Skip to main content
Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America logoLink to Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America
. 2017 Sep 27;66(1):140–148. doi: 10.1093/cid/cix687

Call for Action: Invasive Fungal Infections Associated With Ibrutinib and Other Small Molecule Kinase Inhibitors Targeting Immune Signaling Pathways

Georgios Chamilos 1,2, Michail S Lionakis 3, Dimitrios P Kontoyiannis 4,
PMCID: PMC5850040  PMID: 29029010

An increasing number of invasive fungal infections (IFIs) has been recently reported in patients with lymphoid malignancies treated with BTK inhibitors. The surge in development of small molecule kinase inhibitors requires increased awareness and novel preventive strategies for IFIs..

Keywords: ibrutinib, small molecule tyrosine kinase inhibitors, invasive fungal infections, opportunistic infections

Abstract

Opportunistic infections caused by Pneumocystis jirovecii, Cryptococcus neoformans, and ubiquitous airborne filamentous fungi have been recently reported in patients with hematological cancers historically considered at low risk for invasive fungal infections (IFIs), after receipt of the Bruton tyrosine kinase inhibitor ibrutinib. The spectrum and severity of IFIs often observed in these patients implies the presence of a complex immunodeficiency that may not be solely attributed to mere inhibition of Bruton tyrosine kinase. In view of the surge in development of small molecule kinase inhibitors for treatment of malignant and autoimmune diseases, it is possible that there would be an emergence of IFIs associated with the effects of these molecules on the immune system. Preclinical assessment of the immunosuppressive effects of kinase inhibitors and human studies aimed at improving patient risk stratification for development of IFIs could lead to prevention, earlier diagnosis, and better outcomes in affected patients.

SMALL MOLECULE KINASE INHIBITORS: A BREAKTHROUGH IN THE TREATMENT OF HEMATOLOGICAL CANCERS

To overcome the bottleneck of toxicity and the lack of a specific mechanistic target with conventional chemotherapeutic agents [1–3], major emphasis has been placed on the development of precision “design” compounds that inhibit the signaling pathways implicated in the pathobiology of malignant and autoimmune diseases [4, 5]. The sophisticated mechanism-based design of these compounds has led to unprecedented treatment success in a range of hematological cancers [4, 5].

The most successful story has been the accelerated approval of ibrutinib by the US Food and Drug Administration in 2013. Ibrutinib, an irreversible inhibitor of Bruton tyrosine kinase (BTK) [4, 6, 7], is considered a “game-changing drug” for the treatment of B-cell cancers, including mantle cell lymphoma, chronic lymphocytic leukemia, Waldenström macroglobulinemia, diffuse large B-cell lymphoma, and primary central nervous system (CNS) lymphoma, with durable responses reported even in patients with refractory disease [4, 6–12].

EMERGENCE OF IBRUTINIB-RELATED IFIS

Since the introduction of ibrutinib, there have been increasing reports of opportunistic fungal infections caused by Pneumocystis jirovecii, Cryptococcus neoformans, and ubiquitous airborne filamentous fungi (Aspergillus, Fusarium, and Mucorales); these often present with atypical manifestations, including CNS aspergillosis, extrapulmonary P. jirovecii, and disseminated cryptococcosis, and are associated with substantially increased mortality rates (Table 1) [6, 7, 9–25]. Although there is little information regarding the incidence of invasive fungal infections (IFIs) in patients with chronic lymphoproliferative disorders, such infections are considered uncommon in patients who have not undergone hematopoietic stem cell transplantation [26]. In contrast, in some studies of ibrutinib-based treatment for chronic lymphocytic leukemia [15] and primary CNS lymphoma [12], P. jirovecii pneumonia (PJP) and invasive aspergillosis (IA) occurred in 5% and 39% of patients, respectively. However, the frequency of IFIs in all other studies on patients with hematological cancer treated with ibrutinib was relatively low, ranging from 0% to 11% (Table 2).

Table 1.

Reported Cases of IFI During Ibrutinib Therapy (n = 41)a

Pathogen or Type of IFI Time After Treatment Site of infection Type and Status of Underlying Cancer Age, y/Sex Concomitant Use of Corticosteroids/ Prior Treatment Outcome Reference
Cryptococcosis NA NA MCL/PD NA NA/NA NA Wang et al [7]
Cryptococcus neoformans 1 mo Lungs, blood CLL/PR 68/M No/chlorambucil and prednisone Alive Okamoto et al [22]
C. neoformans 1 mo CNS, lungs Lymphoplasmacytic lymphoma/PD 88/M No/rituximab, benadmustine Alive Messina et al [19]
C. neoformans 3 wk CNS, lungs, blood CLL/PD 54/M No/fludarabine, ritiximab, cyclophosphamide Dead Messina et al [19]
C. neoformans 2 mo CNS, skin WM/PD 74/F No/fludarabine, ritiximab, cyclophosphamide, dexamethasone, and idelasilib Dead Baron et al [18]
PJP NA NA MCL/PD NA NA/yes (NA) NA Wang et al [7]
PJP 1.9 mo Lungs CLL/SD 69/M No/no (frontline) Alive Ahn et al [15]
PJP 23.6 mo Lungs CLL/SD 68/M Yesb/no (frontline) Alive Ahn et al [15]
PJP 1.9 mo Lungs CLL/SD 72/M No/no (frontline) Alive Ahn et al [15]
PJP 6 mo Lungs CLL/PD 78/M No/yes (NA) Alive Ahn et al [15]
PJPb 11.6 mo Lungs CLL/SD 70/M No/no (frontline) Alive Ahn et al [15]
Histoplasmosis NA NA MCL/PD NA NA/NA NA Wang et al [7]
IAc 1 month CNS CLL/PD NA Yes/NA Dead Ruchlemer et al [13]
IA 2 mo CNS CLL/PD NA Yes/NA Alive Ruchlemer et al [13]
IA 2 mo CNS CLL/PD NA Yes/NA Alive Ruchlemer et al [13]
IA 6 wk Lungs CLL/PD 62/M No/fludarabine and rituximab Alive Arthus et al [14]
IA 2 wk CNS, lungs PCNSL 76/F Yes/no (frontline) Dead Lionakis et al [12]
IA 2 wk CNS, lungs PCNSL 65/M Yes/no (frontline) Dead Lionakis et al [12]
IA 3 mo CNS, lungs PCNSL 87/F No/no (frontline) Dead Lionakis et al [12]
IAd 4 mo Lungs PCNSL 60/M Yes/no (frontline) Alive Lionakis et al [12]
IAd 2 mo Lungs PCNSL 53/M No/no (frontline) Alive Lionakis et al [12]
IAd [1 mo Lungs PCNSL 64/M Yes/no (frontline) Alive Lionakis et al [12]
IA 2 wk CNS, lungs PCNSL 49/M Yes/no (frontline) Alive Lionakis et al [12]
Aspergillus nidulans 3 wk Sinusitis, CNS CLL/PD 75/F No/fludarabine, ritiximab, cyclophosphamide Alive Baron et al [18]
Extensive aspergillosis 2.1 mo Lungs, brain CLL/PD 76/NA No/rituximab Dead Jain et al [6]
Mucormycosis, aspergillosis 7 mo Lungs CLL/PD 67/M No/rituximab, fludarabine Dead Kreiniz et al [23]
Mucormycosis NA Skin CLL/NA NA NA/no Dead c Figuera Castro et al [17]
Fusarium solani 1.5 mo Disseminated CLL/PD 46/M No/anti-CD20 mAb and bendamustin Alive Chan et al [20]

