Immune Dysfunction and Consequences in Chronic Lymphocytic Leukemia

Authors:
Mayur Narkhede O’Neal Comprehensive Cancer Center at UAB, Birmingham, AL

Search for other papers by Mayur Narkhede in
Current site
Google Scholar
PubMed
Close
 MD
and
Chaitra S. Ujjani Fred Hutchinson Cancer Center, University of Washington, Seattle, WA

Search for other papers by Chaitra S. Ujjani in
Current site
Google Scholar
PubMed
Close
 MD
Full access

Infectious complications are among the leading causes of mortality in chronic lymphocytic leukemia (CLL). Over the past decade, several advances have been made in treating CLL through inhibition of Bruton tyrosine kinase and the antiapoptotic protein BCL-2. As mortality from CLL progression is expected to decline in the next several years, mortality from severe infections is anticipated to increase. Therefore, understanding the nature of immune defects in CLL and developing strategies to augment the impaired immune system are needed to keep pace with advancements in treatment. This review article summarizes the available data on immune dysfunctions, their clinical consequences, therapeutic implications, and current strategies to enhance immune function in patients with CLL.

Chronic lymphocytic leukemia (CLL) is the most common leukemia in North America and Europe, with approximately 18,740 new cases diagnosed annually in the United States.1 CLL primarily affects an aging population, with a median age at diagnosis of approximately 70 years. The disease is characterized by clonal expansion of B cells, exhibiting monoclonality with either kappa or lambda light chain restriction on the cell surface membrane. These abnormal cells accumulate in the peripheral blood, bone marrow, lymph nodes, liver, and spleen.2,3 The clinical course is highly variable. Currently, disease progression is the leading cause of mortality in CLL, followed by infectious complications.4 Over the past decade, several advancements in treatment have been made, including novel agents targeting B-cell receptor (BCR) signaling via Bruton tyrosine kinase (BTK) inhibition and promoting apoptosis through the inhibition of the antiapoptotic protein BCL-2. As a result, overall survival for patients with CLL appears to be improving.5 However, mortality from severe infections remains a significant concern.

Infectious complications in CLL arise due to underlying deficiencies in immune function, resulting from either the primary disease or the therapies used. CLL affects both the innate and adaptive immune systems, impairing the host’s ability to respond effectively to invading pathogens. The spectrum and severity of infections have changed over the years. Treatment approaches have shifted from inducing absolute lymphodepletion with either bendamustine or fludarabine-based chemotherapy to selectively targeting B-cell functioning by blocking BCR signaling. The COVID-19 pandemic further perturbated the intricate balance between watch-and-wait strategies and active treatment in CLL, presenting new challenges in determining optimal treatment regimens and vaccination strategies for patients living with CLL. Therefore, understanding the nature of immune defects in CLL and developing strategies to augment the impaired immune system are needed to keep pace with advancements in treatment approaches. This review article summarizes available data on immune dysfunctions, their clinical consequences, therapeutic implications, and current strategies to enhance immune function in patients with CLL.

Deciphering the Mechanisms: Understanding How CLL Cells Induce Immune Dysfunction

During CLL, from diagnosis to relapsed disease, there is progressive impairment of the immune system, leading to episodes of recurrent infections. Before the availability of newer diagnostic tools, frequent and severe infections from common pathogens were often the presenting symptom in patients.6 Although the spectrum of infections varies throughout the clinical course, common bacterial pathogens such as Staphylococcus aureus, Streptococcus pneumoniae, and Haemophilus influenzae—which cause lower respiratory tract infections and urinary tract infections—frequently contribute to increased morbidity and mortality. The increased incidence of infections is observed in both treatment-naïve and treatment-exposed patients, suggesting an underlying primary immune dysfunction that is further compromised by immunosuppressive antileukemia therapies.

CLL affects both the innate and adaptive immune systems, altering all aspects of a functioning immune system. Innate immunity, the initial response to invading pathogens, is compromised by qualitative and quantitative deficiencies in normal natural killer (NK) cells, neutrophils, monocytes/macrophages, and the complement system (Figure 1). Additionally, components of the adaptive response system that are dependent on humoral and cellular immunity are impaired due to ineffective antigen recognition and subsequent antibody responses. These dysfunctions result from bidirectional interactions between normal tumor microenvironment (TME) bystander cells and CLL cells, either through direct interactions or via cytokine secretion, which causes effects throughout the lymphoid system. With the treatment of CLL using targeted therapies such as BTK or BCL-2 inhibitors, there is an improvement in the functioning of the innate immune system, leading to a reduction in the severity and frequency of infections.7,8

Figure 1.
Figure 1.

Innate immune system. Inner circle (pretreatment): deficiencies in the quality and quantity of key innate immune cells, such as NK cells, neutrophils, monocytes/macrophages, and the complement system, weaken the body’s initial response to invading pathogens.6772 Outer circle (prolonged BTK ± BTK inhibitor treatment): qualitative and quantitative changes among innate immune system components, reducing infection severity and frequency.7,38,39

Abbreviations: BTK, Bruton tyrosine kinase; CLL, chronic lymphocytic leukemia; mMDSC, monocytic myeloid-derived suppressor cells; NK, natural killer.

Citation: Journal of the National Comprehensive Cancer Network 23, 3; 10.6004/jnccn.2024.7090

Replacement of Normal B Cells With Neoplastic Clonal B Cells

Because CLL is a clonal and neoplastic process, prosurvival signals are essential for the proliferation and survival of the malignant B cells. Normal B cells express BCRs composed of surface immunoglobulin with a cytoplasmic signaling domain, which is responsible for appropriate response to an antigenic epitope. In CLL, chronic tonic signaling occurs due to autoantigen-mediated BCR signaling of the stereotyped BCRs, resulting in the selection and proliferation of neoplastic B cells over normal B cells.9 Additionally, the CD5+ lymphocytes in CLL resist apoptosis through interactions with CD40L+ T cells in the TME, leading to their survival.10,11 These neoplastic cells slowly infiltrate primary and secondary lymphoid tissues, replacing functional, normal B cells. Because these neoplastic cells recognize only specific antigens, they cannot produce effective antibodies against external pathogens. However, despite its appealing simplicity, this model does not fully explain immune dysfunction during the early stages of CLL, when tumor burden is low, suggesting that alternative etiologies may contribute to B-cell impairment.

