Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Brief Communication
Case Report
Case Series
Current Issue
Editorial
Editorial Note
FOREWORD
From the Editor's Desk
Invited Editorial
Letter to Editor
Letter to the Editor
Original Article
Original Research
Photoessay
Pictorial Review
Point of View
Review Article
Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Search in posts
Search in pages
Filter by Categories
Brief Communication
Case Report
Case Series
Current Issue
Editorial
Editorial Note
FOREWORD
From the Editor's Desk
Invited Editorial
Letter to Editor
Letter to the Editor
Original Article
Original Research
Photoessay
Pictorial Review
Point of View
Review Article
View/Download PDF

Translate this page into:

Review Article
ARTICLE IN PRESS
doi:
10.25259/FH_11_2025

The complex interplay between interleukins, biomarkers, and lymphoma: Unravelling the molecular relationships, diagnostics, and clinical implications

Department of Medical Laboratory Science, School of Basic Medical Sciences, College of Medical Sciences, Benin city, Nigeria

* Corresponding author: Dr. Nosa Terry Omorodion, Department of Medical Laboratory Science, School of Basic Medical Sciences, College of Medical Sciences, Benin city, Nigeria. terry.omorodion@uniben.edu

Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Omorodion NT, Obazelu P. The complex interplay between interleukins, biomarkers, and lymphoma: Unravelling the molecular relationships, diagnostics, and clinical implications. Future Health. doi: 10.25259/FH_11_2025

Abstract

Lymphoma, a complex group of blood cancers, varies widely in its molecular and clinical characteristics. This study delves into the critical relationship between specific interleukins and biomarkers of lymphoma, emphasizing their roles in disease progression, diagnosis, and treatment. Interleukins like IL-6, IL-10, and IL-17, along with key biomarkers such as CD20 and MYC, play crucial roles in the lymphoma’s development and its response to different treatments. By understanding how these molecular components interact, we can achieve more precise diagnoses and develop tailored treatment plans for patients. Advances in technology, including next-generation sequencing and liquid biopsies, provide exciting opportunities to enhance disease management. As the field continues to grow, future research should aim to identify new interleukins and biomarkers, which could lead to better patient classification and optimum therapies. This ongoing progress highlights the importance of sustained research and innovation in both diagnosing and treating lymphoma. Key limitations include the molecular and clinical diversity of lymphoma, incomplete understanding of interleukin and biomarker interactions, and the need for further validation through larger studies. The clinical integration of advanced technologies is still being evaluated.

Keywords

Biomarkers
Interleukins
Lymphoma
Molecular diagnostics
Personalized medicine

INTRODUCTION

Overview of lymphoma

Lymphoma encompasses a broad range of cancers that arise from the lymphatic system, which plays a vital role in the body’s immune defense. These cancers originate from the malignant transformation of lymphocytes, a type of white blood cell. Lymphomas are primarily divided into two main categories: Hodgkin lymphoma (HL) and non-Hodgkin lymphoma (NHL). Hodgkin lymphoma is characterised by the presence of Reed-Sternberg cells and tends to follow a more predictable progression.1 In contrast, non-Hodgkin lymphoma comprises various subtypes, such as B-cell and T-cell lymphomas, each with distinct clinical features and molecular characteristics.2

Among these, non-Hodgkin lymphoma is the more common form, representing approximately 4.3% of all cancer cases worldwide and a continuous increase in incidence rates.3 The diverse nature of lymphoma makes it particularly challenging to treat and manage, as its progression can vary significantly between patients. Its impact on public health is profound, not only due to its potential for widespread metastasis but also because of the difficulties it presents in treatment strategies. While lymphoma can develop in individuals of any age, its occurrence becomes more prevalent with advancing age.4 This growing incidence highlights the urgent need for further research, improved diagnostic methods, and more effective therapies tailored to the complexities of lymphoma, especially NHL.