Abbreviations: CLL, chronic lymphocytic leukemia; CNS, central nervous system; F, female; IA, invasive aspergillosis; IFI, invasive fungal infection; M, male; mAb, monoclonal antibody; MCL, mantle cell lymphoma; NA, not available; PCNSL, primary central nervous system lymphoma; PD, progressive disease; PJP, Pneumocystis jirovecii pneumonia; PR, partial response; SD, stable disease; WM, Waldenström macroglobulinemia.

aFive additional cases of IFIs in patients with LLC and MCL were reported at a single university cancer center during a 1-year study period, including 2 cases of IA and 1 case each of mucormycosis, PJP, and cryptococcosis, all occurring within 3 months of ibrutinib therapy (Protin et al [16]). Dimopoulos et al [9] reported 1 case of IA in a patient with WM; Byrd et al [10], 2 cases of pulmonary IA; Byrd et al [11], 1 case of cryptococcal infection in a patient with CLL; O’Brien et al [21], 1 case of PJP in a patient with CLL; Choquet et al [24], 2 cases of pulmonary IA in patients with PCNSL; and Grommes et al [25], 1 case of CNS IA in a patient with PCNSL. Information on infectious disease outcome and patient clinical characteristics was not available for these cases.

bThe patient received an intermittent course of low-dose oral corticosteroids.

cDeath due to unrelated causes.

dPossible IA.

Table 2.

Frequency of Reported IFIs in Clinical Studies of Ibrutinib Treatment for Hematological Cancer

Type and Status of Cancer Type of IFI (No. of Cases) Frequency of IFI, % Patients, No. Median Follow-up, mo Study Timing, Month/ Year Reference
Relapsed CLL Cryptococcosis (1) 1.2 85 20.9 5/2010–2/2013 Byrd et al [11]
Relapsed CLL/SLL IA (2) 0.5 391 9.4 6/2012–11/2013 Byrd et al [10]
Relapsed WM IA (1) 3.2 31 18.1 8/2014–2/2015 Dimopoulos et al [9]
Relapsed MCL Cryptococcosis (1), PJP (1), histoplasmosis (1) 2.7 111 26.7 2/2011–1/2014 Wang et al [7]
CLL 1 multifocal IA, 1 fungal pneumonia 1.6 127 13 7/2010–5/2014 Jain et al [6]
Relapsed/refractory DLBCL None 0 80 11.5 5/2012–5/2013 Wilson et al [8]
Refractory CLL/SLL PJP (1) 0.7 145 27.6 1/2013–6/2013 O’Brien et al [21]
Refractory PCNSLa IA (7), PJP (1) 44 18 15.5 8/2014–3/2016 Lionakis et al 12]
Refractory PCNSL IA (2) 11 18 NA 9/2015–8/2016 Choquet et al [24]
Refractory PCNSL IA (1) 5 20 NA NA Grommes et al [25]

Abbreviations: CLL, chronic lymphocytic leukemia; CNS, central nervous system; DLBCL, diffuse large B cell lymphoma; IA, invasive aspergillosis; IFI, invasive fungal infection; MCL, mantle cell lymphoma; NA, not available; PCNSL, primary central nervous system lymphoma; PJP, Pneumocystis jirovecii pneumonia; SLL, small lymphocytic lymphoma; WM, Waldenström macroglobulinemia.

aOf enrolled patients, 28% were previously untreated.

Pneumocystis jirovecii pneumonia and Cryptococcosis

It is of interest that these opportunistic infections were unanticipated on the basis of the phenotype observed in patients with X-linked agammaglobulinemia, who display a genetic deficiency of BTK [27]. In fact, the only reported case of opportunistic IFI in X-linked agammaglobulinemia children is a case of PJP [28]. Because Pneumocystis infections occur sporadically in patients with acquired or congenital defects in B-cell function, selective inhibition of BTK in B lymphocytes could partially explain the association between ibrutinib and PJP [27, 29]. In fact, recent evidence suggests that immunocompetent mice treated with a BTK inhibitor become susceptible to PJP [30]. Nonetheless, PJP is typically observed in patients with acquired CD4 T-cell immunodeficiency (human immunodeficiency virus [HIV]/AIDS), severe lymphopenia due to high doses of corticosteroids, idiopathic CD4 lymphopenia, severe combined immunodeficiency, X-linked recessive anhidrotic ectodermal dysplasia with immunodeficiency caused by NF-kappa-B essential modulator (NEMO) or IKBA deficiency, X-linked CD40L deficiency, or GATA2 deficiency, indicating that defective CD4 T-lymphocyte–macrophage cross-talk has an important role in the pathogenesis of PJP [27, 29].

Similarly, although cryptococcosis has been reported in a broad range of patients with acquired immunodeficiency (eg, solid organ or stem cell transplant recipients, patients with hematological cancer, and patients receiving corticosteroids), debilitated patients with metabolic abnormalities (eg, cirrhosis), and isolated patients with no apparent acquired or genetic immune defects, this IFI has been classically associated with acquired CD4 T-cell immune defects in the setting of HIV/AIDS or idiopathic CD4 lymphopenia [27]. In addition, cryptococcosis develops in patients with X-linked CD40L deficiency or interleukin 12 receptor mutations and those with autoantibodies to interferon-γ and granulocyte macrophage colony-stimulating factor, revealing an important role for CD4 T-cell–macrophage cross-talk in the host’s defense against this intracellular pathogen [27]. Notably, BTK is expressed in all bone marrow cell lineages, with the exception of T cells and plasma cells [31].

Therefore, given the importance of the T cell-macrophage axis in the pathogenesis of cryptococcosis/PJP, it is plausible that ibrutinib has off-target effects on other kinases that affect CD4 T-cell function. Alternatively, in view of the major role of macrophage activation defects in non-HIV cryptococcosis [32], a direct inhibitory effect of ibrutinib on BTK signaling in monocytes and macrophages could affect susceptibility to infection. In support of this hypothesis, BTK signaling regulates the development and a range of effector functions of myeloid cells, including chemotaxis, adhesion, transmigration, reactive oxygen species production, cytokine response, and inflammasome activation [33–37].

Invasive Mold Infections

There have been sporadic reports of invasive mold infections in patients receiving ibrutinib, either alone or in combination with other immune modulators, raising important questions about the interplay of events that underlie this observation. In particular, IFIs caused by molds, including Aspergillus spp., are typically observed in patients with severe and complex quantitative or qualitative innate immune defects; so far, they have not been associated with B- or T-cell acquired or congenital immunodeficiency [26, 38]. IA typically occurs in patients with hematological cancers and chemotherapy-related prolonged neutropenia or those receiving high-dose corticosteroids and other immunosuppressive regimens for graft-vs-host disease [38]. Indeed, mice with neutrophil or mononuclear phagocyte depletion, but not those with lymphocyte deficiency, are susceptible to IA [39].

