Asher, T.E.; Silvestri, G.; Rao, S.; Kazzaz, Z.; Bornstein, E.; Lam-botte, O.; Altmann, D.; et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 2006, 12, 1365–1371. [Google Scholar] [CrossRef]
  111. Shive, C.L.; Jiang, W.; Anthony, D.D.; Lederman, M.M. Soluble CD14 is a nonspecific marker of monocyte activation. AIDS 2015, 29, 1263–1265. [Google Scholar] [CrossRef]
  112. Wright, S.D.; Ramos, R.A.; Tobias, P.S.; Ulevitch, R.J.; Mathison, J.C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249, 1431–1433. [Google Scholar] [CrossRef]
  113. Zanoni, I.; Granucci, F. Role of CD14 in host protection against infections and in metabolism regulation. Front. Cell Infect. Microbiol. 2013, 3, 32. [Google Scholar] [CrossRef]
  114. Funaoka, H.; Kanda, T.; Fujii, H. Intestinal fatty acid-binding protein (I-FABP) as a new biomarker for intestinal diseases. Rinsho Byori. 2010, 58, 162–168. [Google Scholar]
  115. Pelsers, M.M.A.L.; Namiot, Z.; Kisielewski, W.; Namiot, A.; Januszkiewicz, M.; Hermens, W.T.; Glatz, J.F.C. Intestinal-type and liver-type fatty acid-binding protein in the intestine. Tissue distribution and clinical utility. Clin. Biochem. 2003, 36, 529–535. [Google Scholar] [CrossRef]
  116. Uddin, M.I.; Hossain, M.; Islam, S.; Akter, A.; Nishat, N.S.; Nila, T.A.; Rafique, T.A.; Leung, D.T.; Calderwood, S.B.; Ryan, E.T.; et al. An assessment of potential biomarkers of environment enteropathy and its association with age and microbial infections among children in Bangladesh. PLoS ONE 2021, 16, e0250446. [Google Scholar] [CrossRef]
  117. Sandler, N.G.; Wand, H.; Roque, A.; Law, M.; Nason, M.C.; Nixon, D.E.; Pedersen, C.; Ruxrungtham, K.; Lew-in, S.R.; Emery, S.; et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J. Infect. Dis. 2011, 203, 780–790. [Google Scholar] [CrossRef]
  118. Papasavvas, E.; Pistilli, M.; Reynolds, G.; Bucki, R.; Azzoni, L.; Chehimi, J.; Janmey, P.A.; DiNubile, M.J.; Ondercin, J.; Kostman, J.R.; et al. Delayed loss of control of plasma lipopolysaccharide levels after therapy interruption in chronically HIV-1-infected patients. AIDS 2009, 23, 369–375. [Google Scholar] [CrossRef]
  119. Negi, N.; Singh, R.; Sharma, A.; Das, B.K.; Vajpayee, M. Comparative evaluation of microbial translocation products (LPS, sCD14, IgM Endocab) in HIV-1 infected Indian individuals. Microb. Pathog. 2017, 111, 331–337. [Google Scholar] [CrossRef]
  120. Schroeder, H.W.; Cavacini, L. Structure and function of immunoglobulins. J. Allergy Clin. Immunol. 2010, 125, S41–S52. [Google Scholar] [CrossRef]
  121. Guevara-Hoyer, K.; Ochoa-Grullón, J.; Fernández-Arquero, M.; Cárdenas, M.; Pérez de Diego, R.; Sánchez-Ramón, S. Serum Free Immunoglobulins Light Chains: A Common Feature of Common Variable Immunodeficiency? Front. Immunol. 2020, 11, 2004. [Google Scholar] [CrossRef]
  122. Morabito, F.; De Filippi, R.; Laurenti, L.; Zirlik, K.; Recchia, A.G.; Gentile, M.; Morelli, E.; Vigna, E.; Gigliotti, V.; Calemma, R.; et al. The cumulative amount of serum-free light chain is a strong prognosticator in chronic lymphocytic leukemia. Blood 2011, 118, 6353–6361. [Google Scholar] [CrossRef]
  123. Witzig, T.E.; Maurer, M.J.; Habermann, T.M.; Link, B.K.; Micallef, I.N.M.; Nowakowski, G.S.; Ansell, S.M.; Colgan, J.P.; Inwards, D.J.; Porrata, L.F.; et al. Elevated monoclonal and polyclonal serum immunoglobulin free light chain as prognostic factors in B- and T-cell non-Hodgkin lymphoma. Am. J. Hematol. 2014, 89, 1116–1120. [Google Scholar] [CrossRef]
  124. Maurer, M.J.; Micallef, I.N.M.; Cerhan, J.R.; Katzmann, J.A.; Link, B.K.; Colgan, J.P.; Habermann, T.M.; In-wards, D.J.; Markovic, S.N.; Ansell, S.M.; et al. Elevated serum free light chains are associated with event-free and overall survival in two independent cohorts of patients with diffuse large B-cell lymphoma. J. Clin. Oncol. 2011, 29, 1620–1626. [Google Scholar] [CrossRef]
  125. Thompson, C.A.; Maurer, M.J.; Cerhan, J.R.; Katzmann, J.A.; Ansell, S.M.; Habermann, T.M.; Macon, W.R.; Weiner, G.J.; Link, B.K.; Witzig, T.E. Elevated serum free light chains are associated with inferior event free and overall survival in Hodgkin lymphoma. Am. J. Hematol. 2011, 86, 998–1000. [Google Scholar] [CrossRef]
  126. Landgren, O.; Goedert, J.J.; Rabkin, C.S.; Wilson, W.H.; Dunleavy, K.; Kyle, R.A.; Katzmann, J.A.; Rajkumar, S.V.; Engels, E.A. Circulating serum free light chains as predictive markers of AIDS-related lymphoma. J. Clin. Oncol. 2010, 28, 773–779. [Google Scholar] [CrossRef]
  127. Shepherd, L.; Borges, Á.H.; Harvey, R.; Bower, M.; Grulich, A.; Silverberg, M.; Weber, J.; Ristola, M.; Viard, J.-P.; Bogner, J.R.; et al. The extent of B-cell activation and dysfunction preceding lymphoma development in HIV-positive people. HIV Med. 2018, 19, 90–101. [Google Scholar] [CrossRef]
  128. Bibas, M.; Trotta, M.P.; Cozzi-Lepri, A.; Lorenzini, P.; Pinnetti, C.; Rizzardini, G.; Angarano, G.; Caramello, P.; Sighinolfi, L.; Mastroianni, C.M.; et al. Role of serum free light chains in predicting HIV-associated non-Hodgkin lymphoma and Hodgkin’s lymphoma and its correlation with antiretroviral therapy. Am. J. Hematol. 2012, 87, 749–753. [Google Scholar] [CrossRef]
  129. Bibas, M.; Lorenzini, P.; Cozzi-Lepri, A.; Calcagno, A.; di Giambenedetto, S.; Costantini, A.; Castagna, A.; Manfrin, V.; Monforte, A.D.; Antinori, A.; et al. Polyclonal serum-free light chains elevation in HIV-infected patients. AIDS 2012, 26, 2107–2110. [Google Scholar] [CrossRef] [PubMed]
  130. Grulich, A.E.; Wan, X.; Law, M.G.; Milliken, S.T.; Lewis, C.R.; Garsia, R.J.; Gold, J.; Finlayson, R.J.; Cooper, D.A.; Kaldor, J.M. B-cell stimulation and prolonged immune deficiency are risk factors for non-Hodgkin’s lymphoma in people with AIDS. AIDS 2000, 14, 133–140. [Google Scholar] [CrossRef] [PubMed]
  131. Engels, E.A.; Pfeiffer, R.M.; Landgren, O.; Moore, R.D. Immunologic and virologic predictors of AIDS-related non-hodgkin lymphoma in the highly active antiretroviral therapy era. J. Acquir. Immune Defic. Syndr. 2010, 54, 78–84. [Google Scholar] [CrossRef]
  132. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef]
  133. Yi, M.; Zheng, X.; Niu, M.; Zhu, S.; Ge, H.; Wu, K. Combination strategies with PD-1/PD-L1 blockade: Current advances and future directions. Mol. Cancer 2022, 21, 28. [Google Scholar] [CrossRef]
  134. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
  135. Epeldegui, M.; Conti, D.V.; Guo, Y.; Cozen, W.; Penichet, M.L.; Martínez-Maza, O. Elevated numbers of PD-L1 expressing B cells are associated with the development of AIDS-NHL. Sci. Rep. 2019, 9, 9371. [Google Scholar] [CrossRef] [PubMed]
  136. Zhao, H.; Cai, S.; Xiao, Y.; Xia, M.; Chen, H.; Xie, Z.; Tang, X.; He, H.; Peng, J.; Chen, J. Expression and prognostic significance of the PD-1/PD-L1 pathway in AIDS-related non-Hodgkin lymphoma. Cancer Med. 2024, 13, e7195. [Google Scholar] [CrossRef]
  137. Yu, X.; Qian, J.; Ding, L.; Yin, S.; Zhou, L.; Zheng, S. Galectin-1: A Traditionally Immunosuppressive Protein Displays Context-Dependent Capacities. Int. J. Mol. Sci. 2023, 24, 6501. [Google Scholar] [CrossRef]
  138. Cedeno-Laurent, F.; Watanabe, R.; Teague, J.E.; Kupper, T.S.; Clark, R.A.; Dimitroff, C.J. Galectin-1 inhibits the viability, proliferation, and Th1 cytokine production of nonmalignant T cells in patients with leukemic cutaneous T-cell lymphoma. Blood 2012, 119, 3534–3538. [Google Scholar] [CrossRef] [PubMed]
  139. Vase, M.Ø.; Ludvigsen, M.; Bendix, K.; Dutoit, S.H.; Hjortebjerg, R.; Petruskevicius, I.; Møller, M.B.; Pedersen, G.; Denton, P.W.; Honoré, B.; et al. Predictive value of galectin-1 in the development and progression of HIV-associated lymphoma. AIDS 2017, 31, 2311–2313. [Google Scholar] [CrossRef]
  140. Peterson, P.A.; Cunningham, B.A.; Berggård, I.; Edelman, G.M. 2-Microglobulin—A free immunoglobulin domain. Proc. Natl. Acad. Sci. USA 1972, 69, 1697–1701. [Google Scholar] [CrossRef]
