• Open Access
    Case Report

    How to treat a patient with psoriatic arthritis and chronic lymphocytic leukemia?

    Jürgen Braun 1,2*
    Kirsten Karberg 1
    Denis Poddubnyy 3

    Explor Musculoskeletal Dis. 2024;2:235–241 DOI: https://doi.org/10.37349/emd.2024.00051

    Received: March 06, 2024 Accepted: April 05, 2024 Published: June 20, 2024

    Academic Editor: Ivan Castellví Barranco, Hospital Universitari de la Santa Creu i Sant Pau, Spain

    Abstract

    A 76-year-old male patient who has been suffering from psoriatic arthritis (PsA) for 15 years was diagnosed with chronic lymphocytic leukemia (CLL) 18 months ago. He has been treated him with a Bruton’s tyrosine kinase (BTK) inhibitor (ibrutinib) at a dose of 420 mg once daily (q.d.) for his CLL. For about two years, he received a quite successful treatment with methotrexate and the subcutaneously administered tumor necrosis factor (TNF) inhibitor (adalimumab) for his PsA, until his plaque psoriasis worsened. He consulted us when the severity of his skin condition necessitated a change in his treatment regimen. In the following discussion, we explore treatment options for this clinical scenario, with a particular focus on managing PsA in the context of CLL as a comorbidity. Additionally, we report on the initial phase of treatment with an anti-interleukin-23 (IL-23) inhibitor (guselkumab), specifically targeting his aggravated psoriasis.

    Keywords

    Tumor necrosis factor inhibitors, interleukin-17, Th17, interleukin-23, tyrosine kinase inhibitors

    Introduction

    The function of a healthy immune system involves CD4+ and CD8+ T cells which assist B cells to make antibodies and fight microbes together with natural killer cells and macrophages. The production of cytokines and chemokines is a critical element for this function. Depending on the cytokine milieu, previously uncommitted CD4+ T cells differentiate into Th1 or Th2 cells and, if activated, they produce distinct cytokine patterns [1]. Th1 cells can eradicate intracellular pathogens whereas Th2 cells rather fight extracellular pathogens. While the Th1 system plays a role in autoimmunity, aberrant Th2 responses are associated with allergy. A third subset of CD4+ T cells are Th17 cells which secrete interleukin-17 (IL-17). These are pro-inflammatory and have a key role in the defense against extracellular bacteria and fungi but also in the development of autoimmunity. The secretion of IL-23 by antigen-presenting cells can activate Th17 cells [2, 3].

    IL-23 and Th17 cells play an important role in immune-mediated diseases such as psoriasis [4, 5], PsA, and axial spondyloarthritis (axSpA). In chronic inflammatory bowel disease (IBD) drugs targeting the cytokine IL-23 work well but not agents blocking IL-17 [6, 7]. This is in contrast to axSpA, where only anti-IL-17 works, while both agents are efficacious in psoriasis and PsA [810]. In rheumatology, these agents are grouped under biologic disease modifying anti-rheumatic drugs (bDMARDs) while the Janus kinase inhibitors (JAKi) are targeted synthetic (ts) DMARDs.

    The anti-IL-17 agents approved for psoriasis and PsA are secukinumab, ixekizumab, and bimekizumab, while the approved drugs targeting IL-23 are ustekinumab, risankizumab, tildrakizumab and guselkumab. The first IL-23 inhibitor (IL-23i), ustekinumab, targets the p40 subunit shared by IL-23 and IL-12, the other IL-23i are directed against the p19 subunit of IL-23. Anti-IL-23 agents are known to be efficacious also for nails, scalp, and hands [11].

    Among the many JAKi, four currently play a role in rheumatology: tofacitinib, upadacitinb, baricitinib, and filgotinib. There are four JAKs to be inhibited: JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2), and the JAKi have different selectivity [12].