In the setting of primary immunodeficiency, IA occurs almost exclusively in patients with genetic defects that affect effector mechanisms, numbers, or chemotaxis of phagocytes, including (1) patients with chronic granulomatous disease and genetic defects in NADPH oxidase-dependent reactive oxygen species production; (2) patients with autosomal-dominant mutations in the transcription factor GATA2 (monoMAC syndrome) who display profound monocytopenia, B-cell and natural killer cell lymphocytopenia, decreased numbers of circulating and tissue-resident dendritic cells, and various neutrophil functional abnormalities; and (3) rarely, patients with severe congenital neutropenia and type I leukocyte adhesion deficiency [27, 38]. Importantly, in the congenital immunodeficiency setting, cases of extrapulmonary IA with CNS involvement that resemble ibrutinib-associated IA have been reported only in patients with a deficiency in caspase recruitment domain-containing protein 9 (CARD9), a key downstream signaling component of C-type lectin receptors that is critical for neutrophil recruitment and effector function [40, 41].

Several lines of evidence, including the variability in frequency of reported infections across different studies (Table 2), indicate that ibrutinib-associated IFIs result from a complex immunodeficiency that affects a broad range of immune cells in the adaptive and innate immune systems. Therefore, disease-related factors, including type and status of underlying cancer, aging, and other comorbid conditions, seem to play a critical role in the ibrutinib-related risk for the development of IA and other IFIs. The “net state of immunosuppression” in such patients is differentially influenced by (1) environmental exposures to fungal conidia; (2) synergistic effects of ibrutinib with other immunosuppressive therapies, especially corticosteroids [42], as shown elsewhere with mucormycosis in patients with chronic granulomatous disease [43]; (3) BTK-independent immunogenetic factors associated with an increased risk of IA in some patients [44, 45]; (4) direct effects of acute pharmacological inhibition of BTK on macrophages and monocytes [12, 46, 47]; and (5) off-target effects of ibrutinib on other kinases [35, 48, 49], especially when the drug is given at higher doses [12], or theoretically excessive ibrutinib exposure resulting from drug-drug interactions or pharmacogenetic differences in ibrutinib metabolism.

For example, TLR-9/BTK signaling in murine macrophages regulates calcineurin-nuclear factor of activated T cells-mediated tumor necrosis factor production and neutrophil chemotaxis and partially accounts for the increased susceptibility to IA in solid organ transplant recipients receiving calcineurin inhibitors [48]. The increased risk of mold infections with pharmacological BTK inhibition is not evident in the context of the genetic deficiency of BTK in patients with X-linked agammaglobulinemia; this could be related to residual BTK activity on myeloid cells [31, 33–37] or the development of compensatory immune mechanisms over the course of long-term (congenital) immunodeficiency.

With regard to its potential off-target effects, ibrutinib binds covalently to several other homologous cysteine-containing kinases of the TEC family, including ITK, BMX, TXK, and TEC [49–51]. In fact, ibrutinib BTK-independent actions that are associated with off-target effects on ITK and other kinases of the TEC family result in the activity of this inhibitor in a wide range of cancers, including breast cancer and T-cell and acute myelogenous leukemia [52–54]. Furthermore, ibrutinib has a very broad kinome [55] and binds noncovalently to several other kinases that have pivotal roles in normal immune signaling and malignant B-cell signaling, including kinases of the Src family [52–54]. In fact, off-target binding to other kinases has been already implicated in noninfectious toxic effects, including cardiac arrhythmias (PI3K/Akt signaling) and bleeding [56–58], consistent with the important role of SRC family kinases on platelet activation. Specifically, platelet signaling through phospho-SRC/spleen tyrosine kinase (SYK) signaling downstream of C-type lectin-like receptor 2 (CLEC-2) receptor is required for stable platelet adhesion to lymphatic endothelial cells and is inhibited by ibrutinib [56]. Notably, platelets participate in immune responses against Aspergillus fumigatus [59].

PRECLINICAL EVALUATION OF THE INFECTIOUS RISK OF SMALL MOLECULE KINASE INHIBITORS: A CALL TO ACTION

We have been witnessing an unprecedented rate of development of compounds that target immune-signaling pathways in malignant and autoimmune diseases (Table 3 and Table 4) [60, 61]. In fact, some of these molecules target the essential pathways of antifungal innate immunity, including C-type lectin receptor signaling (Src and SYK inhibitors), reactive oxygen species production in phagocytes (NOX inhibitors), Janus kinase-signal tranducer and activator of transcription (JAK-STAT) signaling, interleukin 17 signaling, and other proinflammatory cytokine pathways (tumor necrosis factor and interleukin 6) with a prominent role in antifungal immunity [59, 60].

Table 3.

FDA-Approved SMKIs Associated With Severe Bacterial Infections and/or OIsa

Compound (Trade name; Manufacturer) Mode of Action Targets Indication Year of Approval Comments
Sunitinib (Sutent; Pfizer) Multiple TKIs PDGFRα/β, VEGFR1/2/3, Kit, Flt3, CSF-1R, RET RCC, GIST, PNET 2006 Postmarketing reports of severe bacterial infections (sepsis, UTI, SSTI, respiratory); no evidence of OI
Dasatinib (Sprycel; Bristol-Myers Squibb) TKI BCR-Abl, Src, Lck, Yes, Fyn, Kit, EphA2, PDGFRβ Ph+ CML, ALL 2006 Bacterial infections, including sepsis, pneumonia, several cases of PJP, possible fungal pneumonia, candidemia, and CMV reactivation
Pazopanib (Votrient; GlaxoSmithKline) Multiple TKIs VEGFR1/2/3, PDGFRα/β, FGFR1/3, Kit, Lck, Fms, Itk RCC, soft-tissue sarcomas 2009 Warnings for serious bacterial infections; no evidence of OI
Ruxolitinib (Jakafil; Incyte) TKI JAK1/2 Myelofibrosis, PV 2011 Warning box: serious bacterial (UTI), mycobacterial, fungal, viral (VZV, PML) infections
Tofacitinib (Xeljanz; Pfizer) TKI JAK 1/3 Rheumatoid arthritis 2012 Warning box: serious infections, including significant risk for OIs; significant risk for disseminated tuberculosis and IFIs, including cryptococcosis, PJP, Candida esophagitis ; disseminated VZV; CMV
Cabozantinib (Cometriq/ Cabometyx; Exelixis) TKI RET, Met, VEGFR1/2/3, Kit, TrkB, Flt3, Axl, Tie2 Metastatic medullary thyroid cancer, advanced RCC 2012, 2016 No evidence of OI; sepsis reported
Ponatinib (Iclusig; Ariad) Multiple TKIs BCR-Abl, BCR-Abl T315I, VEGFR, PDGFR, FGFR, EphR, Src family kinases, Kit, RET, Tie2, Flt3 Ph+ CML or ALL 2012 No evidence of OI; occasional febrile neutropenia and sepsis
Dabrafenib (Tafinar; GSK) Serine/threonine kinase inhibitor B-Raf Melanoma with BRAF mutations 2013 UTI, cellulitis; no evidence of OI
Ibrutinib (Imbruvica; Pharmacyclics and Johnson & Johnson) TKI Bruton kinase MCL, CLL, WM 2013 Reported SSTIs; no evidence of OI b
Idelalisib (Zydelig; Gilead) Lipid kinase inhibitor PI3Kδ Small lymphocytic lymphoma, NHL, CLL 2014 Black-box warning for serious infections in 21% of patients receiving monotherapy, including CMV reactivation, PJP