Tumor Microenvironment in CLL

The CLL TME consists of interactions between soluble components such as cytokines and chemokines and between CLL cells and bystander cells such as T cells.1114 Patients undergoing expectant observation (“watch and wait”) continue to show signs of immune dysfunction, despite slow or no progression of CLL. This suggests that the presence of CLL cells in lymphoid tissues is sufficient to suppress the immune system naturally. They achieve this indirectly through the excess production of anti-inflammatory cytokines such as IL-10 and continuous secretion of proinflammatory cytokines such as IL-6. This leads to an eventual anti-inflammatory environment that limits the host’s ability to respond to pathogens and vaccines.1518 Additionally, chemokines such as CCL17 and CCL22 secreted by CD5+ CLL cells chemo-attract CD4+ T cells, which express CD40L, aiding the neoplastic cells in resisting apoptosis.19,20

Bystander cells such as T cells, NK cells, mesenchymal stromal cells, and nurse-like cells (NLCs), which are tumor-associated macrophages/monocytes, play a critical role in the survival and chemoresistance of tumor cells and form a nourishing environment in lymphoid tissues. The bidirectional interaction between these neoplastic B cells and bystander cells drives tumor progression and recruitment of additional cells to the TME (Figure 2). There is a skewing of CD4+ helper T cells and CD8+ cytotoxic cells in CLL in favor of the latter at the expense of reduced naïve T cells, which are necessary for anti-CLL immunity.21 Despite an increase in the number of cytotoxic CD8+ T cells, their ability to exert cytotoxic effects is severely compromised. CLL cells express inhibitory surface molecules such as CD200, PD-L1, CD276, and CD270, affecting T-cell synapse formation and producing a T-cell exhausted phenotype.21,22 CD200 expressed on CLL cells impairs T-cell proliferation by inhibiting IL-2 and IFN-γ production by CD4+ T cells and promotes the differentiation of CD4+ T cells into CD4+/CD25high/FOXP3+ regulatory T cells (Tregs).23 These Treg cells secrete IL-4, which leads to overexpression of BCL-2, an antiapoptotic protein, in CLL cells, promoting their survival and increasing susceptibility to BCL-2 inhibitors such as venetoclax.24

Figure 2.
Figure 2.

Tumor microenvironment in CLL. CLL cells upregulate CD200, HVEM, and PD-1, impairing synapse formation in T cells. CD200 and IL-10 promote the differentiation of CD4+ T cells into CD4+/CD25high/FOXP3+ Tregs. These Tregs secrete IL-4, which upregulates BCL-2 expression in CLL cells, promoting prosurvival signaling. HVEM and PD-L1 interact with CD160 and PD-1 on T cells, leading to T-cell exhaustion.73 In vitro, NLCs differentiate following long-term culture with PBMCs from patients with CLL. These NLCs in the microenvironment support CLL cell survival via BAFF, CXCL12, and APRIL.74 Small-molecule inhibitors, such as BTK inhibitors and venetoclax, alter the tumor microenvironment by interacting with T cells and NLCs and reducing the CLL disease burden.7578

Abbreviations: APRIL, A proliferation-inducing ligand; BAFF, B-cell activating factor; BTK, Bruton tyrosine kinase; CLL, chronic lymphocytic leukemia; CXCL12, C-X-C motif chemokine 12; HVEM, herpes virus entry mediator; NLC, nurse-like cell; PBMC, peripheral blood mononuclear cell; Treg, regulatory T cell.

Citation: Journal of the National Comprehensive Cancer Network 23, 3; 10.6004/jnccn.2024.7090

Other cells in the TME include monocyte-derived cells (MDC), such as monocytes, macrophages, and dendritic cells. Increased levels of MDCs in the TME correlate with worse prognosis in CLL and are primarily involved in promoting CLL survival and migration through chemokine secretion.25 NLCs, which are derived from monocytes, support CLL survival by secreting factors such as B-cell activating factor (BAFF), C-X-C motif chemokine 12 (CXCL12), and A proliferation-inducing ligand (APRIL).26 Overall, CLL cells potentiate an anti-inflammatory milieu, causing migration of bystander cells into the TME, which further promotes CLL cell survival and proliferation, leading to a vicious cycle of immunosuppression and tumor proliferation.

Hypogammaglobulinemia

An objective measure of humoral immunity in CLL is the assessment of serum levels of immunoglobulins IgA, IgG, and IgM. Hypogammaglobulinemia, particularly affecting the IgG3 and IgG4 subtypes, is a prominent defect in CLL, contributing to increased susceptibility to infections.27 Decreased IgA levels are observed in up to 68% of patients with CLL, leading to recurrent respiratory tract infections and serving as a positive predictor of shorter time to infection development, the need for treatment, and decreased survival.2830 The etiology of reduced immunoglobulin secretion in CLL involves nonclonal B cells and T cells in the TME. The interactions between CD70 on clonal and nonclonal B cells and its ligand CD27 present on bystander T cells in the TME lead to decreased IgG production and plasma cell differentiation via PI3K and MEK signaling.31 CLL clonal B cells suppress plasma cell function and induce direct apoptosis of plasma cells via CD95L(FasL)/CD95(Fas) interactions between CLL cells and plasma cells, leading to hypogammaglobulinemia.32 Therefore, the overall consequence is reduced differentiation toward plasma cells and reduced production of all classes of immunoglobulins.

Clinical Ramifications of Immune Dysfunction in CLL

The intricate interplay between CLL and the immune system is complex and cannot be elucidated in a simplified model. However, its effects are routinely experienced by patients, affecting both their quality and quantity of life. These effects can be grouped into 3 major areas: increased susceptibility to infections, development of secondary primary malignancies, and the emergence of autoimmune disorders associated with CLL.

Infection Risk and Spectrum in CLL

Infections are the second most common cause of mortality in CLL, with their incidence increasing based on disease stage, serum IgG levels, mutations in CLL cells, and the treatments used.5,33 The 5-year risk of developing severe infections increases up to 68% from a baseline risk of 26% in patients with IgG levels <650 mg/dL and Rai stage C disease.34 The most frequent severe infections include pneumonia, followed by sepsis and urinary tract infections caused by encapsulated bacteria such as Streptococcus pneumoniae, Haemophilus influenzae, and Neisseria meningitides, as well as Staphylococcus aureus and Enterobacteriaceae.35 Viral agents, including herpes simplex virus (HSV) and varicella-zoster virus, become more prevalent during antileukemia treatment and occasionally require chronic suppressive therapy during the treatment course.

In 2019, the COVID-19 pandemic challenged health care systems and public policies worldwide, with a particularly severe impact on elderly patients and those with comorbidities or suppressed immune systems. SARS-CoV-2 infection has a variable clinical course but is associated with a high case fatality rate, approaching 33% in patients with CLL.16 This high fatality rate was observed in patients undergoing watch-and-wait observation as well as those undergoing treatment. A high CD8+ T-cell count is associated with lower mortality in SARS-CoV-2 infections. However, in CLL, the CD8+ T cells exhibit functional defects in proliferation and cytotoxicity, making them functionally incompetent.36,37 Prolonged viremia with SARS-CoV-2 is observed in patients with CLL with impaired ability to produce pathogen-specific antibodies, as seen in treatment-naïve patients and those receiving B-cell–depletion therapy with anti-CD20 monoclonal antibodies such as rituximab or obinutuzumab. This prolonged viremia contributes to increased hospital readmissions and higher mortality in patients with CLL and SARS-CoV-2 infection. Multiple resurgences of SARS-CoV-2 infection with newer strains have caused patients to experience crippling fear and anxiety about developing severe infection, often leading to prolonged periods of social isolation and increased rates of depression.

The challenges faced by patients with CLL during the COVID-19 pandemic, particularly the heightened risk of severe infection and mortality, underscore the critical need for therapeutic strategies that can bolster the immune system and reduce susceptibility to infections. Long-term treatment with venetoclax and ibrutinib has been shown to result in immune recovery.38,39 This recovery is dependent on the eradication of CLL cells and restoration of normal B cells, T-cell subtypes, monocytes, and dendritic cells.7 The effect of this immune system recovery on infection risk and mortality is currently being investigated in the PreVent-ACaLL phase II trial of acalabrutinib combined with venetoclax for high-risk CLL (ClinicalTrials.gov identifier: NCT03868722). These findings suggest that targeted therapies not only have the potential to induce remission but may also create an opportunity to enhance treatment responses with immunotherapy and reduce infection risk in this patient population.