    However, there are more options for the treatment of PsA including tumor necrosis factor inhibitors (TNFi), abatacept, and apremilas. The reintroduction of TNFi after the development of paradoxical psoriasis known to occur in < 5% of cases [13] treated with TNFi was not found to be very successful in a larger case series [14]. Abatacept has no strong effects on the skin in PsA. Of note, in patients with a history of malignancy, apremilast seems to safely work in patients with PsA [15].

    Case report

    A 76-year-old male patient had longstanding PsA for 15 years. The proximal interphalangeal (PIP) and the distal interphalangeal (DIP) joints, the fingernails, and the skin (plaque psoriasis) were involved. The patient had been treated with methotrexate 15 mg s.c./week and non-steroidal anti-inflammatory drugs (NSAIDs) such as ibuprofen 400–1,200 mg on demand for 10 years, and in the last 2 years also with adalimumab 40 mg every 2 weeks. In October 2023 he experienced worsening of the nail and skin lesions—a problem known to occur with TNFi [13], while the joints hadn’t worsened much.

    In March 2022 chronic lymphocytic leukemia (CLL) had been diagnosed. The disease was treated with Bruton’s tyrosine kinase inhibitor (BTKi) ibrutinib, dosed 420 mg once daily (q.d.) [16]. The treatment with TNFi was not considered as a risk factor for malignancy [17].

    Taken all considerations together (see details in the discussion), we decided to treat our PsA patient with the serious comorbidity of CLL with the novel anti-IL-23 antibody guselkumab [9, 12].

    Since, after the first two doses given 4 weeks apart there was only limited success, the hematologist decided to change the CLL treatment to zanubrutinib, a second-generation BTKi with enhanced target selectivity and occupancy of the kinase binding site, and superior efficacy for CLL [18]—also because ibrutinib may cause or worsen arthralgias [19].

    When we saw the patient 3 months after the start of this regimen—the dosage of zanubrutinib was only 2 × 80 mg/day (approved dose 160 mg twice daily), ongoing methotrexate s.c. he reported having only minor joint symptoms using 400 mg of ibuprofen 2–3 times a week, no more skin lesions, and largely improved finger and toenails.

    Discussion

    This case is of interest because we had to search the literature for arguments in favor or against the available DMARDs.

    In this regard, the role of Th17 cells in cancer has not been really clarified to date. Depending on the form of cancer, their activities can be either pro- or anti-tumorigenic. As the most predominant form of leukemia in the developed world, the B-cell malignancy CLL accounts for about 11% of all hematologic neoplasms [20, 21]. Complex interactions in the microenvironment support the survival of malignant B cells [22, 23]. However, dysfunctions in the T-cell compartment contribute to disease progression [24]. A terminally differentiated phenotype develops which is characterized by a low proliferation capacity and impaired immune functions. This defect in the T-cell immune response in CLL is considered a major factor for the development of secondary malignancies and infections, which contribute to the increased morbidity and mortality in this disease [25]. The increase in IL-17 serum levels and/or Th17 cells in CLL compared to healthy individuals shows some correlation with prognostic markers and changes in the tumor microenvironment [26]. However, the exact mechanisms how this contributes to CLL progression are not clear. The effects of CLL-targeted therapies on Th17 cells and their potential in therapies are incompletely understood. Nevertheless, Th17 cells are elevated in CLL patients with better prognostic markers and this correlated with longer survival [27]. There are also non-Th17 IL-17A-expressing cells in CLL such as maturing granulocytes and mature mast cells which may have an influence on the microenvironmental milieu in leukemic spleens hereby promoting the recruitment and/or expansion of Th17 and other IL-17-expressing cells [28]. In that study with many untreated CLL patients in different stages, higher Th17 and IL-17A values were associated with less progression of the disease, and lower Th17 and IL-17A values were seen in patients with unfavorable prognostic factors [27]. In addition, IL-17 was shown to be also able to induce IL-6 production [29]. Furthermore, the ability of Th17 cells to neutralize tumors was shown to be greater than that of Th1 polarized cells which was attributed to Th17 cells acquiring a Th17/Th1 phenotype that secretes interferon-γ (IFN-γ) [21, 30]. However, there are also somewhat conflicting data showing that the situation is not entirely clear yet [31, 32]. Nevertheless, in conclusion, we found that anti-IL-17 therapy was not a preferable option for our patient.