Abbreviations: ALL, acute lymphoblastic leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; CMV, cytomegalovirus; FDA, Food and Drug Administration; FGFR, fibroblast growth factor; GIST, gastrointestinal stromal tumor; MCL, mantle cell lymphoma; NHL, non-Hodgkin lymphoma; OI, opportunistic infection; PDGFR, platelet-derived growth factor receptor; Ph+, Philadelphia chromosome positive; PJP, Pneumocystis jirovecii pneumonia; PML, progressive multifocal leukoencephalopathy; PNET, progressive neuroendocrine tumors of pancreatic origin; PV, polycythemia vera; RCC, renal cell carcinoma; SMKIs, small molecular kinase inhibitors; SSTI, skin and soft-tissue infection; TKI, tyrosine kinase inhibitor; UTI, urinary tract infection; VEGFR, vascular endothelial growth factor receptor; VZV, varicella zoster virus; WM, Waldenström macroglobulinemia.

aMajor infectious complications of SMKIs associated with OI and black-box warnings are presented in bold. The full list of FDA-approved SMKIs and associated infectious episodes is presented in Table 4. Temsirolimus (Torsiel; Wyeth, 2007) and everolimus (Afinitor; Novartis, 2009) are 2 recently approved inhibitors of mechanistic target of rapamycin (mTOR) serine/threonine kinase approved for the treatment of RCC and human epidermal growth factor receptor 2–negative breast cancer, PNET, RCC, renal angiomyolipoma, and subependymal giant cell astrocytoma, respectively. Both mTOR inhibitors are immunosuppressive agents associated with an increased risk for severe infections and OIs, including VZV and herpes simplex virus reactivation, PJP, and invasive aspergillosis.

All FDA-approved SMKIs were retrieved from the FDA’s official Web site (https://www.fda.gov/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologic ApprovalReports/NDAandBLAApprovalReports/ucm373420.htm). Information on the pharmacological targets of SMKIs was obtained from the Web site of the Blue Ridge Institute for Medical Research (http://www.brimr.org/PKI/PKIs.htm).

Table 4.

Full List of FDA-Approved SMKIs and Relative Risk for Development of IFIsa

Compound (Trade Name; Manufacturer) Mode of Action Targets Indication Year of Approval Comments
Imatinib (Gleevec; Novartis) TKI BCR-abl, Kit, PDGFR Ph+ CML or ALL, aggressive systemic mastocytosis, CEL, DFSP, HES, GIST, MDS/ MPD 2001 No evidence of OI
Gefitinib (Iressa; Astrazeneca) TKI EGFR NSCLC 2003–2005, 2015 No evidence of infection
Erlotinib (Tarceva; OSI Pharmaceuticals) TKI EGFR NSCLC, pancreatic cancer 2004 No evidence of OI; minimal risk for infection
Sorafenib (Nexavar; Bayer) Dual TKIs B/C-Raf, B-Raf (V600E), Kit, Flt3, RET, VEGFR1/2/3, PDGFRβ Hepatocellular carcinoma, RCC, DTC 2005 No evidence of infection
Sunitinib (Sutent; Pfizer) Multiple TKIs PDGFRα/β, VEGFR1/2/3, Kit, Flt3, CSF-1R, RET RCC, GIST, PNET 2006 Postmarketing reports of severe bacterial infections (sepsis, UTI, SSTI, respiratory); no evidence of OI
Dasatinib (Sprycel, Bristol-Myers Squibb) TKI BCR-Abl, Src, Lck, Yes, Fyn, Kit, EphA2, PDGFRβ Ph+ CML, ALL 2006 Bacterial infections, including sepsis, pneumonia, several cases of PJP, possible fungal pneumonia, candidemia, and CMV reactivation
Lapatinib (Tykerb; GlaxoSmithKline) Dual TKIs EGFR, ErbB2 Breast cancer 2007 No evidence of infection
Nilotinib (Tasigna; Novartis) TKI BCR-Abl, PDGFR, DDR1 Ph+ CML 2007 URTIs, no evidence of OI
Pazopanib (Votrient; GlaxoSmithKline) Multiple TKIs VEGFR1/2/3, PDGFRα/β, FGFR1/3, Kit, Lck, Fms, Itk RCC, soft-tissue sarcomas 2009 Warnings for serious bacterial infections; no evidence of OI
Vandetanib (Caprelsa; IPR Pharms) Multiple TKIs EGFRs, VEGFRs, RET, Brk, Tie2, EphRs, Src family kinases Medullary thyroid cancer 2011 URTIs, no evidence of OI
Vemurafenib (Zelboraf; Hoffmann–La Roche) TKI A/B/C-Raf, B-Raf (V600E), SRMS, ACK1, MAP4K5, FGR Melanoma with BRAFV600E mutation 2011 No evidence of infection
Crizotinib (Xalkori; Pfizer) Multiple TKIs ALK, c-Met (HGFR), ROS, MST1R ALK-positive NSCLC (2011), ROS-1–positive NSCLC (2016) 2011, 2016 URTIs, no evidence of OI
Ruxolitinib (Jakafi; Incyte) TKI JAK1/2 Myelofibrosis, PV 2011 Warning box: serious bacterial (UTI), mycobacterial, fungal, viral (VZV, PML) infections
Axitinib (Inlyta; Pfizer) TKI VEGFR1/2/3, PDGFRβ RCC 2012 No evidence of infection
Bosutinib (Bosulif; Wyeth) TKI BCR-Abl, Src, Lyn, Hck CML 2012 Respiratory tract infections; no evidence of OI
Regorafenib (Stivarga; Bayer) Multiple TKIs VEGFR1/2/3, BCR-Abl, B-Raf, B-Raf (V600E), Kit, PDGFRα/β, RET, FGFR1/2, Tie2, Eph2A CRC 2012 No evidence of OI
Tofacitinib (Xeljanz; Pfizer) TKI JAK 1/3 Rheumatoid arthritis 2012 Warning box: serious infections, including significant risk for OIs; significant risk for disseminated tuberculosis and IFIs, including cryptococcosis, PJP, Candida esophagitis; disseminated VZV; CMV
Cabozantinib (Cometriq/ Cabometyx; Exelixis) TKI RET, Met, VEGFR1/2/3, Kit, TrkB, Flt3, Axl, Tie2 Metastatic medullary thyroid cancer, advanced RCC 2012, 2016 No evidence of OI; sepsis reported
Ponatinib (Iclusig; Ariad) Multiple TKIs BCR-Abl, BCR-Abl T315I, VEGFR, PDGFR, FGFR, EphR, Src family kinases, Kit, RET, Tie2, Flt3 Ph+ CML or ALL 2012 No evidence of OI; occasional febrile neutropenia and sepsis
Trametinib (Mekinist; GlaxoSmithKline) Serine/threonine kinase inhibitor MEK1/2 Melanoma 2013 No evidence of OI; secondary skin infections
Dabrafenib (Tafinar; GlaxoSmithKline) Serine/threonine kinase inhibitor B-Raf Melanoma with BRAF mutations 2013 UTI, cellulitis, no evidence of OI
Afatinib (Gilotrif; Boehringer Ingelheim) TKI EGFR, ErbB2, ErbB4 NSCLC, squamous NSCLC 2013, 2016 No evidence of OI
Ibrutinib (Imbruvica; Pharmacyclics and Johnson & Johnson) TKI Bruton kinase MCL, CLL, WM 2013 Reported SSTIs, no evidence of OI b
Ceritinib (Zykadia; Novartis) TKI ALK, IGF-1R, InsR, ROS1 ALK-positive NSCLC after crizotinib resistance 2014 No evidence of infection
Idelalisib (Zydelig; Gilead) Lipid kinase inhibitor PI3Kδ Small lymphocytic lymphoma, NHL, CLL 2014 Black-box warning for serious infections in 21% of patients receiving monotherapy, including CMV reactivation, PJP
Nintedanib (Ofev; Boehringer Ingelheim) TKI FGFR1/2/3, PDGFRα/β, VEGFR1/2/3, Flt3 Idiopathic pulmonary fibrosis 2014 No evidence of infection
Palbociclib (Ibrance; Park Davis) Serine/threonine kinase CDK 4/6 ER+ and HER2 breast cancer as first- line (2015) and second-line (2016) therapy 2015 URTIs; less commonly, pneumonia, UTIs; no evidence of OI
Lenvatinib (Lenvima; Easai) TKI VEGFRs, FGFRs, PDGFR, Kit, RET Thyroid cancer 2015 Dental/oral infections, UTIs, no evidence of OI
Cobimetinib (Cotellic; Genetech) TKI MEK Melanoma with BRAF V600E/K mutations with vemurafenib 2015 No evidence of infection
Osimetinib (Tagrisso; AstraZeneca) TKI EGFRT790M NSCLC 2015 Pneumonia; no evidence of other infections, including OI
Alectinib (Alecensa; Hoffman–La Roche) TKI ALK, RET ALK-positive NSCLC 2015 No evidence of infection

