Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Sep 29.
Published in final edited form as: Curr Opin Pharmacol. 2022 Dec 27;68:102320. doi: 10.1016/j.coph.2022.102320

Pharmacological strategies for mitigating anti-TNF biologic immunogenicity in rheumatoid arthritis patients

Christian A Fernandez 1
PMCID: PMC10540078  NIHMSID: NIHMS1931652  PMID: 36580770

Abstract

Tumor necrosis factor alpha (TNFα) inhibitors are a mainstay of treatment for rheumatoid arthritis (RA) patients after failed responses to conventional disease-modifying antirheumatic drugs (DMARDs). Despite the clinical efficacy of TNFα inhibitors (TNFi), many RA patients experience TNFi treatment failure due to the development of anti-drug antibodies (ADAs) that can neutralize drug levels and lead to RA disease relapse. Methotrexate (MTX) therapy with concomitant TNFα inhibitors decreases the risk of TNFi immunogenicity, but additional and/or alternative strategies are needed to reduce MTX-associated toxicities and to further increase its potency for preventing TNFα inhibitor immunogenicity. In this review, we highlight the limitations of MTX for mitigating TNFα inhibitor immunogenicity, and we discuss potential alternative pharmacological targets for decreasing the risk of immunogenicity during TNFα inhibitor therapy based on the key kinases, second messengers, and shared signaling mechanisms of lymphocyte receptor signaling.

BACKGROUND

Rheumatoid arthritis (RA) is the most common type of chronic inflammatory disease affecting the joints. The goals of RA treatment are to achieve disease remission by attenuating joint inflammation and damage [1]. Low-dose methotrexate (MTX, 5–25 mg/week) is the first-line therapy for RA [2], whereas biological disease-modifying antirheumatic drugs (DMARDs), such as tumor necrosis factor alpha (TNFα) inhibitors are effective in reducing inflammation after failed or inadequate responses to conventional DMARDs [3]. Currently, five TNFα inhibitors (TNFi) have been approved for RA treatment: adalimumab, infliximab, etanercept, golimumab and certolizumab pegol [4]. To overcome the high-cost limitation associated with TNFi therapy and to increase access to these agents, several biosimilars have been introduced that exhibit similar efficacy as the original TNFi but are more cost-effective [5]. Despite the remarkable improvement in disease outcomes, a significant fraction of patients experience TNFi treatment failure due to the development of immune responses to the biologic [6]. Interestingly, it has been demonstrated that concomitant MTX decreases the risk of TNFi immunogenicity [7]. Nevertheless, additional and/or alternative strategies are needed to reduce toxicities associated with MTX therapy and to further decrease the frequency of TNFi immunogenicity. In this review we highlight the limitations of strategies currently used to mitigate TNFi immunogenicity, and we discuss potential alternative pharmacological targets for mitigating the risk of immunogenicity during TNFi biologic therapy.

Immunogenicity to TNFα inhibitors leads to treatment failure.

Immunogenicity is a common limitation associated with the clinical use of biologics or protein-based therapies from non-human and human sources. Biologics induce immune responses that can lead to the development of anti-drug antibodies (ADAs), which can have several clinical consequences ranging from RA disease relapse to severe life-threatening conditions [8]. Generally, ADA development involves a T cell-dependent antibody response that is initiated by the uptake and presentation of the biologic, in the form of antigenic peptides, on the major histocompatibility complex (MHC) class II molecules expressed on the cell surface of antigen-presenting cells (APCs), such as dendritic cells. Two essential steps during antigen sensitization involve T cell and B cell activation. Naïve T cells are activated upon antigen recognition by the T cell receptor (TCR, signal 1) and CD28 costimulation by B7 molecules (signal 2) [9]. Activated T cells can stimulate the differentiation of B cells to antibody-secreting plasma cells upon T cell receptor recognition of an antigen presented on the MHC II molecules of B cells [10]. Repeated antigen exposure can lead to B cell IgG isotype switching [11], where IgG1 and IgG4 ADAs are common to TNFi biologics [12,13].

ADA generation can be affected by host and drug related factors, including the frequency and duration of the treatment regimen [14,15]. Among patients on TNFi therapy, most develop antibodies within the first year of treatment resulting in loss of clinical response and discontinuation of treatment [1618]. Since ADAs play such an important role in the prediction of treatment efficacy and failure, monitoring of ADAs and drug levels is key for attaining disease remission. Several methods of ADA detection have been developed [19], many of which are limited with regards to detecting broad immunogenicity [20,21]. Nevertheless, several association studies investigating ADAs have attempted to identify biomarkers that predict TNFi immunogenicity. Recently, anti-adalimumab levels were found to be negatively correlated with anti-hinge antibodies [22]. Furthermore, anti-TROVE2 antibody levels and baseline BAFF levels have been identified as a biomarker capable of independently predicting TNFi immunogenicity [23,24]. Despite these positive contributions, the prediction of TNFi immunogenicity is challenging and requires larger prospective studies for validation. Therefore, there remains an urgent need to identify and develop prevention strategies to attenuate the risk of TNFi immunogenicity in RA patients.

MTX is the standard of care for TNFi immunogenicity, but additional or better options are needed for optimal RA treatment.

Concomitant administration of low-dose MTX with TNFi biologics decreases the risk or incidence of TNFi ADAs, rescues TNFi drug levels, and decreases the risk of TNFi therapy discontinuation [2529]. Interestingly, protection against TNFi immunogenicity by MTX is more pronounced compared to other DMARDs or corticosteroids in RA patients [30]. This underscores the need to better understand the molecular mechanisms of biologic immunogenicity for achieving optimal RA efficacy while minimizing the risk of toxicities that can adversely impact the quality of life of RA patients.

MTX is a folic acid antimetabolite used for the treatment of hematologic malignancies that potently inhibits dihydrofolate reductase (DHFR) when used at high doses. DHFR inhibition leads to diminished de novo purine and pyrimidine biosynthesis via preventing the synthesis of tetrahydrofolate [31]. However, because MTX is used at much lower doses for the treatment of RA, it is unlikely that this mechanism provides protection against ADAs [32]. Leucovorin, also known as folinic acid, is a reduced folate that rescues from MTX toxicities during leukemia therapy by bypassing DHFR and directly restoring the conversion of deoxyuridine monophosphate (dUMP) to deoxythymidine monophosphate (dTMP) by thymidylate synthase [32]. Consistent with the effect of MTX on immunogenicity being independent of DHFR inhibition, clinical studies in RA patients receiving folinic acid supplementation have shown that DHFR inhibition rescue protects from MTX toxicities without compromising RA efficacy [33]. Rather, MTX (i.e., MTX polyglutamates) has been shown to inhibit 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) transformylase, resulting in increased intracellular AICAR levels and thereby extracellular adenosine [34]. Several studies have validated the role of adenosine on the anti-inflammatory effects of MTX, including those demonstrating that the effects of MTX are dependent on A2A and A3 adenosine receptors and cell surface CD39 and CD73 enzymes, which convert adenine nucleotides to adenosine [33]. Preclinical studies support that the effect of MTX on extracellular adenosine is essential for protection against TNFi immunogenicity, where BAFF transgenic mice overexpressing CD73 and CD39 were protected from TNFi ADAs by MTX, whereas wild-type mice were not [35]. Based on studies elucidating the effect of MTX on extracellular adenosine levels, there has been a call to identify strategies that can further enhance or improve protection against TNFi immunogenicity [36]. Furthermore, while MTX is suitable for protecting against TNFi immunogenicity, its safety profile can lead to concerns among RA patients and thereby MTX discontinuation [37]. Common toxicities with low-dose MTX include gastrointestinal, hepatic, and hematologic adverse events [38]. MTX therapy may also require lifestyle modifications including changes in contraception use and alcohol consumption habits due to MTX teratogenicity and risk of hepatoxicity, respectively [39]. Altogether, because of the need for enhanced MTX efficacy and decreased toxicities, there remains a need to identify alternative pharmacological targets that can safely mitigate TNFi biologic immunogenicity.