    The literature on IL-23 is, in comparison to IL-17, sparse. Nevertheless, CLL cells activated in vitro have been shown to produce IL-23 and the presence of IL-23 in lymphoid tissues of CLL patients suggests that an autocrine/paracrine loop may induce cell proliferation in CLL. In accordance, interference with the IL-23 receptor (IL-23R)/IL-23 axis using an anti-IL-23p19 antibody showed effects in xenografted mice—suggesting potential therapeutic strategies [33]. In conclusion, we found no relevant arguments against anti-IL-23 treatment in our patients.

    Tyrosine kinases (TKs) are enzymes that can transfer a phosphate group from adenosine triphosphate (ATP) to the tyrosine residues of specific intracellular proteins; they work as an on/off switching of cellular functions. Phosphorylation of proteins by kinases is a centrally important mechanism for signal transduction and regulating cellular activity. Accordingly, there are more than 50 FDA-approved TK inhibitors (TKi) [34]. A total of 90 human genes contain 94 protein TK domains. Four of those genes contain both, an active kinase and a so called pseudokinase domain—domains without catalytic activity: JAK domains JAK1, JAK2, JAK3, and the tyrosine-protein kinase domain TYK2 [35]. These proteins are associated with type I and type II cytokine receptors. The regulation of JAK involves cytokines including ILs, IFNs, and hemopoietins which can activate JAK to regulate the JAK-STAT pathway [14, 35]. The JAK1-dependent pathways include IFN-α/pSTAT5, IL-6/pSTAT, and JAK2-dependent and JAK3-dependent pathways include IL-2, IL-15, IL-4 (JAK1/JAK3), IFN-γ (JAK1/JAK2), granulocyte colony stimulating factor, IL-12, IL-23 (JAK2/TYK2) and granulocyte-macrophage colony-stimulating factor (JAK2/JAK2) [36]. Of note, a small-molecule TYK2 inhibitor, deucravacitinib, has recently been approved for moderate-to-severe plaque psoriasis [37].

    BTKi play a crucial role in B-cell development, differentiation, proliferation, and survival [38]. Ibrutinib, an irreversible BTKi, is an approved oral targeted therapy for CLL but patients may have adverse events and, therefore, may need to reduce the dose or even have to stop treatment [39]. Another JAKi, ruxolitinib, with selectivity for JAK1 and JAK2, approved for more than 10 years, is used for the treatment of myelofibrosis, polycythemia vera, and acute graft-versus-host disease but failed to control and even accelerated CLL in clinical trials. Indeed, ruxolitinib may even release activating signals for CLL cells. Furthermore, JAK inhibition provided no added value for disease control in CLL patients [40].

    Finally, there has been some discussion on the role of JAKi regarding the risk of malignancy [41] and major cardiovascular events (MACE) [42]. However, the risk of developing cancer in patients treated with JAKi was only increased relative to TNFi—agents which rather seem to reduce the risk [41]. Of note, cancers were rare in all comparisons. Furthermore, the risk of MACE was mainly increased in patients who already had cardiovascular events [42]. Taken together, we decided that therapy with a JAKi was not a preferable option for our patient. In conclusion, the treatment of our patient with PsA and CLL with the IL-23 inhibitor guselkumab had relatively good short-term efficacy after 3 months. The influence of co-treatment with BTKi needs further study in PsA and likely also many other diseases. We hope that it may be helpful for colleagues in a similar clinical situation to know what we have determined and considered in this case.