Importance of understanding molecular mechanisms in lymphoma

Understanding the molecular mechanisms behind lymphoma is key to improving how we diagnose, treat, and predict the outcomes of the disease. Interleukins, which are signaling proteins that help regulate immune responses, play a significant role in lymphoma development and progression.5 For example, interleukin-6 (IL-6) contributes to tumor growth by increasing inflammation and supporting cell multiplication.6 On the other hand, interleukin-10 (IL-10) has immune-suppressing effects that help tumors avoid detection and attack by the immune system.7

In addition, biomarkers—such as genetic, protein-based, and epigenetic indicators—are important tools for understanding how lymphoma works and for guiding treatment decisions.8 Certain biomarkers, like CD20 and MYC, are especially important for diagnosing and managing lymphoma. For instance, CD20 is targeted by therapies in B-cell lymphomas, while changes in the MYC gene can indicate more aggressive forms of the disease.9

By understanding how interleukins and biomarkers interact in lymphoma, we can better predict disease progression and develop treatments tailored to individual patients.10 This approach supports personalized medicine, where treatment plans are customized to fit the unique needs of each patient

Interleukins and their role in lymphoma

Interleukins are proteins that act as key messengers in the immune system, regulating immune responses and inflammation. They influence the behavior of immune cells and are integral to the development and progression of lymphoma.11,12 Various interleukins play critical roles in lymphoma; for example, IL-6 levels are commonly increased in lymphoma, correlating with tumor growth and poor prognosis.13 IL-10 helps tumors survive by dampening the immune response,14 while IL-17 promotes chronic inflammation, aiding lymphoma development. IL-21 influences tumor growth and immune responses, further affecting disease progression.15,16

Mechanisms of action

Interleukins exert their effects by binding to specific receptors on target cells, triggering pathways that alter cell behavior. For instance, IL-6 binds to its receptor on immune cells, activating the JAK/STAT pathway, which promotes inflammation and helps tumor cells survive.17 Similarly, IL-10 suppresses the activation of macrophages and T cells, fostering an environment that favors tumor growth.18 Interleukins also affect the tumor microenvironment, with IL-6 altering the extracellular matrix to support tumor growth and enhance resistance to treatment.19 IL-17, through inflammation, contributes to angiogenesis in tumors, facilitating their growth and spread.20

Methodologies used in biomarker analysis

Enzyme-linked immunosorbent assay (ELISA)

ELISA is a commonly employed method for detecting and quantifying soluble proteins, including cytokines such as IL-6, IL-10, and IL-17.21 This assay operates on the principle of antigen-antibody interactions, where the antigen (e.g., a cytokine) binds to a specific antibody linked to an enzyme. The enzyme catalyzes a reaction that results in a color change, which can be measured quantitatively. One key advantage of ELISA is its high sensitivity, which allows the detection of low-abundance proteins in biological samples like blood, plasma, or serum. This makes ELISA particularly useful for profiling cytokines, as their levels can offer insights into disease progression or the effectiveness of treatments.22 This technique has become essential in both clinical and research settings due to its reliability and reproducibility in generating quantitative data.

Flow cytometry

Flow cytometry is a highly effective technique for examining cellular markers, intracellular proteins, and cytokine production on a single-cell level. Using fluorescently tagged antibodies, this method enables the rapid analysis of a large population of cells, providing a detailed profile of cellular subsets and their functional characteristics. For lymphomas, flow cytometry is commonly used to assess the expression of surface markers such as CD20, as well as intracellular cytokines like IL-6, IL-10, and IL-17 after cell stimulation. Its ability to measure multiple markers simultaneously on individual cells makes it an invaluable tool for understanding immune responses and lymphoma cell behavior. It is particularly beneficial for immunophenotyping and monitoring minimal residual disease.23

Polymerase chain reaction

PCR is a powerful technique for detecting specific genetic mutations, translocations, and biomarkers at the DNA or RNA level. In lymphoma, PCR can be used to identify genetic rearrangements, such as for MYC or BCL2, which are critical for accurate diagnosis and classification of lymphoma subtypes. Additionally, quantitative PCR (qPCR) can measure cytokine mRNA levels, offering insights into the molecular mechanisms that regulate the tumor microenvironment. PCR is a highly sensitive method, allowing the detection of minute amounts of DNA or RNA, making it an indispensable tool for understanding the genetic and transcriptomic profile of lymphoma.24