PHARMACOLOGICAL STRATEGIES FOR MITIGATING BIOLOGIC IMMUNOGENICITY

Strategies for mitigating biologic immunogenicity require attenuating antigen-specific CD4+ T and B cell activation.

Because CD4+ T and B cells are essential for the development of biologic immunogenicity, suppressing their activation and proliferation during biologic exposure is essential for attenuating drug-induced immunogenicity. Unlike immunosuppressive agents used for the prevention of graft-versus-host-disease (e.g., tacrolimus and cyclosporine A or CsA), optimal immunosuppressive strategies will have minimal toxicities, limit the risk of neutropenia and infections, and will not attenuate RA efficacy. Below we review pharmacological strategies that can potentially modulate CD4+ T cell and/or B cell receptor signaling for attenuating biologic immunogenicity (Figure 1). While cytotoxic antiproliferative agents, such as azathioprine and cyclophosphamide, have been previously used for immunosuppression [40]; these agents will not be considered due to concerns regarding their toxicity and adverse effects.

Figure 1. There are several pharmacological targets of lymphocyte signaling that can be inhibited for the prevention of TNFi immunogenicity.

Figure 1.

B (BCR) and T cell receptor (TCR) ligation leads to a cascade of signaling events culminating in the activation of transcription factors that drive lymphocyte activation and function. TCR signaling is initiated by co-receptor (e.g., CD4)-associated lymphocyte-specific protein tyrosine kinase (LCK) phosphorylation of tyrosine residues located on the CD3-ζ subunits of the TCR. Zeta-chain-associated protein kinase-70 (ZAP-70) docks to the phosphorylated sites, phosphorylates linker for the activation of T cells (LAT), resulting in activation of phospholipase C-ɣ (PLC-ɣ) and generation of inositol triphosphate (IP3) and diacylglycerol (DAG). Similarly, BCR crosslinking leads to phosphorylation of CD79 and activation of spleen tyrosine kinase (SYK). SYK phosphorylates several targets, including Bruton’s tyrosine kinase (BTK), which results in the generation of secondary messengers, similar to TCR signaling. IP3 and DAG generated during lymphocyte receptor signaling results in activation of calcineurin, protein kinase C (PKC), and mitogen-activated protein (MAP) kinases, which initiate the transcription of genes involved in lymphocyte activation via nuclear factor of activated T cells (NFAT), nuclear factor kappa B (NF-κb), and activator protein-1 (AP-1) transcription factors. Adenosine signaling can mitigate proinflammatory responses by increasing cyclic 3′,5′-adenosine monophosphate (cAMP) levels and activating protein kinase A (PKA), which phosphorylates cAMP responsive element binding protein (CREB). Activated CREB can lead to anti-inflammatory signaling by modulating NF-κB-driven transcription. Potential pharmacological targets for the prevention of biologic immunogenicity are indicated in red font. Created with BioRender.com

T cell receptor (TCR) and B cell receptor (BCR) signaling: potential therapeutic targets.

Naïve T cell activation requires antigen-specific signaling through the TCR and secondary costimulatory signaling through CD28 from a professional APC. TCR cross-linking and co-stimulation leads to a cascade of intracellular signaling involving several enzymes and transcription factors that promote cytokine secretion and proliferation. Briefly (Figure 1), TCR signaling leads to phosphorylation of tyrosine residues located in the cytosolic domains of the CD3-ζ subunits of the TCR by lymphocyte-specific protein tyrosine kinase (LCK). These phosphorylated residues are docked by zeta-chain-associated protein kinase-70 (ZAP-70), which phosphorylates linker for the activation of T cells (LAT), resulting in activation of phospholipase C-ɣ (PLC-ɣ) and generation of inositol triphosphate (IP3) and diacylglycerol (DAG). These second messengers lead to the activation of protein kinase C (PKC)-Ɵ, mitogen-activated protein (MAP) kinases, and Ca2+-calcineurin which collectively initiate the transcription of genes involved in T cell activation via nuclear factor of activated T cells (NFAT), activator protein-1 (AP-1), and nuclear factor kappa B (NF-κb) transcription factors. Similarly, B cell activation signaling via the B cell receptor (BCR) involves several kinases, including Src family kinases (e.g., Lyn, Fyn), spleen tyrosine kinase (SYK), Bruton tyrosine kinase (BTK), AKT, and phosphoinositide 3-kinases (PI3K) [41]. BCR crosslinking leads to Lyn-dependent PI3K activation, which phosphorylates phosphatidylinositol 4,5-bisphophate (PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3) [41]. PIP3 recruits BTK and AKT to the plasma membrane [42]. The secondary messengers DAG and IP3 are produced from a signaling complex of SYK, BTK, and PLC-γ resulting in PKC-β and calcineurin activation [41]. Activated AKT phosphorylates Forkhead Box class O (Foxo) transcription factors, which are involved in regulating apoptosis [43]. Both NF-κB and NFAT transcription factors are important regulators of BCR-induced gene expression changes during B cell activation [44]. Based on the key kinases, second messengers, and shared signaling mechanisms between lymphocyte receptor signaling, it is plausible that strategies mitigating key known signaling events during T/B cell activation can attenuate unwanted drug-induced immunogenicity.

NFAT inhibition can attenuate drug-induced immunogenicity

Tacrolimus (FK506) and CsA prevent allograft rejection via inhibiting the enzyme activity of calcineurin, which is required for activation of NFAT family members during TCR signaling and activation [45]. NFAT regulates the expression of several genes involved in T cell activation and Th2 cell differentiation, including IL-2, IL-4, and IFN-γ [46]. Consistent with NFAT playing a role in the development of biologic immunogenicity, a genome-wide association study (GWAS) of asparaginase immunogenicity identified an association between a polymorphism in NFATC2 (rs6021191) and the risk of developing asparaginase immunogenicity, where asparaginase is a chemotherapeutic biologic (i.e., enzyme) used for the treatment of pediatric acute lymphoblastic leukemia [47]. The identified NFAT variant results in a gain-of-function phenotype, and, in agreement with the clinical discovery, a subsequent preclinical study using wild type and NFATC2-deficient mice demonstrated that genetic NFATC2 inhibition or pharmacological inhibition of NFAT using the 11R-VIVIT peptide resulted in attenuated anti-drug antibody and Th2 cytokine levels [48]. 11R-VIVIT inhibits NFAT activation via attenuating calcineurin-mediated NFAT dephosphorylation but does not inhibit calcineurin enzyme activity, unlike CsA and tacrolimus [49]. The peptide NFAT inhibitor has also demonstrated efficacy in lowering arthritis disease severity [50], but there are limitations associated with the delivery of the peptide and its protection from proteolytic degradation. Among clinically available pharmacological agents, only the antiplatelet agent dipyridamole has been reported to inhibit NFAT activation [51]. Dipyridamole has been shown preclinically and clinically to have anti-inflammatory properties [52,53]; however, dipyridamole has multiple pharmacological targets apart from NFAT that may contribute to its ability to modulate immune responses [52]. Therefore, the development of additional small molecule NFAT inhibitors are required to evaluate whether targeting NFAT activation is suitable for protecting against biologic immunogenicity.

