Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Subcutaneous immunoglobulin replacement for treatment of humoral immune dysfunction in patients with chronic lymphocytic leukemia

  • S. Shahzad Mustafa ,

    Roles Conceptualization, Investigation, Writing – original draft

    shahzad.mustafa@rochesterregional.org

    Affiliations Division of Allergy, Immunology, Rheumatology, Rochester Regional Health, Rochester, New York, United States of America, Division of Allergy, Immunology, Rheumatology, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America

  • Saad Jamshed,

    Roles Conceptualization, Writing – review & editing

    Affiliation Division of Hematology and Oncology, Rochester Regional Health, Rochester, New York, United States of America

  • Karthik Vadamalai,

    Roles Data curation, Formal analysis

    Affiliation Division of Critical Care, Mercy Hospital, Springfield, Missouri, United States of America

  • Allison Ramsey

    Roles Conceptualization, Writing – review & editing

    Affiliations Division of Allergy, Immunology, Rheumatology, Rochester Regional Health, Rochester, New York, United States of America, Division of Allergy, Immunology, Rheumatology, University of Rochester School of Medicine & Dentistry, Rochester, New York, United States of America

Abstract

Background

Patients with chronic lymphocytic leukemia (CLL) experience hypogammaglobinemia and non-neutropenic infections. In this exploratory proof of concept study, our objective was to determine the prevalence of humoral immunodeficiency in patients with CLL and serum IgG ≥ 400 mg/dL, and to evaluate the efficacy of subcutaneous immunoglobulin (SCIG) in this population.

Patients and methods

Patients with CLL with serum IgG ≥ 400 mg/dL were evaluated for serum IgG, IgM, IgA, along with pre/post vaccine IgG titers to diphtheria, tetanus, and Streptococcus pneumoniae. Patients with evidence of humoral dysfunction were treated with SCIG with Hizentra every 7±2 days for 24 weeks.

Results

Fifteen patients enrolled with median IgG = 782 mg/dL [IQR: 570 to 827], and 6/15 (40%) responded to vaccination with Td, while 5/15 (33%) responded to vaccination with PPV23. 14/15 (93.3%) demonstrated humoral immunodeficiency as evidenced by suboptimal vaccine responses, and were treated with SCIG. In patients treated with SCIG, serum IgG increased from 670 mg/dL [IQR: 565 to 819] to 1054 mg/dL [IQR: 1040 to 1166] after 24 weeks (95% CI: 271–540). For streptococcus pneumoniae, the median protective serotypes at baseline was 8 [IQR: 4 to 9] and increased to 17 [IQR: 17 to 19] after 24 weeks (95% CI: 6.93–13.72). Non-neutropenic infections (NNI) decreased from 14 to 5 during treatment with SCIG.

Conclusions

Patients with CLL demonstrate humoral immunodeficiency despite IgG > 400 mg/dL. For these patients, SCIG is well tolerated and efficacious in improving serum IgG, specific IgG to streptococcus pneumoniae, and may decrease reliance on antibiotics for the treatment of NNIs.

Clinical trials registration

NCT 03730129.

Introduction

Chronic lymphocytic leukemia (CLL) is the most common adult leukemia with approximately 21,000 new patients diagnosed annually in the United States [1]. Patients with CLL are at increased risk of infections [2] and up to one-half of patients with CLL experience an infectious complication during their disease course, with up to 17% to 50% of these being fatal [3]. Infections are typically bacterial in nature and most commonly affect the sino-pulmonary tract. Hypogammaglobinemia is the primary cause of increased susceptibility to infections in CLL and is present in up to 85% of CLL patients. Hypogammaglobinemia is due to both disease progression and the use of anti-neoplastic agents [4]. CLL decreases the ability of B lymphocytes to produce immunoglobulins due to defective functioning of non-clonal CD5-negative B cells, and also increases apoptosis of normal immunoglobulin (Ig)-producing plasma cells [5]. Mutations in the immunoglobulin heavy chain variable region may also contribute to abnormal Ig levels, although there is conflicting data on the clinical consequences of these mutations. Additionally, abnormal function of B lymphocytes can also adversely affect cross-talk with T-helper cells, thus further decreasing an adequate immune response.