Why IL-6, IL-10, and IL-17 were chosen for discussion

The cytokines IL-6, IL-10, and IL-17 were specifically selected due to their significant roles in the pathogenesis of lymphoma and their potential as therapeutic targets. However, other interleukins, such as IL-12 and IL-23, also play important roles in immune modulation and tumor progression but were not included in this discussion. The selection of IL-6, IL-10, and IL-17 is based on their well-documented involvement in lymphoma biology.

IL-6 is a key cytokine that regulates inflammation and immune responses. It is a major driver of lymphoma progression, particularly in cases like diffuse large B-cell lymphoma (DLBCL), where elevated IL-6 levels are associated with poor prognosis. IL-6 promotes lymphoma cell survival and resistance to chemotherapy by activating pathways such as JAK/STAT, which in turn upregulate genes like MYC and BCL2. Thus, IL-6 serves as a biomarker for disease progression and a therapeutic target, with monoclonal antibodies like tocilizumab being explored for treatment.25

IL-10 is known for its immunosuppressive effects, which can enhance lymphoma survival by inhibiting anti-tumor immunity. High IL-10 levels are observed in several lymphoma subtypes, including follicular lymphoma, and contribute to the creation of an immunosuppressive microenvironment, making it difficult for the immune system to mount an effective anti-tumor response. Given its role in immune evasion, IL-10 is considered an important biomarker and a potential therapeutic target in lymphoma treatment.26

IL-17, produced by T-helper 17 (Th17) cells, is involved in inflammation and has been implicated in promoting lymphoma growth and metastasis. IL-17 levels are elevated in certain lymphoma subtypes and contribute to tumorigenesis by creating a pro-inflammatory environment that supports tumor cell proliferation and survival. Targeting IL-17 could help reduce lymphoma progression by modulating the tumor-associated inflammation.27

Although IL-12 and IL-23 are also important in the immune response, they were not the primary focus of this discussion. These cytokines are involved in the differentiation of Th1 and Th17 cells, respectively, and may influence the anti-tumor immune response. However, their role in lymphoma is still being actively studied, and clinical applications in lymphoma treatment have not been as widely explored as those of IL-6 and IL-10.28

Integrating biomarkers and interleukins in treatment

By combining profiles of interleukins with genetic and protein biomarkers, clinicians can develop more precise treatment strategies. For example, elevated IL-6 levels could lead to the use of IL-6-targeted therapies in conjunction with traditional treatments. This integrated approach provides a more comprehensive understanding of tumor biology and enables more tailored therapies. Monitoring both cytokine levels and biomarker expression during treatment also offers valuable feedback on therapeutic efficacy, which can help fine-tune treatment plans for better outcomes.29

Ongoing research into interleukins like IL-12 and IL-23 holds great promise for improving lymphoma treatment. IL-12, in particular, has been studied for its potential to enhance anti-tumor immunity through its effects on T-cells, making it an exciting target for immunotherapy. Similarly, IL-23’s role in chronic inflammation and Th17 responses could have therapeutic implications, especially in lymphoma subtypes characterized by immune dysregulation.30

With advancements in technologies like next-generation sequencing (NGS) and liquid biopsy, there are growing opportunities to better our understanding of the molecular and immune landscape of lymphoma. These methods enable continuous, non-invasive monitoring of tumor dynamics and biomarkers, offering new avenues for personalized treatment strategies. Real-time adjustments based on biomarker and interleukin profiles could pave the way for more effective and individualized lymphoma therapies.31

Case studies and research findings

Research highlights the importance of interleukins in lymphoma. McCarthy et al.32 demonstrated that inhibiting IL-6 reduced tumor growth in multiple myeloma, while Hong et al.33 found that elevated IL-10 levels are associated with poor outcomes in diffuse large B-cell lymphomas. Additionally, Wang et al.34,35 revealed how IL-17 promotes T-cell lymphoma by fostering an inflammatory environment.

