Tyrosine kinase inhibitors (TKIs) are used to target dysregulated signaling pathways in virtually all hematologic malignancies. Many of the targeted signaling pathways are also essential in nonmalignant immune cells. The current coronavirus severe acute respiratory syndrome coronavirus 2 pandemic catalyzed clinical exploration of TKIs in the treatment of the various stages of COVID-19, which are characterized by distinct immune-related complications. Most of the reported effects of TKIs on immune regulation have been explored in vitro, with different class-specific drugs having nonoverlapping target affinities. Moreover, many of the reported in vivo effects are based on artificial animal models or on observations made in symptomatic patients with a hematologic malignancy who often already suffer from disturbed immune regulation. Based on in vitro and clinical observations, we attempt to decipher the impact 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.

Over the last decades, dysregulation of signaling pathways involved in proliferation, activation, and survival have been identified in virtually all hematologic malignancies. This has led to the development and application of a diverse set of tyrosine kinase inhibitors (TKIs) that have drastically improved the clinical outcomes of these diseases.1 

In addition to their role in tumorigenesis, many of the targeted signaling pathways are essential in nonmalignant immune cells. Furthermore, many TKIs have significant off-target effects.1 

Their immunomodulatory effects led to an increased interest in TKIs developed for hematologic malignancies in applications other than cancer. The current coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic catalyzed clinical exploration of TKIs in the treatment of the various stages of COVID-19.2  Because a standard treatment for COVID-19 is still lacking, drug repurposing strategies for TKIs are being considered in the management of this potentially lethal disease. Despite the vast number of publications on TKIs, an up-to-date systematic review on the presently exploited immunomodulatory effects of TKIs is lacking. We focus on the main TKIs approved or in late-stage development for the treatment of hematological malignancies: Bruton’s tyrosine kinase (BTK), spleen tyrosine kinase (SYK), BCR-Abl, phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR), JAK/STAT, and FMS-like tyrosine kinase 3 (FLT3). First, we summarize the putative roles of these kinases in innate (neutrophils, dendritic cells [DCs], monocytes, and natural killer [NK] cells) and adaptive (B cells and T cells) immune cells and then systematically discuss in vitro and in vivo evidence for their effects in these cells to provide a rationale for how such inhibitors could modify clinical courses of diseases, such as COVID-19. COVID-19 has major global impacts on health and economy, and our understanding of its underlying pathophysiological mechanisms has progressed with unprecedented speed. The pandemic has also triggered a storm of studies on treatment options and vaccine development. Amid this ongoing research, any review has the obvious limitation that current knowledge may quickly be outdated. Nevertheless, current and future research on COVID-19 and other diseases characterized by immune-related complications could benefit from an overview of the immunomodulatory effects of TKIs provided in this review.

TKIs, their indications, and the clinically relevant (off-target) kinases that they inhibit, will be discussed are summarized in Table 1. The available data on their immunomodulatory effects are summarized by TKI class in supplemental Tables 1 through 6.

Table 1.

TKIs, their indications, and the (off-target) kinases they inhibit

Class/drugsIndication (T)rial/(R)egistration/(O)ff-labelRelevant immunomodulatory off-target molecules
BTK*   
 Ibrutinib CLL/SLL(R), WM(R), MCL(R), MZL(R), BCL(R), cGvHD(R)103  ITK, EGFR, JAK3103  
 Acalabrutinib CLL/SLL(R), NHL(R), MCL(R)104   
 Zanubrutinib MZL(R), CLL/SLL(T), WM(T), MCL(T)105   
 Tirabrutinib CLL(T), BCL(T), WM(T), PCNSL(T)104   
SYK   
 Fostamatinib ITP(R), RA(T), CLL(T), BCL(T)106,107  FLT3, JAK263,108  
 Entospletinib AML(T), CLL(T), BCL(T), GvHD(T), MCL(T), NHL(T)106  JAK2106  
 Cerdulatinib DLBCL(T), CLL(T), NHL(T)106  JAK1/JAK3, AKT, NF-κB106,107  
BCR-Abl   
 Imatinib Ph+CML(R)109   
 Nilotinib Ph+CML(R)110   
 Dasatinib Ph+CML(R)58  BTK58  
 Bosutinib Ph+CML(R), Ph+ALL(R)111   
 Ponatinib Ph+CML(R), Ph+ALL(R)111  Abl, FLT3111  
 Radotinib Ph+CML(R)112   
PI3K/mTOR§   
 Idelalisib (p110δ) ALL(R), CLL/SLL(R), FL(R)113   
 Copanlisib (PI3Kδ & PI3Kδ) FL(R), CLL/SLL(R)114,115   
 Duvelisib FL(R), CLL/SLL(R)115   
 Umbralisib (PI3Kδ) MZL(T), CLL(T), FL(T), DLBCL(T)116   
 Temsirolimus NHL(R), MCL(R).117   
JAK   
 Ruxolitinib (JAK1/2) PV(R), MF(R), GvHD(R)118   
 Fedratinib (JAK2) MF(R)119  FLT3, BCR-Abl120  
 Momelotinib (JAK1/2) MPN(T)121   
 Pacritinib (JAK2) MPN(T)122  FLT3122  
FLT3||   
 Midostaurin FLT3-mutated AML(R), ASM(R), MC(R)123  SYK123  
 Sunitinib RCC(R), GI(R), FLT3-mutated AML(O)124   
 Sorafenib HCC(R), RCC(R), FLT3-mutated AML(O)125   
 Gilteritinib FLT3-mutated AML(R)125   
 Crenolanib FLT3-mutated AML(T)125   
 Quizartinib FLT3-mutated AML(T)125   
Class/drugsIndication (T)rial/(R)egistration/(O)ff-labelRelevant immunomodulatory off-target molecules
BTK*   
 Ibrutinib CLL/SLL(R), WM(R), MCL(R), MZL(R), BCL(R), cGvHD(R)103  ITK, EGFR, JAK3103  
 Acalabrutinib CLL/SLL(R), NHL(R), MCL(R)104   
 Zanubrutinib MZL(R), CLL/SLL(T), WM(T), MCL(T)105   
 Tirabrutinib CLL(T), BCL(T), WM(T), PCNSL(T)104   
SYK   
 Fostamatinib ITP(R), RA(T), CLL(T), BCL(T)106,107  FLT3, JAK263,108  
 Entospletinib AML(T), CLL(T), BCL(T), GvHD(T), MCL(T), NHL(T)106  JAK2106  
 Cerdulatinib DLBCL(T), CLL(T), NHL(T)106  JAK1/JAK3, AKT, NF-κB106,107  
BCR-Abl   
 Imatinib Ph+CML(R)109   
 Nilotinib Ph+CML(R)110   
 Dasatinib Ph+CML(R)58  BTK58  
 Bosutinib Ph+CML(R), Ph+ALL(R)111   
 Ponatinib Ph+CML(R), Ph+ALL(R)111  Abl, FLT3111  
 Radotinib Ph+CML(R)112   
PI3K/mTOR§   
 Idelalisib (p110δ) ALL(R), CLL/SLL(R), FL(R)113   
 Copanlisib (PI3Kδ & PI3Kδ) FL(R), CLL/SLL(R)114,115   
 Duvelisib FL(R), CLL/SLL(R)115   
 Umbralisib (PI3Kδ) MZL(T), CLL(T), FL(T), DLBCL(T)116   
 Temsirolimus NHL(R), MCL(R).117   
JAK   
 Ruxolitinib (JAK1/2) PV(R), MF(R), GvHD(R)118   
 Fedratinib (JAK2) MF(R)119  FLT3, BCR-Abl120  
 Momelotinib (JAK1/2) MPN(T)121   
 Pacritinib (JAK2) MPN(T)122  FLT3122  
FLT3||   
 Midostaurin FLT3-mutated AML(R), ASM(R), MC(R)123  SYK123  
 Sunitinib RCC(R), GI(R), FLT3-mutated AML(O)124   
 Sorafenib HCC(R), RCC(R), FLT3-mutated AML(O)125   
 Gilteritinib FLT3-mutated AML(R)125   
 Crenolanib FLT3-mutated AML(T)125   
 Quizartinib FLT3-mutated AML(T)125   

AKT, protein kinase B; ALL, acute lymphoid leukemia; AML, acute myeloid leukemia; ASM, aggressive systemic mastocytosis; BCL, B-cell lymphoma; cGvHD, chronic graft-versus-host disease; CLL, chronic lymphocytic leukemia; CML, chronic myeloid leukemia; DLBCL, diffuse large B-cell lymphoma; EGFR, epidermal growth factor receptor; FL, follicular non-Hodgkin lymphoma; GI, gastrointestinal tumors; GvHD, graft-versus-host disease; HCC, hepatocellular carcinoma; ITP, idiopathic thrombocytopenic purpura; ITK, interleukin-2–inducible T-cell kinase; MC, mast cell leukemia; MCL, mantle cell lymphoma; MF, myelofibrosis; MPN, myeloproliferative neoplasm; MZL, marginal zone lymphoma; NHL, non-Hodgkin lymphoma; PCSNL, primary central nervous system lymphoma; Ph+, Philadelphia chromosome positive; PV, polycythemia vera; RA, rheumatoid arthritis; RCC, renal cell carcinoma; SLL, small lymphocytic leukemia; WM, Waldenström macroglobulinemia.

*

Summarized in supplemental Table 1.

Summarized in supplemental Table 2.

Summarized in supplemental Table 3.

§

Summarized in supplemental Table 4.

Summarized in supplemental Table 5.

||

Summarized in supplemental Table 6.

The innate immune system

Cells of the innate immune system act as first responders for the detection and clearance of (viral) infections.3  They secrete proinflammatory cytokines that inhibit viral replication, stimulate the adaptive immune response, and recruit other immune cells to the site of infection.3  A schematic overview of a normal immune response during a viral infection can be found in Figure 1.

Figure 1.