Strategies to decrease infectious complications in patients with CLL include routine vaccination, use of prophylactic antibiotics, and immunoglobulin replacement (IgR) therapy. Although no clinical studies have rigorously evaluate the use of prophylactic antibiotics, this strategy is commonly implemented and recommended for prophylaxis against infections for patients with CLL [6]. IgR has been studied for infection prophylaxis in CLL, and its use is universally supported in patients with CLL with hypogammaglobinemia and associated infections [713]. As demonstrated by the international survey by Na et al., roughly 30% of patients with CLL are treated with IgR at some point during their disease management, and this proportion is similar to other hematological malignancies such as multiple myeloma and non-Hodgkin’s lymphoma [14]. Nevertheless, there remains heterogeneity in practice of when clinicians initiate IgR therapy in CLL, and guidelines vary from country to country [15]. Although IgR has been shown to decrease rates of bacterial infection in some studies, the optimal application of this practice remains unclear. A Cochrane meta-analysis concluded that the use of intravenous IgR may be considered in patients with CLL and other lymphoproliferative diseases (LPD) who have hypogammaglobinemia and recurrent infection, but acknowledged that the studies are heterogeneous [16].

Previous studies have shown that patients with CLL have impaired humoral immunity as defined by vaccine responses to both polysaccharide (e.g. pneumococcal) and peptide (e.g. tetanus or diphtheria) antigens [16]. Patients with primary immunodeficiency are routinely evaluated for their humoral antibody response before IgR is considered. However, the practice of checking vaccine responses prior to starting immunoglobulin replacement has been advocated, but not extensively studied in patients with CLL [4, 1719]. Data has shown that low titers to pneumococcal vaccination had a better association with risk of infection compared to serum IgG [20]. Thus, an immune evaluation including vaccine titers may help to identify CLL patients most at risk for life threatening infection as compared to patients with hypogammaglobinemia but preserved humoral immunity, or patients with clinical infections despite preserved immunoglobulin levels and function. Patients with impaired humoral immunity, regardless of Ig level, may benefit most from IgR.

Previous trials and clinical practice have primarily used intravenous immunoglobulin (IVIG) in patients with CLL with recurrent infections and hypogammaglobinemia [9, 13, 21]. As compared to subcutaneous immunoglobulin (SCIG), IVIG carries a higher risk of an infusion reaction, aseptic meningitis, renal dysfunction, hypercoagulability, and requires a trained medical personnel to provide intravenous access [22, 23]. SCIG mitigates these risks and can also more easily be administered at home, providing much greater patient autonomy [2325]. There is limited data on the use of SCIG in patients with secondary immunodeficiency due to CLL, but one center has published experience with SCIG in 61 patients with LPD, including patients with CLL [26].

This study is novel because it stratified patients with CLL according to their humoral response to peptide and polysaccharide vaccines, and then utilized SCIG in those with an impaired humoral response.

Patients and methods

Patients were referred for immunologic evaluation for this prospective case series from local hematology/oncology offices in Rochester, NY. All study procedures were conducted at the Rochester Regional Health allergy/immunology practice in Rochester, NY from March 2019 through June 2020. The authors confirm that all ongoing and related trials for this drug/intervention were registered prior to patient enrollment (NCT 03730129). The study was approved by the institutional review board at Rochester Regional Health on October 10, 2018 (CIC 1850-A-18), and all patients signed informed consent.

Patients with a confirmed diagnosis of CLL per flow cytometry either on peripheral blood or bone marrow biopsy were eligible for enrollment in the study (Fig 1). Exclusion criteria included previously diagnosed primary immunodeficiency, additional immunosuppressive states as defined by the investigators, serum IgG < 400 mg/dL, and previous or ongoing therapy with IgR. Patients underwent a laboratory evaluation for immunodeficiency, including baseline serum IgG, IgM, and IgA, along with pre and post-vaccination IgG titers for peptide antigens (diphtheria, tetanus) with Td and polysaccharide antigens (Streptococcus pneumoniae) with pneumococcus polyvalent vaccine-23 (PPV23) 28±7 days following vaccination. Normal vaccine responses were defined by the following criteria [27]:

  1. Diphtheria: 2-fold increase, and must be into protective range (0.1 IU/ml)
  2. Tetanus: 2-fold increase, and must be in protective range (0.1 IU/ml)
  3. Streptococcus pneumoniae
    1. If < 1.3 μg/ml, 2-fold increase to > 1.3 μg/ml OR 4-fold increase
    2. If > 1.3 μg/ml, 2-fold increase
    3. Responses must be demonstrated by 70% of serotypes
  4. Impaired vaccine response is defined at abnormal response to any of the above antigens.