Biomarkers in lymphoma

Biomarkers are biological indicators that help diagnose, monitor, and predict the progression of diseases like lymphoma.36,37 They provide insights into the molecular and cellular characteristics of lymphoma, assisting in diagnosis and personalized treatment.

Types of biomarkers

Genetic Biomarkers: These include mutations in oncogenes (like MYC) or tumor suppressor genes (like BCL2), which are linked to various lymphoma types (2). Gene expression profiling helps classify lymphoma subtypes.38

Protein Biomarkers: Proteins such as CD20 and cytokines like IL-6 indicate disease state and progression. CD20 is targeted by therapies for B-cell lymphomas, while elevated cytokines reflect immune responses.39

Epigenetic Biomarkers: Changes in gene regulation, such as DNA methylation and histone modifications, can reveal lymphoma vulnerabilities.40

Key biomarkers in lymphoma

CD20: Found on B cells, CD20 is targeted by monoclonal antibodies like rituximab in B-cell lymphomas.41

BCL2: This protein helps lymphoma cells evade apoptosis, important for diagnosis and therapy.42

MYC: An oncogene associated with aggressive lymphoma and poor prognosis.43

Clinical use of biomarkers

Diagnosis: Biomarkers like CD20 and BCL2 help differentiate lymphoma types (2).

Prognosis: Biomarkers like MYC and BCL2 levels indicate the aggressiveness of the disease.44

Treatment Monitoring: Changes in cytokine levels or tumor-specific proteins indicate treatment efficacy and disease relapse.40

Interleukins and their implications

Interleukins play a central role in lymphoma progression by modulating immune responses and tumor cell survival. IL-6, for instance, facilitates lymphoma cell proliferation and resistance to apoptosis, contributing to aggressive disease.23 IL-10 creates an immunosuppressive environment, allowing lymphoma cells to avoid immune detection.13 IL-17 maintains an inflammatory environment, supporting tumor growth, especially in T-cell lymphomas.15

Moreover, interleukins interact with biomarkers to amplify signals that drive lymphoma progression. For example, IL-6 activates the JAK/STAT pathway, which upregulates proteins like BCL2 and MYC, aiding tumor survival.45

Biomarkers and their role in treatment improvement

Biomarkers are essential in lymphoma diagnosis, prognosis, and treatment monitoring. Genetic biomarkers like MYC and BCL2 help clinicians differentiate between aggressive and indolent lymphomas.38 Proteins like CD20 are targeted in B-cell lymphomas with monoclonal antibodies like rituximab.41 Elevated cytokines such as IL-6 not only reflect lymphoma progression but also indicate inflammatory responses within the tumor.40 By integrating these biomarkers, healthcare providers can personalize treatment to improve effectiveness and minimize toxicity.

Biomarkers also assist in assessing prognosis. For example, high MYC expression is linked to more aggressive disease, guiding treatment decisions.43 Changes in cytokine levels during treatment help gauge therapy efficacy and detect early relapse.40

The interactions between interleukins and biomarkers

The relationship between interleukins and biomarkers is complex and interdependent. For example, IL-6-induced activation of the JAK/STAT pathway promotes the expression of MYC and BCL2, enhancing tumor survival and resistance to therapy.45 This creates a feedback loop where the expression of biomarkers like MYC and BCL2 further stimulates interleukin production, exacerbating tumor inflammation and growth.44 Such interactions underscore the importance of targeting both interleukins and biomarkers in treatment strategies to disrupt the tumor’s growth and inflammatory environment.

Personalization of treatment and prognosis

By incorporating interleukin and biomarker profiles, clinicians can personalize lymphoma treatment more effectively. For instance, targeting IL-6 with monoclonal antibodies or small molecule inhibitors shows promise in reducing tumor growth and inflammation.34 Combining biomarkers like MYC and IL-10 can help predict treatment responses, optimizing efficacy while minimizing side effects. This precision approach enables clinicians to select therapies tailored to each patient’s molecular landscape.

