Skip to main content
Medicine logoLink to Medicine
. 2020 Jul 10;99(28):e21167. doi: 10.1097/MD.0000000000021167

The impact of immune checkpoint inhibitors in patients with chronic lymphocytic leukemia (CLL)

A protocol for a systematic review and meta-analysis of randomized controlled trials

Aviwe Ntsethe a, Phiwayinkosi Vusi Dludla c,d, Tawanda Maurice Nyambuya a,b, Siphamandla Raphael Ngcobo a, Bongani Brian Nkambule a,
PMCID: PMC7360255  PMID: 32664154

Abstract

Introduction:

The global burden of chronic lymphocytic leukemia (CLL) has constantly increased over the years, with a current incidence of 3.5 cases per 100,000 people. Although the conventional drugs used to treat CLL patients have been effective treatment failure rate in some of the patients is alarming. Therefore, as a result, novel treatment strategies with improved outcomes such as the blockade of immune checkpoints have emerged. However, consensus on the risk-benefit effects of the using these drugs in patients with CLL is controversial and has not been comprehensively evaluated. This systemic review and meta-analysis provide a comprehensive synthesis of available data assessing adverse events associated with the use of immune checkpoint inhibitors in patients with CLL as well as their influence on the overall survival rate.

Methods:

This protocol for a systematic review and meta-analysis has been prepared in accordance with Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols 2015 guidelines. A search strategy will be developed using medical subject headings words in PubMed search engine with MEDLINE database. The search terms will also be adapted for gray literature, Embase, and Cochrane Central Register of Controlled Trials electronic databases. Two reviewers (AN and SRN) will independently screen studies, with a third reviewer consulted in cases of disagreements using a defined inclusion and exclusion criteria. Data items will be extracted using a predefined data extraction sheet. Moreover, the risk of bias and quality of the included studies will be appraised using the Downs and Black checklist and the quality and strengths of evidence across selected studies will be assessed using the Grading of Recommendations Assessment Development and Evaluation approach. The Cochran's Q statistic and the I2 statistics will be used to analyze statistical heterogeneity across studies. If the included studies show substantial level of statistical heterogeneity (I2 > 50%), a random-effects meta-analysis will be performed using R statistical software.

Ethics and dissemination:

The review and meta-analysis will not require ethical approval and the findings will be published in peer-reviewed journals and presented at local and international conferences. This review may help provide clarity on the risk-benefit effects of using immune checkpoint inhibitors in patients with CLL.

Systematic review registration:

International prospective Register of Systematic Reviews (PROSERO) number: CRD42020156926.

Keywords: adverse events, chronic lymphocytic leukemia, immune checkpoint inhibitors

1. Introduction

The global incidence of leukemia has significantly increased over the years, with chronic lymphocytic leukemia (CLL) cases having a higher prevalence compared to all other lymphoid malignancies.[1] Although the exact aetiology remains elusive, age, lifestyle and environmental factors have been identified as some of the major consequences implicated in the development of CLL.[2,3] To date, it is well established that CLL is the most common type of leukemia, accounting for approximately 37% of all cases of blood malignancies,[4] with an average global prevalence of about 3.5 cases per 100,000 people.[5] In Africa, statistics on the incidence of CLL is very limited with isolated studies reporting on this form of leukemia.[611] Nonetheless, various therapeutic drugs including those that modulate the function of immune checkpoints receptors are continuously being developed and their effectiveness tested in the management of patients with CLL worldwide.[1214]

Immune checkpoints regulate immune function and play a crucial role in preventing autoimmunity.[1517] However, in CLL, the signaling of immune checkpoint receptors is dysregulated which results in immune dysfunction.[18,19] Briefly, CLL is a monoclonal disorder that is characterized by the accumulation of functionally incompetent B-cells with a distinctive CD19+, CD20+, CD5+, CD23+ lymphocyte surface markers and surface immunoglobulin-positive phenotype in the peripheral blood, bone marrow, and lymph nodes.[20,21] Hence, anti-CD20 monoclonal-based drugs such as rituximab and ofatumumab are used as standard treatment for CLL.[12,22,23] However, these drugs are associated with severe adverse events such as neutropenia and thrombocytopenia,[2426] with others reporting on their ineffectiveness as monotherapy.[27] Thus, the need to urgently broaden our understanding of the pathophysiological mechanisms implicated in the aggravation of CLL.