Patients with humoral immunodeficiency (as evidenced by an abnormal vaccine response) despite serum IgG ≥ 400 mg/dL were offered therapy with 20% liquid SCIG (Hizentra, CSL Behring, King of Prussia, PA) for 24 weeks regardless of infection history. SCIG was given at a fixed dose of 0.13 g/kg/week, and administered every 7±2 days. Patients did not receive a loading dose of immunoglobulin. Patients were not pre-medicated prior to SCIG therapy unless they experienced infusion-related side effects. Patients received instruction on SCIG administration for 3–4 sessions from a research coordinator, and then transitioned to independent administration at home. The number of infusion sites and time for infusion was tracked for each patient, as were adverse events. SCIG was provided by the sponsor, CSL Behring.

The following data was collected for each patient: demographic data (age, race, and gender), time since initial diagnosis of CLL, chemotherapeutic regimen and other therapies used for management of CLL, and number of non-neutropenic infections (NNI) requiring therapy with antibiotics in the previous 6 months (per patient recall in combination with a review of the electronic medical record). All patients underwent a laboratory evaluation for humoral immunodeficiency as defined above. For patients who were treated with SCIG the number of infusion sites and duration of infusion was tracked throughout the study period, as were adverse events. Patients on SCIG completed a quality of life survey (Short Form 36) every 28±7 days. Ig levels and IgG titers for diphtheria, tetanus, and streptococcus pneumoniae were checked 4, 12, and 24 weeks after starting IgR. A repeat evaluation for humoral immunodeficiency, including pre and post-vaccination IgG titers was completed 3 months after the last dose of SCIG.

Statistical analysis

Statistical analysis was performed using STATA software (StataCorp LLC, College Station, Texas). Baseline characteristics along with both the primary and secondary endpoints are reported in medians and interquartile (IQR) ranges. A non-parametric, Wilcoxon sign ranked test was used to compare study outcomes at baseline and the completion of the study. Due to the exploratory nature of this proof of concept study, a formal power calculation was not completed prior to study initiation, and the study was not adequately powered to test a specific hypothesis. Microsoft Excel software (Office 365, Microsoft Corporation, Redmond, WA 98052) was used to create the figures.

Results

A total of 18 patients underwent evaluation, with 15 enrolled in the study (Table 1). Three patients were excluded due to IgG < 400 mg/dL. There were 13 (86.7%) males and 2 (13.3%) females. The median age of the cohort was 69 years [IQR: 67 to 75]. The median time since diagnosis of CLL was 4.4 years [IQR: 2.6 to 15.2]. The median number of NNI treated with antibiotics was 1 [IQR: 0 to 2]. Previous to enrollment, four patients had undergone treatment regimens for CLL. At the time of the study, one patient was being treated with ibrutinib, while 14 of 15 patients were managed with close observation.

The baseline immune evaluation was as follows: median IgG = 782 mg/dL [IQR: 570 to 827], median IgM = 44 mg/dL [IQR: 37 to 68], and median IgA = 138 mg/dL [IQR: 81 to 171] (Table 2). At enrollment, the median IgG for tetanus = 1.21 IU/ml [IQR: 0.85 to 1.50] increased to a median of 2.03 IU/ml [IQR: 1.01 to 2.76] following vaccination. Six of the 15 (40%) patients responded to vaccination with tetanus. The median IgG for diphtheria = 0.16 IU/ml [IQR: 0.05 to 0.27] decreased to a median of 0.14 IU/ml [IQR: 0.1 to 0.34] following vaccination. Six of the 15 (40%) patients responded to vaccination with diphtheria. For baseline IgG for streptococcus pneumoniae, the median number of serotypes with protective titers of ≥ 1.3 mcg/ml was 9/23 [IQR: 5 to 12], and this changed to 9/23 [IQR: 6 to 17] following vaccination. Five of the 15 (33%) patients responded to vaccination with PPV23.