Abbreviations: ALK, anaplastic lymphoma kinase; ALL, acute lymphoblastic leukemia; CDK, cyclin-dependent kinase; CEL, chronic eosinophilic leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; CMV, cytomegalovirus; CRC, colorectal cancer; DDR1, discoidin domain receptor family, member 1; DFSP, dermatofibrosarcoma protuberans; DTC, differentiated thyroid carcinoma; EGFR, epidermal growth factor receptor; ER, estrogen receptor; FDA, Food and Drug Administration; FGFR, fibroblast growth factor; GIST, gastrointestinal stromal tumor; HER, human epidermal growth factor receptor; HES, hypereosinophilic syndrome; HGFR, hepatocyte growth factor receptor; IFIs, invasive fungal infections; IGF, insulinlike growth factor; MCL, mantle cell lymphoma; MDS/MPD, myelodysplastic/myeloproliferative diseases; MEK, mitogen-activated protein kinase; MST1R, macrophage-stimulating protein receptor aka RON; NHL, non-Hodgkin lymphoma; NSCLC, non–small cell lung cancer; OI, opportunistic infection; PDGFR, platelet-derived growth factor receptor; Ph+, Philadelphia chromosome positive; PI3K, phosphoinositide 3-kinase; PJP, Pneumocystis jirovecii pneumonia; PML, progressive multifocal leukoencephalopathy; PNET, progressive neuroendocrine tumors of pancreatic origin; PV, polycythemia vera; RCC, renal cell carcinoma; ROS, reactive oxygen species; SMKIs, small molecular kinase inhibitors; SSTI, skin and soft-tissue infection; TKI, tyrosine kinase inhibitor; URTIs, upper respiratory tract infections; UTI, urinary tract infection; VEGFR, vascular endothelial growth factor receptor; VZV, varicella zoster virus; WM, Waldenström macroglobulinemia.

aMajor infectious complications of SMKIs associated with OI and black-box warnings are presented in bold. Temsirolimus (Torsiel; Wyeth, 2007) and everolimus (Afinitor; Novartis, 2009) are 2 recently approved inhibitors of mechanistic target of rapamycin (mTOR) serine/threonine kinase approved for the treatment of RCC and HER2-negative breast cancer, PNET, RCC, renal angiomyolipoma, and subependymal giant cell astrocytoma, respectively. Both mTOR inhibitors are immunosuppressive agents associated with an increased risk for severe infections and OI, including VZV and herpes simplex virus reactivation, PJP, and invasive aspergillosis.

All FDA-approved SMKIs were retrieved from the FDA’s official Web site (https://www.fda.gov/Drugs/DevelopmentApprovalProcess/HowDrugsareDevelopedandApproved/DrugandBiologic ApprovalReports/NDAandBLAApprovalReports/ucm373420.htm). Information on the pharmacological targets of SMKIs was obtained from the Web site of the Blue Ridge Institute for Medical Research (http://www.brimr.org/PKI/PKIs.htm).

bInformation obtained from the official Web site of FDA.

There is a paucity of preclinical data on the immunosuppressive potential of small molecule kinase inhibitors (SMKIs). Knockout mice have been bred with selective genetic defects on these pathways and molecular targets [3]. However, pathogenesis studies of the role of SMKIs in targeting immune signaling pathways in clinically relevant models of fungal diseases and cross-validation in knockout mice is needed; such findings would complement the information on the predisposition of patients with congenital immunodeficient to opportunistic IFIs in various components of the corresponding pathway. They would also help predict the excess risk for IFIs that are associated with the clinical use of compounds that block these pathways. As an example, increased susceptibility to PJP [30] and cryptococcosis [62] has been observed in mice with BTK deficiency. The authors of one of these studies study insightfully predicted that the use of BTK inhibitors would result in cryptococcosis in humans [62]. In addition, recent preclinical studies have confirmed the direct impact of BTK inhibition on susceptibility to IA [12].

Another interesting conclusion from the ibrutinib story is that the effects of SMKIs on the immune system and the associated risk for the development of IFIs cannot always be predicted on the basis of the drug target or the phenotype of the related congenital immunodeficiency in humans. This is probably related to a constellation of predisposing factors that are implicated in the pathogenesis of IFIs in cancer patients receiving SMKIs and are virtually impossible to predict (Box 1) [1–3, 44, 45, 60, 63–67].

Overall, in view of the relative rarity of IFIs in patients receiving SMKIs, it is likely that genetic predisposition has a critical role in the development of infection [45, 46]. Lately, significant progress has been made in characterizing the genetic defects that predispose patients to the development of IFIs. In addition, polymorphisms in immune-related genes may predispose patients to IFIs, even patients with severe acquired immunodeficiencies [44, 45].