Adenosine via the adenosine receptors leads to anti-inflammatory signaling

In addition to inhibiting NFAT, dipyridamole is a known equilibrative nucleoside transporter (ENT1/ENT2) inhibitor, which leads to increased extracellular adenosine by blocking the transport or intracellular reuptake of adenosine [54]. Adenosine signaling via the adenosine A2A receptor increases cyclic 3′,5′-adenosine monophosphate (cAMP) levels and activates PKA, which can phosphorylate and activate cAMP responsive element binding protein (CREB). Phosphorylated CREB can inhibit proinflammatory cytokines by modulating NF-κB-driven transcription (Figure 1) [55]. Consistent with the putative mechanisms of action of dipyridamole and MTX, selective adenosine A2A receptor agonists, such as CGS 21680 and regadenoson also exhibit anti-inflammatory properties [56,57]. Adenosine signaling is a compelling strategy for mitigating undesirable immune responses because the A2A receptor is expressed by various immune cells and because it can attenuate both T and B cell function [55,5861]. However, adenosine signaling can cause substantial side effects including dizziness, headaches, hypotension, nausea, and tachycardia due to the ubiquitous expression of adenosine receptors and the widespread effect of signaling through these receptors [62]. Therefore, despite many years of research towards the development of adenosine receptor agonist, only regadenoson has been approved by the FDA for clinical use [63].

PDE inhibitors can suppress immune cell function

In addition to its effects on endogenous adenosine, dipyridamole can also inhibit the aggregation of platelets through non-selective inhibition of phosphodiesterase (PDE) enzyme. PDE enzymes play a role in immune cell function by degrading cAMP and cGMP [64]. PDE inhibitors can thereby suppress immune cell function by increasing cAMP/cGMP and activating PKA/PKG, which phosphorylates CREB [64]. PKA and PKG can also phosphorylate several other targets, including the c-Src tyrosine kinase (Csk) and NFAT, thus PDE inhibition can attenuate lymphocyte signaling via multiple mechanisms [65,66]. The PDE enzyme family includes 11 subfamilies with different substrate specificities [67]. T lymphocytes express PDE1, PDE2, PDE3, PDE4, PDE5, PDE7, and PDE8, whereas B cells express PDE3, PDE4, and PDE7 [64]. PDE inhibition has been demonstrated to decrease T cell activation and IL-2 production [68]. However, while PDE4 inhibition in PBMCs has demonstrated decreased IgE production [69], the effect of PDE inhibition on B cell function is unclear [64].

Dipyridamole inhibits PDE5 [70,71], and similar to the anti-inflammatory properties of dipyridamole, the PDE5 inhibitor sildenafil can decrease cytokine levels in a model of multiple sclerosis [72], inhibit neuroinflammation [73], and prevent experimental autoimmune encephalomyelitis progression [74]. In patients, sildenafil has been shown to increase the suppressive effects of regulatory T cell on effector T cell function [74]. Several PDE inhibitors have been investigated for RA efficacy [64], including the clinically available PDE4 inhibitors roflumilast and apremilast. Altogether, while PDE inhibition is a viable strategy for attenuating biologic immunogenicity and controlling disease progression, the PDE family members required for TNFi immunogenicity are unclear.

Kinases involved in TCR and BCR signaling can modulate proinflammatory responses

Common transcription factors that mediate TCR and BCR signaling include NFAT, NF-κB, and AP-1 (Figure 1) [75]. The signaling that leads to activation of these transcription factor is initiated by receptor-ligand interaction with many common signaling molecules that can be consider as therapeutic targets for mitigating biologic immunogenicity. TCR and BCR signaling both include cleavage of phosphatidylinositol 4, 5 biphosphate (PIP2) by PLCγ to IP3 and DAG, which leads to activation of calcineurin, PKC, and mitogen-activated protein (MAP) kinases. Small molecules have been developed to inhibit IP3-mediated calcium mobilization; however, toxicity concerns have limited their clinical potential [76]. The development of PKC-Ɵ inhibitors is an area of active research with CC-90005 identified as a selective, safe, and in vivo-compatible small molecule that can mitigate IL-2 production and protect from graft versus host disease [77]. However, the lack of a clinically available PKC inhibitor and concerns of compensation by other PKC family members limit the current translational potential of these agents for biologic immunogenicity [78].

The AP-1 transcription factor consists of a Fos-Jun heterodimer that is regulated by the MAP kinase family of enzymes, which are involved in vital signal transduction pathways [79]. The MAPK kinase extracellular signal-regulated kinase (ERK1/2) is activated by MAPK/ERK kinase (MEK) subsequent to PLCγ-mediated DAG production and phosphorylates the ETS-like 1 (ELK-1) transcription factor. Phosphorylated Elk-1 induces expression of Fos, thereby contributing to the formation of AP-1, which, along with NFAT, drives IL-2 expression and T cell proliferation [80]. Consistent with the crucial role AP-1 plays in the induction of IL-2, MEK inhibitors have been shown to decrease the secretion of IL-2 from donor CD4+ and CD8+ T cell in dendritic cell co-cultures [81], and also decrease anti-CD3/CD28-induced T cell proliferation [82]. However, while the MEK inhibitor trametinib attenuates naïve and memory T-cell function in vitro, it paradoxically does not limit the effectiveness of adoptive cell therapy or checkpoint blockade in mice [83]. While preclinical studies also support that MEK inhibition can attenuate antigen-specific B cell activation, antibody production, and Th2 cytokine secretion [84], toxicities of MEK inhibition, which include rash, dermatitis, diarrhea, and fatigue [85], may preclude its use versus MTX or other agents for biologic immunogenicity.

Additional kinases that are critical for initiating TCR signaling include LCK, ZAP-70, and (IL-2 inducible T-cell kinase (ITK) (Figure 1). However, clinical pharmacological agents with high target selectivity and that have suitable safety profiles are not currently available to assess the utility of these targets for preventing biologic immunogenicity [8688]. In contrast, several kinase inhibitors targeting essential signaling molecules involved in initiating BCR signaling are clinically available for the treatment of hematological malignancies, including inhibitors of BTK, SYK, and PI3K (Figure 1) [89]. However, clinical studies investigating the safety of pharmacologically inhibiting these kinases have indicated that only BTK or SYK inhibition has suitable safety profiles for potentially mitigating biologic immunogenicity [9092]. Furthermore, both BTK and SYK inhibitors have demonstrated their potential for attenuating inflammatory disease, including to RA [93,94]. Pharmacological BTK inhibition has been demonstrated to block B but not T cell proliferation and modulate proinflammatory cytokine secretion [95,96]. In contrast, SYK inhibition decreases CD4+ T and B cell proliferation and proinflammatory cytokine levels [97100]. Therefore, based on the suitable safety profiles, immunomodulatory effects, and potential benefit of BTK or SYK inhibition on RA disease progression, additional studies should be considered to directly assess their ability to attenuate biologic immunogenicity relative to MTX.