Of the 15 patients in the cohort, 14 (93.3%) met criteria for humoral immunodeficiency due to poor vaccine responses to peptides and/or polysaccharides, and were offered therapy with SCIG. One patient declined due to deterioration in clinical status and enrollment into an alternative clinical study. Three patients declined because they did not wish to pursue IgR given their currently stable health status. A total of 10 agreed to pursue therapy, with 9 completing 24 weeks of therapy. One patient discontinued IgR early due to reported fatigue (Table 3). The median weekly dose was 11.5 g [IQR: 11 to 12], with each infusion utilizing a median of 2.5 sites, and taking 61 minutes [IQR: 56 to 64]. Nine of the 10 patients did not require pre-treatment with antihistamines, acetaminophen, NSAIDs, or systemic steroids prior to SCIG infusions. There were no localized or systemic reactions. One patient reported fatigue for 2–3 days after SCIG infusions and was pre-treated with diphenhydramine and acetaminophen with no improvement of symptoms. This patient subsequently discontinued therapy with SCIG.

thumbnail
Table 3. Dosing and adverse events related to subq IgR in study population.

https://doi.org/10.1371/journal.pone.0258529.t003

In the 9 patients who completed 24 weeks of IgR, the median baseline IgG of 670 mg/dL [IQR: 565 to 819] increased to 838 mg/dL [IQR: 749 to 1038] after 4 weeks (95% CI: 154–291), 1001 mg/dL [IQR: 922 to 1173] after 12 weeks (95% CI: 319–432), and 1054 mg/dL [IQR: 1040 to 1166] after 24 weeks (95% CI: 271–540) (Fig 2). Three months after discontinuation of IgR, IgG = 701 mg/dL [IQR: 635 to 823], which was similar to baseline (95% CI: 21–88). For tetanus, the median baseline IgG of 1.07 IU/ml [IQR: 0.72 to 1.54] increased to 1.93 IU/mL [IQR: 1.29 to 3.00] after 4 weeks (95% CI: 0.42–2.17), 2.56 IU/mL [IQR: 2.23 to 4.47] after 12 weeks (95% CI: 0.94–3.67), and 2.14 IU/mL [IQR: 1.93 to 2.62] after 24 weeks (95% CI: 0.8–1.6). Three months after discontinuation of IgR, IgG for tetanus was 1.54 IU/mL [IQR: 1.04 to 1.81], which was still higher than baseline (95% CI: 0.19–1.08). For diphtheria, the median baseline IgG of 0.16 IU/ml [IQR: 0.10 to 0.27] increased to 0.27 IU/mL [IQR: 0.18 to 0.60] after 4 weeks (95% CI: -0.17–1.0), 0.35 IU/mL [IQR: 0.27 to 0.99] after 12 weeks (95% CI: -0.03–1.57), and 0.41 IU/mL [IQR: 0.31 to 0.69] after 24 weeks (95% CI: -0.01–1.09). Three months after discontinuation of IgR, IgG for diphtheria was 0.23 IU/mL [IQR: 0.19 to 0.42], which was still higher than baseline (95% CI: -0.08–0.59). For streptococcus pneumoniae, the median number of serotypes in the protective range (> 1.3 mcg/ml) at baseline was 8 [IQR: 4 to 9] and increased to 13 [IQR: 9 to 15] after 4 weeks (95% CI: 2.59–6.29), 12 [IQR: 11 to 16] after 12 weeks (95% CI: 4.37–8.73), and 17 [IQR: 17 to 19] after 24 weeks (95% CI: 6.93–13.72) (Fig 3). Three months after discontinuation of IgR, protective serotypes for streptococcus pneumoniae = 7 [IQR: 5 to 10], which was similar to baseline (95% CI: 0.39–2.94).

thumbnail
Fig 3. Number of protective serotypes for streptococcus pneumonia.

https://doi.org/10.1371/journal.pone.0258529.g003

The 9 patients who completed 24 weeks of IgR experienced 14 NNI in the 6 months prior to IgR (one requiring inpatient hospitalization), and this decreased to 5 NNI during 6 months of therapy with IgR, with no hospitalizations (Fig 4). Additionally, within the 3 months following IgR, these patients experienced 7 NNI. There was no significant change in the Short Form 36 values during the study period.

thumbnail
Fig 4. NNI prior to, during subq IgR, and after subq IgR.