Incidence of Second Primary Malignancies in CLL

The risk of developing subsequent primary malignancies (SPMs) in patients with CLL is 3 times higher compared with age- and sex-matched controls.40,41 Among SPMs, the highest risk is observed for nonmelanoma skin cancers, with up to an 8-fold increase, followed by a 2-fold increase in other solid tumors, such as malignant melanoma, soft tissue sarcomas, and lung cancer.42,43 The incidence of these SPMs matches that seen in renal transplant recipients, suggesting immunodeficiency as a primary etiology. High-count monoclonal B-cell lymphocytosis, a precursor to CLL, is also associated with an increased risk of nonhematologic cancers compared with healthy individuals, suggesting that the immune dysfunction contributing to the development of SPMs occurs early in the course of CLL.44 The risk of SPMs further increases in patients who receive chemotherapy compared with those who remain untreated, suggesting a model of baseline immune dysfunction in CLL that is further aggravated by subsequent therapies. Even with newer targeted therapies, such as BTK inhibitors and BCL-2 inhibitors, the risk of SPM is as high as 9%.41,4547 Furthermore, the presence of preexisting CLL in patients with SPMs is associated with inferior overall survival and cancer-specific survival, indicating a general state of impaired cancer immunosurveillance in CLL.48

Autoimmune Diseases in CLL

Autoimmunity is a relatively common complication in CLL, with 20% to 25% of patients developing hematologic or nonhematologic autoimmunity over the disease course.49 Autoimmune hemolytic anemia (AIHA) is the most common autoimmune manifestation, followed by immune thrombocytopenia, pure red cell aplasia, and autoimmune neutropenia. Nonhematologic autoimmune disorders, such as paraneoplastic pemphigus, angioedema, and paraneoplastic glomerular disease with proteinuria, occur less frequently and are often observed in patients with stable CLL.50,51 The pathogenesis of paraneoplastic and autoimmune disorders in CLL is complex. It is thought to result from a combination of abnormal antigen presentation by CLL cells during their interactions with helper T cells, which misidentify self-antigens as foreign, and the failure of Tregs to regulate an autoimmune response to self-antigens.52 In the past, treatment with purine analogs such as chlorambucil or fludarabine also led to an increased incidence of AIHA, possibly due to their lymphodepleting effects on Tregs.53 Autoimmune cytopenias (AICs) have also been reported with novel agents, occurring in 9% of patients, typically within the first 6 months of treatment.53 The German CLL Study Group reported AIC incidences of 8% with venetoclax, 2% with BTK inhibitors, and 15% with venetoclax in combination with BTK inhibitor. Regardless of the underlying etiology, the treatment of autoimmune disorders in CLL requires both suppression of interactions between autoantigens and the innate immune system using corticosteroids and, in some cases, eradication of malignant B-cell clones through antileukemia therapies.25

Navigating Implications of Immune Challenges in CLL

In navigating the complex landscape of immune dysfunction and its clinical consequences in CLL, specific strategies have been explored to minimize the effects of a suppressed immune system and even use immunotherapy to develop newer antileukemia treatments. Current approaches include reducing infectious complications through vaccination against preventable infections, repleting protective antibodies using donor-derived antibodies from pooled plasma of healthy donors, and using chronic suppressive anti-infective therapy during antileukemia treatment. Although these strategies show promise, each comes with its own limitations.

Vaccinations in CLL

The potential to enhance adaptive immunity in CLL through vaccinations against pathogens such as pneumococcal, Haemophilus influenzae type b, tetanus, varicella, and SARS-CoV-2 has been extensively investigated.54 However, due to defective antigen presentation and reduced antibody production in CLL, seroconversion rates are poor. Furthermore, patients with advanced disease or low IgG levels have suboptimal responses to immunization, indicating the need to vaccinate early at diagnosis.55 Anti-CD20 monoclonal antibodies and BTK inhibitor therapy further decrease rates of seroconversion.56,57 With SARS-CoV-2 mRNA vaccines, treatment-naïve patients and those receiving B-cell–directed therapy, including BTK and BCL-2 inhibitors, demonstrate poor seroconversion rates, decreased neutralizing antibodies against SARS-CoV-2 spike proteins, and diminished CD4 T-cell responses.58,59 However, no studies have shown that vaccinations cause harm by worsening disease or triggering new, untoward adverse events. Therefore, vaccinations should be discussed at diagnosis and administered ≥2 weeks before treatment initiation.60 Influenza, SARS-CoV-2, pneumococcal, and respiratory syncytial virus vaccines are recommended for all patients. Additionally, recombinant zoster vaccines are indicated for those receiving a BTK inhibitor.61 Live virus vaccines should be avoided.

Immunoglobulin Replacement Therapy

As mentioned previously, hypogammaglobulinemia is a common occurrence in patients with CLL, present either at diagnosis or as a result of B-cell–directed therapy. The use of prophylactic intravenous immunoglobin (IVIG) replacement therapy has been extensively studied and is used in selected patients at risk for severe infections. In a randomized placebo-controlled trial involving patients with CLL and ≥3 sinus infections or 1 episode of pneumonia and low IgG serum levels, administration of 400 mg/kg IVIG every 3 weeks for at least 1 year reduced the incidence of minor to moderate bacterial infections but had no effect on severe infections or mortality rates.62 Similarly, a more recent trial of subcutaneous immunoglobulin replacement therapy showed improved serum IgG levels specific to Streptococcus pneumoniae.63 Other studies continue to show minor benefits without effect on quality of life or increase in overall survival. Clinical studies suggest that lower IgA levels, rather than IgG, are a better predictor of infection risk, raising further questions about the value of IgG replacement therapy.29,30 Therefore, the use of IVIG is controversial and recommended only in cases of recurrent infections with low serum IgG levels.61

Antimicrobial Prophylaxis

Antimicrobial prophylaxis should be individualized based on patient history and risk factors. In general, primary prophylaxis is not indicated for patients receiving BTK or BCL-2 inhibitors, although prophylaxis for Pneumocystis jiroveci pneumonia (PJP) and HSV can be considered for select patients.61 Treatment with BTK inhibitors may increase the risk of invasive fungal infections, such as aspergillosis; however, this risk is low and does not warrant antifungal prophylaxis for all patients. If a patient receiving a BTK inhibitor develops respiratory decompensation or other symptoms suggestive of a fungal infection, testing for serum galactomannan and a chest CT scan should be considered as part of the diagnostic workup. Because venetoclax is associated with significant neutropenia, typically within the first few months of initiation, we recommend early use of granulocyte colony-stimulating factor (G-CSF) and, for patients with prolonged neutropenia (>7 days), consideration of fluoroquinolone and fungal prophylaxis. Entecavir prophylaxis and close monitoring of hepatitis B viral load by quantitative RT-PCR are recommended for patients at high risk for reactivation (eg, hepatitis B surface antigen–positive or hepatitis B core antibody–positive). Although less frequently used in CLL, current or prior treatment with chemoimmunotherapy (purine analogs or bendamustine), phosphoinositide 3-kinase (PI3K) inhibitors, or alemtuzumab warrants HSV and PJP prophylaxis. Additionally, patients receiving PI3K inhibitors or alemtuzumab have a high risk of cytomegalovirus reactivation and should be monitored closely; ganciclovir and consultation with infectious disease specialists may be indicated for managing viremia.