Schematic overview of the normal immune after a viral infection. Phase 0 consists of epithelial infection with the virus, after which monocytes detect the pathogen and subsequent cytokine production and differentiate into cytokine-producing macrophages and other cell types. Phase 1 entails the attraction and activation of other innate immune cells, such as NK cells, which kill infected cells, and DCs, which then travel to naive T cells and lymph nodes. Phase 2 consists of the subsequent activation of T cells and B cells by DCs (and the cytokines produced) into activated and differentiated T cells and antibody-producing plasma cells, respectively. This results in phase 3, in which antibodies and T cells attack the virus and virus-infected cells, respectively. Image created with Biorender.com.

Figure 1.

Schematic overview of the normal immune after a viral infection. Phase 0 consists of epithelial infection with the virus, after which monocytes detect the pathogen and subsequent cytokine production and differentiate into cytokine-producing macrophages and other cell types. Phase 1 entails the attraction and activation of other innate immune cells, such as NK cells, which kill infected cells, and DCs, which then travel to naive T cells and lymph nodes. Phase 2 consists of the subsequent activation of T cells and B cells by DCs (and the cytokines produced) into activated and differentiated T cells and antibody-producing plasma cells, respectively. This results in phase 3, in which antibodies and T cells attack the virus and virus-infected cells, respectively. Image created with Biorender.com.

Close modal

Granulocytes and monocytes detect extracellular pathogens via Toll-like receptors (TLRs).4  Granulocytes will release enzymes and toxic proteins at the site of infection, whereas monocytes traffic to inflamed tissues and differentiate into (cytokine-producing) macrophages and monocyte-derived DCs.3  Macrophages and neutrophils phagocytose and destroy pathogens, as well as infected cells. These sequential processes within neutrophils and monocytes require complex signaling cascades that involve BTK, SYK, Abl, PI3K/mTOR, and JAK/STAT.3,5-8  BTK is involved in granulocyte maturation, and SYK signaling is required for leukocyte adhesion, pathogen recognition, and phagocytosis.8-10  Abl is critical in podosome formation and the function of macrophages and is required for neutrophil recruitment and migration.6,7  PI3K/mTOR modulate oxidative bursts in neutrophils and limit TLR signaling in macrophages.11  JAK/STAT is involved in cytokine production of macrophages, as well as in the differentiation, survival, and activation of neutrophils.8,12 

Activated DCs present pathogen-derived antigens to naive helper T (Th) cells to initiate the adaptive immune response.3  BTK mediates maturation and antigen presentation, whereas SYK is involved in the production of reactive oxygen species and recognition of tissue damage, and PI3K negatively regulates TLR signaling in DCs.5,10,11  JAK/STAT signaling is involved in their maturation, antigen presentation, and cytokine production.11,12 

NK cells kill infected cells via degranulation, receptor-mediated apoptosis, and antibody-dependent cell-mediated cytotoxicity (ADCC). Their maturation and activation are mediated by BTK and PI3K/mTOR, whereas PI3K/mTOR and JAK/STAT are involved in cytokine production.11,13,14  Their cytolytic function is supported by PI3K/mTOR and (indirectly) by FLT3.11,15 

The adaptive immune system

Responses of the adaptive immune system are based on antigen-specific receptors expressed on T- and B-cell surfaces. T-cell receptors recognize peptide fragments when presented by antigen-presenting cells, such as DCs.3  The (cytotoxic) CD8+ subset primarily kills infected cells, whereas the CD4+ subset regulates cellular and humoral responses and contains the immunosuppressive regulatory T-cell (Treg) subset.3,11  CD4+ cells differentiate into Th1, Th2, and Th17 cells. Th1 cells lead to an increased cell-mediated response, and Th2 cells lead to a humoral immune response. Th17 cells are developmentally distinct from Th1 and Th2 lineages and specifically produce proinflammatory interleukin-17 (IL-17).3  SYK and Abl are involved in early T-cell development and maturation, whereas PI3K signaling is involved in the proliferative responses of all T cells and the survival of Tregs.10,16  JAK/STAT signaling plays a key role in T-cell responses, specifically with respect to differentiation and cytokine production.12 

B cells express (and produce) different classes of immunoglobulins based on their maturation and activation status.3  After activation by binding of antigen to the B-cell receptor, B cells undergo affinity-based differentiation into plasma cells that produce specific antibodies, or into memory cells that can rapidly respond following reencounter of their specific antigen.3  BTK and PI3K/mTOR form fundamental parts of the B-cell receptor–signaling cascade in which SYK acts as upstream signal transducer. Activation of this cascade leads to key processes that are involved in proliferation, differentiation, and survival.10,17,18  JAK/STAT signaling is involved in the process of class switching.

Based on above-described roles of these kinases in innate and adaptive immunity, it may seem predictable how TKIs will affect immune responses. Be that as it may, effects of TKIs on immune regulation have primarily been explored in vitro with different class-specific drugs having nonoverlapping target affinities. Moreover, many of the reported in vivo effects are based on artificial animal models or observed in symptomatic patients with a hematologic malignancy who often already suffer from disturbed immune regulation. Furthermore, because TKIs have effects on innate and adaptive immune functions, the timing of administration within the infectious cycle and development could greatly influence the response. Effects of a specific inhibitor within a TKI class can also differ greatly, because these inhibitors display distinct drug-protein interactions and off-target kinase-inhibition profiles, also resulting in inconsistent immunomodulatory effects.19  Bearing in mind these caveats, we attempt to delineate the impact of the major TKIs on the frequency and function of each cell type of the innate and the adaptive immune response.

Innate immune cells

Monocytes/neutrophils.

The SYK and JAK inhibitors, fostamatinib and ruxolitinib, respectively, are associated with acquired neutropenia, without neutrophil dysfunctions.20,21  TKI-induced functional alterations of neutrophils and monocyte-derived cells have been widely reported. Duvelisib improves macrophage function by polarizing them to an M1 phenotype, in contrast to the immunosuppressive effects seen with other PI3K inhibitors.22  Most BCR-Abl inhibitors improve monocyte chemotaxis.23  mTOR inhibition by temsirolimus stimulates neutrophil and macrophages infiltration into lung epithelium, possibly explaining the interstitial lung disease seen in temsirolimus-treated patients.24  Impairment of activation and cytotoxicity/phagocytosis of neutrophils and monocytes occur in patients treated with ibrutinib or idelalisib.25-27  Proliferation and cytokine production is reduced by the SYK and JAK inhibitors fostamatinib, ruxolitinib and fedratinib.28-30 

In summary, granulocyte and monocyte function is impaired by inhibitors of BTK, SYK, and JAK and improved by Abl and mTOR inhibitors. PI3K inhibitors seem to have stimulating and suppressive effects in distinct cell types.

DCs.

DC maturation and activation are improved by ibrutinib in vitro.31  However, SYK and JAK inhibition with fostamatinib and ruxolitinib, respectively, decreases signaling, activation, and differentiation.32,33  Because ruxolitinib’s effect on DCs is expected to be due to on-target JAK inhibition, it is expected that all JAK inhibitors impair DC function.32  Midostaurin decreases DC differentiation and activation, and sorafenib limits their migration and T-cell activation, both seen in vitro.34,35  Because sunitinib does not have any effects on the functionality of DCs, it remains questionable whether the aforementioned effects are the actual results of FLT3 inhibition.35  Treatment with BCR-Abl inhibitors can result in suppression and improvement of DC expansion and function (including antitumor effects), depending on the timing and duration of drug exposure.36  This might be explained by the negative impact on DC maturation and differentiation (eg, lower levels of costimulatory molecules) observed with long-term imatinib cultures of progenitor cells in vitro but the increased chemotaxis seen during short-term cultures of mature DCs.37,38 

Thus, DC function is decreased by SYK, JAK, and FLT3 inhibition and increased by the BTK inhibitor ibrutinib, whereas the effects of BCR-Abl inhibitors depend on the timing and duration of drug exposure.

NK cells.

Alterations in the frequency of NK cells occur during treatment with BCR-Abl and JAK inhibitors. An increased NK cell/lymphocyte ratio is observed in chronic myeloid leukemia (CML) patients treated with dasatinib, imatinib, and nilotinib.39  Ruxolitinib treatment leads to an absolute decrease in NK cells in patients with myeloproliferative neoplasms, likely caused by an increased ratio of immature/mature subsets.40,41  At the functional level, sorafenib increases degranulation and interferon-γ (IFN-γ) secretion, possibly leading to more antitumor NK cell activity.42  However, granule exocytosis and the cytotoxic function of NK cells are reduced in patients treated with ibrutinib, as well as in vitro by idelalisib.43-45  Data on the suppression of NK cell activity by the more selective BTK inhibitors are conflicting, with less pronounced effects with acalabrutinib and zanubrutinib but equal suppression of ADCC with tirabrutinib and ibrutinib.45  Whether BTK inhibitors potentiate or suppress anti-CD20–induced ADCC is still a matter of debate, as reviewed recently.46  Because many of the necessary cytokines in NK cell development and maturation exert their impact through JAK/STAT signaling, blocking the effects of IL-2 and IL-15 by ruxolitinib, fedratinib, and momelotinib prevents (in different degrees) activation and therefore IFN-γ and tumor necrosis factor-α (TNF-α) secretion of NK cells in vitro, hampering their killing capacity.40,41,45  NK cell–related effects of BCR-Abl inhibitors are not uniform. Although nilotinib suppresses NK cell reactivity in CML patients, dasatinib treatment results in enhanced reactivity through an unknown mechanism.23 

In summary, NK cell cytotoxicity is impaired by BTK inhibitors, to the greatest extent by ibrutinib. PI3K and JAK/STAT inhibition lower the killing capacity and cytokine production, whereas BCR-Abl inhibition has variable effects. On the other hand, FLT3 inhibition increases NK cell function.

Adaptive immune cells

T cells.