    Abbreviations

    BTKi:

    Bruton’s tyrosine kinase inhibitor

    CLL:

    chronic lymphocytic leukemia

    DMARDs:

    disease modifying anti-rheumatic drugs

    IFN-γ:

    interferon-γ

    IL-17:

    interleukin-17

    JAK:

    Janus kinase

    JAKi:

    Janus kinase inhibitors

    PsA:

    psoriatic arthritis

    TKs:

    tyrosine kinases

    TNFi:

    tumor necrosis factor inhibitors

    TYK2:

    tyrosine kinase 2

    Declarations

    Author contributions

    JB: Conceptualization, Investigation, Writing—original draft, Writing—review & editing. KK: Investigation, Writing—review & editing. DP: Supervision, Writing—review & editing. All authors read and approved the submitted version.

    Conflicts of interest

    Jürgen Braun, who is the Associate Editor of Exploration of Musculoskeletal Diseases, had no involvement in the decision-making or the review process of this manuscript. The other authors declare that they have no conflicts of interest.

    Ethical approval

    The Ethical Committee of the Ärztekammer Berlin declared on February 12th, 2024 that no ethical vote was necessary for this case report.

    Consent to participate

    Informed consent to participate in the study was obtained from the participants.

    Consent to publication

    The patient has been informed about this publication, and he gave written consent.

    Availability of data and materials

    The raw data supporting the conclusions of this manuscript will be made available by the authors, without undue reservation, to any qualified researcher.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2024.