Molecular relationships between interleukins and biomarkers in lymphoma

Interleukin-Induced Biomarkers: Interleukins influence the expression of biomarkers like BCL2 and MYC in lymphoma. IL-6 activates the JAK/STAT3 pathway, enhancing cell survival,43 while IL-10 alters cytokine levels and surface markers, promoting tumor progression.14

Feedback Loops and Interactions: Elevated MYC levels can enhance interleukin production, creating a feedback loop that supports an inflammatory tumor environment.42 Similarly, CD20-positive B cells can influence cytokine levels, affecting the tumor microenvironment.41

Molecular pathways

JAK/STAT Pathway: IL-6 activates this pathway, leading to increased expression of MYC and BCL2, promoting cell survival.17

NF-kB Pathway: Both IL-6 and IL-10 activate this pathway, regulating genes involved in inflammation and immune responses.19

PI3K/Akt Pathway: IL-10 activates this pathway, enhancing cell survival and resistance to chemotherapy, interacting with biomarkers like BCL2 to support tumor growth.45-47

DISCUSSION

Case studies demonstrating diagnostic and therapeutic success

Several case studies have demonstrated the potential of integrating interleukin and biomarker profiles to improve lymphoma treatment.46 Singh et al. (2021)47 examined the use of CD20 expression and IL-6 levels in patients with diffuse large B-cell lymphoma (DLBCL), showing that this combination enhanced diagnostic accuracy and allowed for more precise monitoring of disease progression.47 By tracking individual responses, clinicians could adjust treatment more effectively. Similarly, Patel et al.48 focused on integrating IL-10 measurements with genetic markers in follicular lymphoma, which improved therapeutic decision-making and provided better patient stratification.49 These approaches highlight the importance of personalized care, with integrated biomarker and interleukin analysis offering more targeted treatment options for lymphoma patients.50-51

Limitations and future directions

Despite these promising advances, challenges remain in fully utilizing interleukins and biomarkers.52 Variability in biomarker levels, often caused by treatment effects or individual patient differences, can complicate interpretation and lead to inaccurate diagnoses or suboptimal treatment decisions.53-54 Additionally, resistance to targeted therapies is a major concern. Lymphoma cells may evolve, altering their interleukin production or biomarker expression, which can cause treatment failure and necessitate new strategies.55

The future of lymphoma treatment will depend on the continued exploration of new biomarkers and interleukins to create more dynamic, tailored therapies. Interleukins like IL-12 and IL-23 have shown potential for modulating immune responses and improving anti-tumor immunity,56 and ongoing research is investigating their roles in lymphoma development. Advancements in technologies such as next-generation sequencing (NGS) and liquid biopsies offer the opportunity for non-invasive, real-time disease monitoring. These innovations can significantly enhance clinical decision-making by providing deeper molecular insights and allowing for treatment adjustments based on ongoing disease progression.57

Recent research is expanding the field by investigating novel biomarkers and interleukins for lymphoma treatment. For instance, the combination of MYC expression profiles with interleukin levels has been shown to help predict treatment responses and disease outcomes in aggressive lymphomas, guiding more personalized therapeutic strategies.50 Personalized medicine, which tailors treatment based on individual interleukin and biomarker profiles, offers a more precise approach to lymphoma care. It allows for more effective treatment and minimizes side effects by targeting specific molecular abnormalities in each patient’s lymphoma.51

Targeted therapies have been developed to address specific interleukins and biomarkers in lymphoma treatment. Monoclonal antibodies, such as rituximab, target CD20-expressing lymphoma cells, revolutionizing the treatment of B-cell lymphomas.41 Other targeted therapies, like ibrutinib, inhibit Bruton’s tyrosine kinase and interfere with signaling pathways activated by interleukins that contribute to lymphoma cell survival.52 Drugs like venetoclax that target the overexpressed BCL2 protein, are also proving effective for lymphomas with high BCL2 expression.53 These therapies demonstrate how targeting specific biomarkers and interleukins can lead to more effective, individualized treatments.