T cells (subsets) are frequently altered during TKI treatment. Ibrutinib increases total CD4+ and CD8+ T cells in chronic lymphocytic leukemia (CLL) patients without lymphocytosis; however, in patients with significant tumor burden, ibrutinib decreases total T-cell numbers, which could be viewed as pseudonormalization.47  Ibrutinib and (the more selective) zanubrutinib increase the frequency of Th17 cells and reduce the number of Tregs in CLL patients, possibly resulting in a more immune-supportive phenotype.47,48  Treg frequency is also reduced by idelalisib, which correlates with autoimmune-related toxicities, as reviewed by Greenwell et al.49  Tregs are probably less affected by umbralisib, because it leads to less severe T-cell–mediated toxicity than seen with idelalisib.50  Imatinib, ruxolitinib, and certain FLT3 inhibitors also reduce Treg numbers.35,39,51-53 

Functional T-cell alterations occur with most TKIs, although they vary in effect and degree. Ibrutinib reverses the polarization of CD4+ Th2 cells to a Th1 cell phenotype through off-target ITK inhibition.54  Ibrutinib, as well as the more selective BTK inhibitors, reduces the (over)activated T-cell state of CLL patients, as measured by lower plasma cytokine levels and decreased exhaustion.47,55,56  These observations are probably the indirect result of on-target (BTK) effects on leukemia cells, resulting in decreased suppressive interactions of the malignant B cells with T cells.57  Sorafenib and sunitinib treatment also results in lower PD-1 expression on CD4+ and CD8+ T cells in vivo and in vitro, respectively.53,58 

Low-dose sorafenib also increases activation, whereas high-dose sorafenib decreases T-cell proliferation and augments PD-1 expression.59  Furthermore, low-dose sorafenib increases tumor-infiltrating T cells compared with high-dose sorafenib treatment of hepatocellular carcinoma patients in vivo, consistent with the dose-dependent immunomodulatory effects seen in vitro.58,59  The increased T-effector activation observed during idelalisib and duvelisib treatment is caused, at least in part, by Treg inhibition.49  Furthermore, mTOR inhibition by temsirolimus increases differentiation and enhances the function of Tregs.60  Sunitinib and sorafenib improve the Th1-type response in vivo, most likely through off-target effects.58,61,62 

On the other hand, fostamatinib, entospletinib, and cerdulatinib reduce activation, proliferation, and cytokine production of T cells in vitro.63  All JAK inhibitors lower T-cell proliferation and cytokine secretion, resulting in less proinflammatory signals.64  Th1 cytokine production is inhibited by midostaurin in vitro.65 

BCR-Abl inhibitors again produce various effects; although nilotinib reduces T-cell activation in vivo, dasatinib treatment leads to enhanced proliferation of T cells in leukemia patients, which correlates with improved disease responses.39,58  The effects of imatinib on T cells are conflicting, as reviewed in detail.38,66 

Altogether, BTK inhibitors lead to changes in absolute T-cell numbers (depending on the clinical status of the patient) and skewing of specific subsets within the T-cell population. They also lead to reduced T-cell exhaustion. BTK, BCR-Abl, PI3K, JAK, and FLT3 inhibitors lower Treg numbers. SYK and PI3K inhibition reduces T-cell function, whereas mTOR inhibition promotes it. Effects of BCR-Abl inhibition are inconsistent between available drugs. JAK/STAT inhibition results in diminished T-cell function over multiple aspects. The effect of FLT3 inhibitors is dose dependent, with low doses leading to improved T-cell functions.

B cells.

Because many TKIs are targeted toward hematological malignancies occurring in B cells, they also have various effects on healthy B cells. These are significantly less sensitive to ibrutinib-, momelotinib-, or idelalisib-induced apoptosis than their malignant counterparts, resulting in stable B-cell numbers.45,55  However, B-cell numbers and immunoglobulin levels decrease in CML patients treated with any of the BCR-Abl inhibitors, without changes in B-cell subsets.67  Although fostamatinib clearly reduces the number of class-switched germinal center B cells, contradictory results have been reported with regard to fostamatinib’s effect on the total B-cell number.28,33 

In terms of functional changes, decreased immunoglobulin G (IgG) levels are observed in CLL patients after treatment with ibrutinib.55,68  Likewise, IgM levels are decreased by ibrutinib in the graft-versus-host disease setting.69  However, humoral immunity seems to be positively affected by ibrutinib. Increased IgA has been observed across clinical trials of ibrutinib, and patients with greater improvements in IgA developed fewer infections.70  Similarly, although IgM and IgG remain unaffected, acalabrutinib treatment also augments IgA levels.70  CLL patients on ibrutinib also respond better to influenza vaccination than do untreated patients, but titers are diminished compared with healthy individuals.71  Effects on B cells in CML patients differ among BCR-Abl inhibitors. Although imatinib reduces all immunoglobulin levels, dasatinib only lowers IgM, bosutinib only lowers IgG, and nilotinib does not incite any immunoglobulin decrease.36,67,72 

Fostamatinib and cerdulatinib lower B-cell activation and BCR-mediated antigen presentation in vitro and attenuate autoantibody production by B cells in vivo, most likely through on-target SYK inhibition.33,73  The effect of FLT3 inhibitors on B cells is modest, with only midostaurin showing an in vitro negative effect on the viability of B cells.74  Although proliferation and IgG production in healthy B cells are decreased by idelalisib in vitro, this is not seen upon idelalisib treatment in vivo.45 

In summary, antibody changes are seen after BTK, BCR-Abl, and, possibly, PI3K inhibition. SYK inhibitors lower B-cell (auto)activity.

The high morbidity and mortality of the current COVID-19 pandemic and the lack of effective treatment has driven research toward application of TKIs outside of the malignant spectrum. Immune-related complications by SARS-CoV-2 infection and other inflammatory diseases led to the consideration of these inhibitors as treatment options. The initial “innate” phase of SARS-CoV-2 infection is set in motion following entry in nasal and bronchial epithelial cells through the viral structural spike protein binding to the angiotensin-converting enzyme 2 receptor.75  Like most viral infections, tissue-resident macrophages in close proximity to infected cells will incite a proinflammatory state, resulting in recruitment of innate immune cells and cytokine production, leading to subsequent activation of adaptive immune responses.75  However, in the case of (severe) COVID-19, massive invasion of innate immune cells into the infected tissues leads to hyperproduction of such cytokines, eventually resulting in a so-called “cytokine storm.”76  As a consequence, recruited T cells reach a state of exhaustion, resulting in T-cell dysfunction and depletion that cause lymphopenia.76  These detrimental inflammatory effects can potentially result in acute respiratory distress syndrome (ARDS), secondary infections, multiple organ failure, and death.76  This chain of events might be interrupted by blocking specific kinases at different phases of the disease. Based on the data and considerations summarized above, we attempt to hypothetically assess the immunomodulatory potential of the TKIs in the pathophysiological process of COVID-19. Ongoing trials with these TKIs are summarized in Table 2. A schematic summary of the hypothetical application and timing of TKIs in COVID-19 is shown in Figure 2.

Table 2.