On the other hand, an analysis of the infectious complications of Food and Drug Administration–approved SMKIs (Table 3) reveals that opportunistic infections, including IFIs, have been almost exclusively related to compounds that target immune signaling pathways, including BTK, JAK/STAT, and PI3K signaling [61]. Because clinical trials typically enroll carefully selected patients with fewer comorbid conditions and immunosuppressive conditions than average patients, the true incidence of IFIs associated with the introduction of a new SMKI is anticipated to be higher in clinical practice, especially in view of the expanding indications of these compounds in patients with severe underlying immunodeficiency [68, 69]. Therefore, an important research priority is increased surveillance for the early capturing and reporting of cases of IFIs in clinical studies of compounds that target the immune system. Importantly, the toxicity associated with IFIs must be clearly discriminated from sepsis episodes or other bacterial infections because the former diseases have different natural histories and prognoses. To that end, the diagnosis of an IFI in the context of a clinical trial should trigger a mandatory report, early notification of the medical community via a timely report in the public access health databases and implementation of preemptive diagnostic measures for IFIs.

With a more standardized approach for capturing IFIs and immunogenetic and immunophenotyping strategies that allow for the accurate assessment of infection risk in individual patients, we can provide personalized approaches to prevent opportunistic fungal diseases associated with SMKIs. We hope that the lessons being learned from ibrutinib will pave the way for new strategies to prevent IFIs in these patients and will increase our insight into the nuances of the immunopathogenesis of human fungal diseases.

Box 1. Potentially Predisposing Factors Associated With an Increased Risk of IFIs in Patients Receiving SMKIs

Factor Comments
Immunodeficiency related to status and type of underlying cancer Certain cancers (eg, B-cell CLL) induce a broad spectrum of immune defects that worsen during progression of the underlying disease [63]; anecdotal observations suggest that disease remission has a positive effect on the infectious complications associated with treatment with SMKIs, whereas clinical studies of frontline ibrutinib-based therapy have not revealed any associations between remission and major infectious complications [64]
Patient-specific environmental exposure Complex epidemiological factors that are associated with climatic variables, travel, occupation, nosocomial exposures, and previous lung colonization with molds (eg, Aspergillus) or other fungi (eg, PJP) have an effect on the fungal inoculum’s effect on individual patients [1, 3]
Broad inhibitory effects of SMKIs on the immune system In contrast to patients with primary immunodeficiency, who may have cell-specific residual activity of the affected gene [47], pharmacological inhibition of the corresponding signaling pathway typically affects a broader range of immune cells that could result in a more severe immunosuppressive phenotype
Acute-onset effects of SMKIs on the immune system Immune restoration mechanisms with a protective role against infectious insults are less likely to operate in the acute setting of pharmacologically induced immunodeficiency than in the long-term course of congenital immunodeficiency
Potential off-target effects of SMKIs The off-target effects of kinase inhibitors depend on the structure, dosage, type of action (covalent binding vs noncompetitive inhibition of the target), and possibly other host-related factors that are difficult to assess (eg, PK/PD properties, metabolism, drug-drug interactions, and synergistic effects with other immunosuppressive medications) and could result in broad immunosuppressive effects
Genetic predisposition to IFI Certain polymorphisms in innate immunity genes are associated with an increased risk for the development of IFI in hematopoietic stem cell transplant recipients and patients with hematological cancer and severe underlying immunodeficiency [44, 45]
Immune defects related to previous infectious episodes IFIs are increasingly observed in patients recovering from sepsis or following infections with certain immunosuppressive viruses (eg, influenza and CMV) [1–3]; the complex underlying mechanisms of sepsis-induced immunodeficiency in these patients are incompletely understood [65]
Immunosuppressive effects of other drugs Corticosteroids and other immunosuppressive medications (eg, purine analogues) markedly increase the risk for the development of IFIs [1–3]; previous, concomitant, or sequential receipt of immunosuppressive therapies makes it challenging to assess the relative risk of IFI in patients treated with SMKIs
Pharmacogenomics and drug-drug interactions on compound metabolism The clinical efficacy, development of resistance, and toxicity associated with SMKI treatment largely depend on the PK/PD parameters of these compounds [60]; genetic variation in drug-metabolizing enzymes or transporters involved in the absorption, metabolism, and elimination of these SMKIs are influenced by a constellation of genetic and nongenetic factors (eg, disease, drugs, comorbid conditions, and exposures) [66, 67]
Net state of immunodeficiency related to aging and underlying comorbid conditions Aging, structural lung disease, iron overload, diabetes mellitus or other metabolic abnormalities, chronic inflammatory disease, and comorbid conditions affect systemic and local antifungal host defense mechanisms; collectively, all these factors profoundly affect the net state of immunodeficiency of individual patients and increase the colonization risk for certain fungi (eg, Aspergillus, and PJP) [3]
Uncharacterized pathogen-associated risk factors Dissecting the complex pathogenetic mechanisms of IFIs in immunocompromised patients will require understanding the molecular interactions among fungal virulence attributes and (1) the microbiome, (2) other copathogens, (3) local and systemic host defense pathways, and (4) immunosuppression related to underlying diseases, medications, and incompletely characterized environmental factors

Abbreviations: CLL, chronic lymphocytic leukemia; CMV, cytomegalovirus; IFI, invasive fungal infection; PJP, Pneumocystis jirovecii pneumonia; PK/PD, pharmacokinetic/pharmacodynamic; SMKIs, small molecule kinase inhibitors.

Notes

Financial support. This work was supported in part by the Division of Intramural Research of the National Institute of Allergy and Infectious Diseases, National Institutes of Health (M. S. L.), the Greek State Scholarship Foundation (IKY) (G. C.), and the Texas 4000 Distinguished Professorship Endowment for Cancer Research (D. P. K.).

Potential conflicts of interest. All authors: No reported conflicts of interest. All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

