Skip to main content

MINI REVIEW article

Front. Pharmacol., 18 September 2019
Sec. Pharmacology of Anti-Cancer Drugs
This article is part of the Research Topic Updated and New Applications of Src Tyrosine Kinase Inhibitors View all 5 articles

Src Tyrosine Kinase Inhibitors: New Perspectives on Their Immune, Antiviral, and Senotherapeutic Potential

  • Department of Pharmacy, Biotechnology, Nutrition, Optics and Optometry, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid. Madrid, Spain

Deregulated activity of the Src tyrosine kinases leads to malignant transformation. Since the FDA approval of the tyrosine kinase inhibitor, imatinib, in 2001 for the treatment of chronic myeloid leukemia (CML), the number of these inhibitors together with Src tyrosine kinase inhibitors (STKIs) has increased notably due to their beneficial effects. Dasatinib, a second-generation STKI inhibitor widely studied, proved high efficiency in CML patients resistant to imatinib. In the last decade STKIs have also been implicated and showed therapeutic potential for the treatment of diverse pathologies other than cancer. In this regard, we review the properties of STKIs, dasatinib in particular, including its immunomodulatory role. Similarly, the potential benefits, adverse effects, and safety concerns of these inhibitors regarding viral infections are considered. Moreover, since life expectancy has increased in the last decades accompanied by age-related morbidity, the reduction of undesirable effects associated to aging has become a powerful therapeutic target. Here, we comment on the ability of STKIs to alleviate age-associated physical dysfunction and their potential impact in the clinic.

Introduction

Src is the prototypical member of a of nonreceptor protein–tyrosine kinases family, which in humans is composed of 11 members (reviewed in Manning et al. (2002). This family has been involved in a wide variety of essential functions to sustain cellular homeostasis where they regulate cell cycle progression, motility, proliferation, differentiation, and survival, among other cellular processes (Roskoski, 2015). As a consequence, their deregulated activity has been linked to malignant transformation, and small tyrosine kinase inhibitors have been indicated for the treatment of certain blood malignancies, including chronic myeloid leukemia (CML).

The tyrosine kinase inhibitor dasatinib demonstrated high efficacy in CML patients resistant to imatinib treatment (Weisberg et al., 2007) by targeting several Src kinases involved in the activation of the immune system (Blake et al., 2008). Importantly, emerging data from basic research and clinical trials suggest a wider potential role, for Src tyrosine kinase inhibitors (STKIs) in general and dasatinib in particular, beyond its antitumoral effect. Next, we review the novel perspectives of these inhibitors as immunotherapeutic, antiviral, and geroprotective drugs.

Immunotherapy

Dasatinib as Immunosuppressor

Ligand binding to TCR/CD3 complex activates diverse Src kinase family members, leading to T-cell activation. Patients with CML showing aberrant expression of the ABL-BCR Src kinase are efficiently treated with dasatinib, a broader and higher specific pan-Src kinase inhibitor (Das et al., 2006).

In fact, proteomic analyses revealed that dasatinib bound to 30 different kinases (Bantscheff et al., 2007; Rix et al., 2007) in contrast to other tyrosine kinase inhibitors such as imatinib or nilotinib, which showed a more restrictive target. These differences in the binding profile correlated with key events in T-cell activation (TCR signaling, expression of activation markers, cytokine production, and proliferation) exclusively inhibited by dasatinib.

Furthermore, the inhibitory effects of dasatinib on natural killer (NK) (Blake et al., 2008), CD4+, and naive T cells (Weichsel et al., 2008) suggest a potential immunomodulatory role directly targeting the TCR/CD3 complex. The possibility of combining dasatinib with other immunosuppressive agents, such as CsA and rapamycin, could represent a new way to treat CML patients and other immune-related diseases where the immune system is activated. In fact, it has been proposed very recently the use of dasatinib as a pharmacologic on/off switch for CAR T cells, an adoptive immunotherapy based on the expression of chimeric antigen receptors against cancer (Mestermann et al., 2019). This therapy has been associated with the emergence of cytokine release syndrome. Therefore, blocking Lck, as dasatinib does, could be an excellent and specific treatment for those patients (Mestermann et al., 2019)

Nevertheless, dasatinib has demonstrated to be safe and efficient upon treatment of CML patients, although side effects, such as myelosuppression, bleeding, fluid retention (Brave et al., 2008), body weight loss, severe pleural effusions, or increased risk of infections (Sillaber et al., 2009), have been reported. These adverse effects could be explained by the wider targets of dasatinib (de Lavallade et al., 2008), but the commonest are mostly moderate and usually self-limited or easy to control (Steegmann et al., 2016).