Inhibition of cytokine signaling may also be a potential strategy for preventing biologic immunogenicity, where Janus kinase (JAK) / signal transducer and activator of transcription 1 (STAT1) signaling mediates cytokine responses, including to IL-2 [101]. Consistent with the key role of JAKs and STATs on signal transduction after cytokines induction by transcription factors, the JAK1/3 inhibitor tofacitinib, which is approved for the treatment of RA, has been shown to attenuate T cell proliferation and cytokine secretion [89,102]. Furthermore, tofacitinib has been shown to modulate B cell activation and reduce antibody secretion of peripheral blood B cells [103]. In agreement, clinical tofacitinib studies in RA patients have demonstrated that the JAK inhibitor reduces IgG and RF circulating levels [104]. Importantly, tofacitinib has an acceptable safety profile among RA patients that make the JAK/STAT pathway a potential clinical therapeutic target for improving biologic immunogenicity while limiting the toxicities associated with MTX [105].

CONCLUSIONS AND FUTURE PERSPECTIVES

The success of MTX in reducing TNFi immunogenicity has indicated that strategies leading to adenosine signaling can protect from this toxicity, presumably via attenuating NF-κB. Mechanistically, strategies that can attenuate the transcriptional activity of NFAT and/or AP-1 should enhance the anti-inflammatory properties of MTX. It is also possible that adenosine receptor agonist or inhibitors that increase extracellular adenosine levels may lead to similar effects as MTX or allow for lower MTX doses if used in combination. Ideally, alternative targets will have strong anti-inflammatory effects, limit toxicities, and contribute to RA efficacy. Therefore, pharmacological targets that have high specificity for lymphocyte activation or function may limit adverse drug reactions and enhance efficacy. In accordance, inhibitors of BTK, SYK, or JAK have suitable safety profiles and potent anti-inflammatory properties. Nevertheless, additional preclinical and clinical studies are required to determine the most cost- and treatment-effective strategy for preventing biologic immunogenicity.

ACKNOWLEDGEMENTS

This work was supported by the University of Pittsburgh School of Pharmacy and NIH Grant R01 CA216815.