    References

    Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol. 1986;136:234857. [PubMed]
    Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol. 2005;6:112332. [DOI] [PubMed]
    Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. 2005;6:113341. [DOI] [PubMed] [PMC]
    Schett G, McInnes IB, Neurath MF. Reframing Immune-Mediated Inflammatory Diseases through Signature Cytokine Hubs. N Engl J Med. 2021;385:62839. [DOI] [PubMed]
    Braun J, Kiltz U, Baraliakos X. Emerging therapies for the treatment of spondyloarthritides with focus on axial spondyloarthritis. Expert Opin Biol Ther. 2023;23:195206. [DOI] [PubMed]
    Schmitt H, Neurath MF, Atreya R. Role of the IL23/IL17 Pathway in Crohn’s Disease. Front Immunol. 2021;12:622934. [DOI] [PubMed] [PMC]
    Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PD, et al.; Secukinumab in Crohn's Disease Study Group. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012;61:1693700. [DOI] [PubMed] [PMC]
    Reich K, Warren RB, Lebwohl M, Gooderham M, Strober B, Langley RG, et al. Bimekizumab versus Secukinumab in Plaque Psoriasis. N Engl J Med. 2021;385:14252. [DOI] [PubMed]
    Coates LC, Gossec L, Theander E, Bergmans P, Neuhold M, Karyekar CS, et al. Efficacy and safety of guselkumab in patients with active psoriatic arthritis who are inadequate responders to tumour necrosis factor inhibitors: results through one year of a phase IIIb, randomised, controlled study (COSMOS). Ann Rheum Dis. 2022;81:35969. [DOI] [PubMed] [PMC]
    Gossec L, Baraliakos X, Kerschbaumer A, de Wit M, McInnes I, Dougados M, et al. EULAR recommendations for the management of psoriatic arthritis with pharmacological therapies: 2019 update. Ann Rheum Dis. 2020;79:70012. [DOI] [PubMed] [PMC]
    Orbai AM, Chakravarty SD, You Y, Shawi M, Yang YW, Merola JF. Efficacy of Guselkumab in Treating Nails, Scalp, Hands, and Feet in Patients with Psoriasis and Self-reported Psoriatic Arthritis. Dermatol Ther (Heidelb). 2023;13:285968. [DOI] [PubMed] [PMC]
    Tanaka Y, Luo Y, O’Shea JJ, Nakayamada S. Janus kinase-targeting therapies in rheumatology: a mechanisms-based approach. Nat Rev Rheumatol. 2022;18:13345. [DOI] [PubMed] [PMC]
    Laga AC, Vleugels RA, Qureshi AA, Velazquez EF. Histopathologic Spectrum of Psoriasiform Skin Reactions Associated With Tumor Necrosis Factor-α Inhibitor Therapy. A Study of 16 Biopsies. Am J Dermatopathol. 2010;32:56873. [DOI] [PubMed]
    Mazloom SE, Yan D, Hu JZ, Ya J, Husni ME, Warren CB, et al. TNF-α inhibitor–induced psoriasis: A decade of experience at the Cleveland Clinic. J Am Acad Dermatol. 2020;83:15908. [DOI] [PubMed]
    Sabella DVA, Venerito V, Fornaro M. AB0920 Safety of Apremilast in PsA patients with history of malignancies or active cancer: a retrospective study. EULAR 2022 AB0920. Ann Rheum Dis. 2022;81:1589. [DOI]
    Burger JA, Tedeschi A, Barr PM, Robak T, Owen C, Ghia P, et al.; RESONATE-2 Investigators. Ibrutinib as Initial Therapy for Patients with Chronic Lymphocytic Leukemia. N Engl J Med. 2015;373:242537. [DOI] [PubMed] [PMC]
    Hellgren K, Dreyer L, Arkema EV, Glintborg B, Jacobsson LT, Kristensen LE, et al.; ARTIS Study Group; For the DANBIO Study Group. Cancer risk in patients with spondyloarthritis treated with TNF inhibitors: a collaborative study from the ARTIS and DANBIO registers. Ann Rheum Dis. 2017;76:10511. [DOI] [PubMed]
    Molica S, Tam C, Allsup D, Polliack A. Advancements in the Treatment of CLL: The Rise of Zanubrutinib as a Preferred Therapeutic Option. Cancers (Basel). 2023;15:3737. [DOI] [PubMed] [PMC]
    Rhodes JM, LoRe VA 3rd, Mato AR, Chong EA, Barrientos JC, Gerson JN, et al. Ibrutinib-associated Arthralgias/Myalgias in Patients With Chronic Lymphocytic Leukemia: Incidence and Impact on Clinical Outcomes. Clin Lymphoma Myeloma Leuk. 2020;20:43844. [DOI] [PubMed] [PMC]
    Hallek M, Al Sawaf O. Chronic lymphocytic leukemia: 2022 update on diagnostic and therapeutic procedures. Am J Hematol. 2021;96:1679705. [DOI] [PubMed]
    Gamal W, Sahakian E, Pinilla-Ibarz J. The role of Th17 cells in chronic lymphocytic leukemia: friend or foe? Blood Adv. 2023;7:240117. [DOI] [PubMed] [PMC]
    Woyach JA, Johnson AJ, Byrd JC. The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood. 2012;120:117584. [DOI] [PubMed] [PMC]