  141. Shi, C.; Zhu, Y.; Su, Y.; Chung, L.W.K.; Cheng, T. Beta2-microglobulin: Emerging as a promising cancer therapeutic target. Drug Discov. Today 2009, 14, 25–30. [Google Scholar] [CrossRef]
  142. Zhuang, K.; Zhang, Y.; Mo, P.; Deng, L.; Jiang, Y.; Yu, L.; Mei, F.; Huang, S.; Chen, X.; Yan, Y.; et al. Plasma proteomic analysis reveals altered protein abundances in HIV-infected patients with or without non-Hodgkin lymphoma. J. Med. Virol. 2022, 94, 3876–3889. [Google Scholar] [CrossRef]
  143. Vargas, J.C.; de Marques, M.O.; Pereira, J.; Braga, W.M.T.; Hamerschlak, N.; Tabacof, J.; Ferreira, P.R.A.; Col-leoni, G.W.B.; Baiocchi, O.C.G. Factors associated with survival in patients with lymphoma and HIV. AIDS 2023, 37, 1217–1226. [Google Scholar] [CrossRef]
  144. Botafogo, V.; Usas, A.; Verdú-Bou, M.; Moreno, M.; Fernández-Rivas, G.; Cardona, P.-J.; Fondelli, F.; Mar-tínez-Cáceres, E.; Doladé, M.; Blanco, I.; et al. Profile of Inflammatory Biomarkers in Hodgkin Lymphoma. Differences between HIV-Negative and HIV-Positive Patients. Blood 2024, 144, 6249. [Google Scholar] [CrossRef]
  145. Szymulewska-Konopko, K.; Reszeć-Giełażyn, J.; Małeczek, M. Ferritin as an Effective Prognostic Factor and Potential Cancer Biomarker. Curr. Issues Mol. Biol. 2025, 47, 60. [Google Scholar] [CrossRef] [PubMed]
  146. Matzner, Y.; Konijn, A.M.; Hershko, C. Serum ferritin in hematologic malignancies. Am. J. Hematol. 1980, 9, 13–22. [Google Scholar] [CrossRef] [PubMed]
  147. Kim, D.J.; Kim, T.; Jeong, J.-Y.; Jo, J.-C.; Lee, W.S.; Shin, H.-J.; Lee, J.H.; Lee, H.S. Poor prognostic impact of high serum ferritin levels in patients with a lower risk of diffuse large B cell lymphoma. Int. J. Hematol. 2020, 111, 559–566. [Google Scholar] [CrossRef]
  148. Sehn, L.H.; Salles, G. Diffuse Large B-Cell Lymphoma. N. Engl. J. Med. 2021, 384, 842–858. [Google Scholar] [CrossRef]
  149. Shen, Z.; Zhang, S.; Zhang, M.; Hu, L.; Sun, Q.; He, C.; Yan, D.; Ye, J.; Zhang, H.; Wang, L.; et al. The Addition of Ferritin Enhanced the Prognostic Value of International Prognostic Index in Diffuse Large B-Cell Lymphoma. Front. Oncol. 2021, 11, 823079. [Google Scholar] [CrossRef]
  150. Vaughan, J.; Wiggill, T.; Mia, Z.; Patel, M. Tumour-associated macrophages in diffuse large B-cell lymphoma: The prognostic and therapeutic impact in a South African centre with high HIV seroprevalence. Immunol. Res. 2024, 72, 1393–1403. [Google Scholar] [CrossRef]
  151. Claps, G.; Faouzi, S.; Quidville, V.; Chehade, F.; Shen, S.; Vagner, S.; Robert, C. The multiple roles of LDH in cancer. Nat. Rev. Clin. Oncol. 2022, 19, 749–762. [Google Scholar] [CrossRef]
  152. Ji, H.; Niu, X.; Yin, L.; Wang, Y.; Huang, L.; Xuan, Q.; Li, L.; Zhang, H.; Li, J.; Yang, Y.; et al. Ratio of Immune Response to Tumor Burden Predicts Survival Via Regulating Functions of Lymphocytes and Monocytes in Diffuse Large B-Cell Lymphoma. Cell Physiol. Biochem. 2018, 45, 951–961. [Google Scholar] [CrossRef]
  153. Rusconi, C.; Re, A.; Bandiera, L.; Crucitti, L.; Spina, M.; Gini, G.; Paulli, M.; Lucioni, M.; Facchetti, F.; Goteri, G.; et al. Cell-of-Origin Identification and Prognostic Correlation in HIV-Associated Diffuse Large B-Cell Lymphomas: Results of an Italian Multicentric Study. Blood 2018, 132, 5294. [Google Scholar] [CrossRef]
  154. Wang, C.; Liu, J.; Lei, H.; Li, Y.; Wu, J.; Guo, B.; Hu, R.; Liu, T.; Wu, J.; Ding, Y.; et al. Clinical characteristics and outcomes of newly diagnosed patients with HIV-associated aggressive B-cell NHL in China. J. Cell Mol. Med. 2022, 26, 5067–5077. [Google Scholar] [CrossRef]
  155. Shen, Y.; Zhang, R.; Liu, L.; Shen, Y.; Song, W.; Qi, T.; Tang, Y.; Wang, Z.; Guan, L.; Lu, H. Clinical and prognostic analysis of 78 patients with human immuno-deficiency virus associated non-Hodgkin’s lymphoma in Chinese population. Infect. Agent. Cancer 2017, 12, 7. [Google Scholar] [CrossRef]
  156. Painschab, M.S.; Kasonkanji, E.; Zuze, T.; Kaimila, B.; Tomoka, T.; Nyasosela, R.; Nyirenda, R.; Dhungel, B.M.; Mulenga, M.; Chikasema, M.; et al. Mature outcomes and prognostic indices in diffuse large B-cell lymphoma in Malawi: A prospective cohort. Br. J. Haematol. 2019, 184, 364–372. [Google Scholar] [CrossRef]
  157. Wang, C.; Wu, Y.; Liu, J.; Min, H.; Huang, Y.; Wei, G.; Zhang, W.; Wang, M.; Tang, X.; Zhou, H.; et al. Impact of initial chemotherapy cycles and clinical characteristics on outcomes for HIV-associated diffuse large B cell lymphoma patients: The Central and Western China AIDS Lymphoma League 001 study (CALL-001 study). Front. Immunol. 2023, 14, 1153790. [Google Scholar]
  158. Zhao, H.; Liu, R.; Tao, Y.; Bertero, L.; Feng, L.; Liu, B.; Chen, Z.; Guan, J.; Liao, B.; Li, L.; et al. Construction and validation of prognostic scoring models to risk stratify patients with acquired immune deficiency syndrome-related diffuse large B cell lymphoma. J. Med. Virol. 2023, 95, e28821. [Google Scholar] [CrossRef] [PubMed]
  159. Wu, D.; Chen, C.; Zhang, M.; Li, Z.; Wang, S.; Shi, J.; Zhang, Y.; Yao, D.; Hu, S. The clinical features and prognosis of 100 AIDS-related lymphoma cases. Sci. Rep. 2019, 9, 5381. [Google Scholar] [CrossRef]
  160. Alderuccio, J.P.; Olszewski, A.J.; Evens, A.M.; Collins, G.P.; Danilov, A.V.; Bower, M.; Jagadeesh, D.; Zhu, C.; Sperling, A.; Kim, S.-H.; et al. HIV-associated Burkitt lymphoma: Outcomes from a US-UK collaborative analysis. Blood Adv. 2021, 5, 2852–2862. [Google Scholar] [CrossRef]
  161. Liu, R.; Zhao, H.; Xiao, G.; Tao, Y.; Tang, X.; Feng, L.; Liao, B.; Liu, B.; Guan, J.; Li, L.; et al. Clinical Characteristics and Outcomes of AIDS-Related Burkitt Lymphoma in China: A Retrospective Single-Center Study. Technol. Cancer Res. Treat. 2024, 23, 15330338231214236. [Google Scholar] [CrossRef]
  162. Mai, B.; Wang, W.; Lin, M.; Hu, S.; Wang, X.I.; Chen, L.; Wahed, A.; Nguyen, A.; Ma, H.Y.; Medeiros, L.J.; et al. HIV-associated plasmablastic lymphoma in the era of HAART: A single-center experience of 21 patients. AIDS 2020, 34, 1735–1743. [Google Scholar] [CrossRef]
  163. Li, M.; Chen, J.; Wang, P.; Feng, E.; Chen, X.; Gao, H.; Ma, Z.; Zhou, X. Clinicopathological analysis of primary central nervous system lymphoma in patients with or without HIV infection. Ann. Diagn. Pathol. 2024, 73, 152383. [Google Scholar] [CrossRef]
  164. Liang, Y.; Chang, J.; Gao, Y.; Zhang, L.; Chen, X.; Zheng, C.; Sun, Y.; Zhang, X.; Guo, C.; Zhang, Y. Development of a novel prognostic nomogram for AIDS-associated diffuse large B-cell lymphoma: A retrospective study from northern China. Clin. Exp. Med. 2025, 25, 62. [Google Scholar] [CrossRef] [PubMed]
  165. Stein, D.R.; Burgener, A.; Ball, T.B. Proteomics as a novel HIV immune monitoring tool. Curr. Opin. HIV AIDS 2013, 8, 140–146. [Google Scholar] [CrossRef]
  166. Abu Sabaa, A.; Shen, Q.; Lennmyr, E.B.; Enblad, A.P.; Gammelgård, G.; Molin, D.; Hein, A.; Freyhult, E.; Ka-mali-Moghaddam, M.; Höglund, M.; et al. Plasma protein biomarker profiling reveals major differences between acute leukaemia, lymphoma patients and controls. New Biotechnol. 2022, 71, 21–29. [Google Scholar] [CrossRef] [PubMed]
  167. Varnum, S.M.; Webb-Robertson, B.-J.M.; Hessol, N.A.; Smith, R.D.; Zangar, R.C. Plasma biomarkers for detecting Hodgkin’s lymphoma in HIV patients. PLoS ONE 2011, 6, e29263. [Google Scholar] [CrossRef] [PubMed]
  168. Vase, M.Ø.; Ludvigsen, M.; Bendix, K.; Hamilton-Dutoit, S.; Mller, M.B.; Pedersen, C.; Pedersen, G.; Obel, N.; Larsen, C.S.; d’Amore, F.; et al. Proteomic profiling of pretreatment serum from HIV-infected patients identifies candidate markers predictive of lymphoma development. AIDS 2016, 30, 1889–1898. [Google Scholar] [CrossRef]
  169. Lu, T.X.; Rothenberg, M.E. MicroRNA. J. Allergy Clin. Immunol. 2018, 141, 1202–1207. [Google Scholar] [CrossRef]
  170. Krol, J.; Loedige, I.; Filipowicz, W. The widespread regulation of microRNA biogenesis, function and decay. Nat. Rev. Genet. 2010, 11, 597–610. [Google Scholar] [CrossRef]