Unexpected Effect of Dasatinib in Lymphocytosis

Monoclonal and oligoclonal expansions of LGLs, known as lymphocytosis, have been linked with a positive immune effect on tumor surveillance (Raitakari et al., 2000; Epling-Burnette et al., 2007). Large Granular Lymphocytes, encompassing CD8+ T lymphocytes (CD3+/CD8+effector memory cells) and NK cells (CD3CD16/CD56+), represent 10% to 15% of the peripheral blood mononuclear cells (PBMCs).

Ten years ago, a pioneering study described a significant expansion of LGLs in PBMCs of a group of CML patients treated with dasatinib (Mustjoki et al., 2009). This unexpected outcome was further confirmed by others (Kim et al., 2009; Nagata et al., 2010; Lee et al., 2011; Tanaka et al., 2012) and correlated with patient’s good prognosis. Although the percentage of LGLs shifts from 27% to 73% depending on the study (reviewed in (Qiu et al., 2014)), generally the effect was dose-dependent, and importantly, none of the other STKIs demonstrated a similar ability.

The expansion of dasatinib-dependent LGLs could lead to a long-term cure in CML patients. In fact, the presence of LGLs at the diagnosis phase was increased during treatment (Kreutzman et al., 2010; Kreutzman et al., 2011; Tanaka et al., 2012) and accompanied by a deep molecular response, by measuring ABL-BCR transcript (Hughes et al., 2017). Furthermore, a case report showed extended lymphocytosis up to 2.4 years after treatment cessation accompanied by presence of memory and effector Cytotoxic T Lymphocytes and NK cells (Jo et al., 2018).

On the other hand, studies with allogenic stem cell–transplanted patients suffering from various malignant hematological diseases confirmed the emergence of NK cells together with γδ T cells and favored patient prognosis (de Witte et al., 2018). In fact, it has been reported that LGL+ population in dasatinib-treated CML patients had more TCR δ rearrangements compared with LGL group (90% vs 10%). These rearrangements were specifically of γδ T cells (Kreutzman et al., 2010). Analyses of a large number of human tumor samples, seeking biomarkers of overall survival outcomes, revealed the key role of infiltrated γδ T cells (Gentles et al., 2015). Evidences of the important role of γδ T cells as antitumoral agents are increasingly growing (for review, see Scheper et al., 2013; Halim et al., 2017). Moreover, their ability to eliminate minimal residual disease in pediatric CML patients with some kind of disorders (malignant or not) was shown (Airoldi et al., 2015). Many clinical trials stimulate γδ T cells with aminobisphosphonates (zoledronate in combination with other compounds) or have applied adoptive transfer of enriched specific γδ T cells (Fisher et al., 2014) to increase their antitumoral effect. The possibility to combine dasatinib with other compounds that promote the action of γδ T cells could be a new approach to broad and improve the possibilities of CML patients’ treatments.

Immune Activation Considerations

The different outcomes of dasatinib-treated patients described above in relation to the immunosuppressive and immune-activation effects are apparently opposed, but the clinical results are indisputable. Patients with CML treated with dasatinib have better prognosis due to the presence of immune cells that protect them from a relapse. Several questions arise from these studies as to why only a group of CML patients have lymphocytosis. Therefore, an exhaustive follow-up of the patients should be carried out to elucidate the reasons underneath.

Dasatinib is a multikinase inhibitor molecule of second generation with unique characteristics compared to other STKIs that acts not only on Src family kinases, but also against c-kit, EPHA2, and PDGF receptor. This mechanism of action could confer dasatinib the ability of increased lymphocytosis and activation of the immune system, therefore conferring patients a better prognosis.

Antiviral Effects

The complexity and redundancy of Src kinases signaling pathways provide a broad window where STKIs can be used. Currently, the field of viral infection is attracting special interest and intense research aimed at understanding and implementing their potential benefits as therapeutics.