NNI: non-neutropenic infection, subq: subcutaneous, IgR: immunoglobulin replacement.

https://doi.org/10.1371/journal.pone.0258529.g004

Discussion

Patients with CLL are recognized to be at increased risk of infections due to a myriad of factors, including hypogammaglobinemia [2, 4], and infections remain a major cause of morbidity and mortality in this population [3]. Although IgR has been used as a strategy for infection prophylaxis [713], our study is unique because it proactively completed an immune evaluation by checking vaccine responses to peptide and polysaccharide antigens in patients with CLL regardless of infection history. We demonstrated that patients with CLL are at risk of humoral dysfunction despite relatively normal Ig levels. Furthermore, prophylactic therapy with SCIG increased serum IgG and specific antibody titers, was well tolerated, and showed a trend in decreased NNIs.

There are no consensus guidelines on the evaluation of humoral immunity in patients with CLL. Although the risk of infection in CLL is felt to generally increase with lower IgG levels [28], not all patients with hypogammaglobinemia will experience infectious complications [29]. In contrast, there is a subset of individuals with CLL who experience increased infectious episodes despite having normal or near-normal IgG levels, due to B lymphocytes producing non-functional, monoclonal Ig [3032]. Significant B cell dysfunction likely also arises from aberrant signaling from the B cell receptor (BCR), which is characterized by variable response to antigenic stimulation, thus affecting other immune functions, including T cell and cellular immunity. Due to the possibility of abnormal functional status despite relatively normal IgG levels, and based on our study results, we encourage the evaluation of vaccine responses to peptide and polysaccharide antigens to help determine the function of Ig in CLL patients. Our cohort of CLL patients all with serum IgG > 400 mg/dL, and a median serum IgG of 782 mg/dl [IQR: 570 to 827] showed only 1/15 patients (6.7%) mounted an adequate immune response to vaccination with Td and PPV23. Our findings are in line with previous research showing similarly poor vaccine responses in this population [3336], and highlight the importance of evaluating vaccine responses for a thorough immune evaluation in patients with CLL. Early identification and treatment of secondary immune deficiency may improve morbidity and possibly mortality in these patients. Additionally, three patients (16.7%) who underwent an immune evaluation demonstrated IgG < 400 mg/dl, with one patient having an IgG = 93 mg/dL. We therefore believe risk stratification based on this immune evaluation should be routinely considered in all patients with CLL regardless of previous infection history, since this may reveal severe immune dysfunction, even early in disease.

Immunoglobulin replacement provides passive immunity by providing a diverse antibody repertoire from healthy donors, and is commonly used in CLL for infection prophylaxis. Although IgR is not felt to be immune modulating, with no direct effects on immune dysfunction, replacement of functional antibodies has been shown to decrease infectious complications [26]. We are among the first to show the potential utility of SCIG in CLL patients with secondary immune deficiency, as the majority of CLL patients treated with IgR receive intravenous infusions. Similar to Compagno et al [26], 90% of patients in our cohort reported no adverse effects related to SCIG infusions. Nine of ten patients also did not require pre-medication, and the majority completed infusions independent of medical professionals in one hour while at home. Self-administered at home administration is particularly valuable during the current global pandemic with COVID-19 infection since it allows for minimizing unnecessary interaction with health care facilities. We encourage all clinicians prescribing IgR to engage in shared decision making with their patients regarding the most suited route of IgR. This discussion is also a unique opportunity for collaboration between hematology/oncology and allergy/immunology colleagues, since the latter utilize routinely utilize SCIG in the management of primary immunodeficiency.