  171. Baek, D.; Villén, J.; Shin, C.; Camargo, F.D.; Gygi, S.P.; Bartel, D.P. The impact of microRNAs on protein output. Nature 2008, 455, 64–71. [Google Scholar] [CrossRef]
  172. He, Y.; Lin, J.; Kong, D.; Huang, M.; Xu, C.; Kim, T.-K.; Etheridge, A.; Luo, Y.; Ding, Y.; Wang, K. Current State of Circulating MicroRNAs as Cancer Biomarkers. Clin. Chem. 2015, 61, 1138–1155. [Google Scholar] [CrossRef] [PubMed]
  173. Ho, P.T.B.; Clark, I.M.; Le, L.T.T. MicroRNA-Based Diagnosis and Therapy. Int. J. Mol. Sci. 2022, 23, 7167. [Google Scholar] [CrossRef]
  174. Kosaka, N.; Iguchi, H.; Ochiya, T. Circulating microRNA in body fluid: A new potential biomarker for cancer diagnosis and prognosis. Cancer Sci. 2010, 101, 2087–2092. [Google Scholar] [CrossRef]
  175. Croce, C.M. Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet. 2009, 10, 704–714. [Google Scholar] [CrossRef]
  176. Shen, J.; Stass, S.A.; Jiang, F. MicroRNAs as potential biomarkers in human solid tumors. Cancer Lett. 2013, 329, 125–136. [Google Scholar] [CrossRef]
  177. Gasparello, J.; Papi, C.; Zurlo, M.; Gambari, L.; Manicardi, A.; Rozzi, A.; Ferrarini, M.; Corradini, R.; Gambari, R.; Finotti, A. MicroRNAs miR-584-5p and miR-425-3p Are Up-Regulated in Plasma of Colorectal Cancer (CRC) Patients: Targeting with Inhibitor Peptide Nucleic Acids Is Associated with Induction of Apoptosis in Colon Cancer Cell Lines. Cancers 2022, 15, 128. [Google Scholar] [CrossRef] [PubMed]
  178. Zhu, X.-L.; Ren, L.-F.; Wang, H.-P.; Bai, Z.-T.; Zhang, L.; Meng, W.-B.; Zhu, K.-X.; Ding, F.-H.; Miao, L.; Yan, J.; et al. Plasma microRNAs as potential new biomarkers for early detection of early gastric cancer. World J. Gastroenterol. 2019, 25, 1580–1591. [Google Scholar] [CrossRef] [PubMed]
  179. Jin, Y.; Wong, Y.S.; Goh, B.K.P.; Chan, C.Y.; Cheow, P.C.; Chow, P.K.H.; Lim, T.K.H.; Goh, G.B.B.; Krishna-moorthy, T.L.; Kumar, R.; et al. Circulating microRNAs as Potential Diagnostic and Prognostic Biomarkers in Hepatocellular Carcinoma. Sci. Rep. 2019, 9, 10464. [Google Scholar] [CrossRef]
  180. Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S.; et al. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol. 2008, 141, 672–675. [Google Scholar] [CrossRef]
  181. Jørgensen, S.; Paulsen, I.W.; Hansen, J.W.; Tholstrup, D.; Hother, C.; Sørensen, E.; Petersen, M.S.; Nielsen, K.R.; Rostgaard, K.; Larsen, M.A.H.; et al. The value of circulating microRNAs for early diagnosis of B-cell lymphoma: A case-control study on historical samples. Sci. Rep. 2020, 10, 9637. [Google Scholar] [CrossRef]
  182. Fulci, V.; Chiaretti, S.; Goldoni, M.; Azzalin, G.; Carucci, N.; Tavolaro, S.; Castellano, L.; Magrelli, A.; Citarella, F.; Messina, M.; et al. Quantitative technologies establish a novel microRNA profile of chronic lymphocytic leukemia. Blood 2007, 109, 4944–4951. [Google Scholar] [CrossRef]
  183. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M.; et al. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef] [PubMed]
  184. Drees, E.E.E.; Pegtel, D.M. Circulating miRNAs as Biomarkers in Aggressive B Cell Lymphomas. Trends Cancer 2020, 6, 910–923. [Google Scholar] [CrossRef] [PubMed]
  185. Sekar, D.; Hairul Islam, V.I.; Thirugnanasambantham, K.; Saravanan, S. Relevance of miR-21 in HIV and non-HIV-related lymphomas. Tumour Biol. 2014, 35, 8387–8393. [Google Scholar] [CrossRef] [PubMed]
  186. Thapa, D.R.; Bhatia, K.; Bream, J.H.; D’Souza, G.; Rinaldo, C.R.; Wolinsky, S.; Detels, R.; Martínez-Maza, O. B-cell activation induced microRNA-21 is elevated in circulating B cells preceding the diagnosis of AIDS-related non-Hodgkin lymphomas. AIDS 2012, 26, 1177–1180. [Google Scholar] [CrossRef]
  187. Thapa, D.R.; Hussain, S.K.; Tran, W.-C.; D’souza, G.; Bream, J.H.; Achenback, C.J.; Ayyavoo, V.; Detels, R.; Martínez-Maza, O. Serum microRNAs in HIV-infected individuals as pre-diagnosis biomarkers for AIDS-NHL. J. Acquir. Immune Defic. Syndr. 2014, 66, 229–237. [Google Scholar] [CrossRef]
  188. Sakamoto, K.; Sekizuka, T.; Uehara, T.; Hishima, T.; Mine, S.; Fukumoto, H.; Sato, Y.; Hasegawa, H.; Kuroda, M.; Katano, H. Next-generation sequencing of miRNAs in clinical samples of Epstein-Barr virus-associated B-cell lymphomas. Cancer Med. 2017, 6, 605–618. [Google Scholar] [CrossRef]
  189. Thapa, D.R.; Li, X.; Jamieson, B.D.; Martínez-Maza, O. Overexpression of microRNAs from the miR-17-92 paralog clusters in AIDS-related non-Hodgkin’s lymphomas. PLoS ONE 2011, 6, e20781. [Google Scholar] [CrossRef]
  190. Ramorola, B.R.; Goolam-Hoosen, T.; Alves de Souza Rios, L.; Mowla, S. Modulation of Cellular MicroRNA by HIV-1 in Burkitt Lymphoma Cells-A Pathway to Promoting Oncogenesis. Genes 2021, 12, 1302. [Google Scholar] [CrossRef]
  191. Hill, L.; Browne, G.; Tulchinsky, E. ZEB/miR-200 feedback loop: At the crossroads of signal transduction in cancer. Int. J. Cancer 2013, 132, 745–754. [Google Scholar] [CrossRef]
  192. Park, S.-M.; Gaur, A.B.; Lengyel, E.; Peter, M.E. The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22, 894–907. [Google Scholar] [CrossRef] [PubMed]
  193. Peckham-Gregory, E.C.; Thapa, D.R.; Martinson, J.; Duggal, P.; Penugonda, S.; Bream, J.H.; Chang, P.-Y.; Dandekar, S.; Chang, S.-C.; Detels, R.; et al. MicroRNA-related polymorphisms and non-Hodgkin lymphoma susceptibility in the Multicenter AIDS Cohort Study. Cancer Epidemiol. 2016, 45, 47–57. [Google Scholar] [CrossRef]
  194. Tiirikainen, M.I.; Mullaney, B.P.; Holly, E.A.; Pallavicini, M.G.; Jensen, R.H. DNA copy number alterations in HIV-positive and HIV-negative patients with diffuse large-cell lymphomas. J. Acquir. Immune Defic. Syndr. 2001, 27, 272–276. [Google Scholar] [CrossRef] [PubMed]
  195. Hybel, T.E.; Sørensen, E.F.; Enemark, M.H.; Hemmingsen, J.K.; Simonsen, A.T.; Lauridsen, K.L.; Møller, M.B.; Pedersen, C.; Pedersen, G.; Obel, N.; et al. Characterization of the genomic landscape of HIV-associated lymphoma reveals heterogeneity across histological subtypes. AIDS 2024, 38, 1897–1906. [Google Scholar] [CrossRef]
  196. Baron, M.; Labreche, K.; Veyri, M.; Désiré, N.; Bouzidi, A.; Seck-Thiam, F.; Charlotte, F.; Rousseau, A.; Morin, V.; Nakid-Cordero, C.; et al. Epstein-Barr virus and immune status imprint the immunogenomics of non-Hodgkin lymphomas occurring in immune-suppressed environments. Haematologica 2024, 109, 3615–3630. [Google Scholar] [CrossRef] [PubMed]
  197. Pott, C.; Kotrova, M.; Darzentas, N.; Brüggemann, M.; Khouja, M.; EuroClonality-NGS Working Group. cfDNA-Based NGS IG Analysis in Lymphoma. Methods Mol. Biol. 2022, 2453, 101–117. [Google Scholar]
  198. Vogt, S.L.; Patel, M.; Omar, T.; Pather, S.; Martinson, N.; Ambinder, R. Molecular Diagnostics for AIDS Lymphoma Diagnosis in South Africa and the Potential for Other Low- and Middle-Income Countries. J. Glob. Oncol. 2018, 4, 1–6. [Google Scholar] [CrossRef]
  199. Armand, P.; Oki, Y.; Neuberg, D.S.; Faham, M.; Cummings, C.; Klinger, M.; Weng, L.; Bhattar, S.; Lacasce, A.S.; Jacobsen, E.D.; et al. Detection of circulating tumour DNA in patients with aggressive B-cell non-Hodgkin lymphoma. Br. J. Haematol. 2013, 163, 123–126. [Google Scholar] [CrossRef]
  200. Kurtz, D.M.; Green, M.R.; Bratman, S.V.; Scherer, F.; Liu, C.L.; Kunder, C.A.; Takahashi, K.; Glover, C.; Keane, C.; Kihira, S.; et al. Noninvasive monitoring of diffuse large B-cell lymphoma by immunoglobulin high-throughput sequencing. Blood 2015, 125, 3679–3687. [Google Scholar] [CrossRef]
  201. Ladetto, M.; Brüggemann, M.; Monitillo, L.; Ferrero, S.; Pepin, F.; Drandi, D.; Barbero, D.; Palumbo, A.; Passera, R.; Boccadoro, M.; et al. Next-generation sequencing and real-time quantitative PCR for minimal residual disease detection in B-cell disorders. Leukemia 2014, 28, 1299–1307. [Google Scholar] [CrossRef]
  202. Roschewski, M.; Dunleavy, K.; Pittaluga, S.; Moorhead, M.; Pepin, F.; Kong, K.; Shovlin, M.; Jaffe, E.S.; Staudt, L.M.; Lai, C.; et al. Circulating tumour DNA and CT monitoring in patients with untreated diffuse large B-cell lymphoma: A correlative biomarker study. Lancet Oncol. 2015, 16, 541–549. [Google Scholar] [CrossRef]