The understanding and deciphering of mechanisms through which STKIs might work as antiviral drugs are growing. For instance, dasatinib and saracatinib have demonstrated effectiveness in modulating the life cycle of dengue virus serotype 2 at viral RNA replication by targeting Fyn kinase in vitro (de Wispelaere et al., 2013). Additionally, dasatinib demonstrated cooperative effect in vitro in combination with the viral entry inhibitor sofosbuvir on hepatitis C virus infection (Xiao et al., 2015).

Regarding the mechanisms involved in blocking HIV-1 infection, the picture is even more complex where diverse targeting has been proposed. Dasatinib was reported to control viral replication in vitro through negative regulation of T-cell proliferation, activation, and cytokine production, due to the inhibition of Lck kinase (Blake et al., 2008). Next, its antiviral mechanism, like imatinib and nilotinib, performed in primary and cell lines, was based on its ability to inhibit kinases involved in the virus-cell fusion process, while cell viability and viral receptor expression, required for fusion, remained unaffected. Dasatinib treatment blocked the viral entry at the hemifusion step (Harmon et al., 2010). In contrast, a study investigating whether STKIs—including dasatinib—exerted their inhibitory effect on HIV-1 infection through the interaction between c-SRC and its ligand PTK2B concluded that multiple postentry steps played a major role in blocking HIV-1 infection (McCarthy et al., 2016).

Recent studies in HIV-infected dasatinib-treated PBMCs, from healthy donors, demonstrated that the antiviral effect of dasatinib was exerted not at the viral fusion and entry step as proposed (Harmon et al., 2010) but downstream, targeting the cellular restriction factor SAMHD1 for dephosphorylation (Bermejo et al., 2016).

Antiviral Considerations

The promising results reported on the potential use of STKI to fight against viral infection, in particular HIV-1, have paved the way for an exciting field of research. However, we consider that several issues should be addressed before moving ahead, such as by using humanized preclinical models to optimize both the treatment scheme and the safety of a combined and lifelong therapy based on STKIs and antiretroviral drugs. The mechanisms to be addressed include shifts in apoptosis and/or signaling pathways, susceptibility to opportunistic infections, and the ability to trigger reactivation of other viral coinfections. In fact, it has been reported that some dasatinib-treated CML patients experienced symptomatic cytomegalovirus reactivation (Kreutzman et al., 2011).

A recent detailed review of the potential use of STKIs as well STKI-related safety concerns in HIV-1 infection is recommended for interested readers (Coiras et al., 2017).

Senotherapeutics

Aging constitutes one of the major risks of morbidity and mortality, with senescence being one of the well-established aging hallmarks (Lopez-Otin et al., 2013; McHugh and Gil, 2018). Cell senescence defines an irreversible cell-cycle arrest accompanied by phenotypic changes including resistance to apoptosis and production of senescent-associated secreted phenotype (Coppe et al., 2008). Although this cellular process is beneficial under healthy physiological conditions, the accumulation of senescent cells underlined tissue damage in preclinical models (Kirkland and Tchkonia, 2015). Moreover, the sole presence of these cells is sufficient to produce physical dysfunction and reduced survival rate and life expectancy (Xu et al., 2018). Hence, the proof of concept that elimination of senescent cells delayed age-associated disorders (Baker et al., 2011) established a causal effect between cellular senescence and aging.

Senotherapeutic Considerations and Perspectives of STKIs

Because of the detrimental effect of senescent cells in a plethora of age-related disorders such as atherosclerosis, cachexia, and sarcopenia (McHugh and Gil, 2018), their selective elimination represents an important target for therapeutics development. Among the diverse approaches, senolytics constitute one of the most promising antisenescence therapies. In support of that, and thanks to the use of navitoclax, a first-generation senolytic targeting antiapoptotic proteins, the role of senescent astrocytes and microglia in the etiology of neurodegenerative diseases has been recently stablished (Bussian et al., 2018).

Similarly, STKIs have also been proposed as potential therapeutic agents to fight osteoporosis, another major health problem in the elderly. Since Src kinase is expressed and required for the development of the ruffled border of the osteoclast, the STKI saracatinib has been shown to play a role on bone turnover (Hannon et al., 2010). More recently, the anabolic and antiresorption effects of dasatinib on human osteoblast and osteoclast differentiation and function were reported (Garcia-Gomez et al., 2012).