Our results also show promising efficacy of SCIG in the setting of CLL. All of the patients in our cohort experienced significant increases in their IgG levels, and this was significant as early as four weeks after the initiation of IgR. In addition to an increase in serum IgG, our study is the first to show a significant improvement in specific IgG for peptide antigens such as tetanus and diphtheria, and polysaccharide antigens such as Streptococcus pneumonia. This is likely to be particularly meaningful, since Streptococcus pneumoniae is felt to be one of the most common etiologies of recurrent sino-pulmonary infections in patients with CLL [37]. Most importantly, for our cohort of patients with CLL, these improvement in laboratory findings while on SCIG correlated with a decreased reliance on antibiotics for the treatment of NNI. Although our results did not show a significant change in health-related quality of life scores, we suspect this is because our enrollment criteria did not require a previous history of infection, and many patients reported a high quality of life at enrollment despite an abnormal immune evaluation.

We acknowledge limitations to our study. Most importantly, our study was small in size, and did not require a history of NNIs for enrollment. We suspect a larger cohort with a greater history of NNIs requiring frequent antibiotic therapy would benefit even more from our diagnostic and treatment strategy. Due to our small sample size, we were not able to formally evaluate for improvement in NNIs. Importantly, although patients served as their own control, our study also did not have a control arm, which makes it more difficult to assess the clinical impact of IgR in this patient population. Future, larger studies are warranted to evaluate the efficacy of risk stratification with vaccine response, followed by treatment with SCIG in patients with an abnormal evaluation. We also did not evaluate the potential role of conjugate pneumococcal vaccination (PCV13) as a strategy to improve Streptococcus pneumoniae titers in patients not responding to polysaccharide vaccination (PPV23), as this may serve as adequate protection in a subset of individuals and may avoid the need for IgR. Despite these limitations, we believe our study is novel in its use of vaccine responses to evaluate for humoral immunodeficiency, and provides foundational data for the use of SCIG in the setting of CLL despite adequate IgG levels.

In conclusion, this study suggests that patients with CLL demonstrate significant humoral immunodeficiency as defined by abnormal vaccine responses even in the setting of relatively normal IgG levels (> 400 mg/dL). For these patients, SCIG is likely to be well tolerated and efficacious in improving serum IgG, specific IgG to streptococcus pneumoniae, and may decrease reliance on antibiotics for the treatment of NNIs. Larger studies using vaccine responses as risk stratification for infectious complications are needed, as are randomized controlled trials of subq IgR in patients with CLL.