Although CLL is a B-cell malignancy, recent studies have also described the involvement of T-cells in the pathogenesis and progression of the disease.[2830] In fact in CLL, T-cell exhaustion mediated by an upregulation of coinhibitory receptors such as programmed death-1 (PD-1), lymphocyte-activation gene 3 (LAG-3), T-cell immunoglobulin-3 (TIM-3), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) has been reported.[18,31] Consequently, this has led to the advancement of immune checkpoint inhibitors that targets both B and T-cell function as a treatment strategy for CLL.[32] However, contradictory findings on the effects of using immune checkpoint inhibitors in CLL patients have been reported.[13,3236] Thus, the exact effect of immune checkpoint inhibitors in CLL is contradictory and needs to be investigated further. As a result, due to high quality of evidence reported in randomized controlled trials (RCTs), this review will target such studies to assess and update available literature on the impact immune checkpoint inhibitors in CLL.

2. Research question

What are common adverse events associated with the use of immune checkpoint inhibitors in patients with CLL?

3. Objectives

  • 1.

    To assess the adverse events associated with the use of immune checkpoint inhibitors in patients with CLL.

  • 2.

    To estimate the overall survival rate of patient with CLL on immune checkpoints inhibitors.

4. Methods

This protocol was prepared in accordance with the Preferred Reporting Items for Systematic Review and Meta-analysis Protocols (PRISMA-P) 2015 guidelines.[37] In addition, the protocol has been submitted on PROSPERO for registration.

5. Eligibility

5.1. Study design

This systematic review and meta-analysis will include RCTs with a clearly defined population and interventions used. While, observational studies, reviews, case studies, and animal studies will be excluded in this study.

5.2. Participants

Studies evaluating the use of immune checkpoint inhibitors as a treatment method in adult patients (≥18 years) with CLL, will be included.

5.3. Intervention

We will include studies reporting on the use of immune checkpoint inhibitors targeting PD-1, CTLA-4, LAG-3, and TIM-3 signaling as a therapeutic strategy for CLL.

5.4. Comparators

CLL patients on immune checkpoint inhibitor drugs that did not develop any associated adverse events.

5.5. Outcomes

The primary endpoints will include the following:

  • 1.

    Adverse events that are associated with the use of immune checkpoint inhibitors. These include mortality, endocrinopathies, and dermatitis, autoimmune, gastrointestinal and hematological disorders.

5.6. Surrogate outcomes

  • 1.

    Overall response, progression-free survival, and event-free remission

  • 2.

    Common low-grade adverse events as described by National Cancer Institute grading system[38]

5.7. Search strategy

The search strategy will be developed using medical subheading words on MEDLINE and will be adapted to gray literature, Embase, The Cochrane Central Register of Controlled Trials databases, and ClinicalTrials.gov with the help of an experienced librarian. The search strategy will consist of the following keywords and their respective synonyms; chronic lymphocytic leukemia, anti-PD-1 drugs (nivolumab, Pembrolizumab, Pidilizumab, Atezolizumab, Avelumab), anti-PD-L1 drugs (Atezolizumab, Avelumab, Durvalumab), anti-CTLA-4 drugs (Ipilimumab, Tremelimumab) anti-LAG-3 and anti-TIM-3 drugs and adverse events.

5.8. Study selection

The study screening and selection process will be carried out by 2 independent reviewers (AN and SRN) to eliminate risk of bias and inconsistencies with regards to reviewers’ inclusion and exclusion of studies. Each reviewer will screen tittle, abstract, and full texts in contrast to the inclusion criteria. The exclusion criteria for title and abstract screening phase will include duplicate of the same study, reviews, observation studies, and studies that reported nonimmune checkpoint-related CLL therapeutic drugs. In cases of disagreements, a third reviewer (TMN) will be consulted for arbitration. The level of inter-rater agreement will be determined by using the Cohen's kappa inter-rater reliability.[39] A kappa value of < 0.00 will be interpreted as a poor strength of agreement, 0.00–0.20 as slight agreement, 0.21–0.40 as fair agreement, 0.41–0.60 as moderate agreement, 0.61–0.80 as substantial agreement, and 0.81–1.00 as perfect agreement.

6. Data management

6.1. Data collection process

The reviewers (AN and SRN) will develop a data extraction form that will be used in the collection relevant data items. To reduce data entry errors, selected studies will be independently assessed by two reviewers (AN and TMN), the third reviewer (BBN) will be consulted for arbitration in case of any disagreements.

6.2. Data items

Extracted data items will include the author's name, year of publication, sample size, duration of follow-up, outcome measures, age, gender, immune checkpoint receptors targeted by the drugs, dosage, adverse events reported, and overall survival rate.