REFERENCES

  • 1.Mian A, Ibrahim F, Scott DL: A systematic review of guidelines for managing rheumatoid arthritis. BMC Rheumatol 2019, 3:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Lucas CJ, Dimmitt SB, Martin JH: Optimising low-dose methotrexate for rheumatoid arthritis-A review. Br J Clin Pharmacol 2019, 85:2228–2234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Singh JA, Saag KG, Bridges SL Jr., Akl EA, Bannuru RR, Sullivan MC, Vaysbrot E, McNaughton C, Osani M, Shmerling RH, et al. : 2015 American College of Rheumatology Guideline for the Treatment of Rheumatoid Arthritis. Arthritis Rheumatol 2016, 68:1–26. [DOI] [PubMed] [Google Scholar]
  • 4.Ma X, Xu S: TNF inhibitor therapy for rheumatoid arthritis. Biomed Rep 2013, 1:177–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Dey M, Zhao SS, Moots RJ: Anti-TNF biosimilars in rheumatology: the end of an era? Expert Opinion on Biological Therapy 2021, 21:29–36. This manuscript reviews TNF biosimilars in rheumatic diseases, their role in a rapidly evolving treatment landscape, and speculate about the future for this iconic therapeutic class.
  • 6.Souto A, Maneiro JR, Gómez-Reino JJ: Rate of discontinuation and drug survival of biologic therapies in rheumatoid arthritis: a systematic review and meta-analysis of drug registries and health care databases. Rheumatology 2015, 55:523–534. [DOI] [PubMed] [Google Scholar]
  • 7.Krieckaert CL, Nurmohamed MT, Wolbink GJ: Methotrexate reduces immunogenicity in adalimumab treated rheumatoid arthritis patients in a dose dependent manner. Ann Rheum Dis 2012, 71:1914–1915. [DOI] [PubMed] [Google Scholar]
  • 8.Jani M, Dixon WG, Chinoy H: Drug safety and immunogenicity of tumour necrosis factor inhibitors: the story so far. Rheumatology (Oxford) 2018, 57:1896–1907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Cappell KM, Kochenderfer JN: A comparison of chimeric antigen receptors containing CD28 versus 4–1BB costimulatory domains. Nat Rev Clin Oncol 2021, 18:715–727. This review provides an overview of T cell costimulation by CD28 and 4–1BB and compares the efficacy and toxicity profiles associated with CARs containing either costimulatory domain.
  • 10.Sauerborn M, Brinks V, Jiskoot W, Schellekens H: Immunological mechanism underlying the immune response to recombinant human protein therapeutics. Trends Pharmacol Sci 2010, 31:53–59. [DOI] [PubMed] [Google Scholar]
  • 11.Nielsen SCA, Roskin KM, Jackson KJL, Joshi SA, Nejad P, Lee JY, Wagar LE, Pham TD, Hoh RA, Nguyen KD, et al. : Shaping of infant B cell receptor repertoires by environmental factors and infectious disease. Sci Transl Med 2019, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gorovits B: Current Considerations for Immunoglobulin Isotype Characterization of Antibody Response against Biotherapeutics. AAPS J 2020, 22:144. This manuscript reviews isotype-specific characteristics, methods of detection, and case studies of anti-drug antibody response against biotherapeutics.
  • 13.Hindryckx P, Novak G, Vande Casteele N, Khanna R, Laukens D, Jairath V, Feagan BG: Incidence, Prevention and Management of Anti-Drug Antibodies Against Therapeutic Antibodies in Inflammatory Bowel Disease: A Practical Overview. Drugs 2017, 77:363–377. [DOI] [PubMed] [Google Scholar]
  • 14.Schellekens H: Factors influencing the immunogenicity of therapeutic proteins. Nephrol Dial Transplant 2005, 20 Suppl 6:vi3–9. [DOI] [PubMed] [Google Scholar]
  • 15.Bartelds G, Wijbrandts C, Nurmohamed M, Wolbink G, De Vries N, Tak P, Dijkmans B, Crusius J, vand der Horst-Bruinsma IJA, Rheumatology ROJotACo: Anti-adalimumab antibodies in rheumatoid arthritis patients are associated with interleukin-10 gene polymorphisms. 2009, 60:2541–2542. [DOI] [PubMed] [Google Scholar]
  • 16.Wolbink GJ, Vis M, Lems W, Voskuyl AE, De Groot E, Nurmohamed MT, Stapel S, Tak PP, Aarden L, Dijkmans B: Development of antiinfliximab antibodies and relationship to clinical response in patients with rheumatoid arthritis. 2006, 54:711–715. [DOI] [PubMed] [Google Scholar]
  • 17.Pascual-Salcedo D, Plasencia C, Ramiro S, Nuño L, Bonilla G, Nagore D, Ruiz Del Agua A, Martínez A, Aarden L, Martín-Mola E, et al. : Influence of immunogenicity on the efficacy of long-term treatment with infliximab in rheumatoid arthritis. Rheumatology (Oxford) 2011, 50:1445–1452. [DOI] [PubMed] [Google Scholar]
  • 18.Bartelds GM, Krieckaert CLM, Nurmohamed MT, van Schouwenburg PA, Lems WF, Twisk JWR, Dijkmans BAC, Aarden L, Wolbink GJ: Development of Antidrug Antibodies Against Adalimumab and Association With Disease Activity and Treatment Failure During Long-term Follow-up. JAMA 2011, 305:1460–1468. [DOI] [PubMed] [Google Scholar]
  • 19.Ogric M, Tercelj M, Praprotnik S, Tomsic M, Bozic B, Sodin-Semrl S, Cucnik S: Detection of adalimumab and anti-adalimumab antibodies in patients with rheumatoid arthritis: a comprehensive overview of methodology pitfalls and benefits. Immunol Res 2017, 65:172–185. [DOI] [PubMed] [Google Scholar]
  • 20.Hart MH, de Vrieze H, Wouters D, Wolbink G-J, Killestein J, de Groot ER, Aarden LA, Rispens T: Differential effect of drug interference in immunogenicity assays. Journal of Immunological Methods 2011, 372:196–203. [DOI] [PubMed] [Google Scholar]
  • 21.Bendtzen K: Immunogenicity of Anti-TNF-α Biotherapies: II. Clinical Relevance of Methods Used for Anti-Drug Antibody Detection. 2015, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Hoshitsuki K, Rathod S, Ramsey MJ, Zhu L, Moreland LW, Fernandez CA: Adalimumab Immunogenicity Is Negatively Correlated with Anti-Hinge Antibody Levels in Patients with Rheumatoid Arthritis. 2020, 375:488–497. This research describes a cytometric method for simultaneously detecting biologic immunogenicity and monitoring drug levels to provide clinically meaningful recommendations. The manuscript also demonstrates that patients positive for anti-hinge antibodies have lower anti-adalimumab antibody levels, indicating that anti-hinge antibodies may predict adalimumab immunogenicity.
  • 23. Chen PK, Lan JL, Chen YM, Chen HH, Chang SH, Chung CM, Rutt NH, Tan TM, Mamat RNR, Anuar ND, et al. : Anti-TROVE2 Antibody Determined by Immune-Related Array May Serve as a Predictive Marker for Adalimumab Immunogenicity and Effectiveness in RA. J Immunol Res 2021, 2021:6656121. This manuscript identified anti-TROVE2 antibody as a useful biomarker for predicting the development of anti-drug antibodies and therapeutic response in adalimumab-treated patients.
  • 24. Hernández-Breijo B, Navarro-Compán V, Plasencia-Rodríguez C, Parodis I, Gehin JE, Martínez-Feito A, Novella-Navarro M, Mezcua A, Warren DJ, Nozal P, et al. : BAFF predicts immunogenicity in older patients with rheumatoid arthritis treated with TNF inhibitors. Scientific Reports 2021, 11:11632. This research article suggest that baseline serum BAFF concentrations predict the presence of anti-drug antibodies within 6 months of TNFi therapy in older patients with RA.
  • 25.Weinblatt ME, Keystone EC, Furst DE, Moreland LW, Weisman MH, Birbara CA, Teoh LA, Fischkoff SA, Chartash EK: Adalimumab, a fully human anti–tumor necrosis factor α monoclonal antibody, for the treatment of rheumatoid arthritis in patients taking concomitant methotrexate: The ARMADA trial. 2003, 48:35–45. [DOI] [PubMed] [Google Scholar]
  • 26.Thomas SS, Borazan N, Barroso N, Duan L, Taroumian S, Kretzmann B, Bardales R, Elashoff D, Vangala S, Furst DE: Comparative Immunogenicity of TNF Inhibitors: Impact on Clinical Efficacy and Tolerability in the Management of Autoimmune Diseases. A Systematic Review and Meta-Analysis. BioDrugs 2015, 29:241–258. [DOI] [PubMed] [Google Scholar]