  203. Sarkozy, C.; Huet, S.; Carlton, V.E.H.; Fabiani, B.; Delmer, A.; Jardin, F.; Delfau-Larue, M.-H.; Hacini, M.; Ribrag, V.; Guidez, S.; et al. The prognostic value of clonal heterogeneity and quantitative assessment of plasma circulating clonal IG-VDJ sequences at diagnosis in patients with follicular lymphoma. Oncotarget 2017, 8, 8765–8774. [Google Scholar] [CrossRef] [PubMed]
  204. Wagner-Johnston, N.D.; Gellert, L.; Gocke, C.D.; Lemas, V.M.; Lee, J.; Martínez-Maza, O.; Ambinder, R.F. Clonal immunoglobulin DNA in the plasma of patients with AIDS lymphoma. Blood 2011, 117, 4860–4862. [Google Scholar] [CrossRef] [PubMed]
  205. Kurtz, D.M.; Scherer, F.; Jin, M.C.; Soo, J.; Craig, A.F.M.; Esfahani, M.S.; Chabon, J.J.; Stehr, H.; Liu, C.L.; Tib-shirani, R.; et al. Circulating Tumor DNA Measurements As Early Outcome Predictors in Diffuse Large B-Cell Lymphoma. J. Clin. Oncol. 2018, 36, 2845–2853. [Google Scholar] [CrossRef]
  206. Vogt, S.L.; Patel, M.; Lakha, A.; Philip, V.; Omar, T.; Ashmore, P.; Pather, S.; Haley, L.M.; Zheng, G.; Stone, J.; et al. Feasibility of Cell-Free DNA Collection and Clonal Immunoglobulin Sequencing in South African Patients With HIV-Associated Lymphoma. JCO Glob. Oncol. 2021, 7, 611–621. [Google Scholar] [CrossRef] [PubMed]
  207. van Niel, G.; D’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  208. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef]
  209. Yamamoto, T.; Yamamoto, Y.; Ochiya, T. Extracellular vesicle-mediated immunoregulation in cancer. Int. J. Hematol. 2023, 117, 640–646. [Google Scholar] [CrossRef]
  210. Naito, Y.; Yoshioka, Y.; Yamamoto, Y.; Ochiya, T. How cancer cells dictate their microenvironment: Present roles of extracellular vesicles. Cell Mol. Life Sci. 2017, 74, 697–713. [Google Scholar] [CrossRef]
  211. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  212. Asleh, K.; Dery, V.; Taylor, C.; Davey, M.; Djeungoue-Petga, M.-A.; Ouellette, R.J. Extracellular vesicle-based liquid biopsy biomarkers and their application in precision immuno-oncology. Biomark. Res. 2023, 11, 99. [Google Scholar] [CrossRef] [PubMed]
  213. Ofori, K.; Bhagat, G.; Rai, A.J. Exosomes and extracellular vesicles as liquid biopsy biomarkers in diffuse large B-cell lymphoma: Current state of the art and unmet clinical needs. Br. J. Clin. Pharmacol. 2021, 87, 284–294. [Google Scholar] [CrossRef] [PubMed]
  214. Lee, A.A.; Godwin, A.K.; Abdelhakim, H. The multifaceted roles of extracellular vesicles for therapeutic intervention with non-Hodgkin lymphoma. Extracell. Vesicles Circ. Nucl. Acids 2024, 5, 329–343. [Google Scholar] [CrossRef]
  215. Martínez, L.E.; Lensing, S.; Chang, D.; Magpantay, L.I.; Mitsuyasu, R.; Ambinder, R.F.; Sparano, J.A.; Mar-tínez-Maza, O.; Epeldegui, M. Plasma extracellular vesicles bearing PD-L1, CD40, CD40L or TNF-RII are significantly reduced after treatment of AIDS-NHL. Sci. Rep. 2022, 12, 9185. [Google Scholar] [CrossRef] [PubMed]
  216. Martínez, L.E.; Magpantay, L.I.; Guo, Y.; Hegde, P.; Detels, R.; Hussain, S.K.; Epeldegui, M. Extracellular vesicles as biomarkers for AIDS-associated non-Hodgkin lymphoma risk. Front. Immunol. 2023, 14, 1259007. [Google Scholar] [CrossRef]
  217. Mussini, C.; Lorenzini, P.; Cozzi-Lepri, A.; Lapadula, G.; Marchetti, G.; Nicastri, E.; Cingolani, A.; Lichtner, M.; Antinori, A.; Gori, A.; et al. CD4/CD8 ratio normalisation and non-AIDS-related events in individuals with HIV who achieve viral load suppression with antiretroviral therapy: An observational cohort study. Lancet HIV 2015, 2, e98–e106. [Google Scholar] [CrossRef]
  218. Matthews, G.V.; Bower, M.; Mandalia, S.; Powles, T.; Nelson, M.R.; Gazzard, B.G. Changes in acquired immunodeficiency syndrome-related lymphoma since the introduction of highly active antiretroviral therapy. Blood 2000, 96, 2730–2734. [Google Scholar] [CrossRef]
  219. Stebbing, J.; Gazzard, B.; Mandalia, S.; Teague, A.; Waterston, A.; Marvin, V.; Nelson, M.; Bower, M. Antiretroviral treatment regimens and immune parameters in the prevention of systemic AIDS-related non-Hodgkin’s lymphoma. J. Clin. Oncol. 2004, 22, 2177–2183. [Google Scholar] [CrossRef]
  220. Kirk, O.; Pedersen, C.; Cozzi-Lepri, A.; Antunes, F.; Miller, V.; Gatell, J.M.; Katlama, C.; Lazzarin, A.; Skinhøj, P.; Barton, S.E.; et al. Non-Hodgkin lymphoma in HIV-infected patients in the era of highly active antiretroviral therapy. Blood 2001, 98, 3406–3412. [Google Scholar] [CrossRef]
  221. Guiguet, M.; Boué, F.; Cadranel, J.; Lang, J.-M.; Rosenthal, E.; Costagliola, D.; Clinical Epidemiology Group of the FHDH-ANRS CO4 Cohort. Effect of immunodeficiency, HIV viral load, and antiretroviral therapy on the risk of individual malignancies (FHDH-ANRS CO4): A prospective cohort study. Lancet Oncol. 2009, 10, 1152–1159. [Google Scholar] [CrossRef]
  222. Reekie, J.; Kosa, C.; Engsig, F.; Monforte, A.d.; Wiercinska-Drapalo, A.; Domingo, P.; Antunes, F.; Clumeck, N.; Kirk, O.; Lundgren, J.D.; et al. Relationship between current level of immunodeficiency and non-acquired immunodeficiency syndrome-defining malignancies. Cancer 2010, 116, 5306–5315. [Google Scholar] [CrossRef]
  223. Silverberg, M.J.; Chao, C.; Leyden, W.A.; Xu, L.; Horberg, M.A.; Klein, D.; Towner, W.J.; Dubrow, R.; Quesen-berry, C.P.; Neugebauer, R.S.; et al. HIV infection, immunodeficiency, viral replication, and the risk of cancer. Cancer Epidemiol. Biomark. Prev. 2011, 20, 2551–2559. [Google Scholar] [CrossRef]
  224. Ramaswami, R.; Chia, G.; Dalla Pria, A.; Pinato, D.J.; Parker, K.; Nelson, M.; Bower, M. Evolution of HIV-Associated Lymphoma Over 3 Decades. J. Acquir. Immune Defic. Syndr. 2016, 72, 177–183. [Google Scholar] [CrossRef]
  225. Polesel, J.; Clifford, G.M.; Rickenbach, M.; Dal Maso, L.; Battegay, M.; Bouchardy, C.; Furrer, H.; Hasse, B.; Levi, F.; Probst-Hensch, N.M.; et al. Non-Hodgkin lymphoma incidence in the Swiss HIV Cohort Study before and after highly active antiretroviral therapy. AIDS 2008, 22, 301–306. [Google Scholar] [CrossRef]
  226. Nicolau, I.A.; Moineddin, R.; Brooks, J.D.; Antoniou, T.; Gillis, J.L.; Kendall, C.E.; Cooper, C.; Cotterchio, M.; Salters, K.; Smieja, M.; et al. Associations of CD4 Cell Count Measures With Infection-Related and Infection-Unrelated Cancer Risk Among People With HIV. J. Acquir. Immune Defic. Syndr. 2024, 96, 447–456. [Google Scholar] [CrossRef]
  227. Mathoma, A.; Sartorius, B.; Mahomed, S. The Trends and Risk Factors of AIDS-Defining Cancers and Non-AIDS-Defining Cancers in Adults Living with and without HIV: A Narrative Review. J. Cancer Epidemiol. 2024, 2024, 7588928. [Google Scholar] [CrossRef]
  228. Castilho, J.L.; Bian, A.; Jenkins, C.A.; Shepherd, B.E.; Sigel, K.; Gill, M.J.; Kitahata, M.M.; Silverberg, M.J.; Mayor, A.M.; Coburn, S.B.; et al. CD4/CD8 Ratio and Cancer Risk Among Adults With HIV. J. Natl. Cancer Inst. 2022, 114, 854–862. [Google Scholar] [CrossRef]
  229. Martínez-Sanz, J.; Díaz-Álvarez, J.; Rosas, M.; Ron, R.; Iribarren, J.A.; Bernal, E.; Gutiérrez, F.; Ruiz Sancho, A.; Cabello, N.; Olalla, J.; et al. Expanding HIV clinical monitoring: The role of CD4, CD8, and CD4/CD8 ratio in predicting non-AIDS events. EBioMedicine 2023, 95, 104773. [Google Scholar] [CrossRef]
  230. Chammartin, F.; Mocroft, A.; Egle, A.; Zangerle, R.; Smith, C.; Mussini, C.; Wit, F.; Vehreschild, J.J.; d’Arminio Monforte, A.; Castagna, A.; et al. Measures of Longitudinal Immune Dysfunction and Risk of AIDS and Non-AIDS Defining Malignancies in Antiretroviral-Treated People With Human Immunodeficiency Virus. Clin. Infect. Dis. 2024, 78, 995–1004. [Google Scholar] [CrossRef]
  231. Caby, F.; Guiguet, M.; Weiss, L.; Winston, A.; Miro, J.M.; Konopnicki, D.; Le Moing, V.; Bonnet, F.; Reiss, P.; Mussini, C.; et al. CD4/CD8 Ratio and the Risk of Kaposi Sarcoma or Non-Hodgkin Lymphoma in the Context of Efficiently Treated Human Immunodeficiency Virus (HIV) Infection: A Collaborative Analysis of 20 European Cohort Studies. Clin. Infect. Dis. 2021, 73, 50–59. [Google Scholar] [CrossRef]