Several reports have also established the importance of STKIs as promising agents against a plethora of fibrotic, chronic, and age-related diseases in several tissues (reviewed by Wang and Zhuang (2017). Specifically, treatment of epithelial cells and renal interstitial fibroblasts with dasatinib demonstrated suppression of renal fibrosis by targeting Hck, the sole member of the Src kinase family being upregulated in the kidney from renal-transplanted patients (Wei et al., 2017). The authors confirmed their results in a mouse model of renal interstitial fibrosis based on reduction in fibrotic markers (collagen, vimentin, and MMP-2, among others) and proinflammatory cytokines. Similarly, nintedanib (Awasthi and Schwarz, 2015), PP2 (Mima et al., 2011; Taniguchi et al., 2013; Wu et al., 2015), SU6656 (Das et al., 2016), and bosutinib (Sweeney et al., 2008; Elliott et al., 2011) demonstrated effectiveness as potential chronic kidney disease therapeutics.

Combination of senolytics, targeting different pathways, might expand the range of target cells. Specifically, the first combination reported so far (dasatinib plus quercetin [D+Q]) increased the ablation of senescent cells, compared to the effect demonstrated independently (Zhu et al., 2015; Xu et al., 2018). In fact, this combination is currently being tested in clinical trials. NCT02874989 trial aimed to eliminate proinflammatory cells in patients suffering from idiopathic pulmonary fibrosis (Kirkland and Tchkonia, 2017; Katsuumi et al., 2018; Justice et al., 2019). The ability of D+Q is also being tested in an ongoing clinical trial (NCT02652052), which aims to evaluate age-related changes in transplanted survivors.

Likewise, senescence also represents one of the features of an extremely rare and segmental disease known as Hutchinson–Gilford progeria syndrome (HGPS) (Opresko et al., 2002; Kudlow et al., 2007). Patients with HGPS expressing progerin, an unprocessed nuclear lamin A–dominant mutant, show aging features typical of the elderly and undergo accelerated aging, leading to death caused by cardiovascular disease at an average age of 13.5 years (Trigueros-Motos et al., 2011)

Progerin accelerates cellular stressors, including DNA damage and genomic instability (Gonzalo and Kreienkamp, 2015), upregulation of p53 signaling pathway (Varela et al., 2005; Benson et al., 2010; von Muhlinen et al., 2018), and mitochondrial dysfunction (Rivera-Torres et al., 2013), among others. These stimuli trigger senescence in mouse model of the disease (Liu et al., 2013) as well as in cell lines isolated from HGPS patients (Benson et al., 2010; Wheaton et al., 2017).

Recent studies have demonstrated cardiac electrical defects both in a progeroid mouse model as well as in HGPS patients (Rivera-Torres et al., 2016; Filgueiras-Rama et al., 2018). Moreover, the progeroid mouse model showed a sharp decrease in vascular smooth muscle cells (VSMCs) in the medial layer of the aortic arch (Osorio et al., 2011). Progeroid aortic VSMC had an impaired capacity to inhibit vascular calcification as a result of mitochondrial dysfunction, leading to excessive vascular calcification (Villa-Bellosta et al., 2013). Interestingly, D+Q treatment decreased senescence cell markers in the medial layer of the vasculature, although its effects on calcification have not been analyzed yet (Roos et al., 2016).

Considering the complete absence of any treatment for HGPS patients, and in agreement with a recent report suggesting the feasible role of quercetin in attenuating cellular senescence in HGPS (Geng et al., 2018), it is interesting to speculate whether the senolytic approach could help ameliorate the aged phenotype of progeroid preclinical models and, if so, be transferred to clinics.

Concluding Remarks

Increasing evidence reinforces the potential of STKIs, with dasatinib being the most widely studied, as therapeutics in biomedical fields other than cancer. Herein, we have reviewed their novel role and their future perspectives in immunotherapy, viral infections, and geroprotection.

Despite the reported efficacy and safety of dasatinib, certain questions remained to be addressed before results can be translated from preclinical studies into clinics. Although its long-term effect has been studied as immunotherapeutic, it needs to be implemented for other pathologies. Overall, a further characterization of the mechanisms and functions underlying the use of STKIs in diverse contexts looks essential in order to design specific therapies. Additionally, the identification of potential off-target effects raised during prolonged treatments is required. Such treatments should be tested in order to clearly define the link with an increase in healthy life expectancy as well as in ameliorating the pathology.