6.3. Data simplification

Studies will be grouped according to the type of immune checkpoint inhibitor used. In addition, studies will be grouped based on the immune checkpoint receptor targeted (PD-1, PD-L1, CTLA-4, LAG-3, TIM-3). Studies that report on immune checkpoint inhibitor combined with other conventional drugs will be pooled. The adverse events will be grouped and graded into grades 1 to 4 based on their severity. Group considered 1 and 2 are mild and moderate whilst groups 3, 4, and 5 are severe.[38]

6.4. Risk of bias in individual studies

To evaluate the potential risk of bias in RCTs, Cochrane collaboration tool for assessing bias[40] and Downs and Black checklist[41] will be used. Two independent reviewers (AN and SRN) will appraise all included studies and a third reviewer (PVD) will be consulted in cases of disagreements.

6.5. Data synthesis

A summary of findings table will be used to provide a synthesis of the main outcomes of included studies. Moreover, if the included studies are homogeneous in terms of the type of immune checkpoint inhibitor used and participant characteristics, data will be analyzed with Rev Manager (Version 5.3) to conduct a meta-analysis. To measure statistical heterogeneity between studies, I2 and Chi squared statistical tests will be used.[42,43] An I2 value of > 50% will be considered substantial heterogeneity.[44] To find the sources of heterogeneity within the included studies, a subgroup analysis and meta-regression comparing the study estimates from different study-level characteristics, quality, intervention type (type of immune checkpoint inhibitor), and the reported effect measure of adverse events will be conducted.

7. Quality assessment of the cumulative evidence

The Grading of Recommendations, Assessment, Development and Evaluation assessment tool[45] will be used to assess the overall quality of evidence. Moreover, the quality of each included study will be independently evaluated by 2 authors (AN, SRN). The third author (TMN) will adjudicate in cases of disagreements. The quality of evidence will be assessed based on several factors such as study limitations, indirectness of results, and publication or reporting bias. The evidence of each outcome will be rated as high, moderate, low, or very low.

8. Discussion

Immune checkpoint inhibitors have been shown to be effective in the treatment of CLL, its association with adverse events is controversial[13,32] and has not been critically assessed. Therefore, this systemic review and meta-analysis aims to evaluate the risk-benefit of using immune checkpoint inhibitors as a therapeutic strategy for patients with CLL. Findings from this study will give a better understanding on the effectiveness of immune checkpoint inhibitors as well as paving way for strategic development of effective therapies and management of patients with CLL.

Author contributions

AN, TMN, PVD, and BBN conceptualized, designed, and drafted this manuscript. All authors including SRN wrote and approved the final manuscript. BBN is the guarantor of the review.

Footnotes

Abbreviations: CLL = chronic lymphocytic leukemia, CTLA-4 = cytotoxic T-lymphocyte-associated protein 4, LAG-3 = lymphocyte-activation gene 3, PD-1 = programmed death-1, PD-L1 = programmed death-ligand 1, PRISMA-P = Preferred Reporting Items for Systematic Review and Meta-analysis Protocols, RCTs = randomized controlled trials, TIM-3 = T-cell immunoglobulin 3.

How to cite this article: Ntsethe A, Dludla PV, Nyambuya TM, Ngcobo SR, Nkambule BB. The impact of immune checkpoint inhibitors in patients with chronic lymphocytic leukemia (CLL): A protocol for a systematic review and meta-analysis of randomised controlled trials. Medicine. 2020;99:28(e21167).

BBN is partially funded by the National Research Foundation of South Africa (grant number: 107519). BBN is also University of KwaZulu Natal (UKZN) Developing Research Innovation, Localisation and Leadership in South Africa (DRILL) fellow. DRILL is a HIH D43 grant (D43TW010131) awarded to UKZN in 2015 to support a research training and induction programme for early career academics. PVD was partially supported as a Post-Doctoral Fellow by funding from the South African Medical Research Council (SAMRC) through its division of Research Capacity Development under the Intra-Mural Postdoctoral Fellowship Programme from funding received from the South African Treasury. The content hereof is the sole responsibility of the authors and does not necessary represent the official views of the SAMRC or the funders.

The authors have no conflicts of interest to disclose.

Data sharing not applicable to this article as no datasets were generated or analyzed during the current study.