Ongoing trials with TKIs in treatment of COVID-19

Class/agentGoalComments
BTK   
 Ibrutinib Efficacy in reducing respiratory failure Quadruple-blind RCT, NCT04375397 
 Efficacy in reducing respiratory failure and preventing death Double-blind RCT, NCT04439006 
 Acalabrutinib Efficacy in preventing deterioration, mortality, and respiratory failure or speeding up recovery of hospitalized patients with COVID-19 Multiarm RCT, 2020-001736-95 (ACCORD 2) 
 Zanubrutinib Efficacy in increasing proportion of respiratory failure-free patients Double-blind RCT, NCT04382586 
SYK   
 Fostamatinib Efficacy in reducing mortality, SAEs, inflammatory biomarkers, and hospital admission RCT, 2020-001750-22 
 Efficacy in preventing progression to severe/critical disease Double-blind RCT, NCT04629703 
BCR-Abl   
 Imatinib Efficacy in preventing vascular leak Single blind-RCT, 2020-001236-10 
 Efficacy of baricitinib, imatinib, or lopinavir in combination with hydroxychloroquine RCT, 2020-001321-31 
 Efficacy in preventing need for invasive mechanical ventilation RCT, NCT04422678 
 Safety and efficacy in hospitalized adults Double-blind RCT, NCT04394416 
 Time to clinical improvement with treatment with hydroxychloroquine in combination with baricitinib, imatinib, or lopinavir/ritonavir RCT, 2020-001321-31 
 Time to clinical improvement RCT, NCT04346147 
 Efficacy in preventing severe COVID-19 in hospitalized aged patients RCT, NCT04357613 
PI3K/mTOR   
 Duvelisib Overall survival, length of hospital/ICU stay Triple-blind RCT, NCT04372602 
 Efficacy in requirement for mechanical ventilation or dying Double-blind RCT, NCT04487886 
 Temsirolimus Efficacy in preventing respiratory failure Double-blind RCT, NCT04482712 
 Time to clinical recovery and viral clearance Single-blind RCT, NCT04461340 
 Proportion of patients alive and free from advanced respiratory support measures Quadruple-blind RCT, NCT04341675 
JAK   
 Ruxolitinib Efficacy in reducing mortality, SAEs, inflammatory biomarkers, and hospital admission RCT, 2020-001750-22 
 Incidence of SAEs and severe disease, safety Single arm, NCT04348071 
 Efficacy in preventing clinical worsening of respiratory status Single arm, NCT04414098 
 Treatment of cytokine storm Open label, NCT04355793 
 Efficacy in preventing death and influencing SAEs/ICU-free days Double-blind RCT, NCT04377620 
 Recovery from pneumonia Open label, NCT04334044 
 Efficacy in reducing respiratory failure and ICU care and preventing death Double-blind RCT, NCT04362137, NCT04337359 
 Reduction of cytokine load by single plasma volume exchanges, with or without ruxolitinib Open label, NCT04374149 
 Efficacy in reversing hyperinflammation (CIS) Open label, NCT04338958 
 Efficacy in preventing death, ICU admission, or mechanical ventilation Triple-blind RCT, NCT04477993 
 Efficacy in preventing SAEs and COVID-19 patients from becoming critically ill Single arm, NCT04331665 
 Overall survival Open label, NCT04359290 
 Efficacy in preventing severe respiratory failure and reducing ICU/hospital stay and survival, in combination with simvastatin Open label, NCT04348695 
 Ability to prevent all-cause mortality, noninvasive or invasive ventilation Double-blind RCT, NCT04581954 
 Ability to achieve a change from baseline in the clinical assessment score COVID-19 with colchicine, ruxolitinib, secukinumab, or standard treatment Open label, NCT04403243 
 Safety and efficacy of ruxolitinib combined with mesenchymal stem cells Single-blind RCT, ChiCTR2000029580 
 Efficacy and safety in patients on respiratory support or approaching need for respiratory support Open label, 2020-001459-42 
 Efficacy of anakinra/tocilizumab, alone or in combination with ruxolitinib, in increasing ventilation-free days Open-label RCT, 2020-001754-21, NCT04424056 
 Clinical and biological efficacy of anakinra, alone or in combination with ruxolitinib, in reducing oxygen dependency, need for invasive ventilation, and systemic inflammation Open-label RCT, 2020-001963-10 
 Efficacy in preventing deterioration requiring mechanical ventilation Open label, 2020-001777-71 
 Overall survival after 28 d Single arm, 2020-001732-10 
 Pacritinib Efficacy in preventing mechanical ventilation or ECMO Double-blind RCT, NCT04404361 
Class/agentGoalComments
BTK   
 Ibrutinib Efficacy in reducing respiratory failure Quadruple-blind RCT, NCT04375397 
 Efficacy in reducing respiratory failure and preventing death Double-blind RCT, NCT04439006 
 Acalabrutinib Efficacy in preventing deterioration, mortality, and respiratory failure or speeding up recovery of hospitalized patients with COVID-19 Multiarm RCT, 2020-001736-95 (ACCORD 2) 
 Zanubrutinib Efficacy in increasing proportion of respiratory failure-free patients Double-blind RCT, NCT04382586 
SYK   
 Fostamatinib Efficacy in reducing mortality, SAEs, inflammatory biomarkers, and hospital admission RCT, 2020-001750-22 
 Efficacy in preventing progression to severe/critical disease Double-blind RCT, NCT04629703 
BCR-Abl   
 Imatinib Efficacy in preventing vascular leak Single blind-RCT, 2020-001236-10 
 Efficacy of baricitinib, imatinib, or lopinavir in combination with hydroxychloroquine RCT, 2020-001321-31 
 Efficacy in preventing need for invasive mechanical ventilation RCT, NCT04422678 
 Safety and efficacy in hospitalized adults Double-blind RCT, NCT04394416 
 Time to clinical improvement with treatment with hydroxychloroquine in combination with baricitinib, imatinib, or lopinavir/ritonavir RCT, 2020-001321-31 
 Time to clinical improvement RCT, NCT04346147 
 Efficacy in preventing severe COVID-19 in hospitalized aged patients RCT, NCT04357613 
PI3K/mTOR   
 Duvelisib Overall survival, length of hospital/ICU stay Triple-blind RCT, NCT04372602 
 Efficacy in requirement for mechanical ventilation or dying Double-blind RCT, NCT04487886 
 Temsirolimus Efficacy in preventing respiratory failure Double-blind RCT, NCT04482712 
 Time to clinical recovery and viral clearance Single-blind RCT, NCT04461340 
 Proportion of patients alive and free from advanced respiratory support measures Quadruple-blind RCT, NCT04341675 
JAK   
 Ruxolitinib Efficacy in reducing mortality, SAEs, inflammatory biomarkers, and hospital admission RCT, 2020-001750-22 
 Incidence of SAEs and severe disease, safety Single arm, NCT04348071 
 Efficacy in preventing clinical worsening of respiratory status Single arm, NCT04414098 
 Treatment of cytokine storm Open label, NCT04355793 
 Efficacy in preventing death and influencing SAEs/ICU-free days Double-blind RCT, NCT04377620 
 Recovery from pneumonia Open label, NCT04334044 
 Efficacy in reducing respiratory failure and ICU care and preventing death Double-blind RCT, NCT04362137, NCT04337359 
 Reduction of cytokine load by single plasma volume exchanges, with or without ruxolitinib Open label, NCT04374149 
 Efficacy in reversing hyperinflammation (CIS) Open label, NCT04338958 
 Efficacy in preventing death, ICU admission, or mechanical ventilation Triple-blind RCT, NCT04477993 
 Efficacy in preventing SAEs and COVID-19 patients from becoming critically ill Single arm, NCT04331665 
 Overall survival Open label, NCT04359290 
 Efficacy in preventing severe respiratory failure and reducing ICU/hospital stay and survival, in combination with simvastatin Open label, NCT04348695 
 Ability to prevent all-cause mortality, noninvasive or invasive ventilation Double-blind RCT, NCT04581954 
 Ability to achieve a change from baseline in the clinical assessment score COVID-19 with colchicine, ruxolitinib, secukinumab, or standard treatment Open label, NCT04403243 
 Safety and efficacy of ruxolitinib combined with mesenchymal stem cells Single-blind RCT, ChiCTR2000029580 
 Efficacy and safety in patients on respiratory support or approaching need for respiratory support Open label, 2020-001459-42 
 Efficacy of anakinra/tocilizumab, alone or in combination with ruxolitinib, in increasing ventilation-free days Open-label RCT, 2020-001754-21, NCT04424056 
 Clinical and biological efficacy of anakinra, alone or in combination with ruxolitinib, in reducing oxygen dependency, need for invasive ventilation, and systemic inflammation Open-label RCT, 2020-001963-10 
 Efficacy in preventing deterioration requiring mechanical ventilation Open label, 2020-001777-71 
 Overall survival after 28 d Single arm, 2020-001732-10 
 Pacritinib Efficacy in preventing mechanical ventilation or ECMO Double-blind RCT, NCT04404361 

CIS, chronic inflammation score; ECMO, extracorporeal membrane oxygenation; ICU, intensive care unit; RCT, randomized controlled trial; SAE, serious adverse event.

Figure 2.

Hypothetical application of TKIs during the SARS-COV-2–induced immunopathology. Phase 0 consists of infection with the virus; phase 1 entails the polarization of macrophages to an M1 phenotype (and their subsequent activation) and their production of cytokines to attract other immune cells. Phase 2 is the recruitment and activation of cytokine-producing innate immune cells and T cells and B cells, which (together with the cytokine storm that results from phase 1) results in phase 3: depletion and exhaustion of lymphocytes and, subsequently, secondary bacterial infections. In phase 4, acute respiratory distress syndrome, secondary infections and multiorgan failure arise, leading to respiratory insufficiency and life-threatening situations. The applicable TKIs are indicated below the phases during which their immunomodulatory effects would be the most beneficial. Image created with Biorender.com.

Figure 2.

Hypothetical application of TKIs during the SARS-COV-2–induced immunopathology. Phase 0 consists of infection with the virus; phase 1 entails the polarization of macrophages to an M1 phenotype (and their subsequent activation) and their production of cytokines to attract other immune cells. Phase 2 is the recruitment and activation of cytokine-producing innate immune cells and T cells and B cells, which (together with the cytokine storm that results from phase 1) results in phase 3: depletion and exhaustion of lymphocytes and, subsequently, secondary bacterial infections. In phase 4, acute respiratory distress syndrome, secondary infections and multiorgan failure arise, leading to respiratory insufficiency and life-threatening situations. The applicable TKIs are indicated below the phases during which their immunomodulatory effects would be the most beneficial. Image created with Biorender.com.

Close modal

BTK inhibitors

Ibrutinib reduces macrophage numbers, activation, infiltration, and degranulation.77,78  Moreover, ibrutinib treatment decreases serum levels of inflammatory cytokines associated with cytokine release syndrome, such as IL-1β, IL-6, IL-10, and TNF-α.79  In addition, ibrutinib polarizes the T-cell population toward effector cells and reduces their exhaustion marker expression, which possibly enables the adaptive immune system to a more capable immune response.56,80  B-cell activation and a subsequent increase in IgG levels, which are both inhibited by ibrutinib, are specifically increased in patients with severe COVID-19.45,68,76  Because of these immunomodulatory effects, ibrutinib, and perhaps the more selective BTK inhibitors as well, might be beneficial in the early and late stages of COVID-19.81  However, zanubrutinib also lowers Tregs in CLL patients and could potentially ameliorate immune responses.48  First results of a clinical trial with acalabrutinib in COVID-19 (the CALAVI trial) showed decreased IL-6 levels, reflecting the inhibition of monocytes and attenuation of their cytokine production.82  However, it was recently reported that the trial did not meet its primary end point of increasing the patient proportion remaining alive and free of respiratory failure. It remains to be seen whether specific subgroups of patients benefit from acalabrutinib treatment.83 

SYK inhibitors

The SYK inhibitors affect some of the essential players in the cytokine storm seen in severe COVID-19 patients: IL-6, IL-10, and TNF-α.28,63,84  Entospletinib also reduces pulmonary inflammation by macrophages and increases Treg numbers, which might mitigate the lung damage seen in the later stages of severe COVID-19.63,85,86  In a high-content screen for mucin-1–reducing compounds, a biochemical marker predicting ARDS, fostamatinib reduced mucin-1 in vitro and in vivo.87  However, all SYK inhibitors reduce T- and B-cell frequency and activation, potentially impairing an adequate adaptive immune response.28,33,63,73,88  Therefore, SYK inhibitors are expected to be of therapeutic benefit, primarily in the initial phase of the disease.