The use of dasatinib as immunomodulatory agent has opened a broader window for the treatment of diseases other than CML. On the one hand, it could be possible to use it as an immunosuppressor as recently it has been shown for CAR T-cell immunotherapy or even in autoimmune diseases where a profound deregulation of the immune system takes place. On the other hand, and due to its immune activation effect, the potential immune activation due to the emergence of lymphocyte changes in number and a favorable discontinuation of dasatinib is an area of growing interest, due to prolonged treatments and due to financial issues. The achievement of deep molecular responses together with a profound immunosurveillance in some dasatinib-treated patients demands looking for specific predictive biomarkers to asses which patients could interrupt the treatment without undergoing decay. Several studies have shown up the analysis of putative biomarkers as CTL-A4, TFG-β, PDMPs (platelet-derived microparticles), and PD-1, but more clinical trials should be done to confirm these data (Hughes et al., 2017; Nomura et al., 2019).

Regarding the antiviral effect of dasatinib, it is important to mention that the role of the cellular restriction factor SAMHD1 is not restricted to RNA viruses (Ballana and Este, 2015) but to DNA viruses including poxviruses (Hollenbaugh et al., 2013), herpes simplex 1 (Kim et al., 2013), and hepatitis B (Chen et al., 2014; Jeong et al., 2016; Sommer et al., 2016). Hence, it suggests a potential role of dasatinib in viral infections other than RNA as therapeutic.

The use of dasatinib as senolytic avoids the side effects upon accumulation of senescent cells in specific tissues in preclinical model. However, a recent report casts doubts on its beneficial effects to clear senescent cells upon liver cancer chemotherapy (Kovacovicova et al., 2018).

In summary, we consider that translating outcomes obtained by using preclinical models into clinics is highly challenging and that further research is required to shed light on the precise role of STKIs, especially for the senotherapeutic field, which is still in its infancy.

Author Contributions

JR-T and EM conceived and wrote the manuscript. Both authors contributed equally.

Funding

Publication fee was supported by Universidad Europea de Madrid.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Acknowledgments

The authors thank A. Nuñez-Galindo, N. Martínez-Martin, and Z. Sebestyen for critical reading of the manuscript and Peter Bonney for proofreading the article.

References

Airoldi, I., Bertaina, A., Prigione, I., Zorzoli, A., Pagliara, D., Cocco, C., et al. (2015). gammadelta T-cell reconstitution after HLA-haploidentical hematopoietic transplantation depleted of TCR-alphabeta+/CD19+ lymphocytes. Blood 125 (15), 2349–2358. doi: 10.1182/blood-2014-09-599423

PubMed Abstract | CrossRef Full Text | Google Scholar

Awasthi, N., Schwarz, R. E. (2015). Profile of nintedanib in the treatment of solid tumors: the evidence to date. Onco. Targets Ther. 8, 3691–3701. doi: 10.2147/OTT.S78805

PubMed Abstract | CrossRef Full Text | Google Scholar

Baker, D. J., Wijshake, T., Tchkonia, T., LeBrasseur, N. K., Childs, B. G., de Sluis, B., et al. (2011). Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479 (7372), 232–236. doi: 10.1038/nature10600

PubMed Abstract | CrossRef Full Text | Google Scholar

Ballana, E., Este, J. A. (2015). SAMHD1: at the crossroads of cell proliferation, immune responses, and virus restriction. Trends Microbiol. 23 (11), 680–692. doi: 10.1016/j.tim.2015.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Bantscheff, M., Eberhard, D., Abraham, Y., Bastuck, S., Boesche, M., Hobson, S., et al. (2007). Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 25 (9), 1035–1044. doi: 10.1038/nbt1328

PubMed Abstract | CrossRef Full Text | Google Scholar

Benson, E. K., Lee, S. W., Aaronson, S. A. (2010). Role of progerin-induced telomere dysfunction in HGPS premature cellular senescence. J. Cell Sci. 123 (Pt 15), 2605–2612. doi: 10.1242/jcs.067306