  232. Tedeschi, R.; Bortolin, M.T.; Bidoli, E.; Zanussi, S.; Pratesi, C.; Vaccher, E.; Tirelli, U.; De Paoli, P. Assessment of immunovirological features in HIV related non-Hodgkin lymphoma patients and their impact on outcome. J. Clin. Virol. 2012, 53, 297–301. [Google Scholar] [CrossRef]
  233. Xiong, Y.; Liu, W.; Chen, X.; Mo, P.; Xiong, Y.; Deng, L.; Zhang, Y. Survival of HIV associated diffuse large B-cell lymphoma and Burkitt lymphoma in China. Sci. Rep. 2024, 14, 30397. [Google Scholar] [CrossRef]
  234. Ron, R.; Martínez-Sanz, J.; Herrera, S.; Ramos-Ruperto, L.; Díez, A.; Sainz, T.; Álvarez-Díaz, N.; Correa-Pérez, A.; Muriel, A.; López-Alcalde, J.; et al. CD4/CD8 ratio and CD8+ T-cell count as prognostic markers for non-AIDS mortality in people living with HIV. A systematic review and meta-analysis. Front. Immunol. 2024, 15, 1343124. [Google Scholar]
  235. Xiao, Q.; Quan, X.; Chen, S.; Wang, C.; Jun, L.; Zhang, X.; Yang, Z.; Liu, Y. The Disparity of Circulating Immune Cell Subsets and Inflammatory Status between HIV-Positive and HIV-Negative Patients with Lymphoma. Blood 2024, 144, 6271. [Google Scholar] [CrossRef]
  236. Barta, S.K.; Xue, X.; Wang, D.; Lee, J.Y.; Kaplan, L.D.; Ribera, J.-M.; Oriol, A.; Spina, M.; Tirelli, U.; Boue, F.; et al. A new prognostic score for AIDS-related lymphomas in the rituximab-era. Haematologica 2014, 99, 1731–1737. [Google Scholar] [CrossRef]
  237. Chen, J.; Liu, X.; Qin, S.; Ruan, G.; Lu, A.; Zhang, J.; Wu, Y.; Xie, Z.; Peng, J. A Novel Prognostic Score Including the CD4/CD8 for AIDS-Related Lymphoma. Front. Cell Infect. Microbiol. 2022, 12, 919446. [Google Scholar] [CrossRef]
  238. Matsuki, E.; Bohn, O.L.; El Jamal, S.; Pichardo, J.D.; Zelenetz, A.D.; Younes, A.; Teruya-Feldstein, J. Lymphocyte-to-Monocyte Ratio May Serve as a Better Prognostic Indicator Than Tumor-associated Macrophages in DLBCL Treated With Rituximab. Appl. Immunohistochem. Mol. Morphol. 2019, 27, 572–580. [Google Scholar] [CrossRef]
  239. Deng, Q.; He, B.; Liu, X.; Yue, J.; Ying, H.; Pan, Y.; Sun, H.; Chen, J.; Wang, F.; Gao, T.; et al. Prognostic value of pre-operative inflammatory response biomarkers in gastric cancer patients and the construction of a predictive model. J. Transl. Med. 2015, 13, 66. [Google Scholar] [CrossRef]
  240. Zeng, J.; Zhang, X.; Jia, L.; Wu, Y.; Tian, Y.; Zhang, Y. Pretreatment lymphocyte-to-monocyte ratios predict AIDS-related diffuse large B-cell lymphoma overall survival. J. Med. Virol. 2021, 93, 3907–3914. [Google Scholar] [CrossRef]
  241. Vaughan, J.; Wiggill, T.; Lawrie, D.; Machaba, M.; Patel, M. The prognostic impact of monocyte fluorescence, immunosuppressive monocytes and peripheral blood immune cell numbers in HIV-associated Diffuse Large B-cell Lymphoma. PLoS ONE 2023, 18, e0280044. [Google Scholar] [CrossRef]
  242. Borges, Á.H.; Silverberg, M.J.; Wentworth, D.; Grulich, A.E.; Fätkenheuer, G.; Mitsuyasu, R.; Tambussi, G.; Sabin, C.A.; Neaton, J.D.; Lundgren, J.D.; et al. Predicting risk of cancer during HIV infection: The role of inflammatory and coagulation biomarkers. AIDS 2013, 27, 1433–1441. [Google Scholar] [CrossRef]
  243. Borges, A.H.; Dubrow, R.; Silverberg, M.J. Factors contributing to risk for cancer among HIV-infected individuals, and evidence that earlier combination antiretroviral therapy will alter this risk. Curr. Opin. HIV AIDS 2014, 9, 34–40. [Google Scholar] [CrossRef]
  244. Deeks, S.G. HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med. 2011, 62, 141–155. [Google Scholar] [CrossRef]
  245. Collaboration of Observational HIV Epidemiological Research Europe (COHERE) Study Group; Bohlius, J.; Schmidlin, K.; Costagliola, D.; Fätkenheuer, G.; May, M.; Caro-Murillo, A.M.; Mocroft, A.; Bonnet, F.; Clifford, G.; et al. Incidence and risk factors of HIV-related non-Hodgkin’s lymphoma in the era of combination antiretroviral therapy: A European multicohort study. Antivir. Ther. 2009, 14, 1065–1074. [Google Scholar]
  246. Zoufaly, A.; Stellbrink, H.-J.; der Heiden, M.A.; Kollan, C.; Hoffmann, C.; van Lunzen, J.; Hamouda, O.; Clin-Surv Study Group. Cumulative HIV viremia during highly active antiretroviral therapy is a strong predictor of AIDS-related lymphoma. J. Infect. Dis. 2009, 200, 79–87. [Google Scholar] [CrossRef]
  247. Achenbach, C.J.; Buchanan, A.L.; Cole, S.R.; Hou, L.; Mugavero, M.J.; Crane, H.M.; Moore, R.D.; Haubrich, R.H.; Gopal, S.; Eron, J.J.; et al. HIV viremia and incidence of non-Hodgkin lymphoma in patients successfully treated with antiretroviral therapy. Clin. Infect. Dis. 2014, 58, 1599–1606. [Google Scholar] [CrossRef]
  248. Ruffieux, Y.; Mwansa-Kambafwile, J.; Metekoua, C.; Tombe-Nyahuma, T.; Bohlius, J.; Muchengeti, M.; Egger, M.; Rohner, E. HIV-1 Viremia and Cancer Risk in 2.8 Million People: The South African HIV Cancer Match Study. Clin. Infect. Dis. 2024, ciae652. [Google Scholar] [CrossRef] [PubMed]
  249. Lee, J.S.; Cole, S.R.; Achenbach, C.J.; Dittmer, D.P.; Richardson, D.B.; Miller, W.C.; Mathews, C.; Althoff, K.N.; Moore, R.D.; Eron, J.J.; et al. Cancer risk in HIV patients with incomplete viral suppression after initiation of antiretroviral therapy. PLoS ONE 2018, 13, e0197665. [Google Scholar] [CrossRef]
  250. Elvstam, O.; Marrone, G.; Medstrand, P.; Treutiger, C.J.; Svedhem, V.; Gisslén, M.; Björkman, P. Associations Between Plasma Human Immunodeficiency Virus (HIV) Ribonucleic Acid Levels and Incidence of Invasive Cancer in People With HIV After Initiation of Combination Antiretroviral Therapy. Open Forum Infect Dis. 2021, 8, ofab131. [Google Scholar] [CrossRef]
  251. Gopal, S.; Patel, M.R.; Yanik, E.L.; Cole, S.R.; Achenbach, C.J.; Napravnik, S.; Burkholder, G.A.; Reid, E.G.; Ro-driguez, B.; Deeks, S.G.; et al. Association of early HIV viremia with mortality after HIV-associated lymphoma. AIDS 2013, 27, 2365–2373. [Google Scholar] [CrossRef]
  252. Trifone, C.; Richard, C.; Pagliuzza, A.; Dufour, C.; Lemieux, A.; Clark, N.M.; Janaka, S.K.; Fennessey, C.M.; Keele, B.E.; Fromentin, R.; et al. Contribution of intact viral genomes persisting in blood and tissues during ART to plasma viral rebound in SHIV-infected rhesus macaques. iScience 2025, 28, 111998. [Google Scholar] [CrossRef]
  253. Amand, M.; Adams, P.; Schober, R.; Iserentant, G.; Servais, J.-Y.; Moutschen, M.; Seguin-Devaux, C. The anti-caspase 1 inhibitor VX-765 reduces immune activation, CD4+ T cell depletion, viral load, and total HIV-1 DNA in HIV-1 infected humanized mice. Elife 2023, 12, e83207. [Google Scholar] [CrossRef]
  254. Wang, J.; Xiao, N.; Zhu, Z.; Qiao, H.; Zhao, F.; Zhang, L.; Gou, J.; Lu, M.; He, Y.; Lu, H.; et al. Comparing acute versus AIDS ART initiation on HIV-1 integration sites and clonal expansion. Signal Transduct. Target. Ther. 2025, 10, 23. [Google Scholar] [CrossRef]
  255. Liu, Y.; Xie, X.; Li, J.; Xiao, Q.; He, S.; Fu, H.; Zhang, X.; Liu, Y. Immune Characteristics and Immunotherapy of HIV-Associated Lymphoma. Curr. Issues Mol. Biol. 2024, 46, 9984–9997. [Google Scholar] [CrossRef]
  256. Shmakova, A.; Hugot, C.; Kozhevnikova, Y.; Schwager Karpukhina, A.; Tsimailo, I.; Gérard, L.; Boutboul, D.; Oksenhendler, E.; Szewczyk-Roszczenko, O.; Roszczenko, P.; et al. Chronic HIV-1 Tat action induces HLA-DR downregulation in B cells: A mechanism for lymphoma immune escape in people living with HIV. J. Med. Virol. 2024, 96, e29423. [Google Scholar] [CrossRef]
  257. El-Amine, R.; Germini, D.; Zakharova, V.V.; Tsfasman, T.; Sheval, E.V.; Louzada, R.A.N.; Dupuy, C.; Bil-hou-Nabera, C.; Hamade, A.; Najjar, F.; et al. HIV-1 Tat protein induces DNA damage in human peripheral blood B-lymphocytes via mitochondrial ROS production. Redox Biol. 2018, 15, 97–108. [Google Scholar] [CrossRef]
  258. Germini, D.; Tsfasman, T.; Klibi, M.; El-Amine, R.; Pichugin, A.; Iarovaia, O.V.; Bilhou-Nabera, C.; Subra, F.; Bou Saada, Y.; Sukhanova, A.; et al. HIV Tat induces a prolonged MYC relocalization next to IGH in circulating B-cells. Leukemia 2017, 31, 2515–2522. [Google Scholar] [CrossRef]