Infection Management

BTK inhibitors and venetoclax-based regimens can typically be continued during the treatment of low-grade infections, with the caveat that antimicrobial therapies should be checked for potential drug–drug interactions. Therapy interruption should be considered for patients who develop severe infections (grade ≥3), with resumption once clinical improvement is evident.6466 The timing of resumption is individualized, depending on the patient and the type of infection. Caution should be taken with patients receiving a BTK inhibitor, particularly early in their course, as dose interruptions may lead to disease progression. The development of multiple significant infections may warrant a dose reduction. In addition to appropriate antimicrobial therapy, other supportive measures such as G-CSF or IVIG may be considered if supported by the patient’s laboratory values and clinical history.

Conclusions

CLL presents significant challenges due to its impact on the immune system, leading to increased susceptibility to infections, a higher risk of secondary malignancies, and the development of autoimmune disorders. Despite advancements in treatment, mortality from infections remains a prominent concern, especially in the context of the COVID-19 pandemic. The intricate interactions between CLL cells, the immune system, and the TME contribute to immune dysfunction, affecting both innate and adaptive immunity. The clinical consequences of these dysfunctions underscore the importance of comprehensively addressing immune challenges in CLL. Strategies such as vaccinations, immunoglobulin replacement therapy, and antimicrobial prophylaxis have been explored to mitigate infectious complications, but their efficacy is variable. Navigating the implications of immune challenges in CLL requires a nuanced understanding of the disease’s immune dynamics and ongoing research to develop more effective therapeutic approaches. Despite the complexities involved, efforts to enhance immune function and minimize associated risks remain crucial for improving the quality of life and clinical outcomes for individuals living with CLL.

References

  • 1.

    Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin 2023;73:1748.

  • 2.

    Eichhorst B, Robak T, Montserrat E, et al. Chronic lymphocytic leukaemia: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2021;32:2333.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood 2018;131:27452760.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Strati P, Parikh SA, Chaffee KG, et al. Relationship between co-morbidities at diagnosis, survival and ultimate cause of death in patients with chronic lymphocytic leukaemia (CLL): a prospective cohort study. Br J Haematol 2017;178:394402.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5.

    Muthiah C, Narra R, Atallah E, et al. Evaluating population-level outcomes in chronic lymphocytic leukemia in the era of novel therapies using the SEER registry. Leuk Res 2024;140:107496.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6.

    Forconi F, Moss P. Perturbation of the normal immune system in patients with CLL. Blood 2015;126:573581.

  • 7.

    Svanberg Teglgaard R, Marquart HV, Hartling HJ, et al. Improved innate immune function in patients with chronic lymphocytic leukemia treated with targeted therapy in clinical trials. Clin Cancer Res 2024;30:19591971.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8.

    Purroy N, Tong YE, Lemvigh CK, et al. Single-cell analysis reveals immune dysfunction from the earliest stages of CLL that can be reversed by ibrutinib. Blood 2022;139:22522256.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Burger JA, Chiorazzi N. B cell receptor signaling in chronic lymphocytic leukemia. Trends Immunol 2013;34:592601.

  • 10.

    Furman RR, Asgary Z, Mascarenhas JO, et al. Modulation of NF-κB activity and apoptosis in chronic lymphocytic leukemia B cells. J Immunol 2000;164:22002206.

  • 11.

    Herishanu Y, Katz BZ, Lipsky A, Wiestner A. Biology of chronic lymphocytic leukemia in different microenvironments: clinical and therapeutic implications. Hematol Oncol Clin North Am 2013;27:173206.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Shain K, Dalton W, Tao J. The tumor microenvironment shapes hallmarks of mature B-cell malignancies. Oncogene 2015;34:46734682.

  • 13.

    Dubois N, Crompot E, Meuleman N, et al. Importance of crosstalk between chronic lymphocytic leukemia cells and the stromal microenvironment: direct contact, soluble factors, and extracellular vesicles. Front Oncol 2020;10:1422.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Landeira-Viñuela A, Arias-Hidalgo C, Juanes-Velasco P, et al. Unravelling soluble immune checkpoints in chronic lymphocytic leukemia: physiological immunomodulators or immune dysfunction. Front Immunol 2022;13:965905.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    DiLillo DJ, Weinberg JB, Yoshizaki A, et al. Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence and immunosuppressive function. Leukemia 2013;27:170182.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Garaud S, Morva A, Lemoine S, et al. CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation. J Immunol 2011;186:48354844.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Herishanu Y, Avivi I, Aharon A, et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood 2021;137:31653173.

  • 18.

    Whitaker JA, Parikh SA, Shanafelt TD, et al. The humoral immune response to high-dose influenza vaccine in persons with monoclonal B-cell lymphocytosis (MBL) and chronic lymphocytic leukemia (CLL). Vaccine 2021;39:11221130.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Ghia P, Strola G, Granziero L, et al. Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22. Eur J Immunol 2002;32:14031413.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Zaaboub R, Vimeux L, Contremoulins V, et al. Nurselike cells sequester B cells in disorganized lymph nodes in chronic lymphocytic leukemia via alternative production of CCL21. Blood Adv 2022;6:46914704.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Palma M, Gentilcore G, Heimersson K, et al. T cells in chronic lymphocytic leukemia display dysregulated expression of immune checkpoints and activation markers. Haematologica 2017;102:562572.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Ramsay AG, Clear AJ, Fatah R, Gribben JG. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood 2012;120:14121421.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Mpakou VE, Ioannidou HD, Konsta E, et al. Quantitative and qualitative analysis of regulatory T cells in B cell chronic lymphocytic leukemia. Leuk Res 2017;60:7481.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Jak M, Mous R, Remmerswaal E, et al. Enhanced formation and survival of CD4+ CD25hi Foxp3+ T-cells in chronic lymphocytic leukemia. Leuk Lymphoma 2009;50:788801.

  • 25.

    Gustafson MP, Abraham RS, Lin Y, et al. Association of an increased frequency of CD14+HLA-DRlo/neg monocytes with decreased time to progression in chronic lymphocytic leukaemia (CLL). Br J Haematol 2012;156:674676.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Talbot H, Saada S, Barthout E, et al. BDNF belongs to the nurse-like cell secretome and supports survival of B chronic lymphocytic leukemia cells. Sci Rep 2020;10:12572.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Sourmerai J, Yousif Z, Gift T, et al. Patterns of IgG testing and rates of hypogammaglobulinemia in patients with chronic lymphocytic leukemia or small lymphocytic lymphoma. Clin Immunol 2023;250:109601.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Ishdorj G, Streu E, Lambert P, et al. IgA levels at diagnosis predict for infections, time to treatment, and survival in chronic lymphocytic leukemia. Blood Adv 2019;3:21882198.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Andersen MA, Eriksen CT, Brieghel C, et al. Incidence and predictors of infection among patients prior to treatment of chronic lymphocytic leukemia: a Danish nationwide cohort study. Haematologica 2018;103:e300303.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Sun C, Tian X, Lee YS, et al. Partial reconstitution of humoral immunity and fewer infections in patients with chronic lymphocytic leukemia treated with ibrutinib. Blood 2015;126:22132219.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Arens R, Nolte MA, Tesselaar K, et al. Signaling through CD70 regulates B cell activation and IgG production. J Immunol 2004;173:39013908.

  • 32.