References

  • [1].Hao T, Li-Talley M, Buck A, et al. An emerging trend of rapid increase of leukemia but not all cancers in the aging population in the United States. Sci Rep 2019;9:12070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Slager SL, Rabe KG, Achenbach SJ, et al. Genome-wide association study identifies a novel susceptibility locus at 6p21. 3 among familial CLL. Blood 2011;117:1911–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Miranda-Filho A, Piñeros M, Ferlay J, et al. Epidemiological patterns of leukaemia in 184 countries: a population-based study. Lancet Haematol 2018;5:e14–24. [DOI] [PubMed] [Google Scholar]
  • [4].DeSantis CE, Miller KD, Dale W, et al. Cancer statistics for adults aged 85 years and older, 2019. CA A Cancer J Clin 2019;69:452–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Combest AJ. Overview of the recent developments in chronic lymphocytic leukemia, Part 1. J Hematol Oncol Pharm 2016;2:54–6. [Google Scholar]
  • [6].Sall A, Toure AO, Sall FB, et al. Characteristics of chronic lymphocytic leukemia in Senegal. BMC Hematol 2016;16:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Koffi KG, Nanho DC, Tolo A, et al. [Chronic lymphocytic leukemia in Subsaharian Africa: clinical outcome experience of Cote d’Ivoire]. Bull Cancer 2009;96:901–6. [DOI] [PubMed] [Google Scholar]
  • [8].Malam-Abdou B, Brah S, Djibrilla A, et al. Leucémie Lymphoïde Chronique au Niger: une Étude de 99 cas au Service d’Onco-Hématologie de l’Hôpital National de Niamey. Health Sci Dis 2018;19:93–6. [Google Scholar]
  • [9].Omoti C, Awodu O, Bazuaye G. Chronic lymphoid leukaemia: clinico-haematological correlation and outcome in a single institution in Niger Delta region of Nigeria. Int J Lab Hematol 2007;29:426–32. [DOI] [PubMed] [Google Scholar]
  • [10].Salawu L, Bolarinwa R, Durosinmi M. Chronic lymphocytic leukaemia: a-twenty-years experience and problems in Ile-Ife, South-Western Nigeria. Afr Health Sci 2010;10:187–92. [PMC free article] [PubMed] [Google Scholar]
  • [11].Fall AS, Dieye TN, Ndiaye FS, et al. Characteristics of chronic lymphocytic leukemia (CLL) in senegal. clinical features, cytology, immunophenotype, cytogenetic abnormalities and altered expression of micro-RNA. Blood 2013;122:5283. [Google Scholar]
  • [12].Hallek M. Chronic lymphocytic leukemia: 2017 update on diagnosis, risk stratification, and treatment. Am J Hematol 2017;92:946–65. [DOI] [PubMed] [Google Scholar]
  • [13].Jain N, Basu S, Thompson PA, et al. Nivolumab combined with Ibrutinib for CLL and Richter transformation: a phase II trial. Blood Cells Molecules Dis 2016;128. [Google Scholar]
  • [14].Hallek M. Chronic lymphocytic leukemia: 2020 update on diagnosis, risk stratification and treatment. Am J Hematol 2019;94:1266–87. [DOI] [PubMed] [Google Scholar]
  • [15].Hogg SJ, Vervoort SJ, Deswal S, et al. BET-bromodomain inhibitors engage the host immune system and regulate expression of the immune checkpoint ligand PD-L1. Cell Rep 2017;18:2162–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Joller N, Peters A, Anderson AC, et al. Immune checkpoints in central nervous system autoimmunity. Immunol Rev 2012;248:122–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Dyck L, Mills KH. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur J Immunol 2017;47:765–79. [DOI] [PubMed] [Google Scholar]
  • [18].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;102:562–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Yi JS, Cox MA, Zajac AJ. T-cell exhaustion: characteristics, causes and conversion. Immunology 2010;129:474–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Awan FT, Byrd JC. Chronic lymphocytic leukemia. Abeloff's Clinical Oncology. 2019;Amsterdam, Netherlands: Elsevier, 1850–1871.e5. [Google Scholar]
  • [21].Lanasa MC. Novel insights into the biology of CLL. Hematology Am Soc Hematol Educ Program 2010;2010:70–6. [DOI] [PubMed] [Google Scholar]
  • [22].Coiffier B, Lepretre S, Pedersen LM, et al. Safety and efficacy of ofatumumab, a fully human monoclonal anti-CD20 antibody, in patients with relapsed or refractory B-cell chronic lymphocytic leukemia: a phase 1-2 study. Blood 2008;111:1094–100. [DOI] [PubMed] [Google Scholar]