  259. Sall, F.B.; El Amine, R.; Markozashvili, D.; Tsfasman, T.; Oksenhendler, E.; Lipinski, M.; Vassetzky, Y.; Germi-ni, D. HIV-1 Tat protein induces aberrant activation of AICDA in human B-lymphocytes from peripheral. Blood J. Cell Physiol. 2019, 234, 15678–15685. [Google Scholar] [CrossRef]
  260. Akbay, B.; Germini, D.; Bissenbaev, A.K.; Musinova, Y.R.; Sheval, E.V.; Vassetzky, Y.; Dokudovskaya, S. HIV-1 Tat Activates Akt/mTORC1 Pathway and AICDA Expression by Downregulating Its Transcriptional Inhibitors in B Cells. Int. J. Mol. Sci. 2021, 22, 1588. [Google Scholar] [CrossRef]
  261. Wang, X.; Duan, Z.; Yu, G.; Fan, M.; Scharff, M.D. Human Immunodeficiency Virus Tat Protein Aids V Region Somatic Hypermutation in Human B Cells. mBio 2018, 9, e02315–e02317. [Google Scholar] [CrossRef]
  262. Fritsch, L.; Marechal, V.; Schneider, V.; Barthet, C.; Rozenbaum, W.; Moisan-Coppey, M.; Coppey, J.; Nicolas, J.C. Production of HIV-1 by human B cells infected in vitro: Characterization of an EBV genome-negative B cell line chronically synthetizing a low level of HIV-1 after infection. Virology 1998, 244, 542–551. [Google Scholar] [CrossRef]
  263. Valyaeva, A.A.; Tikhomirova, M.A.; Feng, J.; Zharikova, A.A.; Potashnikova, D.M.; Musinova, Y.R.; Mironov, A.A.; Vassetzky, Y.S.; Sheval, E.V. Compensatory reactions of B cells in response to chronic HIV-1 Tat exposure. J. Cell Physiol. 2025, 240, e31459. [Google Scholar] [CrossRef]
  264. Carroll, V.A.; Lafferty, M.K.; Marchionni, L.; Bryant, J.L.; Gallo, R.C.; Garzino-Demo, A. Expression of HIV-1 matrix protein p17 and association with B-cell lymphoma in HIV-1 transgenic mice. Proc. Natl. Acad. Sci. USA 2016, 113, 13168–13173. [Google Scholar] [CrossRef]
  265. Caccuri, F.; Giagulli, C.; Bugatti, A.; Benetti, A.; Alessandri, G.; Ribatti, D.; Marsico, S.; Apostoli, P.; Slevin, M.A.; Rusnati, M.; et al. HIV-1 matrix protein p17 promotes angiogenesis via chemokine receptors CXCR1 and CXCR2. Proc. Natl. Acad. Sci. USA 2012, 109, 14580–14585. [Google Scholar] [CrossRef]
  266. Caccuri, F.; Rueckert, C.; Giagulli, C.; Schulze, K.; Basta, D.; Zicari, S.; Marsico, S.; Cervi, E.; Fiorentini, S.; Slevin, M.; et al. HIV-1 matrix protein p17 promotes lymphangiogenesis and activates the endothelin-1/endothelin B receptor axis. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 846–856. [Google Scholar] [CrossRef]
  267. Fiorentini, S.; Marini, E.; Caracciolo, S.; Caruso, A. Functions of the HIV-1 matrix protein p17. New Microbiol. 2006, 29, 1–10. [Google Scholar]
  268. Popovic, M.; Tenner-Racz, K.; Pelser, C.; Stellbrink, H.-J.; van Lunzen, J.; Lewis, G.; Kalyanaraman, V.S.; Gallo, R.C.; Racz, P. Persistence of HIV-1 structural proteins and glycoproteins in lymph nodes of patients under highly active antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2005, 102, 14807–14812. [Google Scholar] [CrossRef]
  269. Feng, Y.; Wang, Z.; Zeng, D.; Song, S.; Yang, Y.; Wang, A.; Xu, J.; Guo, W.; Wu, M.; Shi, Y.; et al. High expression of HIV-1 matrix protein p17 in both lymphoma and lymph node tissues of AIDS patients. Pathol. Res. Pract. 2022, 237, 154061. [Google Scholar] [CrossRef]
  270. Giagulli, C.; Marsico, S.; Magiera, A.K.; Bruno, R.; Caccuri, F.; Barone, I.; Fiorentini, S.; Andò, S.; Caruso, A. Opposite effects of HIV-1 p17 variants on PTEN activation and cell growth in B cells. PLoS ONE 2011, 6, e17831. [Google Scholar] [CrossRef]
  271. Dolcetti, R.; Giagulli, C.; He, W.; Selleri, M.; Caccuri, F.; Eyzaguirre, L.M.; Mazzuca, P.; Corbellini, S.; Campi-longo, F.; Marsico, S.; et al. Role of HIV-1 matrix protein p17 variants in lymphoma pathogenesis. Proc. Natl. Acad. Sci. USA 2015, 112, 14331–14336. [Google Scholar] [CrossRef]
  272. Caccuri, F.; Messali, S.; Zani, A.; Campisi, G.; Giovanetti, M.; Zanussi, S.; Vaccher, E.; Fabris, S.; Bugatti, A.; Focà, E.; et al. HIV-1 mutants expressing B cell clonogenic matrix protein p17 variants are increasing their prevalence worldwide. Proc. Natl. Acad. Sci. USA 2022, 119, e2122050119. [Google Scholar] [CrossRef]
  273. Ding, X.; Liang, T.; Liang, B.; Gao, H.; Wang, J.; Liu, H.; Wang, F.; Zheng, X.; Li, P.; Chen, S.; et al. Diagnostic value of EBV-DNA in CSF for PCNSL in AIDS patients with focal brain lesions: A meta-analysis of diagnostic test. Medicine 2022, 101, e31793. [Google Scholar] [CrossRef]
  274. MacMahon, E.M.; Glass, J.D.; Hayward, S.D.; Mann, R.B.; Becker, P.S.; Charache, P.; McArthur, J.C.; Ambinder, R.F. Epstein-Barr virus in AIDS-related primary central nervous system lymphoma. Lancet 1991, 338, 969–973. [Google Scholar] [CrossRef]
  275. Ul-Haq, I.; Dalla Pria, A.; Suardi, E.; Pinato, D.J.; Froeling, F.; Forni, J.; Randell, P.; Bower, M. Blood Epstein-Barr virus DNA does not predict outcome in advanced HIV-associated Hodgkin lymphoma. Med. Oncol. 2018, 35, 53. [Google Scholar] [CrossRef]
  276. Corcoran, C.; Rebe, K.; van der Plas, H.; Myer, L.; Hardie, D.R. The predictive value of cerebrospinal fluid Epstein-Barr viral load as a marker of primary central nervous system lymphoma in HIV-infected persons. J. Clin. Virol. 2008, 42, 433–436. [Google Scholar] [CrossRef]
  277. Cinque, P.; Brytting, M.; Vago, L.; Castagna, A.; Parravicini, C.; Zanchetta, N.; D’Arminio Monforte, A.; Wah-ren, B.; Lazzarin, A.; Linde, A. Epstein-Barr virus DNA in cerebrospinal fluid from patients with AIDS-related primary lymphoma of the central nervous system. Lancet 1993, 342, 398–401. [Google Scholar] [CrossRef] [PubMed]
  278. Montgomery, N.D.; Randall, C.; Painschab, M.; Seguin, R.; Kaimila, B.; Kasonkanji, E.; Zuze, T.; Krysiak, R.; Sanders, M.K.; Elliott, A.; et al. High pretreatment plasma Epstein-Barr virus (EBV) DNA level is a poor prognostic marker in HIV-associated, EBV-negative diffuse large B-cell lymphoma in Malawi. Cancer Med. 2020, 9, 552–561. [Google Scholar] [CrossRef] [PubMed]
  279. Carbone, A.; Cesarman, E.; Spina, M.; Gloghini, A.; Schulz, T.F. HIV-associated lymphomas and gamma-herpesviruses. Blood 2009, 113, 1213–1224. [Google Scholar] [CrossRef]
  280. da Silva, S.R.; de Oliveira, D.E. HIV, EBV and KSHV: Viral cooperation in the pathogenesis of human malignancies. Cancer Lett. 2011, 305, 175–185. [Google Scholar] [CrossRef] [PubMed]
  281. Lurain, K.; Yarchoan, R.; Uldrick, T.S. Treatment of Kaposi Sarcoma Herpesvirus-Associated Multicentric Castleman Disease. Hematol. Oncol. Clin. N. Am. 2018, 32, 75–88. [Google Scholar] [CrossRef]
  282. Uldrick, T.S.; Polizzotto, M.N.; Yarchoan, R. Recent advances in Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Curr. Opin. Oncol. 2012, 24, 495–505. [Google Scholar] [CrossRef]
  283. Carbone, A.; Vaccher, E.; Gloghini, A. Hematologic cancers in individuals infected by HIV. Blood 2022, 139, 995–1012. [Google Scholar] [CrossRef]
Figure 1. The pathogenesis of HIV-associated lymphoma (HAL). (a) HIV infection targets the CD4+ T cells, entering to replicate internally, leading to cell depletion and eventually triggering a severe immunodeficiency in the body that drives lymphomas; (b) HIV infection destabilizes the genomic instability, leading to chromosomal aberrations, DNA recombination, and ultimately driving lymphomagenesis; (c) HIV itself and various HIV-encoded proteins, including p17, nef, tat, gp120, act directly on follicular germinal center aggregates in lymph nodes to drive lymphomas; (d) impaired immune surveillance of T cells after HIV infection increases the chances of tumorigenesis driven by oncogenic viruses such as EBV and KSHV, leading to lymphomagenesis; (e) HIV infection causes chronic antigen activation and B-cell activation, long-term chronic inflammation, and the overproduction of various cytokines that promote the development and growth of lymphoma. The solid arrows (↑) represent elevated. Created in BioRender. Xuejiao Shu. (2025) https://app.biorender.com/illustrations/67f232faa3efd15ebf9d87de.