    Sampalo A, Navas G, Medina F, et al. Chronic lymphocytic leukemia B cells inhibit spontaneous Ig production by autologous bone marrow cells: role of CD95-CD95L interaction. Blood 2000;96:31683174.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Else M, Blakemore SJ, Strefford JC, Catovsky D. The association between deaths from infection and mutations of the BRAF, FBXW7, NRAS and XPO1 genes: a report from the LRF CLL4 trial. Leukemia 2021;35:25632569.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Molica S, Levato D, Levato L. Infections in chronic lymphocytic leukemia. Analysis of incidence as a function of length of follow-up. Haematologica 1993;78:374377.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Soumerai J, Gift T, Yousif Z, et al. Infection outcomes and hypogammaglobulinemia in patients with chronic lymphocytic leukemia treated with immunoglobulin replacement therapy. Blood 2023;142:3280.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Bange EM, Han NA, Wileyto P, et al. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer. Res Sq. Preprint posted online February 2, 2021. doi: 10.21203/rs.3.rs-162289/v1

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Riches JC, Davies JK, McClanahan F, et al. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood 2013;121:16121621.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Moreno C, Solman IG, Tam CS, et al. Immune restoration with ibrutinib plus venetoclax in first-line chronic lymphocytic leukemia: the phase 2 CAPTIVATE study. Blood Adv 2023;7:52945303.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Davis JE, Handunnetti SM, Ludford-Menting M, et al. Immune recovery in patients with mantle cell lymphoma receiving long-term ibrutinib and venetoclax combination therapy. Blood Adv 2020;4:48494859.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Manusow D, Weinerman BH. Subsequent neoplasia in chronic lymphocytic leukemia. JAMA 1975;232:267269.

  • 41.

    Bond DA, Huang Y, Fisher JL, et al. Second cancer incidence in CLL patients receiving BTK inhibitors. Leukemia 2020;34:31973205.

  • 42.

    Tsimberidou AM, Wen S, McLaughlin P, et al. Other malignancies in chronic lymphocytic leukemia/small lymphocytic lymphoma. J Clin Oncol 2009;27:904910.

  • 43.

    Kumar V, Ailawadhi S, Bojanini L, et al. Trends in the risk of second primary malignancies among survivors of chronic lymphocytic leukemia. Blood Cancer J 2019;9:75.

  • 44.

    Solomon BM, Chaffee KG, Moreira J, et al. Risk of non-hematologic cancer in individuals with high-count monoclonal B-cell lymphocytosis. Leukemia 2016;30:331336.

  • 45.

    Shen Y, Coyle L, Kerridge I, et al. Second primary malignancies in chronic lymphocytic leukaemia: skin, solid organ, haematological and Richter’s syndrome. EJHaem 2022;3:129138.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Al-Sawaf O, Zhang C, Jin HY, et al. Transcriptomic profiles and 5-year results from the randomized CLL14 study of venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab in chronic lymphocytic leukemia. Nat Commun 2023;14:2147.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Byrd JC, Hillmen P, O’Brien S, et al. Long-term follow-up of the RESONATE phase 3 trial of ibrutinib vs ofatumumab. Blood 2019;133:20312042.

  • 48.

    Solomon BM, Rabe KG, Slager SL, et al. Overall and cancer-specific survival of patients with breast, colon, kidney, and lung cancers with and without chronic lymphocytic leukemia: a SEER population-based study. J Clin Oncol 2013;31:930937.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Hamblin TJ. Autoimmune complications of chronic lymphocytic leukemia. Semin Oncol 2006;33:230239.

  • 50.

    Cao L, Wang F, Du XY, et al. Chronic lymphocytic leukemia-associated paraneoplastic pemphigus: potential cause and therapeutic strategies. Sci Rep 2020;10:16357.

  • 51.

    Strati P, Nasr SH, Leung N, et al. Renal complications in chronic lymphocytic leukemia and monoclonal B-cell lymphocytosis: the Mayo Clinic experience. Haematologica 2015;100:11801188.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Hall AM, Vickers MA, McLeod E, Barker RN. Rh autoantigen presentation to helper T cells in chronic lymphocytic leukemia by malignant B cells. Blood 2005;105:20072015.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53.

    Borthakur G, O’Brien S, Wierda WG, et al. Immune anaemias in patients with chronic lymphocytic leukaemia treated with fludarabine, cyclophosphamide and rituximab–incidence and predictors. Br J Haematol 2007;136:800805.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54.

    Wang KY, Shah P, Skavla B, et al. Vaccination efficacy in patients with chronic lymphocytic leukemia. Leuk Lymphoma 2023;64:4256.

  • 55.

    Mauro FR, Giannarelli D, Galluzzo CM, et al. Response to the conjugate pneumococcal vaccine (PCV13) in patients with chronic lymphocytic leukemia (CLL). Leukemia 2021;35:737746.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56.

    Vijenthira A, Gong I, Betschel SD, et al. Vaccine response following anti-CD20 therapy: a systematic review and meta-analysis of 905 patients. Blood Adv 2021;5:26242643.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57.

    Andrick B, Alwhaibi A, DeRemer DL, et al. Lack of adequate pneumococcal vaccination response in chronic lymphocytic leukaemia patients receiving ibrutinib. Br J Haematol 2018;182:712714.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58.

    Ujjani C, Gooley TA, Spurgeon SE, et al. Diminished humoral and cellular responses to SARS-CoV-2 vaccines in patients with chronic lymphocytic leukemia. Blood Adv 2023;7:47284737.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 59.

    Qin K, Honjo K, Sherrill-Mix S, et al. Exposure of progressive immune dysfunction by SARS-CoV-2 mRNA vaccination in patients with chronic lymphocytic leukemia: a prospective cohort study. PLoS Med 2023;20:e1004157.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 60.

    Baden LR, Swaminathan S, Almyroudis NG, et al. NCCN Clinical Practice Guidelines in Oncology: Prevention and Treatment of Cancer-Related Infections. Version 3.2024. To view the most recent version, visit https://www.nccn.org/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 61.

    Wierda WG, Brown J, Abramson JS, et al. NCCN Clinical Practice Guidelines in Oncology: Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. Version 1.2025. To view the most recent version, visit https://www.nccn.org/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 62.

    Gale RP, Chapel HM, Bunch C, et al. Intravenous immunoglobulin for the prevention of infection in chronic lymphocytic leukemia. A randomized, controlled clinical trial. N Engl J Med 1988;319:902907.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 63.

    Mustafa SS, Jamshed S, Vadamalai K, Ramsey A. Subcutaneous immunoglobulin replacement for treatment of humoral immune dysfunction in patients with chronic lymphocytic leukemia. PLoS One 2021;16:e0258529.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 64.

    Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): a randomised, controlled, phase 3 trial. Lancet 2020;395:12781291.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65.

    Tam CS, Brown JR, Kahl BS, et al. Zanubrutinib versus bendamustine and rituximab in untreated chronic lymphocytic leukaemia and small lymphocytic lymphoma (SEQUOIA): a randomised, controlled, phase 3 trial. Lancet Oncol 2022;23:10311043.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66.

    Al-Sawaf O, Zhang C, Tandon M, et al. Venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab for previously untreated chronic lymphocytic leukaemia (CLL14): follow-up results from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2020;21:11881200.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67.

    Dearden C. Disease-specific complications of chronic lymphocytic leukemia. Hematology Am Soc Hematol Educ Program 2008;2008:450456.

  • 68.