BCR-Abl inhibitors

Prior to injection of viral RNA into the host cytoplasm for replication, all enveloped viruses, including coronaviruses, must fuse with cellular membranes or with endosomes, which is mediated by Abl proteins.89  Indeed, in the early phases of infection, imatinib prevents viral entry via Abl-mediated cytoskeletal rearrangement, whereas in later infection phases, imatinib and dasatinib inhibit SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV) replication through blocking of the Abl2 protein.89  In addition to its effects on viral replication, BCR-Abl inhibitor’s immunomodulatory effects include attenuation of proinflammatory cytokine release (among them IL-1β and TNF-α) and reduction of macrophage development and M1 polarization, which means that their application could result in a decrease in, or perhaps even prevention of, severe cases of COVID-19.23,39,90,91  Nevertheless, BCR-Abl inhibitors also cause a decrease in B-cell numbers, antibody production, and Treg viability.39,67,72  Therefore, application of BCR-Abl inhibitors could be tested at early stages of the disease, but not at later stages, to preserve long-term memory and adequate antibody formation.39,67,72  The wide variation in the immunomodulatory effects within this class of TKIs must always be considered carefully when choosing a specific inhibitor.

PI3K/mTOR inhibitors

PI3K/mTOR signaling was reported to be overactivated in MERS-CoV–infected cell lines, and treatment with the nonselective PI3K inhibitor wortmannin and the mTOR inhibitor rapamycin led to inhibited MERS-CoV infection.92  The PI3K inhibitors described in this review decrease multiple inflammatory cytokines, including IL-1, TNF-α, IL-10, and IFN-γ.43,93  However, idelalisib reduces cytokine production in macrophages, whereas duvelisib polarizes them to an M1 phenotype, leading to a more proinflammatory state.22,50,55  Idelalisib and umbralisib reduce exhaustion marker expression on CD8+ T cells, which may assist in preventing lymphocyte dysfunction and depletion.93  Therefore, PI3K inhibitors could be useful in innate response modulation by inhibiting viral replication, improving early macrophage activation, and ameliorating the T-cell dysfunction seen in later stages. However, the last should be approached cautiously because hyperinflammatory side effects (due to a decrease in Tregs) are relatively common.94  Because mTOR inhibitors are known to cause interstitial lung disease, possibly caused by increased monocyte infiltration in lung epithelium, their application in this ARDS-prone disease seems counterintuitive.24  However, multiple trials of rapamycin are underway to assess the possibility that it might decrease cytokine release and T-cell dysfunction (Table 2).

JAK inhibitors

The key role of JAK-STAT–mediated cytokines, such as IL-6, IL-10, and TNF-α, in the severity of COVID-19 suggests a role for JAK-STAT inhibitors in ameliorating disease severity.95  However, blocking the JAK-STAT pathway strongly inhibits IFN signaling and decreases T-cell proliferation and response.45,96  Because IFN signaling is instrumental in initiating the innate immune response and in preventing viral replication, JAK-STAT inhibitors should be avoided during the very early stages of the disease.97  Because preventing the (IFN-triggered) cytokine storm would be highly desirable, attenuating JAK-STAT signaling could very well improve disease outcome when such symptoms are occurring. Indeed, preliminary data from COVID-19 patients treated with the JAK inhibitor baricitinib in the early phases of their disease demonstrated improvements in inflammatory symptoms and in pulmonary function tests.95  Ruxolitinib treatment was also proven to reduce severe systemic hyperinflammation in a pilot study, with more extensive phase 2 testing underway.98  Because JAK inhibitors affect T-cell skewing and function, inhibition of an adequate T-cell response to secondary infections might be expected upon prolonged treatment.45  The JAK inhibitors baricitinib, ruxolitinib, and tofacitinib are being tested in clinical trials (Table 2).

FLT3 inhibitors

Although FLT3 signaling seems to have a less distinctive role in cytokine production compared with the other pathways mentioned, possible helpful immunomodulatory effects could be expected upon administration of FLT3 inhibitors to COVID-19 patients. Midostaurin treatment leads to macrophage apoptosis and lowers IL-6, IL-10, and TNF-α.52,65,99  Quizartinib also lowers IL-6 and TNF-α, suggesting that these TKIs might be useful in attenuating the cytokine storm.100  However, midostaurin also decreases the proliferation and activation of T and B cells. On the other hand, sorafenib and sunitinib increase IFN-γ levels, lower PD-1 and CTLA4 expression on T cells, and enhance T-cell responses (primarily Th1 cells) in general, which could hypothetically strengthen the primary immune response against the virus.42,53,58,62 

Because most kinases play a role in >1 cell type, and because most inhibitors have promiscuous effects, the use of these drugs will affect immune responses at multiple levels. However, depending on the timing, method, and state of cellular development in which cells are exposed to TKIs, specific skewing could be feasible. Use of kinase inhibitors to mediate specific immune alterations is becoming more widespread, as seen in the treatment of graft-versus-host disease, rheumatoid arthritis, and other immune-mediated diseases, although the specific way in which these TKIs perturb the pathophysiology is often not completely understood.

The recent widespread adoption of dexamethasone treatment in patients with severe COVID-19, based on the RECOVERY trial, will change the need for some of the immunomodulatory schemes described here.101  Specific subgroups of COVID-19 patients could very well benefit from immunosuppression through the downregulation of proinflammatory cytokine production achieved by low doses of corticosteroids.102  Because corticosteroids could also reduce the initial immune responses needed to induce pathogen recognition and control viral replication, the timing of treatment seems to be of paramount importance.101,102  Thus, well-timed corticosteroid treatment could potentially be combined with, or followed by, regimens with some of the TKIs mentioned in this review. It could also limit the need for TKIs in the later (phase 3 and 4) stages of immunopathological developments, in COVID-19 and other inflammatory diseases.

Expectations are high for the application of TKIs in the treatment of various diseases that display imbalances in immune functions. Investigating their effects in clinical trials necessitates careful timing of treatment initiation and treatment duration, as well as close immune monitoring using biomarkers of the innate and adaptive immune response. The severity of the current pandemic will no doubt accelerate clinical testing, which can provide the requisite insight, hopefully in the nearby future.

Send data sharing request via email to the corresponding author, Arnon P. Kater, a.p.kater@amsterdamumc.nl.

Contribution: C.F.J., E.E., and A.P.K. selected and collected data and wrote the manuscript.

Conflict-of-interest disclosure: A.P.K. receives research funding from Janssen, AbbVie, Roche/Genentech, Astra Zeneca, and Celgene and is a member of advisory boards for Janssen, AbbVie, Roche/Genentech, and Juno. E.E. receives research funding from Janssen, AbbVie, Roche/Genentech, Astra Zeneca, and Celgene. C.F.J. declares no competing financial interests.

Correspondence: Arnon P. Kater, Department of Hematology, D3-221.1, Amsterdam University Medical Center, Location AMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; e-mail: a.p.kater@amsterdamumc.nl.