  • [23].Actis Dato V, Chiabrando GA. The role of low-density lipoprotein receptor-related protein 1 in lipid metabolism, glucose homeostasis and inflammation. Int J Mol Sci 2018;19:1780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Cartron G, de Guibert S, Dilhuydy MS, et al. Obinutuzumab (GA101) in relapsed/refractory chronic lymphocytic leukemia: final data from the phase 1/2 GAUGUIN study. Blood 2014;124:2196–202. [DOI] [PubMed] [Google Scholar]
  • [25].Hillmen P, Robak T, Janssens A, et al. Chlorambucil plus ofatumumab versus chlorambucil alone in previously untreated patients with chronic lymphocytic leukaemia (COMPLEMENT 1): a randomised, multicentre, open-label phase 3 trial. Lancet 2015;385:1873–83. [DOI] [PubMed] [Google Scholar]
  • [26].Zaja F, Vianelli N, Sperotto A, et al. Anti-CD20 therapy for chronic lymphocytic leukemia-associated autoimmune diseases. Leuk Lymphoma 2003;44:1951–5. [DOI] [PubMed] [Google Scholar]
  • [27].Huhn D, von Schilling C, Wilhelm M, et al. Rituximab therapy of patients with B-cell chronic lymphocytic leukemia. Blood 2001;98:1326–31. [DOI] [PubMed] [Google Scholar]
  • [28].Gassner FJ, Zaborsky N, Catakovic K, et al. Chronic lymphocytic leukaemia induces an exhausted T cell phenotype in the TCL 1 transgenic mouse model. Br J Haematol 2015;170:515–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Lad D, Hoeppli R, Huang Q, et al. Regulatory T-cells drive immune dysfunction in CLL. Leuk Lymphoma 2018;59:486–9. [DOI] [PubMed] [Google Scholar]
  • [30].De Matteis S, Molinari C, Abbati G, et al. Immunosuppressive Treg cells acquire the phenotype of effector-T cells in chronic lymphocytic leukemia patients. J Transl Med 2018;16:172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].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;121:1612–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Younes A, Brody J, Carpio C, et al. Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: a phase 1/2a study. Lancet Haematol 2019;6:e67–78. [DOI] [PubMed] [Google Scholar]
  • [33].Ding W, LaPlant BR, Call TG, et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 2017;129:3419–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Arenbergerova M, Fialova A, Arenberger P, et al. Killing two birds with one stone: response to pembrolizumab in a patient with metastatic melanoma and B-cell chronic lymphocytic leukaemia. J Eur Acad Dermatol Venereol 2018;32:e72–4. [DOI] [PubMed] [Google Scholar]
  • [35].Mato AR, Svoboda J, Prak ETL, et al. Phase I/II study of Umbralisib (TGR-1202) in combination with Ublituximab (TG-1101) and Pembrolizumab in patients with relapsed/refractory CLL and Richter's transformation. Am Soc Hematol 2018;37:119–20. [Google Scholar]
  • [36].Archibald WJ, Meacham PJ, Williams AM, et al. Management of melanoma in patients with chronic lymphocytic leukemia. Leuk Res 2018;71:43–6. [DOI] [PubMed] [Google Scholar]
  • [37].Moher D, Shamseer L, Clarke M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev 2015;4:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Health UDo, Services H. Common terminology criteria for adverse events v 4.0 (CTCAE). National Institutes of Health. National Cancer Care Institute 2010. [Google Scholar]
  • [39].Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics 1977;33:159–74. [PubMed] [Google Scholar]
  • [40].Higgins JP, Altman DG, Gotzsche PC, et al. The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. BMJ 2011;343:d5928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Downs SH, Black N. The feasibility of creating a checklist for the assessment of the methodological quality both of randomised and non-randomised studies of health care interventions. J Epidemiol Community Health 1998;52:377–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Cochran WG. The combination of estimates from different experiments. Biometrics 1954;10:101–29. [Google Scholar]
  • [43].Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med 2002;21:1539–58. [DOI] [PubMed] [Google Scholar]
  • [44].Schroll JB, Moustgaard R, Gotzsche PC. Dealing with substantial heterogeneity in Cochrane reviews. Cross-sectional study. BMC Med Res Methodol 2011;11:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Balshem H, Helfand M, Schunemann HJ, et al. GRADE guidelines: 3. Rating the quality of evidence. J Clin Epidemiol 2011;64:401–6. [DOI] [PubMed] [Google Scholar]

Articles from Medicine are provided here courtesy of Wolters Kluwer Health

RESOURCES