    Arruga F, Gyau BB, Iannello A, et al. Immune response dysfunction in chronic lymphocytic leukemia: dissecting molecular mechanisms and microenvironmental conditions. Int J Mol Sci 2020;21:1825.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69.

    Jurado-Camino T, Cordoba R, Esteban-Burgos L, et al. Chronic lymphocytic leukemia: a paradigm of innate immune cross-tolerance. J Immunol 2015;194:719727.

  • 70.

    Manukyan G, Papajik T, Gajdos P, et al. Neutrophils in chronic lymphocytic leukemia are permanently activated and have functional defects. Oncotarget 2017;8:8488984901.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71.

    Toniolo PA, Liu S, Yeh JE, et al. Deregulation of SOCS5 suppresses dendritic cell function in chronic lymphocytic leukemia. Oncotarget 2016;7:4630146314.

  • 72.

    MacFarlane IA, Jillab M, Smith MR, et al. NK cell dysfunction in chronic lymphocytic leukemia is associated with loss of the mature cells expressing inhibitory killer cell Ig-like receptors. Oncoimmunology 2017;6:e1330235.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 73.

    Roessner PM, Seiffert M. T-cells in chronic lymphocytic leukemia: guardians or drivers of disease? Leukemia 2021;35:36342024.

  • 74.

    Puente XS, Jares P, Campo E. Chronic lymphocytic leukemia and mantle cell lymphoma: crossroads of genetic and microenvironment interactions. Blood 2018;131:22832296.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 75.

    Long M, Beckwith K, Do P, et al. Ibrutinib treatment improves T cell number and function in CLL patients. J Clin Invest 2017;127:30523064.

  • 76.

    Zou YX, Zhu HY, Li XT, et al. The impacts of zanubrutinib on immune cells in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma. Hematol Oncol 2019;37:392400.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 77.

    Niemann CU, Herman SE, Maric I, et al. Disruption of in vivo chronic lymphocytic leukemia tumor–microenvironment interactions by ibrutinib–findings from an investigator-initiated phase II study. Clin Cancer Res 2016;22:15721582.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78.

    de Weerdt I, Hofland T, de Boer R, et al. Distinct immune composition in lymph node and peripheral blood of CLL patients is reshaped during venetoclax treatment. Blood Adv 2019;3:26422652.

    • PubMed
    • Search Google Scholar
    • Export Citation

Submitted February 23, 2024; final revision received October 20, 2024; accepted for publication November 18, 2024.

Disclosures: Dr. Narkhede has disclosed receiving grant/research support from Natera, Genmab, Genentech, Gilead, Gilead/Forty-Seven, EUSA Pharma, Caribou Biosciences, BeiGene, and Cullinan Therapeutics; and serving as a scientific advisor for ADC Therapeutics, Kite Pharma, T.G Therapeutics, BeiGene, AstraZeneca, Adaptive Biotechnologies, AbbVie, and Pharmacyclics. Dr. Ujjani has disclosed receiving grant/research support from AbbVie, AstraZeneca, Lilly, and Gilead; and serving as a consultant for AbbVie, AstraZeneca, Allogene Therapeutics, Ascentage Pharma, Atara Biotherapeutics, Bristol Myers Squibb, BeiGene, Genentech, Janssen Pharmaceuticals, and Pharmacyclics.

Correspondence: Chaitra S. Ujjani, MD, Fred Hutchinson Cancer Center, University of Washington, 825 Eastlake Avenue E, CE3-300, Seattle, WA 98109. Email: Ujjani@uw.edu
  • Collapse
  • Expand
  • Figure 1.

    Innate immune system. Inner circle (pretreatment): deficiencies in the quality and quantity of key innate immune cells, such as NK cells, neutrophils, monocytes/macrophages, and the complement system, weaken the body’s initial response to invading pathogens.6772 Outer circle (prolonged BTK ± BTK inhibitor treatment): qualitative and quantitative changes among innate immune system components, reducing infection severity and frequency.7,38,39

    Abbreviations: BTK, Bruton tyrosine kinase; CLL, chronic lymphocytic leukemia; mMDSC, monocytic myeloid-derived suppressor cells; NK, natural killer.

  • Figure 2.

    Tumor microenvironment in CLL. CLL cells upregulate CD200, HVEM, and PD-1, impairing synapse formation in T cells. CD200 and IL-10 promote the differentiation of CD4+ T cells into CD4+/CD25high/FOXP3+ Tregs. These Tregs secrete IL-4, which upregulates BCL-2 expression in CLL cells, promoting prosurvival signaling. HVEM and PD-L1 interact with CD160 and PD-1 on T cells, leading to T-cell exhaustion.73 In vitro, NLCs differentiate following long-term culture with PBMCs from patients with CLL. These NLCs in the microenvironment support CLL cell survival via BAFF, CXCL12, and APRIL.74 Small-molecule inhibitors, such as BTK inhibitors and venetoclax, alter the tumor microenvironment by interacting with T cells and NLCs and reducing the CLL disease burden.7578

    Abbreviations: APRIL, A proliferation-inducing ligand; BAFF, B-cell activating factor; BTK, Bruton tyrosine kinase; CLL, chronic lymphocytic leukemia; CXCL12, C-X-C motif chemokine 12; HVEM, herpes virus entry mediator; NLC, nurse-like cell; PBMC, peripheral blood mononuclear cell; Treg, regulatory T cell.

  • 1.

    Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin 2023;73:1748.

  • 2.

    Eichhorst B, Robak T, Montserrat E, et al. Chronic lymphocytic leukaemia: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2021;32:2333.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3.

    Hallek M, Cheson BD, Catovsky D, et al. iwCLL guidelines for diagnosis, indications for treatment, response assessment, and supportive management of CLL. Blood 2018;131:27452760.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4.

    Strati P, Parikh SA, Chaffee KG, et al. Relationship between co-morbidities at diagnosis, survival and ultimate cause of death in patients with chronic lymphocytic leukaemia (CLL): a prospective cohort study. Br J Haematol 2017;178:394402.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5.

    Muthiah C, Narra R, Atallah E, et al. Evaluating population-level outcomes in chronic lymphocytic leukemia in the era of novel therapies using the SEER registry. Leuk Res 2024;140:107496.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6.

    Forconi F, Moss P. Perturbation of the normal immune system in patients with CLL. Blood 2015;126:573581.

  • 7.

    Svanberg Teglgaard R, Marquart HV, Hartling HJ, et al. Improved innate immune function in patients with chronic lymphocytic leukemia treated with targeted therapy in clinical trials. Clin Cancer Res 2024;30:19591971.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8.

    Purroy N, Tong YE, Lemvigh CK, et al. Single-cell analysis reveals immune dysfunction from the earliest stages of CLL that can be reversed by ibrutinib. Blood 2022;139:22522256.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Burger JA, Chiorazzi N. B cell receptor signaling in chronic lymphocytic leukemia. Trends Immunol 2013;34:592601.

  • 10.

    Furman RR, Asgary Z, Mascarenhas JO, et al. Modulation of NF-κB activity and apoptosis in chronic lymphocytic leukemia B cells. J Immunol 2000;164:22002206.

  • 11.

    Herishanu Y, Katz BZ, Lipsky A, Wiestner A. Biology of chronic lymphocytic leukemia in different microenvironments: clinical and therapeutic implications. Hematol Oncol Clin North Am 2013;27:173206.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Shain K, Dalton W, Tao J. The tumor microenvironment shapes hallmarks of mature B-cell malignancies. Oncogene 2015;34:46734682.