1.
Jiao
Q
,
Bi
L
,
Ren
Y
,
Song
S
,
Wang
Q
,
Wang
YS
.
Advances in studies of tyrosine kinase inhibitors and their acquired resistance
.
Mol Cancer
.
2018
;
17
(
1
):
36
.
2.
Guidelines
N-C-T
. Kinase inhibitors: Bruton’s tyrosine kinase inhibitors and Janus kinase inhibitors. Available at: https://www.covid19treatmentguidelines.nih.gov/immune-based-therapy/immunomodulators/kinase-inhibitors. Accessed 17 July 2020.
3.
Chaplin
DD
.
Overview of the immune response
.
J Allergy Clin Immunol
.
2010
;
125
(
suppl 2
):
S3
-
S23
.
4.
Kawai
T
,
Akira
S
.
The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors
.
Nat Immunol
.
2010
;
11
(
5
):
373
-
384
.
5.
Weber
ANR
,
Bittner
Z
,
Liu
X
,
Dang
TM
,
Radsak
MP
,
Brunner
C
.
Bruton’s tyrosine kinase: an emerging key player in innate immunity
.
Front Immunol
.
2017
;
8
:
1454
.
6.
Baruzzi
A
,
Berton
G
.
The tyrosine kinase Abl is a component of macrophage podosomes and is required for podosome formation and function
.
Eur J Immunol
.
2012
;
42
(
10
):
2720
-
2726
.
7.
Tong
H
,
Zhao
B
,
Shi
H
, et al
.
c-Abl tyrosine kinase plays a critical role in β2 integrin-dependent neutrophil migration by regulating Vav1 activity
.
J Leukoc Biol
.
2013
;
93
(
4
):
611
-
622
.
8.
Futosi
K
,
Fodor
S
,
Mócsai
A
.
Neutrophil cell surface receptors and their intracellular signal transduction pathways
.
Int Immunopharmacol
.
2013
;
17
(
3
):
638
-
650
.
9.
Fiedler
K
,
Sindrilaru
A
,
Terszowski
G
, et al
.
Neutrophil development and function critically depend on Bruton tyrosine kinase in a mouse model of X-linked agammaglobulinemia
.
Blood
.
2011
;
117
(
4
):
1329
-
1339
.
10.
Mócsai
A
,
Ruland
J
,
Tybulewicz
VL
.
The SYK tyrosine kinase: a crucial player in diverse biological functions
.
Nat Rev Immunol
.
2010
;
10
(
6
):
387
-
402
.
11.
Weichhart
T
,
Säemann
MD
.
The PI3K/Akt/mTOR pathway in innate immune cells: emerging therapeutic applications
.
Ann Rheum Dis
.
2008
;
67
(
suppl 3
):
iii70
-
iii74
.
12.
Tundwal
K
,
Alam
R
.
JAK and Src tyrosine kinase signaling in asthma
.
Front Biosci
.
2012
;
17
(
7
):
2107
-
2121
.
13.
Bao
Y
,
Zheng
J
,
Han
C
, et al
.
Tyrosine kinase Btk is required for NK cell activation [published correction appears in J Biol Chem. 2020;295(10):3389]
.
J Biol Chem
.
2012
;
287
(
28
):
23769
-
23778
.
14.
Gotthardt
D
,
Trifinopoulos
J
,
Sexl
V
,
Putz
EM
.
JAK/STAT cytokine signaling at the crossroad of NK cell development and maturation
.
Front Immunol
.
2019
;
10
:
2590
.
15.
Tsapogas
P
,
Mooney
CJ
,
Brown
G
,
Rolink
A
.
The cytokine Flt3-ligand in normal and malignant hematopoiesis
.
Int J Mol Sci
.
2017
;
18
(
6
):
1115
.
16.
Silberman
I
,
Sionov
RV
,
Zuckerman
V
, et al
.
T cell survival and function requires the c-Abl tyrosine kinase
.
Cell Cycle
.
2008
;
7
(
24
):
3847
-
3857
.
17.
Puri
KD
,
Gold
MR
.
Selective inhibitors of phosphoinositide 3-kinase delta: modulators of B-cell function with potential for treating autoimmune inflammatory diseases and B-cell malignancies
.
Front Immunol
.
2012
;
3
:
256
.
18.
Herman
SEM
,
Gordon
AL
,
Hertlein
E
, et al
.
Bruton tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765
.
Blood
.
2011
;
117
(
23
):
6287
-
6296
.
19.
Rix
U
,
Hantschel
O
,
Dürnberger
G
, et al
.
Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets
.
Blood
.
2007
;
110
(
12
):
4055
-
4063
.
20.
Genovese
MC
,
Kavanaugh
A
,
Weinblatt
ME
, et al
.
An oral Syk kinase inhibitor in the treatment of rheumatoid arthritis: a three-month randomized, placebo-controlled, phase II study in patients with active rheumatoid arthritis that did not respond to biologic agents
.
Arthritis Rheum
.
2011
;
63
(
2
):
337
-
345
.
21.
Shi
JG
,
Chen
X
,
McGee
RF
, et al
.
The pharmacokinetics, pharmacodynamics, and safety of orally dosed INCB018424 phosphate in healthy volunteers
.
J Clin Pharmacol
.
2011
;
51
(
12
):
1644
-
1654
.
22.
Horwitz
SM
,
Koch
R
,
Porcu
P
, et al
.
Activity of the PI3K-δ,γ inhibitor duvelisib in a phase 1 trial and preclinical models of T-cell lymphoma
.
Blood
.
2018
;
131
(
8
):
888
-
898
.
23.
Hayashi
Y
,
Nakamae
H
,
Katayama
T
, et al
.
Different immunoprofiles in patients with chronic myeloid leukemia treated with imatinib, nilotinib or dasatinib
.
Leuk Lymphoma
.
2012
;
53
(
6
):
1084
-
1089
.
24.
Washino
S
,
Ando
H
,
Ushijima
K
, et al
.
Temsirolimus induces surfactant lipid accumulation and lung inflammation in mice
.
Am J Physiol Lung Cell Mol Physiol
.
2014
;
306
(
12
):
L1117
-
L1128
.
25.
Prezzo
A
,
Cavaliere
FM
,
Bilotta
C
, et al
.
Ibrutinib-based therapy impaired neutrophils microbicidal activity in patients with chronic lymphocytic leukemia during the early phases of treatment
.
Leuk Res
.
2019
;
87
:
106233
.
26.
Papin
A
,
Tessoulin
B
,
Bellanger
C
, et al
.
CSF1R and BTK inhibitions as novel strategies to disrupt the dialog between mantle cell lymphoma and macrophages
.
Leukemia
.
2019
;
33
(
10
):
2442
-
2453
.
27.
Alflen
A
,
Stadler
N
,
Aranda Lopez
P
, et al
.
Idelalisib impairs TREM-1 mediated neutrophil inflammatory responses
.
Sci Rep
.
2018
;
8
(
1
):
5558
.
28.
Le Huu
D
,
Kimura
H
,
Date
M
, et al
.
Blockade of Syk ameliorates the development of murine sclerodermatous chronic graft-versus-host disease
.
J Dermatol Sci
.
2014
;
74
(
3
):
214
-
221
.
29.
Pattison
MJ
,
Mackenzie
KF
,
Arthur
JS
.
Inhibition of JAKs in macrophages increases lipopolysaccharide-induced cytokine production by blocking IL-10-mediated feedback
.
J Immunol
.
2012
;
189
(
6
):
2784
-
2792
.
30.
Pardanani
A
,
Tefferi
A
,
Jamieson
C
, et al
.
A phase 2 randomized dose-ranging study of the JAK2-selective inhibitor fedratinib (SAR302503) in patients with myelofibrosis
.
Blood Cancer J
.
2015
;
5
(
8
):
e335
.
31.
Natarajan
G
,
Oghumu
S
,
Terrazas
C
,
Varikuti
S
,
Byrd
JC
,
Satoskar
AR
.
A Tec kinase BTK inhibitor ibrutinib promotes maturation and activation of dendritic cells
.
OncoImmunology
.
2016
;
5
(
6
):
e1151592
.
32.
Heine
A
,
Held
SA
,
Daecke
SN
, et al
.
The JAK-inhibitor ruxolitinib impairs dendritic cell function in vitro and in vivo
.
Blood
.
2013
;
122
(
7
):
1192
-
1202
.
33.
Colonna
L
,
Catalano
G
,
Chew
C
, et al
.
Therapeutic targeting of Syk in autoimmune diabetes
.
J Immunol
.
2010
;
185
(
3
):
1532
-
1543
.
34.
Huang
YC
,
Shieh
HR
,
Chen
YJ
.
Midostaurin (PKC412) modulates differentiation and maturation of human myeloid dendritic cells
.
Toxicol In Vitro
.
2010
;
24
(
6
):
1705
-
1710
.
35.
Hipp
MM
,
Hilf
N
,
Walter
S
, et al
.
Sorafenib, but not sunitinib, affects function of dendritic cells and induction of primary immune responses
.
Blood
.
2008
;
111
(
12
):
5610
-
5620
.
36.
Larmonier
N
,
Janikashvili
N
,
LaCasse
CJ
, et al
.
Imatinib mesylate inhibits CD4+ CD25+ regulatory T cell activity and enhances active immunotherapy against BCR-ABL- tumors
.
J Immunol
.
2008
;
181
(
10
):
6955
-
6963
.
37.
Appel
S
,
Boehmler
AM
,
Grünebach
F
, et al
.
Imatinib mesylate affects the development and function of dendritic cells generated from CD34+ peripheral blood progenitor cells
.
Blood
.
2004
;
103
(
2
):
538
-
544
.
38.
Wolf
D
,
Tilg
H
,
Rumpold
H
,
Gastl
G
,
Wolf
AM
.
The kinase inhibitor imatinib–an immunosuppressive drug?
Curr Cancer Drug Targets
.
2007
;
7
(
3
):
251
-
258
.
39.
Marinelli Busilacchi
E
,
Costantini
A
,
Viola
N
, et al
.
Immunomodulatory effects of tyrosine kinase inhibitor in vitro and in vivo study
.
Biol Blood Marrow Transplant
.
2018
;
24
(
2
):
267
-
275
.
40.
Schönberg
K
,
Rudolph
J
,
Vonnahme
M
, et al
.
JAK inhibition impairs NK cell function in myeloproliferative neoplasms
.
Cancer Res
.
2015
;
75
(
11
):
2187
-
2199
.
41.
Curran
SA
,
Shyer
JA
,
St Angelo
ET
, et al
.
Human dendritic cells mitigate NK-cell dysfunction mediated by nonselective JAK1/2 blockade
.
Cancer Immunol Res
.
2017
;
5
(
1
):
52
-
60
.
42.
Sprinzl
MF
,
Reisinger
F
,
Puschnik
A
, et al
.
Sorafenib perpetuates cellular anticancer effector functions by modulating the crosstalk between macrophages and natural killer cells
.
Hepatology
.
2013
;
57
(
6
):
2358
-
2368
.
43.
Herman
SE
,
Gordon
AL
,
Wagner
AJ
, et al
.