However, variability in biomarkers and the potential for resistance to these therapies remain significant obstacles. Lymphoma cells can adapt to treatments by modifying their biomarker expression or developing mutations that reduce the effectiveness of targeted therapies, leading to relapse.54,55

The rapid advancements in molecular diagnostics, such as NGS and liquid biopsies, are reshaping how lymphoma is diagnosed and treated. NGS allows for a comprehensive analysis of genetic mutations and epigenetic changes, offering a detailed molecular landscape of lymphoma.43 Liquid biopsies, which analyze circulating tumor DNA or RNA, provide a non-invasive method for monitoring disease progression and treatment response.57 These technologies offer great promise for improving the precision of lymphoma diagnostics and monitoring, ensuring more effective management of the disease.

Future directions in clinical research

Future clinical trials should explore the potential of combining novel interleukin inhibitors with existing treatments to improve patient outcomes.58 Moreover, studies should focus on validating emerging biomarkers for predicting treatment responses and refining personalized medicine approaches. Incorporating advanced diagnostic tools, such as liquid biopsies and imaging techniques, into clinical practice will further enhance patient management and decision-making.59

Research into interleukins like IL-12 and IL-23, as well as newly discovered biomarkers, continues to hold promise for improving lymphoma diagnostics and treatment. Understanding how these molecular components interact with tumor progression will be essential for developing new therapeutic strategies. Ultimately, the goal is to integrate these insights into clinical practice, allowing for more dynamic, precise, and effective treatment options for lymphoma patients.

CONCLUSION

The integration of interleukins and biomarkers into lymphoma diagnostics and treatment represents a significant step forward in personalized medicine. These molecular tools help to refine treatment strategies, making them more targeted and effective. By analyzing interleukin profiles alongside biomarkers, clinicians can better classify lymphoma, predict treatment responses, and ultimately improve therapeutic outcomes. As research continues to uncover new interleukins and biomarkers, and as technological advancements in molecular diagnostics progress, lymphoma treatments will become increasingly individualized, leading to better patient care and improved survival rates.

Author contributions

NTO: Conceptualization, experimentation, funding acquisition, writing – original draft (introduction, results, methodology and discussion), proofreading; PO: Experimentation, funding acquisition, writing – original draft (methodology), proofreading.

Ethical approval

Institutional Review Board approval is not required.

Declaration of patient consent

Patient’s consent not required as there are no patients in this study.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

Use of artificial intelligence (AI)-assisted technology for manuscript preparation

The authors confirm that they have used Artificial Intelligence (AI)-Assisted Technology for assisting in the writing or editing of the manuscript or image creations.