    Burger JA. The CLL cell microenvironment. Adv Exp Med Biol. 2013;792:2545. [DOI] [PubMed]
    Riches JC, Davies JK, McClanahan F, Fatah R, Iqbal S, Agrawal S, et al. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood. 2013;121:1612. [DOI] [PubMed] [PMC]
    The BW, Tam CS, Handunnetti S, Worth LJ, Slavin MA. Infections in patients with chronic lymphocytic leukaemia: mitigating risk in the era of targeted therapies. Blood Rev. 2018;32:499507. [DOI] [PubMed]
    Gorczynski RM. IL-17 Signaling in the Tumor Microenvironment. Adv Exp Med Biol. 2020;1240:4758. [DOI] [PubMed]
    Hus I, Bojarska-Junak A, Chocholska S, Tomczak W, Woś J, Dmoszyńska A, et al. Th17/IL-17A might play a protective role in chronic lymphocytic leukemia immunity. PLoS One. 2013;8:e78091. [DOI] [PubMed] [PMC]
    Jain P, Javdan M, Feger FK, Chiu PY, Sison C, Damle RN, et al. Th17 and non-Th17 interleukin-17-expressing cells in chronic lymphocytic leukemia: delineation, distribution, and clinical relevance. Haematologica. 2012;97:599607. [DOI] [PubMed] [PMC]
    Zhu F, McCaw L, Spaner DE, Gorczynski RM. Targeting the IL-17/IL-6 axis can alter growth of Chronic Lymphocytic Leukemia in vivo/in vitro. Leuk Res. 2018;66:2838. [DOI] [PubMed]
    Muranski P, Borman ZA, Kerkar SP, Klebanoff CA, Ji Y, Sanchez-Perez L, et al. Th17 cells are long lived and retain a stem cell-like molecular signature. Immunity. 2011;35:97285. [DOI] [PubMed] [PMC]
    Bankir M, Acik DY. IL-17 and IL-23 levels in patients with early-stage chronic lymphocytic leukemia. North Clin Istanb. 2020;8:2430. [DOI] [PubMed] [PMC]
    Karmali R, Paganessi LA, Frank RR, Jagan S, Larson ML, Venugopal P, et al. Aggressive disease defined by cytogenetics is associated with cytokine dysregulation in CLL/SLL patients. J Leukoc Biol. 2013;93:16170. [DOI] [PubMed] [PMC]
    Cutrona G, Tripodo C, Matis S, Recchia AG, Massucco C, Fabbi M, et al. Microenvironmental regulation of the IL-23R/IL-23 axis overrides chronic lymphocytic leukemia indolence. Sci Transl Med. 2018;10:eaal1571. [DOI] [PubMed]
    Allegra S, Dondi E, De Francia S. Tyrosine kinase inhibitors: the role of pharmacokinetics and pharmacogenetics. Expert Opin Drug Metab Toxicol. 2023;19:7339. [DOI] [PubMed]
    Taylor PC, Choy E, Baraliakos X, Szekanecz Z, Xavier RM, Isaacs JD, et al. Differential properties of Janus kinase inhibitors in the treatment of immune-mediated inflammatory diseases. Rheumatology (Oxford). 2024;63:298308. [DOI] [PubMed] [PMC]
    Traves PG, Murray B, Campigotto F, Galien R, Meng A, Di Paolo JA. JAK selectivity and the implications for clinical inhibition of pharmacodynamic cytokine signalling by filgotinib, upadacitinib, tofacitinib and baricitinib. Ann Rheum Dis. 2021;80:86575. [DOI] [PubMed] [PMC]
    Qiu J, Liu J, Liu W, Lin F, Shi N. The efficacy and safety of tyrosine kinase 2 inhibitor deucravacitinib in the treatment of plaque psoriasis: a systematic review and meta-analysis of randomized controlled trials. Front Med (Lausanne). 2023;10:1264667. [DOI] [PubMed] [PMC]
    Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, et al. Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369:3242. Erratum in: N Engl J Med. 2014;370:786. [DOI] [PubMed] [PMC]
    Shanafelt TD, Wang XV, Kay NE, Hanson CA, O’Brien S, Barrientos J, et al. Ibrutinib–Rituximab or Chemoimmunotherapy for Chronic Lymphocytic Leukemia. N Engl J Med. 2019;381:43243. [DOI] [PubMed] [PMC]
    Spaner DE, Luo Y, Wang G, Gallagher J, Tsui H, Shi Y. Janus kinases restrain chronic lymphocytic leukemia cells in patients on ibrutinib: Results of a phase II trial. Cancer Med. 2021;10:878998. [DOI] [PubMed] [PMC]
    Russell MD, Stovin C, Alveyn E, Adeyemi O, Chan CKD, Patel V, et al. JAK inhibitors and the risk of malignancy: a meta-analysis across disease indications. Ann Rheum Dis. 2023;82:105967. [DOI] [PubMed] [PMC]
    Dougados M, Charles-Schoeman C, Szekanecz Z, Giles JT, Ytterberg SR, Bhatt DL, et al. Impact of cardiovascular risk enrichment on incidence of major adverse cardiovascular events in the tofacitinib rheumatoid arthritis clinical programme. Ann Rheum Dis. 2023;82:57577. [DOI] [PubMed] [PMC]
    • Citation
      Cite this Article
      • Article Metrics
      • View: 116
      • Download: 7
      • Times Cited: 0