Phosphatidylinositol 3-kinase-δ inhibitor CAL-101 shows promising preclinical activity in chronic lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular survival signals
.
Blood
.
2010
;
116
(
12
):
2078
-
2088
.
44.
Da Roit
F
,
Engelberts
PJ
,
Taylor
RP
, et al
.
Ibrutinib interferes with the cell-mediated anti-tumor activities of therapeutic CD20 antibodies: implications for combination therapy
.
Haematologica
.
2015
;
100
(
1
):
77
-
86
.
45.
Hofland
T
,
de Weerdt
I
,
Ter Burg
H
, et al
.
Dissection of the effects of JAK and BTK inhibitors on the functionality of healthy and malignant lymphocytes
.
J Immunol
.
2019
;
203
(
8
):
2100
-
2109
.
46.
Pavlasova
G
,
Mraz
M
.
The regulation and function of CD20: an “enigma” of B-cell biology and targeted therapy
.
Haematologica
.
2020
;
105
(
6
):
1494
-
1506
.
47.
Long
M
,
Beckwith
K
,
Do
P
, et al
.
Ibrutinib treatment improves T cell number and function in CLL patients
.
J Clin Invest
.
2017
;
127
(
8
):
3052
-
3064
.
48.
Zou
YX
,
Zhu
HY
,
Li
XT
, et al
.
The impacts of zanubrutinib on immune cells in patients with chronic lymphocytic leukemia/small lymphocytic lymphoma
.
Hematol Oncol
.
2019
;
37
(
4
):
392
-
400
.
49.
Greenwell
IB
,
Ip
A
,
Cohen
JB
.
PI3K inhibitors: understanding toxicity mechanisms and management
.
Oncology (Williston Park)
.
2017
;
31
(
11
):
821
-
828
.
50.
Maharaj
K
,
Powers
JJ
,
Achille
A
, et al
.
The dual PI3Kδ/CK1ε inhibitor umbralisib exhibits unique immunomodulatory effects on CLL T cells
.
Blood Adv
.
2020
;
4
(
13
):
3072
-
3084
.
51.
Lussana
F
,
Cattaneo
M
,
Rambaldi
A
,
Squizzato
A.
.
Ruxolitinib-associated infections: a systematic review and meta-analysis
.
Am J Hematol
.
2018
;
93
(
3
):
339
-
347
.
52.
Gutierrez
L
,
Jang
M
,
Zhang
T
,
Akhtari
M
,
Alachkar
H
.
Midostaurin reduces regulatory T cell markers in acute myeloid leukemia
.
Sci Rep
.
2018
;
8
(
1
):
17544
.
53.
Kalathil
SG
,
Lugade
AA
,
Miller
A
,
Iyer
R
,
Thanavala
Y
.
PD-1+ and Foxp3+ T cell reduction correlates with survival of HCC patients after sorafenib therapy
.
JCI Insight
.
2016
;
1
(
11
):
e86182
.
54.
Dubovsky
JA
,
Beckwith
KA
,
Natarajan
G
, et al
.
Ibrutinib is an irreversible molecular inhibitor of ITK driving a Th1-selective pressure in T lymphocytes
.
Blood
.
2013
;
122
(
15
):
2539
-
2549
.
55.
Pleyer
C
,
Wiestner
A
,
Sun
C
.
Immunological changes with kinase inhibitor therapy for chronic lymphocytic leukemia
.
Leuk Lymphoma
.
2018
;
59
(
12
):
2792
-
2800
.
56.
Mhibik
M
,
Wiestner
A
,
Sun
C
.
Harnessing the effects of BTKi on T cells for effective immunotherapy against CLL
.
Int J Mol Sci
.
2019
;
21
(
1
):
68
.
57.
Hofland
T
,
Eldering
E
,
Kater
AP
,
Tonino
SH
.
Engaging cytotoxic T and NK cells for immunotherapy in chronic lymphocytic leukemia
.
Int J Mol Sci
.
2019
;
20
(
17
):
4315
.
58.
Heine
A
,
Held
SA
,
Bringmann
A
,
Holderried
TA
,
Brossart
P
.
Immunomodulatory effects of anti-angiogenic drugs
.
Leukemia
.
2011
;
25
(
6
):
899
-
905
.
59.
Iyer
RV
,
Maguire
O
,
Kim
M
, et al
.
Dose-dependent sorafenib-induced immunosuppression is associated with aberrant NFAT activation and expression of PD-1 in T cells
.
Cancers (Basel)
.
2019
;
11
(
5
):
681
.
60.
Beziaud
L
,
Mansi
L
,
Ravel
P
, et al
.
Rapalogs efficacy relies on the modulation of antitumor T-cell immunity
.
Cancer Res
.
2016
;
76
(
14
):
4100
-
4112
.
61.
Wolleschak
D
,
Mack
TS
,
Perner
F
, et al
.
Clinically relevant doses of FLT3-kinase inhibitors quizartinib and midostaurin do not impair T-cell reactivity and function
.
Haematologica
.
2014
;
99
(
6
):
e90
-
e93
.
62.
Finke
JH
,
Rini
B
,
Ireland
J
, et al
.
Sunitinib reverses type-1 immune suppression and decreases T-regulatory cells in renal cell carcinoma patients
.
Clin Cancer Res
.
2008
;
14
(
20
):
6674
-
6682
.
63.
Colado
A
,
Almejún
MB
,
Podaza
E
, 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
(
4
):
461
-
473
.
64.
McLornan
DP
,
Khan
AA
,
Harrison
CN
.
Immunological consequences of JAK inhibition: friend or foe?
Curr Hematol Malig Rep
.
2015
;
10
(
4
):
370
-
379
.
65.
Si
MS
,
Reitz
BA
,
Borie
DC
.
Effects of the kinase inhibitor CGP41251 (PKC 412) on lymphocyte activation and TNF-alpha production
.
Int Immunopharmacol
.
2005
;
5
(
7-8
):
1141
-
1149
.
66.
Appel
S
,
Balabanov
S
,
Brümmendorf
TH
,
Brossart
P
.
Effects of imatinib on normal hematopoiesis and immune activation
.
Stem Cells
.
2005
;
23
(
8
):
1082
-
1088
.
67.
Rajala
HLM
,
Missiry
ME
,
Ruusila
A
, et al
.
Tyrosine kinase inhibitor therapy-induced changes in humoral immunity in patients with chronic myeloid leukemia
.
J Cancer Res Clin Oncol
.
2017
;
143
(
8
):
1543
-
1554
.
68.
Burger
JA
,
Sivina
M
,
Jain
N
, et al
.
Randomized trial of ibrutinib vs ibrutinib plus rituximab in patients with chronic lymphocytic leukemia
.
Blood
.
2019
;
133
(
10
):
1011
-
1019
.
69.
Jaglowski
SM
,
Blazar
BR
.
How ibrutinib, a B-cell malignancy drug, became an FDA-approved second-line therapy for steroid-resistant chronic GVHD
.
Blood Adv
.
2018
;
2
(
15
):
2012
-
2019
.
70.
Sun
C
,
Tian
X
,
Lee
YS
, et al
.
Partial reconstitution of humoral immunity and fewer infections in patients with chronic lymphocytic leukemia treated with ibrutinib [published correction appears in Blood. 2016;128(7):1020]
.
Blood
.
2015
;
126
(
19
):
2213
-
2219
.
71.
Sun
C
,
Gao
J
,
Couzens
L
, et al
.
Seasonal influenza vaccination in patients with chronic lymphocytic leukemia treated with ibrutinib
.
JAMA Oncol
.
2016
;
2
(
12
):
1656
-
1657
.
72.
Steegmann
JL
,
Moreno
G
,
Aláez
C
, et al
.
Chronic myeloid leukemia patients resistant to or intolerant of interferon alpha and subsequently treated with imatinib show reduced immunoglobulin levels and hypogammaglobulinemia
.
Haematologica
.
2003
;
88
(
7
):
762
-
768
.
73.
Coffey
G
,
Betz
A
,
DeGuzman
F
, et al
.
The novel kinase inhibitor PRT062070 (cerdulatinib) demonstrates efficacy in models of autoimmunity and B-cell cancer
.
J Pharmacol Exp Ther
.
2014
;
351
(
3
):
538
-
548
.
74.
Majumder
MM
,
Leppä
AM
,
Hellesøy
M
, et al
.
Multi-parametric single cell evaluation defines distinct drug responses in healthy hematologic cells that are retained in corresponding malignant cell types
.
Haematologica
.
2020
;
105
(
6
):
1527
-
1538
.
75.
Cunha
LL
,
Perazzio
SF
,
Azzi
J
,
Cravedi
P
,
Riella
LV
.
Remodeling of the immune response with aging: immunosenescence and its potential impact on COVID-19 immune response
.
Front Immunol
.
2020
;
11
:
1748
.
76.
Yang
L
,
Liu
S
,
Liu
J
, et al
.
COVID-19: immunopathogenesis and Immunotherapeutics
.
Signal Transduct Target Ther
.
2020
;
5
(
1
):
128
.
77.
Chang
BY
,
Huang
MM
,
Francesco
M
, et al
.
The Bruton tyrosine kinase inhibitor PCI-32765 ameliorates autoimmune arthritis by inhibition of multiple effector cells
.
Arthritis Res Ther
.
2011
;
13
(
4
):
R115
.
78.
Rip
J
,
Van Der Ploeg
EK
,
Hendriks
RW
,
Corneth
OBJ
.
The role of Bruton’s tyrosine kinase in immune cell signaling and systemic autoimmunity
.
Crit Rev Immunol
.
2018
;
38
(
1
):
17
-
62
.
79.
Treon
SP
,
Castillo
JJ
,
Skarbnik
AP
, et al
.
The BTK inhibitor ibrutinib may protect against pulmonary injury in COVID-19-infected patients
.
Blood
.
2020
;
135
(
21
):
1912
-
1915
.
80.
Molina-Cerrillo
J
,
Alonso-Gordoa
T
,
Gajate
P
,
Grande
E
.
Bruton’s tyrosine kinase (BTK) as a promising target in solid tumors
.
Cancer Treat Rev
.
2017
;
58
:
41
-
50
.
81.
McGee
MC
,
August
A
,
Huang
W
.
BTK/ITK dual inhibitors: modulating immunopathology and lymphopenia for COVID-19 therapy [published online ahead of print 8 July 2020]
.
J Leukoc Biol
. .
82.
Roschewski
M
,
Lionakis
MS
,
Sharman
JP
, et al
.
Inhibition of Bruton tyrosine kinase in patients with severe COVID-19
.
Sci Immunol
.
2020
;
5
(
48
):
eabd0110
.
83.
AstraZeneca
.
Update on CALAVI Phase II trials for Calquence in patients hospitalised with respiratory symptoms of COVID-19
. Available at: https://www.astrazeneca.com/media-centre/press-releases/2020/update-on-calavi-phase-ii-trials-for-calquence-in-patients-hospitalised-with-respiratory-symptoms-of-covid-19.html. Accessed 9 December 2020.
84.
McAdoo
SP
,
Reynolds
J
,
Bhangal
G
, et al
.
Spleen tyrosine kinase inhibition attenuates autoantibody production and reverses experimental autoimmune GN