References

  1. 1. Siegel RL, Miller KD, Jemal A. Cancer Statistics 2019. Cancer Journal for Clinicians 2019; 69(1): 7–34.
  2. 2. Twomey JJ, Houston MB. Infections complicating multiple myeloma and chronic lymphocytic leukemia. Arch Int Med 1973; 132: 562–565. pmid:4542660
  3. 3. Morra E, Nosari A, Montillo M. Infectious complications in chronic lymphocytic leukemia. Hematological Cell Therapies 1999; 41: 145–151.
  4. 4. Dhalla F, Lucas M, Schuh A, Bhole M, Jain R, Patel SY, et al. Antibody deficiency secondary to chronic lymphocytic leukemia: Should patients be treated with prophylactic replacement immunoglobulin? Journal of Clinical Immunology 2014; 34(3): 277–282. pmid:24557494
  5. 5. Sampalo A, Navas G, Medina F, Segundo C, Cámara C, Brieva JA. Chronic lymphocytic leukemia B cells inhibit spontaneous Ig production by autologous bone marrow cells: role of CD95-CD95L interaction. Blood 2000; 96(9): 3168–3174. pmid:11049999
  6. 6. Oscier D, Dearden C, Eren E, Fegan C, Follows G, Hillmen P, et al. Guidelines on the diagnosis, investigation and management of chronic lymphocytic leukaemia. British Journal of Haematology 2012; 159(5): 541–564. pmid:23057493
  7. 7. Gale RP, Chapel HM, Bunch C, Rai KR, Foon K, Courter SG, et al. Cooperative Group for the Study of Immunoglobulin in Chronic Lymphocytic Leukemia, Intravenous immunoglobulin for the prevention of infection in chronic lymphocytic leukemia. A randomized, controlled clinical trial. New England Journal of Medicine 1988; 319: 902–907.
  8. 8. Jurlander J, Geisler CH, Hansen MM. Treatment of hypogammaglobulinaemia in chronic lymphocytic leukaemia by low dose intravenous gammaglobulin. European Journal of Haematology 1994; 53: 114–118. pmid:8088382
  9. 9. Chapel H, Dicato M, Gamm H, Brennan V, Ries F, Bunch C, et al. Immunoglobulin replacement in patients with chronic lymphocytic leukaemia: a comparison of two dose regimes. British Journal of Haematology 1994; 88: 209–212. pmid:7803248
  10. 10. Sklenar I, Schiffman G, Jonsson V, Verhoef G, Birgens H, Boogaerts M, et al. Effect of various doses of intravenous polyclonal IgG on in vivo levels of 12 pneumococcal antibodies in patients with chronic lymphocytic leukaemia and multiple myeloma. Oncology 1993; 50: 466–477. pmid:8233289
  11. 11. Griffiths H, Brennan V, Lea J, Bunch C, Lee M, Chapel H. Crossover study of immunoglobulin replacement therapy in patients with low-grade B-cell tumors. Blood 1989; 73: 366–368. pmid:2492832
  12. 12. Boughton BJ, Jackson N, Lim S, Smith N. Randomized trial of intravenous immunoglobulin prophylaxis for patients with chronic lymphocytic leukaemia and secondary hypogammaglobulinemia. Clinical Laboratory Haematology 1995; 17: 75–80. pmid:7621634
  13. 13. Molica S, Musto P, Chiurazzi F, Specchia G, Brugiatelli M, Cicoira L, et al. Prophylaxis against infections with low-dose intravenous immunoglobulins (IVIG) in chronic lymphocytic leukemia. Results of a crossover study. Haematologica 1996; 81: 121–126. pmid:8641639
  14. 14. Na I, Buckland M, Agostini C, Edgar JD, Friman V, Michallet M, et al. Current clinical practice and challenges in the management of secondary immunodeficiency in hematologic malignancies. European Journal of Hematology 2019; 102: 447–456.
  15. 15. Patel SY, Carbone J, Jolles S. The expanding field of secondary antibody deficiency: causes, diagnosis, and management. Frontiers in Immunology 2019; 10(33): 1–22.
  16. 16. Raanani P, Gafter-Gvili A, Paul M, Ben-Bassat I, Leibovici L, Shpiberg O. Immunoglobulin prophylaxis in hematologic malignancies and hematopoietic stem cell transplantation. Cochrane Database Syst Rev 2008. Oct 8(4): CD006501.
  17. 17. Lachance S, Christofides AL, Lee JK, Sehn LH, Ritchie BC, Shustik C, et al. A Canadian perspective on the use of immunoglobulin therapy to reduce infectious complications in chronic lymphocytic leukemia. Cur Onc 2016; 23(1): 42–51. pmid:26966403
  18. 18. Ueda M, Berger M, Gale RP, Lazarus HM. Immunoglobulin therapy in hematologic neoplasms and after hematopoietic cell transplantation. Blood Reviews 2018; 32(2): 106–115. pmid:28958644