References

  1. , , . Integrating MYC expression and interleukin profiles for the diagnosis and management of aggressive lymphomas. J Clin Oncol. 2023;41:2145-53.
    [Google Scholar]
  2. , , . WHO classification of tumours of haematopoietic and lymphoid tissues (4th ed). Lyon: International Agency for Research on Cancer; .
  3. . Lymphoma statistics. Cancer Research UK; . Available from: https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/lymphoma [Last accessed 2024 Mar 01]
  4. . A clinical evaluation of the international prognostic index for Hodgkin’s lymphoma. J Clin Oncol. 2017;35:1852-60.
    [Google Scholar]
  5. . New IL-6 family members. Nat Rev Immunol. 2018;18:217-28.
    [Google Scholar]
  6. , , . The role of IL-6 in lymphoma: A review. Leuk Lymphoma. 2019;60:69-77.
    [CrossRef] [PubMed] [Google Scholar]
  7. , , , . New developments in diagnosis, risk assessment and management in systemic amyloidosis. Blood Rev. 2020;40:100636.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  8. , , . Emerging biomarkers in lymphoma: Implications for treatment and prognosis. Clin Cancer Res. 2021;27:2250-7.
    [Google Scholar]
  9. , , . Targeted therapies for lymphomas: Current and future perspectives. Hematol Oncol Clin North Am. 2022;36:163-80.
    [Google Scholar]
  10. , . Personalized medicine in lymphoma: Translating molecular insights into clinical practice. Cancer Treat Rev. 2021;97:102186.
    [Google Scholar]
  11. . Overview of the interleukin-1 family of cytokines. Methods Mol Biol. 2018;1922:1-10.
    [Google Scholar]
  12. , , . Janus kinase inhibitors in autoimmune disease. Annual Review of Medicine.. 2015;66:55-67.
    [Google Scholar]
  13. , , . IL-6 and its receptors: A new therapeutic target. Nat Rev Drug Discovery. 2011;10:905-17.
    [Google Scholar]
  14. , , , . Interleukin-10 and the interleukin-10 receptor. Annu Rev Immunol. 2001;19:683-765.
    [CrossRef] [PubMed] [Google Scholar]
  15. . Structure and signalling in the IL-17 receptor family. Nat Rev Immunol. 2009;9:556-67.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  16. , , . IL-21 acts directly on B cells to enhance the antibody response. Nat Rev Immunol. 2010;10:301-14.
    [CrossRef] [PubMed] [Google Scholar]
  17. , , . Interleukin-6: A cytokine for the immune system and beyond. J Immunol. 2005;174:13-8.
    [Google Scholar]
  18. , , . Interleukin-10 as a regulatory cytokine in cancer and inflammation. J Clin Invest. 1999;104:1291-6.
    [Google Scholar]
  19. , , . The pro-inflammatory cytokine interleukin-6. Immunol Rev. 2011;239:1-16.
    [Google Scholar]
  20. , , . The role of IL-17 in rheumatoid arthritis and other inflammatory conditions. Eur J Immunol. 2003;33:17-21.
    [Google Scholar]
  21. , , . Interleukins and lymphoma development: The role of IL-6 in tumor proliferation. J Cancer Res.. 2020;45:123-134.
    [Google Scholar]
  22. , , . Enzyme immunoassay: A review. J Clin Pathol. 1994;47:347-53.
    [Google Scholar]
  23. , , . Flow cytometry: from basic principles to clinical applications. J Clin Immunol. 1999;19:337-42.
    [CrossRef] [PubMed] [Google Scholar]
  24. , , . The polymerase chain reaction. Biotechniques. 1991;10:335-8.
    [Google Scholar]
  25. , , . The role of interleukin-6 in the pathogenesis of rheumatoid arthritis. Rheumatol Int. 2008;28:697-702.
    [CrossRef] [PubMed] [Google Scholar]
  26. , . IL-10, cytokine production, and therapeutic intervention in autoimmune diseases. Immunol Res. 1999;20:21-7.
    [Google Scholar]
  27. , , . Janus kinases in immune cell signaling. Immunol Rev. 2009;228:273-87.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  28. , , . The IL-12 family of cytokines. Nat Rev Immunol. 2003;3:133-46.
    [CrossRef] [PubMed] [Google Scholar]
  29. , , . Lymphoma: The role of biomarkers in prognosis and therapy. J Clin Oncol. 2006;24:410-20.
    [Google Scholar]
  30. , , . The role of interleukin-23 in immune responses. Immunol Lett. 2009;126:1-7.
    [CrossRef] [PubMed] [Google Scholar]
  31. , , , , , , et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108-12.
    [CrossRef] [PubMed] [Google Scholar]
  32. , , . IL-6 inhibition in multiple myeloma: A review of current evidence. Hematol Oncol Clin North Am. 2022;34:539-51.