  • 27.Maini RN, Breedveld FC, Kalden JR, Smolen JS, Davis D, MacFarlane JD, Antoni C, Leeb B, Elliott MJ, Woody JNJA, et al. : Therapeutic efficacy of multiple intravenous infusions of anti-tumor necrosis factor α monoclonal antibody combined with low-dose weekly methotrexate in rheumatoid arthritis. 1998, 41:1552–1563. [DOI] [PubMed] [Google Scholar]
  • 28.Vogelzang EH, Pouw MF, Nurmohamed M, Kneepkens EL, Rispens T, Wolbink GJ, Krieckaert CLM: Adalimumab trough concentrations in patients with rheumatoid arthritis and psoriatic arthritis treated with concomitant disease-modifying antirheumatic drugs. 2015, 74:474–475. [DOI] [PubMed] [Google Scholar]
  • 29. Hernández-Breijo B, Brenis CM, Plasencia-Rodríguez C, Martínez-Feito A, Novella-Navarro M, Pascual-Salcedo D, Balsa A: Methotrexate Reduces the Probability of Discontinuation of TNF Inhibitors in Seropositive Patients With Rheumatoid Arthritis. A Real-World Data Analysis. 2021, 8. This retrospective study used real-world data of patients, who started their first TNFi therapy between 1999 and 2018 from the RA-PAZ cohort, to demonstrate that concomitant MTX in seropositive patients prolongs the effectiveness of TNFi therapy.
  • 30.Bendtzen K, Geborek P, Svenson M, Larsson L, Kapetanovic MC, Saxne T: Individualized monitoring of drug bioavailability and immunogenicity in rheumatoid arthritis patients treated with the tumor necrosis factor alpha inhibitor infliximab. Arthritis Rheum 2006, 54:3782–3789. [DOI] [PubMed] [Google Scholar]
  • 31. Maksimovic V, Pavlovic-Popovic Z, Vukmirovic S, Cvejic J, Mooranian A, Al-Salami H, Mikov M, Golocorbin-Kon S: Molecular mechanism of action and pharmacokinetic properties of methotrexate. Mol Biol Rep 2020, 47:4699–4708. This review provides an overview of the molecular mechanism of action and pharmacokinetic properties of methotrexate.
  • 32. Cronstein BN, Aune TM: Methotrexate and its mechanisms of action in inflammatory arthritis. Nature Reviews Rheumatology 2020, 16:145–154. This review describes the pharmacology and metabolic and cellular mechanisms by which methotrexate suppresses inflammation in RA. The manuscript also examines the mechanisms by which methotrexate induces some of its well-known toxic effects.
  • 33.Cronstein BN: Low-dose methotrexate: a mainstay in the treatment of rheumatoid arthritis. Pharmacol Rev 2005, 57:163–172. [DOI] [PubMed] [Google Scholar]
  • 34.Baggott JE, Vaughn WH, Hudson BBJBJ: Inhibition of 5-aminoimidazole-4-carboxamide ribotide transformylase, adenosine deaminase and 5′-adenylate deaminase by polyglutamates of methotrexate and oxidized folates and by 5-aminoimidazole-4-carboxamide riboside and ribotide. 1986, 236:193–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Bitoun S, Nocturne G, Ly B, Krzysiek R, Roques P, Pruvost A, Paoletti A, Pascaud J, Dönnes P, Florence K, et al. : Methotrexate and BAFF interaction prevents immunization against TNF inhibitors. 2018, 77:1463–1470. [DOI] [PubMed] [Google Scholar]
  • 36.Cronstein BN: Methotrexate BAFFles anti-drug antibodies. Nat Rev Rheumatol 2018, 14:505–506. [DOI] [PubMed] [Google Scholar]
  • 37.Michaud K, Vrijens B, Tousset E, Pedro S, Schumacher R, Dasic G, Chen C, Agarwal E, Suarez-Almazor ME: Real-World Adherence to Oral Methotrexate Measured Electronically in Patients With Established Rheumatoid Arthritis. ACR Open Rheumatol 2019, 1:560–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang W, Zhou H, Liu L: Side effects of methotrexate therapy for rheumatoid arthritis: A systematic review. Eur J Med Chem 2018, 158:502–516. [DOI] [PubMed] [Google Scholar]
  • 39. Chat VS, Uppal SK, Kearns DG, Han G, Wu JJ: Translating the 2020 AAD-NPF Guidelines of Care for the Management of Psoriasis With Systemic Nonbiologics to Clinical Practice. Cutis 2021, 107:99–103. This review addresses dosing, efficacy, toxicity, drug-related interactions, and contraindications alongside evidence-based treatment recommendations for acitretin, apremilast, cyclosporine, fumaric acid esters, methotrexate, and tofacitinib.
  • 40. Khurana A, Saxena S: Immunosuppressive agents for dermatological indications in the ongoing COVID-19 pandemic: Rationalizing use and clinical applicability. Dermatol Ther 2020, 33:e13639. The purpose of this review is to provide useful insights on the infection predisposition potential of conventional immunosuppressive agents, including cytotoxic antiproliferative agents, and briefly mention COVID-19 clinical outcomes in patients receiving immunosuppressive agents.
  • 41.Woyach JA, Johnson AJ, Byrd JC: The B-cell receptor signaling pathway as a therapeutic target in CLL. Blood 2012, 120:1175–1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Pal Singh S, Dammeijer F, Hendriks RW: Role of Bruton’s tyrosine kinase in B cells and malignancies. Mol Cancer 2018, 17:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Calissi G, Lam EW, Link W: Therapeutic strategies targeting FOXO transcription factors. Nat Rev Drug Discov 2021, 20:21–38. This review provides an overview of the role of FOXO proteins in health and disease and discuss the pharmacological approaches to modulate FOXO function.
  • 44.Wiestner A: Emerging role of kinase-targeted strategies in chronic lymphocytic leukemia. Blood 2012, 120:4684–4691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rao A, Luo C, Hogan PG: Transcription factors of the NFAT family: regulation and function. Annu Rev Immunol 1997, 15:707–747. [DOI] [PubMed] [Google Scholar]
  • 46.Lee JU, Kim LK, Choi JM: Revisiting the Concept of Targeting NFAT to Control T Cell Immunity and Autoimmune Diseases. Front Immunol 2018, 9:2747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fernandez CA, Smith C, Yang W, Mullighan CG, Qu C, Larsen E, Bowman WP, Liu C, Ramsey LB, Chang TJB, The Journal of the American Society of Hematology: Genome-wide analysis links NFATC2 with asparaginase hypersensitivity. 2015, 126:69–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Rathod S, Ramsey M, Finkelman FD, Fernandez CA: Genetic inhibition of NFATC2 attenuates asparaginase hypersensitivity in mice. Blood Advances 2020, 4:4406–4416. This is a validation study of previous clinical findings associating NFATC2 and the risk of drug-induced immunogenicity. The research shows that genetic or pharmacological inhibition of NFATC2 in mice protects from hypersensitivity reactions to antileukemic asparaginase.
  • 49.Elloumi HZ, Maharshak N, Rao KN, Kobayashi T, Ryu HS, Mühlbauer M, Li F, Jobin C, Plevy SE: A Cell Permeable Peptide Inhibitor of NFAT Inhibits Macrophage Cytokine Expression and Ameliorates Experimental Colitis. PLOS ONE 2012, 7:e34172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Marongiu L, Mingozzi F, Cigni C, Marzi R, Gioia MD, Garrè M, Parazzoli D, Sironi L, Collini M, Sakaguchi R, et al. : Inositol 1,4,5-trisphosphate 3-kinase B promotes Ca2+ mobilization and the inflammatory activity of dendritic cells. 2021, 14:eaaz2120. This study shows that pharmacological inhibition of inositol 1,4,5-trisphosphate 3-kinase B (ITPKB) in mice reduced both LPS-induced tissue swelling and the severity of inflammatory arthritis to a similar extent as that induced by the inhibition of NFAT using nanoparticles that delivered an NFAT-inhibiting peptide (VIVIT) specifically to phagocytic cells.