.
J Am Soc Nephrol
.
2014
;
25
(
10
):
2291
-
2302
.
85.
Yoshimoto
T
,
Hayashi
T
,
Kondo
T
,
Kittaka
M
,
Reichenberger
EJ
,
Ueki
Y
.
Second-generation SYK inhibitor entospletinib ameliorates fully established inflammation and bone destruction in the cherubism mouse model
.
J Bone Miner Res
.
2018
;
33
(
8
):
1513
-
1519
.
86.
Poe
JC
,
Jia
W
,
Di Paolo
JA
, et al
.
SYK inhibitor entospletinib prevents ocular and skin GVHD in mice
.
JCI Insight
.
2018
;
3
(
19
):
e122430
.
87.
Alimova
M
,
Sidhom
EH
,
Satyam
A
, et al
.
A high content screen for mucin-1-reducing compounds identifies fostamatinib as a candidate for rapid repurposing for acute lung injury during the COVID-19 pandemic
.
bioRxiv
. Preprint 20 Jun
2020
.
doi:10.1101/2020.06.30.180380
88.
Zhu
Y
,
Herlaar
E
,
Masuda
ES
, et al
.
Immunotoxicity assessment for the novel spleen tyrosine kinase inhibitor R406
.
Toxicol Appl Pharmacol
.
2007
;
221
(
3
):
268
-
277
.
89.
Sisk
JM
,
Frieman
MB
,
Machamer
CE
.
Coronavirus S protein-induced fusion is blocked prior to hemifusion by Abl kinase inhibitors
.
J Gen Virol
.
2018
;
99
(
5
):
619
-
630
.
90.
Dewar
AL
,
Domaschenz
RM
,
Doherty
KV
,
Hughes
TP
,
Lyons
AB
.
Imatinib inhibits the in vitro development of the monocyte/macrophage lineage from normal human bone marrow progenitors
.
Leukemia
.
2003
;
17
(
9
):
1713
-
1721
.
91.
Baruzzi
A
,
Iacobucci
I
,
Soverini
S
,
Lowell
CA
,
Martinelli
G
,
Berton
G
.
c-Abl and Src-family kinases cross-talk in regulation of myeloid cell migration
.
FEBS Lett
.
2010
;
584
(
1
):
15
-
21
.
92.
Kindrachuk
J
,
Ork
B
,
Hart
BJ
, et al
.
Antiviral potential of ERK/MAPK and PI3K/AKT/mTOR signaling modulation for Middle East respiratory syndrome coronavirus infection as identified by temporal kinome analysis
.
Antimicrob Agents Chemother
.
2015
;
59
(
2
):
1088
-
1099
.
93.
Dwyer
CJ
,
Arhontoulis
DC
,
Rangel Rivera
GO
, et al
.
Ex vivo blockade of PI3K gamma or delta signaling enhances the antitumor potency of adoptively transferred CD8+ T cells
.
Eur J Immunol
.
2020
;
50
(
9
):
1386
-
1399
.
94.
Lampson
BL
,
Kasar
SN
,
Matos
TR
, et al
.
Idelalisib given front-line for treatment of chronic lymphocytic leukemia causes frequent immune-mediated hepatotoxicity
.
Blood
.
2016
;
128
(
2
):
195
-
203
.
95.
Spinelli
FR
,
Conti
F
,
Gadina
M
.
HiJAKing SARS-CoV-2? The potential role of JAK inhibitors in the management of COVID-19
.
Sci Immunol
.
2020
;
5
(
47
):
96.
Massalska
M
,
Maslinski
W
,
Ciechomska
M.
.
Small molecule inhibitors in the treatment of rheumatoid arthritis and beyond: latest updates and potential strategy for fighting COVID-19
.
Cells
.
2020
;
9
(
8
):
1876
.
97.
Walz
L
,
Cohen
AJ
,
Rebaza
AP
, et al
.
Janus kinase-inhibitor and type I interferon ability to produce favorable clinical outcomes in COVID-19 patients: a systematic review and meta-analysis
.
medRxiv
. Preprint 20 Aug
2020
.
98.
La Rosée
F
,
Bremer
HC
,
Gehrke
I
, et al
.
The Janus kinase 1/2 inhibitor ruxolitinib in COVID-19 with severe systemic hyperinflammation
.
Leukemia
.
2020
;
34
(
7
):
1805
-
1815
.
99.
Miyatake
K
,
Inoue
H
,
Hashimoto
K
, et al
.
PKC412 (CGP41251) modulates the proliferation and lipopolysaccharide-induced inflammatory responses of RAW 264.7 macrophages
.
Biochem Biophys Res Commun
.
2007
;
360
(
1
):
115
-
121
.
100.
Wang
R
,
Chen
T
,
Wang
C
, et al
.
Flt3 inhibition alleviates chronic kidney disease by suppressing CD103+ dendritic cell-mediated T cell activation
.
Nephrol Dial Transplant
.
2019
;
34
(
11
):
1853
-
1863
.
101.
Horby
P
,
Lim
WS
,
Emberson
JR
, et al;
RECOVERY Collaborative Group
.
Dexamethasone in hospitalized patients with Covid-19—preliminary report [published online ahead of print 17 July 2020]
.
N Engl J Med
. .
102.
Rizk
JG
,
Kalantar-Zadeh
K
,
Mehra
MR
,
Lavie
CJ
,
Rizk
Y
,
Forthal
DN
.
Pharmaco-immunomodulatory therapy in COVID-19
.
Drugs
.
2020
;
80
(
13
):
1267
-
1292
.
103.
Pal Singh
S
,
Dammeijer
F
,
Hendriks
RW
.
Role of Bruton’s tyrosine kinase in B cells and malignancies [published correction appears in Mol Cancer. 2019;18(1):79]
.
Mol Cancer
.
2018
;
17
(
1
):
57
.
104.
Puła
B
,
Gołos
A
,
Górniak
P
,
Jamroziak
K
.
Overcoming ibrutinib resistance in chronic lymphocytic leukemia
.
Cancers (Basel)
.
2019
;
11
(
12
):
1834
.
105.
Syed
YY
.
Zanubrutinib: first approval
.
Drugs
.
2020
;
80
(
1
):
91
-
97
.
106.
Liu
D
,
Mamorska-Dyga
A
.
Syk inhibitors in clinical development for hematological malignancies
.
J Hematol Oncol
.
2017
;
10
(
1
):
145
.
107.
Bussel
JB
,
Arnold
DM
,
Boxer
MA
, et al
.
Long-term fostamatinib treatment of adults with immune thrombocytopenia during the phase 3 clinical trial program
.
Am J Hematol
.
2019
;
94
(
5
):
546
-
553
.
108.
Rolf
MG
,
Curwen
JO
,
Veldman-Jones
M
, et al
.
In vitro pharmacological profiling of R406 identifies molecular targets underlying the clinical effects of fostamatinib
.
Pharmacol Res Perspect
.
2015
;
3
(
5
):
e00175
.
109.
Salesse
S
,
Verfaillie
CM
.
BCR/ABL: from molecular mechanisms of leukemia induction to treatment of chronic myelogenous leukemia
.
Oncogene
.
2002
;
21
(
56
):
8547
-
8559
.
110.
Sacha
T
,
Saglio
G
.
Nilotinib in the treatment of chronic myeloid leukemia
.
Future Oncol
.
2019
;
15
(
9
):
953
-
965
.
111.
Tan
FH
,
Putoczki
TL
,
Stylli
SS
,
Luwor
RB
.
Ponatinib: a novel multi-tyrosine kinase inhibitor against human malignancies
.
OncoTargets Ther
.
2019
;
12
:
635
-
645
.
112.
Kwak
JY
,
Kim
SH
,
Oh
SJ
, et al
.
Phase III clinical trial (RERISE study) results of efficacy and safety of radotinib compared with imatinib in newly diagnosed chronic phase chronic myeloid leukemia
.
Clin Cancer Res
.
2017
;
23
(
23
):
7180
-
7188
.
113.
Mayer
IA
,
Arteaga
CL
.
The PI3K/AKT pathway as a target for cancer treatment
.
Annu Rev Med
.
2016
;
67
(
1
):
11
-
28
.
114.
Sapon-Cousineau
V
,
Sapon-Cousineau
S
,
Assouline
S
.
PI3K inhibitors and their role as novel agents for targeted therapy in lymphoma
.
Curr Treat Options Oncol
.
2020
;
21
(
6
):
51
.
115.
Patel
K
,
Danilov
AV
,
Pagel
JM
.
Duvelisib for CLL/SLL and follicular non-Hodgkin lymphoma
.
Blood
.
2019
;
134
(
19
):
1573
-
1577
.
116.
Burris
HA
,
Flinn
IW
,
Patel
MR
, et al
.
Umbralisib, a novel PI3Kδ and casein kinase-1ε inhibitor, in relapsed or refractory chronic lymphocytic leukaemia and lymphoma: an open-label, phase 1, dose-escalation, first-in-human study
.
Lancet Oncol
.
2018
;
19
(
4
):
486
-
496
.
117.
Porta
C
,
Paglino
C
,
Mosca
A
.
Targeting PI3K/Akt/mTOR signaling in cancer
.
Front Oncol
.
2014
;
4
:
64
.
118.
Zeiser
R
,
von Bubnoff
N
,
Butler
J
, et al;
REACH2 Trial Group
.
Ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease
.
N Engl J Med
.
2020
;
382
(
19
):
1800
-
1810
.
119.
Harrison
CN
,
Schaap
N
,
Mesa
RA
.
Management of myelofibrosis after ruxolitinib failure
.
Ann Hematol
.
2020
;
99
(
6
):
1177
-
1191
.
120.
Ragheb
M
,
Harrison
CN
,
McLornan
DP
.
Current and future role of fedratinib in the treatment of myelofibrosis
.
Future Oncol
.
2020
;
16
(
6
):
175
-
186
.
121.
Pardanani
A
,
Lasho
T
,
Smith
G
,
Burns
CJ
,
Fantino
E
,
Tefferi
A
.
CYT387, a selective JAK1/JAK2 inhibitor: in vitro assessment of kinase selectivity and preclinical studies using cell lines and primary cells from polycythemia vera patients
.
Leukemia
.
2009
;
23
(
8
):
1441
-
1445
.
122.
Jain
T
,
Mesa
R
.
The development, safety and efficacy of pacritinib for the treatment of myelofibrosis
.
Expert Rev Anticancer Ther
.
2016
;
16
(
11
):
1101
-
1108
.
123.
Daver
N
,
Schlenk
RF
,
Russell
NH
,
Levis
MJ
.
Targeting FLT3 mutations in AML: review of current knowledge and evidence
.
Leukemia
.
2019
;
33
(
2
):
299
-
312
.
124.
Larrosa-Garcia
M
,
Baer
MR
.
FLT3 inhibitors in acute myeloid leukemia: current status and future directions
.
Mol Cancer Ther
.
2017
;
16
(
6
):
991
-1001.
125.
Roskoski
R.
The role of small molecule Flt3 receptor protein-tyrosine kinase inhibitors in the treatment of Flt3-positive acute myelogenous leukemias
.
Pharmacol Res
.
2020
;
155
:
104725
.

Author notes

The full-text version of this article contains a data supplement.

Supplemental data