    [Google Scholar]
  33. , , . The prognostic significance of IL-10 in diffuse large B-cell lymphoma. Int J Hematol. 2011;113:200-9.
    [Google Scholar]
  34. , , . IL-17 and its role in T-cell lymphomas. Front Immunol. 2023;13:807462.
    [Google Scholar]
  35. , . Biomarkers in lymphoma: Current knowledge and future directions. Hematology Am Soc Hematol Educ Program 2020:21-31.
    [Google Scholar]
  36. . The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252-64.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  37. , , , , , , et al. Distinct types of diffuse large b-cell lymphoma identified by gene expression profiling. Nature. 2000;403:503-11.
    [CrossRef] [PubMed] [Google Scholar]
  38. , , . Protein biomarkers in lymphoma: Current applications and future directions. Leuk Lymphoma. 2020;61:1835-46.
    [Google Scholar]
  39. , , . Epigenetic biomarkers in lymphoma: Implications for diagnosis and therapy. J Hematol Oncol. 2021;14:65.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  40. , , . The role of rituximab in the treatment of non-Hodgkin’s lymphoma: A meta-analysis of randomized controlled trials. Blood. 2018;91:3040-9.
    [Google Scholar]
  41. , , . The role of BCL2 in lymphoma: Mechanistic insights and therapeutic opportunities. Blood. 2018;132:1063-74.
    [Google Scholar]
  42. , , . MYC alterations in lymphoma: Clinical implications and mechanistic insights. Blood Adv,. 2020;4:5702-14.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  43. , , . The role of IL-6 in the pathogenesis of lymphoma: Therapeutic implications. Hematol Oncol Clin North Am. 2019;33:555-69.
    [Google Scholar]
  44. , , . The PI3K/Akt pathway in lymphoma: Mechanisms and therapeutic strategies. Cancer Treat Rev. 2014;40:835-47.
    [Google Scholar]
  45. , , . Diagnostic potential of interleukins in lymphoma: Current perspectives and future directions. Oncol Rev. 2019;13:425-34.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  46. , , . The role of IL-10 in the diagnosis and management of follicular lymphoma. Hematol Rep. 2020;12:82-93.
    [Google Scholar]
  47. , , , . The role of interleukins and biomarkers in lymphoma diagnostics and personalized treatment strategies: A case study review. J Mol Diagn Ther. 2021;18:245-59.
    [Google Scholar]
  48. , , . Combined use of IL-10 and genetic biomarkers in differentiating follicular lymphoma from other B-cell lymphomas. Blood Adv. 2022;6:4563-72.
    [Google Scholar]
  49. , , . Pathogenesis of Hodgkin lymphoma: Insights into the disease mechanisms. J Hematol Oncol. 2022;15:7.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  50. , , , . Molecularly guided therapies targeting IL-6 and MYC gene mutations improve clinical outcomes. Mol Oncol. 2021;15:1025-37.
    [Google Scholar]
  51. , , , , , , et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med. 2015;373:2425-37.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  52. , , , , , , et al. Expression leading to significant clinical responses. N Engl J Med. 2016;374:311-22.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  53. , , , . Changes in biomarker expression or mutations render targeted treatments less effective. Nat Commun. 2021;12:1-12.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  54. , , . Emerging roles of interleukin-12 and interleukin-23 in lymphoma: Opportunities for new therapeutic strategies. Cytokine Growth Factor Rev. 2022;63:45-58.
    [Google Scholar]
  55. , , . The role of microRNAs in lymphoma: Current status and future directions. J Hematol Oncol. 2023;16:52.
    [CrossRef] [PubMed] [PubMed Central] [Google Scholar]
  56. , , . Liquid biopsy for cancer monitoring: Advances and challenges. Nat Rev Clin Oncol. 2022;19:484-97.
    [Google Scholar]
  57. , , . Targeting IL-12 and IL-23 pathways in lymphoma: Preclinical studies and clinical applications. J Immunother. 2021;44:155-62.
    [Google Scholar]
  58. , , . Exploring new immunotherapy combinations in lymphoma: Clinical trials and future directions. Cancer Immunol Res. 2022;10:523-36.
    [Google Scholar]
  59. , , . Innovations in molecular diagnostics for lymphoma: The promise of liquid biopsies and next-generation sequencing. Clin Cancer Res. 2023;29:145-57.
    [Google Scholar]
Show Sections