  • 1. Bodey GP. Managing infections in the immunocompromised patient. Clin Infect Dis 2005; 40(suppl 4):S239. [DOI] [PubMed] [Google Scholar]
  • 2. Kontoyiannis DP, Patterson TF. Diagnosis and treatment of invasive fungal infections in the cancer patient: recent progress and ongoing questions. Clin Infect Dis 2014; 59(suppl 5):S356–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Brown GD, Denning DW, Gow NA, Levitz SM, Netea MG, White TC. Hidden killers: human fungal infections. SciTransl Med 2012; 4:165rv13. [DOI] [PubMed] [Google Scholar]
  • 4. Wu P, Nielsen TE, Clausen MH. Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. Drug Discov Today 2016; 21:5–10. [DOI] [PubMed] [Google Scholar]
  • 5. Scott DE, Bayly AR, Abell C, Skidmore J. Small molecules, big targets: drug discovery faces the protein-protein interaction challenge. Nat Rev Drug Discov 2016; 15:533–50. [DOI] [PubMed] [Google Scholar]
  • 6. Jain P, Keating M, Wierda W et al. . Outcomes of patients with chronic lymphocytic leukemia after discontinuing ibrutinib. Blood 2015; 125:2062–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Wang ML, Blum KA, Martin P et al. . Long-term follow-up of MCL patients treated with single-agent ibrutinib: updated safety and efficacy results. Blood 2015; 126:739–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Wilson WH, Young RM, Schmitz R et al. . Targeting B cell receptor signaling with ibrutinib in diffuse large B cell lymphoma. Nat Med 2015; 21:922–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Dimopoulos MA, Trotman J, Tedeschi A et al. ; iNNOVATE Study Group and the European Consortium for Waldenström’s Macroglobulinemia Ibrutinib for patients with rituximab-refractory Waldenström’s macroglobulinaemia (iNNOVATE): an open-label substudy of an international, multicentre, phase 3 trial. Lancet Oncol 2017; 18:241–50. [DOI] [PubMed] [Google Scholar]
  • 10. Byrd JC, Brown JR, O’Brien S et al. ; RESONATE Investigators Ibrutinib versus ofatumumab in previously treated chronic lymphoid leukemia. N Engl J Med 2014; 371:213–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Byrd JC, Furman RR, Coutre SE et al. . Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med 2013; 369:32–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Lionakis MS, Dunleavy K, Roschewski M et al. . Inhibition of B cell receptor signaling by ibrutinib in primary CNS lymphoma. Cancer Cell 2017; 31:833–843.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ruchlemer R, Ben Ami R, Lachish T. Ibrutinib for chronic lymphocytic leukemia. N Engl J Med 2016; 374:1593–4. [DOI] [PubMed] [Google Scholar]
  • 14. Arthurs B, Wunderle K, Hsu M, Kim S. Invasive aspergillosis related to ibrutinib therapy for chronic lymphocytic leukemia. Respir Med Case Rep 2017; 21:27–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Ahn IE, Jerussi T, Farooqui M, Tian X, Wiestner A, Gea-Banacloche J. Atypical Pneumocystis jirovecii pneumonia in previously untreated patients with CLL on single-agent ibrutinib. Blood 2016; 128:1940–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Protin C, Paricaud K, Dutertre M et al. . ID-04—profil des infections survenant sous ibrutinib chez des patients atteints d’hémopathies lymphoïdes B. Med Mal Infect 2016; 46(4 suppl 1):61.26725541 [Google Scholar]
  • 17. CE Figuera Castro. Cutaneous mucormycosis in a patient with chronic lymphocytic leukemia: a pharmacological dilemma. J Invest Med 2016; 64:956–57. [Google Scholar]
  • 18. Baron M, Zini JM, Belval TC, Vignon M, Denis B, Malphettes M. Fungal infections in patients treated with ibrutinib: two unusual cases of invasive aspergillosis and cryptococcal meningoencephalitis. Leuk Lymph 2017; 58:2981–2. [DOI] [PubMed] [Google Scholar]
  • 19. Messina JA, Maziarz EK, Spec A, Kontoyiannis DP, Perfect JR. Disseminated cryptococcosis with brain involvement in patients with chronic lymphoid malignancies on ibrutinib. Open Forum Infect Dis 2017; 4:ofw261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Chan TS, Au-Yeung R, Chim CS, Wong SC, Kwong YL. Disseminated fusarium infection after ibrutinib therapy in chronic lymphocytic leukaemia. Ann Hematol 2017; 96:871–2. [DOI] [PubMed] [Google Scholar]
  • 21. O’Brien S, Jones JA, Coutre SE et al. . Ibrutinib for patients with relapsed or refractory chronic lymphocytic leukaemia with 17p deletion (RESONATE-17): a phase 2, open-label, multicentre study. Lancet Oncol 2016; 17:1409–18. [DOI] [PubMed] [Google Scholar]
  • 22. Okamoto K, Proia LA, Demarais PL. Disseminated cryptococcal disease in a patient with chronic lymphocytic leukemia on ibrutinib. Case Rep Infect Dis 2016; 2016:4642831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Kreiniz N, Bejar J, Polliack A, Tadmor T. Severe pneumonia associated with ibrutinib monotherapy for CLL and lymphoma. Hematol Oncol. 2017. doi:10.1002/hon.2387. [DOI] [PubMed] [Google Scholar]
  • 24. Choquet S, et al. Ibrutinib monotherapy in relapse or refractory primary CNS lymphoma (PCNSL) and primary vitreo-retinal lymphoma (PVRL): result of the interim analysis of the iLOC phase II study from the Lysa and the French LOC network. Abstract 784. Presented at: 58th ASH Annual Meeting and Exposition; 2016. [Google Scholar]
  • 25. Grommes C, et al. Single-agent ibrutinib in recurrent/refractory central nervous system lymphoma. Abstract 783. Presented at: 58th ASH Annual Meeting and Exposition; 2016. [Google Scholar]
  • 26. Tisi MC, Hohaus S, Cuccaro A et al. . Invasive fungal infections in chronic lymphoproliferative disorders: a monocentric retrospective study. Haematologica 2017; 102:e108–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Lionakis MS, Netea MG, Holland SM. Mendelian genetics of human susceptibility to fungal infection. Cold Spring Harb Perspect Med 2014; 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Kanegane H, Nakano T, Shimono Y, Zhao M, Miyawaki T. Pneumocystis jiroveci pneumonia as an atypical presentation of X-linked agammaglobulinemia. Int J Hematol 2009; 89:716–7. [DOI] [PubMed] [Google Scholar]
  • 29. Eddens T, Kolls JK. Pathological and protective immunity to Pneumocystis infection. Semin Immunopathol 2015; 37:153–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Linke M, Cusion M, Ashbaugh A.. Immunocompetent mice treated with a Bruton kinase inhibitor are susceptible to Pneumocystis infection. Abstract HMB04. Presented at: ASM Microbe Meeting; June 2017; New Orleans, LA. [Google Scholar]
  • 31. Stiff A, Trikha P, Wesolowski R et al. . Myeloid-derived suppressor cells express Bruton’s tyrosine kinase and can be depleted in tumor-bearing hosts by ibrutinib treatment. Cancer Res 2016; 76:2125–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Panackal AA, Wuest SC, Lin YC et al. . Paradoxical immune responses in non-HIV cryptococcal meningitis. PLoS Pathog 2015; 11:e1004884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. 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; 117:1329–39. [DOI] [PubMed] [Google Scholar]
  • 34. Mueller H, Stadtmann A, Van Aken H et al. . Tyrosine kinase Btk regulates E-selectin-mediated integrin activation and neutrophil recruitment by controlling phospholipase C (PLC) gamma2 and PI3Kgamma pathways. Blood 2010; 115:3118–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Mangla A, Khare A, Vineeth V et al. . Pleiotropic consequences of Bruton tyrosine kinase deficiency in myeloid lineages lead to poor inflammatory responses. Blood 2004; 104:1191–7. [DOI] [PubMed] [Google Scholar]