  • 51.Mulero MC, Aubareda A, Orzaez M, Messeguer J, Serrano-Candelas E, Martinez-Hoyer S, Messeguer A, Perez-Paya E, Perez-Riba M: Inhibiting the calcineurin-NFAT (nuclear factor of activated T cells) signaling pathway with a regulator of calcineurin-derived peptide without affecting general calcineurin phosphatase activity. J Biol Chem 2009, 284:9394–9401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tan MKX, Heng TYJ, Mak A: The Potential Use of Metformin, Dipyridamole, N-Acetylcysteine and Statins as Adjunctive Therapy for Systemic Lupus Erythematosus. Cells 2019, 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ramakers BP, Riksen NP, Stal TH, Heemskerk S, van den Broek P, Peters WH, van der Hoeven JG, Smits P, Pickkers P: Dipyridamole augments the antiinflammatory response during human endotoxemia. Crit Care 2011, 15:R289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Visser F, Vickers MF, Ng AM, Baldwin SA, Young JD, Cass CE: Mutation of residue 33 of human equilibrative nucleoside transporters 1 and 2 alters sensitivity to inhibition of transport by dilazep and dipyridamole. J Biol Chem 2002, 277:395–401. [DOI] [PubMed] [Google Scholar]
  • 55.Hasko G, Linden J, Cronstein B, Pacher P: Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov 2008, 7:759–770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yu JC, Lin G, Field JJ, Linden J: Induction of antiinflammatory purinergic signaling in activated human iNKT cells. JCI Insight 2018, 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mazzon E, Esposito E, Impellizzeri D, R DIP, Melani A, Bramanti P, Pedata F, Cuzzocrea S: CGS 21680, an agonist of the adenosine (A2A) receptor, reduces progression of murine type II collagen-induced arthritis. J Rheumatol 2011, 38:2119–2129. [DOI] [PubMed] [Google Scholar]
  • 58.Cekic C, Linden J: Purinergic regulation of the immune system. Nat Rev Immunol 2016, 16:177–192. [DOI] [PubMed] [Google Scholar]
  • 59.Romio M, Reinbeck B, Bongardt S, Huls S, Burghoff S, Schrader J: Extracellular purine metabolism and signaling of CD73-derived adenosine in murine Treg and Teff cells. Am J Physiol Cell Physiol 2011, 301:C530–539. [DOI] [PubMed] [Google Scholar]
  • 60.Huang S, Apasov S, Koshiba M, Sitkovsky M: Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood 1997, 90:1600–1610. [PubMed] [Google Scholar]
  • 61. Levack RC, Newell KL, Cabrera-Martinez B, Cox J, Perl A, Bastacky SI, Winslow GM: Adenosine receptor 2a agonists target mouse CD11c(+)T-bet(+) B cells in infection and autoimmunity. Nat Commun 2022, 13:452. This study using the A2A receptor agonist CGS-21680 and regadenoson suggests that targeting the adenosine signaling pathway may provide a method for the treatment of lupus and other autoimmune diseases mediated by T-bet+ B cells.
  • 62.Palani G, Husain Z, Salinas RC, Karthikeyan V, Karthikeyan AS, Ananthasubramaniam K: Safety of regadenoson as a pharmacologic stress agent for myocardial perfusion imaging in chronic kidney disease patients not on hemodialysis. J Nucl Cardiol 2011, 18:605–611. [DOI] [PubMed] [Google Scholar]
  • 63. Huang Z, Xie N, Illes P, Di Virgilio F, Ulrich H, Semyanov A, Verkhratsky A, Sperlagh B, Yu SG, Huang C, et al. : From purines to purinergic signalling: molecular functions and human diseases. Signal Transduct Target Ther 2021, 6:162. This review summarizes recent findings in purine biogenesis and metabolism, with a focus on the role of purinergic signaling in cellular functions and several human diseases including neurological disorders, rheumatic diseases, and cancer.
  • 64. He Y, Huang Y, Mai C, Pan H, Luo HB, Liu L, Xie Y: The immunomodulatory role of PDEs inhibitors in immune cells: therapeutic implication in rheumatoid arthritis. Pharmacol Res 2020, 161:105134. This review summarizes the modulating effects of PDEs on immune cells and describes the molecular underpinnings and potential clinical application of PDEs inhibitors for RA.
  • 65.Vang T, Torgersen KM, Sundvold V, Saxena M, Levy FO, Skalhegg BS, Hansson V, Mustelin T, Tasken K: Activation of the COOH-terminal Src kinase (Csk) by cAMP-dependent protein kinase inhibits signaling through the T cell receptor. J Exp Med 2001, 193:497–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kamel R, Leroy J, Vandecasteele G, Fischmeister R: Cyclic nucleotide phosphodiesterases as therapeutic targets in cardiac hypertrophy and heart failure. Nat Rev Cardiol 2022. The review describes cAMP and cGMP signaling in cardiomyocytes and presents the various PDE families expressed in the heart as well as their modifications in pathological cardiac hypertrophy and heart failure.
  • 67.Omori K, Kotera J: Overview of PDEs and Their Regulation. 2007, 100:309–327. [DOI] [PubMed] [Google Scholar]
  • 68.Li L, Yee C, Beavo JA: CD3- and CD28-dependent induction of PDE7 required for T cell activation. Science 1999, 283:848–851. [DOI] [PubMed] [Google Scholar]
  • 69.Coqueret O, Boichot E, Lagente V: Selective type IV phosphodiesterase inhibitors prevent IL-4-induced IgE production by human peripheral blood mononuclear cells. Clin Exp Allergy 1997, 27:816–823. [PubMed] [Google Scholar]
  • 70.Gamboa A, Abraham R, Diedrich A, Shibao C, Paranjape SY, Farley G, Biaggioni I: Role of adenosine and nitric oxide on the mechanisms of action of dipyridamole. Stroke 2005, 36:2170–2175. [DOI] [PubMed] [Google Scholar]
  • 71.Aktas B, Utz A, Hoenig-Liedl P, Walter U, Geiger J: Dipyridamole enhances NO/cGMP-mediated vasodilator-stimulated phosphoprotein phosphorylation and signaling in human platelets: in vitro and in vivo/ex vivo studies. Stroke 2003, 34:764–769. [DOI] [PubMed] [Google Scholar]
  • 72.Nunes AK, Raposo C, Luna RL, Cruz-Hofling MA, Peixoto CA: Sildenafil (Viagra(R)) down regulates cytokines and prevents demyelination in a cuprizone-induced MS mouse model. Cytokine 2012, 60:540–551. [DOI] [PubMed] [Google Scholar]
  • 73.Zhang J, Guo J, Zhao X, Chen Z, Wang G, Liu A, Wang Q, Zhou W, Xu Y, Wang C: Phosphodiesterase-5 inhibitor sildenafil prevents neuroinflammation, lowers beta-amyloid levels and improves cognitive performance in APP/PS1 transgenic mice. Behav Brain Res 2013, 250:230–237. [DOI] [PubMed] [Google Scholar]
  • 74.Pifarre P, Gutierrez-Mecinas M, Prado J, Usero L, Roura-Mir C, Giralt M, Hidalgo J, Garcia A: Phosphodiesterase 5 inhibition at disease onset prevents experimental autoimmune encephalomyelitis progression through immunoregulatory and neuroprotective actions. Exp Neurol 2014, 251:58–71. [DOI] [PubMed] [Google Scholar]
  • 75.Li J, Luo Y, Wang X, Feng G: Regulatory B cells and advances in transplantation. J Leukoc Biol 2019, 105:657–668. [DOI] [PubMed] [Google Scholar]
  • 76.Stauderman KA: CRAC channels as targets for drug discovery and development. Cell Calcium 2018, 74:147–159. [DOI] [PubMed] [Google Scholar]
  • 77. Papa P, Whitefield B, Mortensen DS, Cashion D, Huang D, Torres E, Parnes J, Sapienza J, Hansen J, Correa M, et al. : Discovery of the Selective Protein Kinase C-theta Kinase Inhibitor, CC-90005. J Med Chem 2021, 64:11886–11903. This manuscript describes the development and discovery of a selective protein kinase C-θ kinase inhibitor, CC-90005, that inhibits T cell activation.
  • 78.Altman A, Kong KF: Protein kinase C inhibitors for immune disorders. Drug Discov Today 2014, 19:1217–1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Eferl R, Wagner EF: AP-1: a double-edged sword in tumorigenesis. Nat Rev Cancer 2003, 3:859–868. [DOI] [PubMed] [Google Scholar]
  • 80.Atsaves V, Leventaki V, Rassidakis GZ, Claret FX: AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers (Basel) 2019, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Hoyer S, Eberlein V, Schuler G, Berking C, Heinzerling L, Schaft N, Dorrie J: BRAF and MEK Inhibitors Affect Dendritic-Cell Maturation and T-Cell Stimulation. Int J Mol Sci 2021, 22. This study examines the influence of BRAF/MEK inhibition, either as single agent or in combination, on the maturation of monocyte-derived dendritic cells and their interaction with T cells, including T cell activation.