  • 13.

    Dubois N, Crompot E, Meuleman N, et al. Importance of crosstalk between chronic lymphocytic leukemia cells and the stromal microenvironment: direct contact, soluble factors, and extracellular vesicles. Front Oncol 2020;10:1422.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14.

    Landeira-Viñuela A, Arias-Hidalgo C, Juanes-Velasco P, et al. Unravelling soluble immune checkpoints in chronic lymphocytic leukemia: physiological immunomodulators or immune dysfunction. Front Immunol 2022;13:965905.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    DiLillo DJ, Weinberg JB, Yoshizaki A, et al. Chronic lymphocytic leukemia and regulatory B cells share IL-10 competence and immunosuppressive function. Leukemia 2013;27:170182.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16.

    Garaud S, Morva A, Lemoine S, et al. CD5 promotes IL-10 production in chronic lymphocytic leukemia B cells through STAT3 and NFAT2 activation. J Immunol 2011;186:48354844.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Herishanu Y, Avivi I, Aharon A, et al. Efficacy of the BNT162b2 mRNA COVID-19 vaccine in patients with chronic lymphocytic leukemia. Blood 2021;137:31653173.

  • 18.

    Whitaker JA, Parikh SA, Shanafelt TD, et al. The humoral immune response to high-dose influenza vaccine in persons with monoclonal B-cell lymphocytosis (MBL) and chronic lymphocytic leukemia (CLL). Vaccine 2021;39:11221130.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Ghia P, Strola G, Granziero L, et al. Chronic lymphocytic leukemia B cells are endowed with the capacity to attract CD4+, CD40L+ T cells by producing CCL22. Eur J Immunol 2002;32:14031413.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 20.

    Zaaboub R, Vimeux L, Contremoulins V, et al. Nurselike cells sequester B cells in disorganized lymph nodes in chronic lymphocytic leukemia via alternative production of CCL21. Blood Adv 2022;6:46914704.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21.

    Palma M, Gentilcore G, Heimersson K, et al. T cells in chronic lymphocytic leukemia display dysregulated expression of immune checkpoints and activation markers. Haematologica 2017;102:562572.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Ramsay AG, Clear AJ, Fatah R, Gribben JG. Multiple inhibitory ligands induce impaired T-cell immunologic synapse function in chronic lymphocytic leukemia that can be blocked with lenalidomide: establishing a reversible immune evasion mechanism in human cancer. Blood 2012;120:14121421.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Mpakou VE, Ioannidou HD, Konsta E, et al. Quantitative and qualitative analysis of regulatory T cells in B cell chronic lymphocytic leukemia. Leuk Res 2017;60:7481.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Jak M, Mous R, Remmerswaal E, et al. Enhanced formation and survival of CD4+ CD25hi Foxp3+ T-cells in chronic lymphocytic leukemia. Leuk Lymphoma 2009;50:788801.

  • 25.

    Gustafson MP, Abraham RS, Lin Y, et al. Association of an increased frequency of CD14+HLA-DRlo/neg monocytes with decreased time to progression in chronic lymphocytic leukaemia (CLL). Br J Haematol 2012;156:674676.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Talbot H, Saada S, Barthout E, et al. BDNF belongs to the nurse-like cell secretome and supports survival of B chronic lymphocytic leukemia cells. Sci Rep 2020;10:12572.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27.

    Sourmerai J, Yousif Z, Gift T, et al. Patterns of IgG testing and rates of hypogammaglobulinemia in patients with chronic lymphocytic leukemia or small lymphocytic lymphoma. Clin Immunol 2023;250:109601.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Ishdorj G, Streu E, Lambert P, et al. IgA levels at diagnosis predict for infections, time to treatment, and survival in chronic lymphocytic leukemia. Blood Adv 2019;3:21882198.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Andersen MA, Eriksen CT, Brieghel C, et al. Incidence and predictors of infection among patients prior to treatment of chronic lymphocytic leukemia: a Danish nationwide cohort study. Haematologica 2018;103:e300303.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30.

    Sun C, Tian X, Lee YS, et al. Partial reconstitution of humoral immunity and fewer infections in patients with chronic lymphocytic leukemia treated with ibrutinib. Blood 2015;126:22132219.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Arens R, Nolte MA, Tesselaar K, et al. Signaling through CD70 regulates B cell activation and IgG production. J Immunol 2004;173:39013908.

  • 32.

    Sampalo A, Navas G, Medina F, et al. Chronic lymphocytic leukemia B cells inhibit spontaneous Ig production by autologous bone marrow cells: role of CD95-CD95L interaction. Blood 2000;96:31683174.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33.

    Else M, Blakemore SJ, Strefford JC, Catovsky D. The association between deaths from infection and mutations of the BRAF, FBXW7, NRAS and XPO1 genes: a report from the LRF CLL4 trial. Leukemia 2021;35:25632569.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Molica S, Levato D, Levato L. Infections in chronic lymphocytic leukemia. Analysis of incidence as a function of length of follow-up. Haematologica 1993;78:374377.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Soumerai J, Gift T, Yousif Z, et al. Infection outcomes and hypogammaglobulinemia in patients with chronic lymphocytic leukemia treated with immunoglobulin replacement therapy. Blood 2023;142:3280.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Bange EM, Han NA, Wileyto P, et al. CD8 T cells compensate for impaired humoral immunity in COVID-19 patients with hematologic cancer. Res Sq. Preprint posted online February 2, 2021. doi: 10.21203/rs.3.rs-162289/v1

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Riches JC, Davies JK, McClanahan F, et al. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood 2013;121:16121621.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38.

    Moreno C, Solman IG, Tam CS, et al. Immune restoration with ibrutinib plus venetoclax in first-line chronic lymphocytic leukemia: the phase 2 CAPTIVATE study. Blood Adv 2023;7:52945303.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39.

    Davis JE, Handunnetti SM, Ludford-Menting M, et al. Immune recovery in patients with mantle cell lymphoma receiving long-term ibrutinib and venetoclax combination therapy. Blood Adv 2020;4:48494859.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Manusow D, Weinerman BH. Subsequent neoplasia in chronic lymphocytic leukemia. JAMA 1975;232:267269.

  • 41.

    Bond DA, Huang Y, Fisher JL, et al. Second cancer incidence in CLL patients receiving BTK inhibitors. Leukemia 2020;34:31973205.

  • 42.

    Tsimberidou AM, Wen S, McLaughlin P, et al. Other malignancies in chronic lymphocytic leukemia/small lymphocytic lymphoma. J Clin Oncol 2009;27:904910.

  • 43.

    Kumar V, Ailawadhi S, Bojanini L, et al. Trends in the risk of second primary malignancies among survivors of chronic lymphocytic leukemia. Blood Cancer J 2019;9:75.

  • 44.

    Solomon BM, Chaffee KG, Moreira J, et al. Risk of non-hematologic cancer in individuals with high-count monoclonal B-cell lymphocytosis. Leukemia 2016;30:331336.

  • 45.

    Shen Y, Coyle L, Kerridge I, et al. Second primary malignancies in chronic lymphocytic leukaemia: skin, solid organ, haematological and Richter’s syndrome. EJHaem 2022;3:129138.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Al-Sawaf O, Zhang C, Jin HY, et al. Transcriptomic profiles and 5-year results from the randomized CLL14 study of venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab in chronic lymphocytic leukemia. Nat Commun 2023;14:2147.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Byrd JC, Hillmen P, O’Brien S, et al. Long-term follow-up of the RESONATE phase 3 trial of ibrutinib vs ofatumumab. Blood 2019;133:20312042.