  • 36. Lougaris V, Baronio M, Vitali M et al. . Bruton tyrosine kinase mediates TLR9-dependent human dendritic cell activation. J Allergy Clin Immunol 2014; 133:1644–50.e4. [DOI] [PubMed] [Google Scholar]
  • 37. Ito M, Shichita T, Okada M et al. . Bruton’s tyrosine kinase is essential for NLRP3 inflammasome activation and contributes to ischaemic brain injury. Nat Commun 2015; 6:7360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Obar JJ, Hohl TM, Cramer RA. New advances in invasive aspergillosis immunobiology leading the way towards personalized therapeutic approaches. Cytokine 2016; 84:63–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Espinosa V, Jhingran A, Dutta O et al. . Inflammatory monocytes orchestrate innate antifungal immunity in the lung. PLoS Pathog 2014; 10:e1003940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Rieber N, Gazendam RP, Freeman AF et al. . Extrapulmonary Aspergillus infection in patients with CARD9 deficiency. JCI Insight 2016; 1:e89890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Drewniak A, Gazendam RP, Tool AT et al. . Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency. Blood 2013; 121:2385–92. [DOI] [PubMed] [Google Scholar]
  • 42. Lionakis MS, Kontoyiannis DP. Glucocorticoids and invasive fungal infections. Lancet 2003; 362:1828–38. [DOI] [PubMed] [Google Scholar]
  • 43. Vinh DC, Freeman AF, Shea YR et al. . Mucormycosis in chronic granulomatous disease: association with iatrogenic immunosuppression. J Allergy Clin Immunol 2009; 123:1411–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Fisher CE, Hohl TM, Fan W et al. . Validation of single nucleotide polymorphisms in invasive aspergillosis following hematopoietic cell transplantation. Blood 2017; 129:2693–2701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Cunha C, Aversa F, Bistoni G et al. . Immunogenetic profiling to predict risk of invasive fungal diseases: where are we now?Immunol Invest 2011; 40:723–34. [DOI] [PubMed] [Google Scholar]
  • 46. Stadler N, Hasibeder A, Aranda Lopez P et al. . The Bruton tyrosine kinase inhibitor ibrutinib abrogates triggering receptor on myeloid cells 1 mediated neutrophil activation. Haematologica 2017; 102:e191–e194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Futatani T, Miyawaki T, Tsukada S et al. . Deficient expression of Bruton’s tyrosine kinase in monocytes from X-linked agammaglobulinemia as evaluated by a flow cytometric analysis and its clinical application to carrier detection. Blood 1998; 91:595–602. [PubMed] [Google Scholar]
  • 48. Herbst S, Shah A, Mazon Moya M et al. . Phagocytosis-dependent activation of a TLR9-BTK-calcineurin-NFAT pathway co-ordinates innate immunity to Aspergillus fumigatus. EMBO Mol Med 2015; 7:240–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Wu P, Nielsen TE, Clausen MH. Small-molecule kinase inhibitors: an analysis of FDA-approved drugs. Drug Discov Today 2016; 21:5–10. [DOI] [PubMed] [Google Scholar]
  • 50. Hutchinson CV, Dyer MJ. Breaking good: the inexorable rise of BTK inhibitors in the treatment of chronic lymphocytic leukaemia. Br J Haematol 2014; 166:12–22. [DOI] [PubMed] [Google Scholar]
  • 51. Sagiv-Barfi I, Kohrt HE, Czerwinski DK, Ng PP, Chang BY, Levy R. Therapeutic antitumor immunity by checkpoint blockade is enhanced by ibrutinib, an inhibitor of both BTK and ITK. Proc Natl Acad Sci U S A 2015; 112:E966–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Rotin LE, Gronda M, MacLean N et al. . Ibrutinib synergizes with poly(ADP-ribose) glycohydrolase inhibitors to induce cell death in AML cells via a BTK-independent mechanism. Oncotarget 2016; 7:2765–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Grabinski N, Ewald F. Ibrutinib (ImbruvicaTM) potently inhibits ErbB receptor phosphorylation and cell viability of ErbB2-positive breast cancer cells. Invest New Drugs 2014; 32:1096–104. [DOI] [PubMed] [Google Scholar]
  • 54. Zaitseva L, Murray MY, Shafat MS et al. . Ibrutinib inhibits SDF1/CXCR4 mediated migration in AML. Oncotarget 2014; 5:9930–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Pan Z, Scheerens H, Li SJ et al. . Discovery of selective irreversible inhibitors for Bruton’s tyrosine kinase. ChemMedChem 2007; 2:58–61. [DOI] [PubMed] [Google Scholar]
  • 56. Levade M, David E, Garcia C, Laurent PA et al. . Ibrutinib treatment affects collagen and von Willebrand factor-dependent platelet functions. Blood 2014; 124:3991–5. [DOI] [PubMed] [Google Scholar]
  • 57. McMullen JR, Boey EJ, Ooi JY, Seymour JF, Keating MJ, Tam CS. Ibrutinib increases the risk of atrial fibrillation, potentially through inhibition of cardiac PI3K-Akt signaling. Blood 2014; 124:3829–30. [DOI] [PubMed] [Google Scholar]
  • 58. Beyer A, Ganti B, Majkrzak A, Theyyunni N. A perfect storm: tyrosine kinase inhibitor-associated polymorphic ventricular tachycardia. J Emerg Med 2016; 52:e123–e127. [DOI] [PubMed] [Google Scholar]
  • 59. Perkhofer S, Kehrel BE, Dierich MP et al. . Human platelets attenuate Aspergillus species via granule-dependent mechanisms. J Infect Dis 2008; 198:1243–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Scott DE, Bayly AR, Abell C, Skidmore J. Small molecules, big targets: drug discovery faces the protein-protein interaction challenge. Nat Rev Drug Discov 2016; 15:533–50. [DOI] [PubMed] [Google Scholar]
  • 61. Reinwald M, Boch T, Hofmann WK, Buchheidt D. Risk of infectious complications in hemato-oncological patients treated with kinase inhibitors. Biomark Insights 2016; 10(suppl 3):55–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Szymczak WA, Davis MJ, Lundy SK, Dufaud C, Olszewski M, Pirofski LA. X-linked immunodeficient mice exhibit enhanced susceptibility to Cryptococcus neoformans infection. MBio 2013; 4:pii: e00265–13. doi:10.1128/mBio.00265-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Forconi F, Moss P. Perturbation of the normal immune system in patients with CLL. Blood 2015; 126:573–81. [DOI] [PubMed] [Google Scholar]
  • 64. Burger JA, Tedeschi A, Barr PM et al. ; RESONATE-2 Investigators Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med 2015; 373:2425–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol 2013; 13:862–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Pander J, Guchelaar HJ, Gelderblom H. Pharmacogenetics of small-molecule tyrosine kinase inhibitors: optimizing the magic bullet. Curr Opin Mol Ther 2010; 12:654–61. [PubMed] [Google Scholar]
  • 67. De Jong J, Hellemans P, De Wilde S et al. . Drug-drug interaction study of ibrutinib and moderate and strong CYP3A inhibitors in patients with B-cell malignancy. Abstract 3964. Presented at: 58th American Society of Hematology Meeting; December 2016; San Diego, CA. [Google Scholar]
  • 68. Dubovsky JA, Flynn R, Du J et al. . Ibrutinib treatment ameliorates murine chronic graft-versus-host disease. J Clin Invest 2014; 124:4867–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Schutt SD, Fu J, Nguyen H et al. . 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]

Articles from Clinical Infectious Diseases: An Official Publication of the Infectious Diseases Society of America are provided here courtesy of Oxford University Press

RESOURCES