Figure 1. The pathogenesis of HIV-associated lymphoma (HAL). (a) HIV infection targets the CD4+ T cells, entering to replicate internally, leading to cell depletion and eventually triggering a severe immunodeficiency in the body that drives lymphomas; (b) HIV infection destabilizes the genomic instability, leading to chromosomal aberrations, DNA recombination, and ultimately driving lymphomagenesis; (c) HIV itself and various HIV-encoded proteins, including p17, nef, tat, gp120, act directly on follicular germinal center aggregates in lymph nodes to drive lymphomas; (d) impaired immune surveillance of T cells after HIV infection increases the chances of tumorigenesis driven by oncogenic viruses such as EBV and KSHV, leading to lymphomagenesis; (e) HIV infection causes chronic antigen activation and B-cell activation, long-term chronic inflammation, and the overproduction of various cytokines that promote the development and growth of lymphoma. The solid arrows (↑) represent elevated. Created in BioRender. Xuejiao Shu. (2025) https://app.biorender.com/illustrations/67f232faa3efd15ebf9d87de.
Biomolecules 15 00993 g001
Figure 2. Panoramic overview map of circulating biomarkers for HAL. This figure categorizes six types of circulating biomarkers involved in the onset, progression, and prognosis of HAL. Red solid arrows (↑) indicate upregulation, while green dashed arrows (⇣) indicate downregulation. Created in BioRender. Xuejiao Shu. (2025) https://app.biorender.com/illustrations/68103ee84139ebc329581dce.
Figure 2. Panoramic overview map of circulating biomarkers for HAL. This figure categorizes six types of circulating biomarkers involved in the onset, progression, and prognosis of HAL. Red solid arrows (↑) indicate upregulation, while green dashed arrows (⇣) indicate downregulation. Created in BioRender. Xuejiao Shu. (2025) https://app.biorender.com/illustrations/68103ee84139ebc329581dce.
Biomolecules 15 00993 g002
Figure 3. The disruption of intestinal barrier integrity and microbial translocation in HIV-positive individuals. (a) In HIV-negative individuals, an intact gut epithelial barrier effectively prevents microbial products from entering the circulation; (b) HIV infection damages the gastrointestinal epithelium, leading to increased permeability and allowing bacterial products such as LPS to translocate into the lamina propria and bloodstream. LPS forms complexes with LBP, which activate the TLR4/NF-κB signaling pathway and trigger the release of inflammatory cytokines including IL-6 and TNF-α, promoting lymphomagenesis.Created in BioRender. Xuejiao Shu. (2025) https://app.biorender.com/illustrations/67f68ca3b9e5bb8ed54bd2ff.
Figure 3. The disruption of intestinal barrier integrity and microbial translocation in HIV-positive individuals. (a) In HIV-negative individuals, an intact gut epithelial barrier effectively prevents microbial products from entering the circulation; (b) HIV infection damages the gastrointestinal epithelium, leading to increased permeability and allowing bacterial products such as LPS to translocate into the lamina propria and bloodstream. LPS forms complexes with LBP, which activate the TLR4/NF-κB signaling pathway and trigger the release of inflammatory cytokines including IL-6 and TNF-α, promoting lymphomagenesis.Created in BioRender. Xuejiao Shu. (2025) https://app.biorender.com/illustrations/67f68ca3b9e5bb8ed54bd2ff.
Biomolecules 15 00993 g003
Table 1. Biomarkers for cytokines
Table 1. Biomarkers for cytokines
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
CytokinesIL-1α
IL-1β
IL-4
IL-5
IL-6
IL-7
IL-8
IL-10
IL10 SNPs
IL-11
IL-12p40
IL-13
IL-15
IL-18
IL-29
GM-CSF
Neopterin
TNF-α
TGF-β
IFN-γ
BAFF/BLyS
HIV-NHL patients
Mice
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients/mice
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients/mice
HIV-NHL patients
HIV-NHL patients
Mice
HIV-NHL patients/mice
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients/mice
HIV-NHL patients
HIV-NHL patients/mice
HIV-DLBCL patients
HIV-NHL patients
HIV-NHL patients
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Predictive
Predictive
Predictive
Predictive
Predictive/Prognostic
Prognostic
Prognostic
Predictive/Prognostic
Predictive
Predictive
Predictive
Predictive
Prognostic
Predictive
Predictive
Predictive
Predictive
Predictive
Prognostic
Prognostic
Prognostic
[9]
[54]
[9]
[9]
[52,54,55,56,59,60,61]
[62]
[62]
[50,52,54,55,56,59,60,61]
[50,51]
[7]
[54]
[9,54]
[62]
[7]
[7]
[9,54]
[55,56]
[54,55,56]
[61]
[62]
[59,60]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; HIV-DLBC, HIV-associated Diffuse large B-cell lymphoma; IL, interleukin; SNPs, single nucleotide polymorphisms; GM-CSF, granulocyte-macrophage colony-stimulating factor; TNF-α, tumor necrosis factor alpha; TGF-β, transforming growth factor-β; IFN-γ, interferon-γ; BAFF/BlyS, B-cell-activating factor of the TNF family/B lymphocyte Stimulator.
Table 2. Biomarkers for chemokines
Table 2. Biomarkers for chemokines
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
ChemokinesCXCL10/IP-10
CXCL11
CXCL13/BCA-1
CXCL13 SNPs
CCL2
CCL8
CCL17
CCL19
MIP-1δ
MIP-1β
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients/mice
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
Mice
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Increased
Predictive/Prognostic
Predictive
Predictive/Prognostic
Predictive
Predictive/Prognostic
Predictive
Predictive
Predictive
Predictive
Predictive
[55,57,60,84]
[57]
[7,56,59,78,79]
[56]
[7,54,60]
[57]
[7]
[57]
[57]
[54]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; IP-10, Interferon-inducible protein 10; BCA-1, B-cell-attracting chemokine 1; SNPs, single nucleotide polymorphisms; MIP, macrophage inflammatory protein.
Table 3. Biomarkers for chemokines
Table 3. Biomarkers for chemokines
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
Soluble receptorssCD23
sCD25
sCD27
sCD30
sCD163
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
Increased
Increased
Increased
Increased
Increased
Predictive
Predictive/Treatment monitoring
Predictive
Predictive
Predictive
[7,52,78,98,99,100,101]
[60]
[7,52,78,98,99,100]
[7,52,78,98,99,100]
[7,60]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; sCD, soluble cluster of differentiation.
Table 4. Microbial translocation biomarkers
Table 4. Microbial translocation biomarkers
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
Microbial translocation biomarkerssCD14
LBP
LPS
FABP2
EndoCAb
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
Increased
Increased
Increased
Increased
Decreased
Predictive/Prognostic
Predictive/Prognostic
Predictive
Predictive
Predictive
[7,8,60]
[7,8,60]
[7,8]
[7,8,60]
[7,8]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; sCD, soluble cluster of differentiation; LBP, lipopolysaccharide-binding protein; LPS, lipopolysaccharides; FABP2, fatty acid-binding protein 2; EndoCAb, Endotoxin core antibody.
Table 5. Protein-related biomarkers.
Table 5. Protein-related biomarkers.
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
Serum Protein biomarkersκ and λ FLC
Total globulin
IgA
IgE
IgG
IgM
PD-1 and PD-L1
Gal-1
β2-M
Ferritin
LDH
SAA
APO C-II
C4-A
C1Q
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HAL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL patients
HIV-NHL/HL patients
HIV-NHL/HL patients
HIV-NHL/HL patients
HIV-NHL patients
Increased
Increased
Unchanged
Unchanged
Unchanged
Unchanged
Increased
Decreased
Increased
Increased
Increased
Increased
Increased
Increased
Decreased
Predictive
Still in dispute
Unrelated
Unrelated
Unrelated
Unrelated
Predictive/Prognostic
Predictive/Prognostic
Predictive/Prognostic
Prognostic
Prognostic
Predictive/Prognostic
Predictive
Predictive
Predictive/Prognostic
[55,61,126,127,128]
[130,131]
[52,126,127]
[52,126,127]
[52,126,127]
[52,126,127]
[135,136]
[139]
[142,143]
[61,150]
[153,154,155,156,157,158,159,160,161,162,163,164]
[167,168]
[167,168]
[167,168]
[142]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; HAL, HIV-associated lymphoma; HIV-HL, HIV-associated Hodgkin’s lymphoma; FLC, free light chains; Ig, Immunoglobulin; Gal-1, Galectin-1; β2-M, beta 2-microglobulin; LDH, lactate dehydrogenase; SAA, serum amyloid A; APO C-II, apolipoprotein C-II; complement 4-A; C1Q, complement 1Q.