  • 48.

    Solomon BM, Rabe KG, Slager SL, et al. Overall and cancer-specific survival of patients with breast, colon, kidney, and lung cancers with and without chronic lymphocytic leukemia: a SEER population-based study. J Clin Oncol 2013;31:930937.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49.

    Hamblin TJ. Autoimmune complications of chronic lymphocytic leukemia. Semin Oncol 2006;33:230239.

  • 50.

    Cao L, Wang F, Du XY, et al. Chronic lymphocytic leukemia-associated paraneoplastic pemphigus: potential cause and therapeutic strategies. Sci Rep 2020;10:16357.

  • 51.

    Strati P, Nasr SH, Leung N, et al. Renal complications in chronic lymphocytic leukemia and monoclonal B-cell lymphocytosis: the Mayo Clinic experience. Haematologica 2015;100:11801188.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52.

    Hall AM, Vickers MA, McLeod E, Barker RN. Rh autoantigen presentation to helper T cells in chronic lymphocytic leukemia by malignant B cells. Blood 2005;105:20072015.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53.

    Borthakur G, O’Brien S, Wierda WG, et al. Immune anaemias in patients with chronic lymphocytic leukaemia treated with fludarabine, cyclophosphamide and rituximab–incidence and predictors. Br J Haematol 2007;136:800805.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54.

    Wang KY, Shah P, Skavla B, et al. Vaccination efficacy in patients with chronic lymphocytic leukemia. Leuk Lymphoma 2023;64:4256.

  • 55.

    Mauro FR, Giannarelli D, Galluzzo CM, et al. Response to the conjugate pneumococcal vaccine (PCV13) in patients with chronic lymphocytic leukemia (CLL). Leukemia 2021;35:737746.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56.

    Vijenthira A, Gong I, Betschel SD, et al. Vaccine response following anti-CD20 therapy: a systematic review and meta-analysis of 905 patients. Blood Adv 2021;5:26242643.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57.

    Andrick B, Alwhaibi A, DeRemer DL, et al. Lack of adequate pneumococcal vaccination response in chronic lymphocytic leukaemia patients receiving ibrutinib. Br J Haematol 2018;182:712714.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58.

    Ujjani C, Gooley TA, Spurgeon SE, et al. Diminished humoral and cellular responses to SARS-CoV-2 vaccines in patients with chronic lymphocytic leukemia. Blood Adv 2023;7:47284737.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 59.

    Qin K, Honjo K, Sherrill-Mix S, et al. Exposure of progressive immune dysfunction by SARS-CoV-2 mRNA vaccination in patients with chronic lymphocytic leukemia: a prospective cohort study. PLoS Med 2023;20:e1004157.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 60.

    Baden LR, Swaminathan S, Almyroudis NG, et al. NCCN Clinical Practice Guidelines in Oncology: Prevention and Treatment of Cancer-Related Infections. Version 3.2024. To view the most recent version, visit https://www.nccn.org/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 61.

    Wierda WG, Brown J, Abramson JS, et al. NCCN Clinical Practice Guidelines in Oncology: Chronic Lymphocytic Leukemia/Small Lymphocytic Lymphoma. Version 1.2025. To view the most recent version, visit https://www.nccn.org/

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 62.

    Gale RP, Chapel HM, Bunch C, et al. Intravenous immunoglobulin for the prevention of infection in chronic lymphocytic leukemia. A randomized, controlled clinical trial. N Engl J Med 1988;319:902907.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 63.

    Mustafa SS, Jamshed S, Vadamalai K, Ramsey A. Subcutaneous immunoglobulin replacement for treatment of humoral immune dysfunction in patients with chronic lymphocytic leukemia. PLoS One 2021;16:e0258529.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 64.

    Sharman JP, Egyed M, Jurczak W, et al. Acalabrutinib with or without obinutuzumab versus chlorambucil and obinutuzmab for treatment-naive chronic lymphocytic leukaemia (ELEVATE TN): a randomised, controlled, phase 3 trial. Lancet 2020;395:12781291.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65.

    Tam CS, Brown JR, Kahl BS, et al. Zanubrutinib versus bendamustine and rituximab in untreated chronic lymphocytic leukaemia and small lymphocytic lymphoma (SEQUOIA): a randomised, controlled, phase 3 trial. Lancet Oncol 2022;23:10311043.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66.

    Al-Sawaf O, Zhang C, Tandon M, et al. Venetoclax plus obinutuzumab versus chlorambucil plus obinutuzumab for previously untreated chronic lymphocytic leukaemia (CLL14): follow-up results from a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2020;21:11881200.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67.

    Dearden C. Disease-specific complications of chronic lymphocytic leukemia. Hematology Am Soc Hematol Educ Program 2008;2008:450456.

  • 68.

    Arruga F, Gyau BB, Iannello A, et al. Immune response dysfunction in chronic lymphocytic leukemia: dissecting molecular mechanisms and microenvironmental conditions. Int J Mol Sci 2020;21:1825.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69.

    Jurado-Camino T, Cordoba R, Esteban-Burgos L, et al. Chronic lymphocytic leukemia: a paradigm of innate immune cross-tolerance. J Immunol 2015;194:719727.

  • 70.

    Manukyan G, Papajik T, Gajdos P, et al. Neutrophils in chronic lymphocytic leukemia are permanently activated and have functional defects. Oncotarget 2017;8:8488984901.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71.

    Toniolo PA, Liu S, Yeh JE, et al. Deregulation of SOCS5 suppresses dendritic cell function in chronic lymphocytic leukemia. Oncotarget 2016;7:4630146314.

  • 72.

    MacFarlane IA, Jillab M, Smith MR, et al. NK cell dysfunction in chronic lymphocytic leukemia is associated with loss of the mature cells expressing inhibitory killer cell Ig-like receptors. Oncoimmunology 2017;6:e1330235.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 73.

    Roessner PM, Seiffert M. T-cells in chronic lymphocytic leukemia: guardians or drivers of disease? Leukemia 2021;35:36342024.

  • 74.

    Puente XS, Jares P, Campo E. Chronic lymphocytic leukemia and mantle cell lymphoma: crossroads of genetic and microenvironment interactions. Blood 2018;131:22832296.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 75.

    Long M, Beckwith K, Do P, et al. Ibrutinib treatment improves T cell number and function in CLL patients. J Clin Invest 2017;127:30523064.

  • 76.

    Zou YX, Zhu HY, Li XT, et al. The impacts of zanubrutinib on immune cells in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma. Hematol Oncol 2019;37:392400.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 77.

    Niemann CU, Herman SE, Maric I, et al. Disruption of in vivo chronic lymphocytic leukemia tumor–microenvironment interactions by ibrutinib–findings from an investigator-initiated phase II study. Clin Cancer Res 2016;22:15721582.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78.

    de Weerdt I, Hofland T, de Boer R, et al. Distinct immune composition in lymph node and peripheral blood of CLL patients is reshaped during venetoclax treatment. Blood Adv 2019;3:26422652.

    • PubMed
    • Search Google Scholar
    • Export Citation

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 1489 1489 1489
PDF Downloads 542 542 542
EPUB Downloads 0 0 0