  19. 19. Sanchez-Ramon S, Dhalla F, Chapel H. Challenges in the role of gammaglobulin replacement therapy and vaccination strategies for hematological malignancy. Frontiers in Immunology 2016; (7)317: 1–11. pmid:27597852
  20. 20. Friman V, Winqvist O, Blimark C, Langerbeins P, Chapel H, Dhalla F. Secondary immunodeficiency in lymphoproliferative malignancies. Hematol Onocol 2016; 34(3): 121–132. pmid:27402426
  21. 21. Gale RP, Chapel H, Bunch C, Rai KR, Foon K, Courter SG, et al. Intravenous immunoglobulin for prevention of infection in chronic lymphocytic leukemia. NEJM 1998; 319(14): 902–907.
  22. 22. Looney RJ, Huggins J. Use of immunoglobulin G (IVIG). Best Prac and Res Clin Haematol 2006; 19(1): 3–25.
  23. 23. Berger M. Adverse effects of IgG therapy. J Allergy Clin Imunol Pract 2013; 1(6): 558–566. pmid:24565701
  24. 24. Gardulf A. Immunoglobulin treatment for primary antibody deficiencies: advantages of the subcutaneous route. Biodrugs 2007; 21(2): 105–116. pmid:17402794
  25. 25. Lingman-Framme J, Fasth A. Subcutaneous immunoglobulin for primary and secondary immunodeficiencies: an evidence-based review. Drugs 2013; 73(12): 1307–1319. pmid:23861187
  26. 26. Compagno N, Cinetto F, Semenzato G, Agostini C. Subcutaneous immunoglobulin in lymphoproliferative disorders and rituximab-related secondary hypogammaglobulinemia: a single center experience in 61 patients. Haematologica 2014; 99(6): 1101–1106. pmid:24682509
  27. 27. Orange JS, Ballow M, Stiehm ER, Ballas ZK, Chinen J, La Morena MD, et al. Use and interpretation of diagnostic vaccination in primary immunodeficiency: A working group report of the basic and clinical immunology interest section of the american academy of allergy, asthma & immunology. J Allergy Clin Immunol. 2012;130(3 Suppl):1–24.
  28. 28. Visentin A, Compagno N, Cinetto F, Imbergamo S, Zambello R, Piazza F, et al. Clinical profile associated with infections in patients with chronic lymphocytic leukemia. Protective role of immunoglobulin replacement therapy. Haematologica 2015; 100: e515–518. pmid:26294735
  29. 29. Griffiths H. Lea J. Bunch C, Lee M, Chapel H. Predictors of infection in chronic lymphocytic leukemia (CLL). Clinical Experimental Immunology 1992; 89: 374–377. pmid:1516254
  30. 30. Martin W, Abraham R, Shanafelt T, Clark RJ, Bone N, Geyer SM, et al. Serum-free light chain—a new biomarker for patients with B-cell non-Hodgkin lymphoma and chronic lymphocytic leukemia. Translational Research 2007; 149: 231–235. pmid:17383597
  31. 31. Bernstein ZP, Fitzpatrick JE, O’Donnell A, Han T, Foon KA, Bhargava A. Clinical significance of monoclonal proteins in chronic lymphocytic leukemia. Leukemia 1992; 6(12): 1243–1245. pmid:1453768
  32. 32. Deegan MJ, Abraham JP, Sawdyk M, Van Slyck EJ. High incidence of monoclonal proteins in the serum and urine of chronic lymphocytic leukemia patients. Blood 1984; 64(6): 1207–1211. pmid:6437461
  33. 33. Svensson T, Kättström M, Hammarlund Y, Roth D, Andersson PO, Svensson M, et al. Pneumococcal conjugate vaccine triggers a better immune response than pneumococcal polysaccharide vaccine in patients with chronic lymphocytic leukemia A randomized study by the Swedish CLL group. Vaccine 2018; 36(25): 3701–3707. pmid:29748028
  34. 34. Sinisalo M, Aittoniemi J, Oivanen P, Käyhty H, Olander RM, Vilpo J. Response to vaccination against different types of antigens in patients with chronic lymphocytic leukaemia. British Journal of Haematology 2001; 114(1): 107–110. pmid:11472353
  35. 35. Hartkamp A, Mulder AH, Rijkers GT, van Velzen-Blad H, Biesma DH. Antibody responses to pneumococcal and haemophilus vaccinations in patients with B-cell chronic lymphocytic leukaemia. Vaccine 2001; 19(13–14): 1671–1677. pmid:11166890
  36. 36. Mustafa SS, Jamshed S, Ramsey A. Humoral immunodeficiency in patients with chronic lymphocytic leukemia. Abstract. ACAAI Annual Meeting 2019.
  37. 37. Wadhwa PD, Morrison VA. Infectious complication of chronic lymphocytic leukemia. Seminars in Oncology 2006; 33(2): 240–249. pmid:16616071