Table 7. Extracellular vesicle biomarkers.
Table 7. Extracellular vesicle biomarkers.
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
Extracellular VesiclesPD-L1
CD40
CD40L
TNF-RII
IL-6Ra
HIV-NHL patientsIncreasedPredictive[215,216]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; TNF-RII, TNF receptors; PD-L1, programmed cell death ligand 1; TNF-RII, TNF receptors-II.
Table 8. Lymphocyte subpopulation.
Table 8. Lymphocyte subpopulation.
Biomarker CategoryRepresentative BiomarkersSample SourceExpression TrendClinical RelevanceReferences
Lymphocyte subpopulationCD4+T cell
CD4/CD8 ratio
LMR
HIV-NHL patients
HIV-NHL patients
HIV-DLBCL patients
Decreased
Decreased
Decreased
Predictive/Prognostic
Predictive/Prognostic
Prognostic
[131,218,219,220,221,222,223,224,225,226,227,232,233,235,236]
[226,227,228,229,230,231,234,235,237]
[14,240,241]
Abbreviations: HIV-NHL, HIV-associated non-Hodgkin’s lymphoma; HIV-DLBCL, HIV-associated diffuse large B-cell lymphoma; LMR, lymphocyte-to-monocyte ratio.
Table 10. Summary table of circulating biomarkers by clinical utility.
Table 10. Summary table of circulating biomarkers by clinical utility.
Clinical UtilityTypeBiomarkersKey Features
Early Risk PredictionCytokinesIL-1α
IL-1β
IL-4
IL-5
IL-6
IL-10
IL10 SNPs
IL-11
IL-12p40
IL-13
IL-18
IL-29
GM-CSF
Neopterin
TNF-α
Associated with immune activation and inflammation; combined monitoring of cytokine levels prior to onset of disease allows early screening of at-risk individuals.
ChemokinesCXCL10/IP-10
CXCL11
CXCL13/BCA-1
CXCL SNPs
CCL2
CCL8
CCL17
CCL19
MIP-1δ
MIP-1β
Involved in B-cell migration and immunomodulation for HAL.
Soluble receptorssCD23
sCD27
sCD30
sCD163
Involved in the regulation of malignant B-cell pathology and associated with B-cell activation. sCD163 may also be involved in virus-driven carcinogenesis, especially in EBV-driven.
Microbial translocation biomarkerssCD14
LBP
LPS
FABP2
EndoCAb
Associated with immune activation and inflammation, also reflects the degree of pathological damage to the intestinal mucosa, the combined assay can be used as a biomarker for PLWH disease monitoring.
Serum protein biomarkersΚ and λ FLC
PD-1 and PD-L1
Gal-1
β2-M
SAA
APO C-II
C4-A
C1Q
Responds to abnormal B-cell activation; involved in immune regulation; predicted time points may be influenced by a variety of factors; β2-M is directly associated with active tumor cell replication and increased tumor load.
Genetic-related biomarkersmiRNA SNPs
miR-17
miR-21
miR-122
miR-106a
miR-106b
miR-223
miR-200c-3p
DNA-CNAs
Clonal Ig DNA
cfDNA
HIV-1 infection may exacerbate lymphoma progression and metastasis by remodeling the miRNA regulatory network and more significant correlation with risk of developing some specific lymphoma subtypes.
Alterations in genes promote lymphoma development.
Extracellular vesiclesPD-L1
CD40
CD40L
TNF-RII
IL-6Ra
Immunomodulatory molecules can be specifically expressed on the surface of EVs, which are involved in intercellular communication and TME regulation through ligand-receptor interactions to promote tumor progression.
Lymphocyte subpopulationCD4+T cell
CD4/CD8 ratio
Sensitive to immune reconstitution and an important independent risk factor for predicting the development of HAL.
Virus-related biomarkersHIV viraemia
p17
p17 gene variation
Tat
Nef
gp120
EBV viraemia
KSHV viraemia
HIV causes CD4+ T-cell depletion and chronic immune activation, indirectly increasing the risk of HAL. It can also directly increase cancer incidence through HIV-1’s own pro-cancer mechanisms. HIV proteins can drive carcinogenesis through multiple mechanisms.
Synergistic carcinogenic effects of EBV and KSHV.
Prognostic StratificationCytokinesIL-6
IL-7
IL-8
IL-15
TGF-β
IFN-γ
BAFF/BLyS
Elevated levels linked to poor prognosis and disease progression; multi-cytokine models offer superior predictive performance.
ChemokinesCXCL10/IP-10
CXCL13/BCA-1
CCL2
Elevated levels linked to poor prognosis and disease progression.
Microbial translocation biomarkerssCD14
LBP
Significantly negatively correlated with overall survival.
Serum protein biomarkersPD-1 and PD-L1
Gal-1
β2-M
Ferritin
LDH
SAA
C1Q
Indicators of poor prognosis. Gal-1 levels in serum are low, whereas in tumors Gal-1 shows relatively high expression and is associated with a better prognosis. Ferritin as an independent prognostic indicator. Elevated LDH is strongly associated with poorer PFS and OS. HAL patients with high SAA level expression also had longer survival periods. The levels of C1Q negatively correlate with HAL survival time.
Lymphocyte subpopulationCD4+T cell
CD4/CD8 ratio
LMR
Associated with shorter OS and PFS in HAL.
CD4/CD8 ratio <0.5 was an independent predictor of non-AIDS-related and all-cause mortality in PLWH.
Combined with IPI can further improve risk prediction and prognostic assessment accuracy.
Virus-related biomarkersHIV viraemia
EBV viraemia
Persistent hyperviremia can increase a patient’s risk of death.
Treatment MonitoringCytokinesBAFF/BLyS
TNF-α
Correlates with clinical response to treatment and survival.
ChemokinesCXCL10/IP-10
CXCL13/BCA-1
CCL2
CCL17
Associated with inflammation, which partially declines or returns to normal after therapeutic intervention, with a potential role as a dynamic prognostic marker.
Monitor disease progression and response to therapy.
Soluble receptorssCD25Dynamic changes may reflect treatment response.
Microbial translocation biomarkerssCD14
LBP
Dynamic post-treatment decline may reflect reversal of partial immune activation status by treatment.
Serum protein biomarkersFerritin
C1Q
Ferritin levels may reflect tumor microenvironmental Status, informing treatment.
C1Q decline after treatment predicts poor prognosis.
Extracellular vesiclesPD-L1
CD40
CD40L
TNF-RII
Significant reduction in plasma EVs harboring PD-L1, CD40, CD40L, or TNF-RII following treatment.
Virus-related biomarkersHIV viraemiaPost-treatment decline reduces risk of HAL development.
Abbreviations: IL, interleukin; SNPs, single nucleotide polymorphisms; GM-CSF, granulocyte-macrophage colony-stimulating factor; TNF-α, tumor necrosis factor alpha; IP-10, interferon-inducible protein 10; BCA-1, B-cell-attracting chemokine 1; MIP, macrophage inflammatory protein; HAL, HIV-associated lymphoma; sCD, soluble cluster of differentiation; EBV, Epstein–Barr virus; LBP, lipopolysaccharide-binding protein; LPS, lipopolysaccharides; FABP2, fatty acid-binding protein 2; EndoCAb, Endotoxin core antibody; PLWH, people living with HIV; FLC, free light chains; Ig, immunoglobulin; PD-1, Programmed cell death protein 1; PD-L1, programmed cell death ligand 1;Gal-1, galectin-1; β2-M, beta 2-microglobulin; SAA, serum amyloid A; APO C-II, apolipoprotein C-II; complement 4-A; complement 1Q; HIV, human immunodeficiency virus; miRNA, microRNA; DNA-CNAs, copy number alterations; cfDNA, circulating free DNA; TME, tumor microenvironment; TNF-RII, TNF receptors-II; Tat, transactivator of transcription; Nef, negative factor; KSHV, Kaposi’s sarcoma-associated herpesvirus; TGF-β, transforming growth factor-β; IFN-γ,interferon-γ; BAFF/BlyS, B-cell-activating factor of the TNF family/B lymphocyte Stimulator; LDH, lactate dehydrogenase; LMR, lymphocyte-to-monocyte ratio.
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

Shu, X.; Xiao, Q.; Liu, Y.; Li, Y.; Xie, X.; He, S.; Li, J.; Zhang, X.; Liu, Y. Circulating Biomarker Panorama in HIV-Associated Lymphoma: A Bridge from Early Risk Warning to Prognostic Stratification. Biomolecules 2025, 15, 993. https://doi.org/10.3390/biom15070993

AMA Style

Shu X, Xiao Q, Liu Y, Li Y, Xie X, He S, Li J, Zhang X, Liu Y. Circulating Biomarker Panorama in HIV-Associated Lymphoma: A Bridge from Early Risk Warning to Prognostic Stratification. Biomolecules. 2025; 15(7):993. https://doi.org/10.3390/biom15070993

Chicago/Turabian Style

Shu, Xuejiao, Qing Xiao, Yi Liu, Ya Li, Xiaoqing Xie, Sanxiu He, Jun Li, Xiaomei Zhang, and Yao Liu. 2025. "Circulating Biomarker Panorama in HIV-Associated Lymphoma: A Bridge from Early Risk Warning to Prognostic Stratification" Biomolecules 15, no. 7: 993. https://doi.org/10.3390/biom15070993

APA Style

Shu, X., Xiao, Q., Liu, Y., Li, Y., Xie, X., He, S., Li, J., Zhang, X., & Liu, Y. (2025). Circulating Biomarker Panorama in HIV-Associated Lymphoma: A Bridge from Early Risk Warning to Prognostic Stratification. Biomolecules, 15(7), 993. https://doi.org/10.3390/biom15070993

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