  • 82.Vella LJ, Pasam A, Dimopoulos N, Andrews M, Knights A, Puaux AL, Louahed J, Chen W, Woods K, Cebon JS: MEK inhibition, alone or in combination with BRAF inhibition, affects multiple functions of isolated normal human lymphocytes and dendritic cells. Cancer Immunol Res 2014, 2:351–360. [DOI] [PubMed] [Google Scholar]
  • 83.Allegrezza MJ, Rutkowski MR, Stephen TL, Svoronos N, Perales-Puchalt A, Nguyen JM, Payne KK, Singhal S, Eruslanov EB, Tchou J, et al. : Trametinib Drives T-cell-Dependent Control of KRAS-Mutated Tumors by Inhibiting Pathological Myelopoiesis. Cancer Res 2016, 76:6253–6265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Duan W, Chan JH, Wong CH, Leung BP, Wong WS: Anti-inflammatory effects of mitogen-activated protein kinase kinase inhibitor U0126 in an asthma mouse model. J Immunol 2004, 172:7053–7059. [DOI] [PubMed] [Google Scholar]
  • 85.Welsh SJ, Corrie PG: Management of BRAF and MEK inhibitor toxicities in patients with metastatic melanoma. Ther Adv Med Oncol 2015, 7:122–136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Hu J, Jarusiewicz J, Du G, Nishiguchi G, Yoshimura S, Panetta JC, Li Z, Min J, Yang L, Chepyala D, et al. : Preclinical evaluation of proteolytic targeting of LCK as a therapeutic approach in T cell acute lymphoblastic leukemia. Sci Transl Med 2022, 14:eabo5228. This study investigates lymphocyte-specific protein tyrosine kinase (LCK) signaling as a common therapeutic vulnerability in T-ALL and describes the development of a series of LCK degraders. The research identifies a lead compound SJ11646 that exhibited marked efficiency in cereblon-mediated LCK degradation in T-ALL cells.
  • 87. Rao D, Li H, Ren X, Sun Y, Wen C, Zheng M, Huang H, Tang W, Xu S: Discovery of a potent, selective, and covalent ZAP-70 kinase inhibitor. Eur J Med Chem 2021, 219:113393. This manuscript describes the development of the first potent and covalent inhibitor of the ZAP-70 kinase.
  • 88. Abdel-Magid AF: Dual Inhibition of IL-2-Inducible T-Cell Kinase (ITK) and Tropomyosin Receptor Kinase A (TRKA) as Potential Treatment for Atopic Dermatitis and Other Inflammatory and Autoimmune Diseases. ACS Med Chem Lett 2021, 12:1889–1891. This letter describes a patent application related to inhibitors of interleukin-2-inducible T-cell kinase (ITK) that may be useful as a treatment for inflammatory and autoimmune diseases including skin diseases such as atopic dermatitis.
  • 89. Jacobs CF, Eldering E, Kater AP: Kinase inhibitors developed for treatment of hematologic malignancies: implications for immune modulation in COVID-19. Blood Adv 2021, 5:913–925. This review describes the immunomodulatory effects of the main TKIs approved or in late-stage development for the treatment of hematological malignancies, including inhibitors of Bruton’s tyrosine kinase, spleen tyrosine kinase, BCR-Abl, phosphatidylinositol 3-kinase/ mammalian target of rapamycin, JAK/STAT, and FMS-like tyrosine kinase 3, to provide a rationale for how such inhibitors could modify clinical courses of diseases, such as COVID-19.
  • 90.Esposito A, Viale G, Curigliano G: Safety, Tolerability, and Management of Toxic Effects of Phosphatidylinositol 3-Kinase Inhibitor Treatment in Patients With Cancer: A Review. JAMA Oncol 2019, 5:1347–1354. [DOI] [PubMed] [Google Scholar]
  • 91. Tam CS, Dimopoulos M, Garcia-Sanz R, Trotman J, Opat S, Roberts AW, Owen R, Song Y, Xu W, Zhu J, et al. : Pooled safety analysis of zanubrutinib monotherapy in patients with B-cell malignancies. Blood Adv 2022, 6:1296–1308. This study constructed a pooled safety analysis to better understand zanubrutinib-associated treatment-emergent adverse events and identify treatment-limiting toxicities.
  • 92.Baluom M, Grossbard EB, Mant T, Lau DT: Pharmacokinetics of fostamatinib, a spleen tyrosine kinase (SYK) inhibitor, in healthy human subjects following single and multiple oral dosing in three phase I studies. Br J Clin Pharmacol 2013, 76:78–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Weinblatt ME, Kavanaugh A, Genovese MC, Musser TK, Grossbard EB, Magilavy DB: An oral spleen tyrosine kinase (Syk) inhibitor for rheumatoid arthritis. N Engl J Med 2010, 363:1303–1312. [DOI] [PubMed] [Google Scholar]
  • 94. Arneson LC, Carroll KJ, Ruderman EM: Bruton’s Tyrosine Kinase Inhibition for the Treatment of Rheumatoid Arthritis. Immunotargets Ther 2021, 10:333–342. This review outlines the mechanisms of BTK inhibition and its potential impact on immune mediated disease, the types of BTK inhibitors being studied for RA, the findings from both preclinical and clinical trials of BTK inhibitors in RA, and directions for future research.
  • 95.Schutt SD, Fu J, Nguyen H, Bastian D, Heinrichs J, Wu Y, Liu C, McDonald DG, Pidala J, Yu XZ: Inhibition of BTK and ITK with Ibrutinib Is Effective in the Prevention of Chronic Graft-versus-Host Disease in Mice. PLoS One 2015, 10:e0137641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Treon SP, Castillo JJ, Skarbnik AP, Soumerai JD, Ghobrial IM, Guerrera ML, Meid K, Yang G: The BTK inhibitor ibrutinib may protect against pulmonary injury in COVID-19-infected patients. Blood 2020, 135:1912–1915. This study investigates the impact of ibrutinib in COVID-19 patients.
  • 97.Colado A, Almejun MB, Podaza E, Risnik D, Stanganelli C, Elias EE, Dos Santos P, Slavutsky I, Fernandez Grecco H, Cabrejo M, et al. : The kinase inhibitors R406 and GS-9973 impair T cell functions and macrophage-mediated anti-tumor activity of rituximab in chronic lymphocytic leukemia patients. Cancer Immunol Immunother 2017, 66:461–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Colonna L, Catalano G, Chew C, D’Agati V, Thomas JW, Wong FS, Schmitz J, Masuda ES, Reizis B, Tarakhovsky A, et al. : Therapeutic targeting of Syk in autoimmune diabetes. J Immunol 2010, 185:1532–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Platt AM, Benson RA, McQueenie R, Butcher JP, Braddock M, Brewer JM, McInnes IB, Garside P: The active metabolite of spleen tyrosine kinase inhibitor fostamatinib abrogates the CD4(+) T cell-priming capacity of dendritic cells. Rheumatology (Oxford) 2015, 54:169–177. [DOI] [PubMed] [Google Scholar]
  • 100.Ying H, Li Z, Yang L, Zhang J: Syk mediates BCR- and CD40-signaling integration during B cell activation. Immunobiology 2011, 216:566–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Ross SH, Cantrell DA: Signaling and Function of Interleukin-2 in T Lymphocytes. Annu Rev Immunol 2018, 36:411–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Park HB, Oh K, Garmaa N, Seo MW, Byoun OJ, Lee HY, Lee DS: CP-690550, a Janus kinase inhibitor, suppresses CD4+ T-cell-mediated acute graft-versus-host disease by inhibiting the interferon-gamma pathway. Transplantation 2010, 90:825–835. [DOI] [PubMed] [Google Scholar]
  • 103.Rizzi M, Lorenzetti R, Fischer K, Staniek J, Janowska I, Troilo A, Strohmeier V, Erlacher M, Kunze M, Bannert B, et al. : Impact of tofacitinib treatment on human B-cells in vitro and in vivo. J Autoimmun 2017, 77:55–66. [DOI] [PubMed] [Google Scholar]
  • 104.Sonomoto K, Yamaoka K, Kubo S, Hirata S, Fukuyo S, Maeshima K, Suzuki K, Saito K, Tanaka Y: Effects of tofacitinib on lymphocytes in rheumatoid arthritis: relation to efficacy and infectious adverse events. Rheumatology (Oxford) 2014, 53:914–918. [DOI] [PubMed] [Google Scholar]
  • 105. Alvaro-Gracia JM, Garcia-Llorente JF, Valderrama M, Gomez S, Montoro M: Update on the Safety Profile of Tofacitinib in Rheumatoid Arthritis from Clinical Trials to Real-World Studies: A Narrative Review. Rheumatol Ther 2021, 8:17–40. The article provides complete and up-to-date information on the safety profile of tofacitinib in RA, from clinical trials to real-world studies.

RESOURCES