PubMed Abstract | CrossRef Full Text | Google Scholar

Bermejo, M., Lopez-Huertas, M. R., Garcia-Perez, J., Climent, N., Descours, B., Ambrosioni, J., et al. (2016). Dasatinib inhibits HIV-1 replication through the interference of SAMHD1 phosphorylation in CD4+ T cells. Biochem. Pharmacol. 106, 30–45. doi: 10.1016/j.bcp.2016.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Blake, S., Hughes, T. P., Mayrhofer, G., Lyons, A. B. (2008). The Src/ABL kinase inhibitor dasatinib (BMS-354825) inhibits function of normal human T-lymphocytes in vitro. Clin. Immunol. 127 (3), 330–339. doi: 10.1016/j.clim.2008.02.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Brave, M., Goodman, V., Kaminskas, E., Farrell, A., Timmer, W., Pope, S., et al. (2008). Sprycel for chronic myeloid leukemia and Philadelphia chromosome–positive acute lymphoblastic leukemia resistant to or intolerant of imatinib mesylate. Clin. Cancer Res. 14 (2), 352–359. doi: 10.1158/1078-0432.CCR-07-4175

PubMed Abstract | CrossRef Full Text | Google Scholar

Bussian, T. J., Aziz, A., Meyer, C. F., Swenson, B. L., van Deursen, J. M., Baker, D. J. (2018). Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 562 (7728), 578–582. doi: 10.1038/s41586-018-0543-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Coiras, M., Ambrosioni, J., Cervantes, F., Miro, J. M., Alcami, J. (2017). Tyrosine kinase inhibitors: potential use and safety considerations in HIV-1 infection. Expert Opin. Drug Saf. 16 (5), 547–559. doi: 10.1080/14740338.2017.1313224

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppe, J. P., Patil, C. K., Rodier, F., Sun, Y., Munoz, D. P., Goldstein, J., et al. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6 (12), 2853–2868. doi: 10.1371/journal.pbio.0060301

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Z., Zhu, M., Pan, X., Zhu, Y., Yan, H., Jiang, T., et al. (2014). Inhibition of hepatitis B virus replication by SAMHD1. Biochem. Biophys. Res. Commun. 450 (4), 1462–1468. doi: 10.1016/j.bbrc.2014.07.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, J., Chen, P., Norris, D., Padmanabha, R., Lin, J., Moquin, R. V., et al. (2006). 2-Aminothiazole as a novel kinase inhibitor template. Structure–activity relationship studies toward the discovery of N-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1- piperazinyl)]-2-methyl-4-pyrimidinyl]amino)]-1,3-thiazole-5-carboxamide (dasatinib, BMS-354825) as a potent pan-Src kinase inhibitor. J. Med. Chem. 49 (23), 6819–6832. doi: 10.1021/jm060727j

PubMed Abstract | CrossRef Full Text | Google Scholar

Das, J. R., Gutkind, J. S., Ray, P. E. (2016). Circulating fibroblast growth factor-2, HIV-Tat, and vascular endothelial cell growth factor-A in HIV-infected children with renal disease activate rho-A and Src in cultured renal endothelial cells. PLoS One 11 (4), e0153837. doi: 10.1371/journal.pone.0153837

PubMed Abstract | CrossRef Full Text | Google Scholar

de Lavallade, H., Punnialingam, S., Milojkovic, D., Bua, M., Khorashad, J. S., Gabriel, I. H., et al. (2008). Pleural effusions in patients with chronic myeloid leukaemia treated with dasatinib may have an immune-mediated pathogenesis. Br. J. Haematol. 141 (5), 745–747. doi: 10.1111/j.1365-2141.2008.07108.x

PubMed Abstract | CrossRef Full Text | Google Scholar

de Wispelaere, M., LaCroix, A. J., Yang, P. L. (2013). The small molecules AZD0530 and dasatinib inhibit dengue virus RNA replication via Fyn kinase. J. Virol. 87 (13), 7367–7381. doi: 10.1128/JVI.00632-13

PubMed Abstract | CrossRef Full Text | Google Scholar

de Witte, M. A., Sarhan, D., Davis, Z., Felices, M., Vallera, D. A., Hinderlie, P., et al. (2018). Early reconstitution of NK and gammadelta T cells and its implication for the design of post-transplant immunotherapy. Biol. Blood Marrow Transplant. 24 (6), 1152–1162. doi: 10.1016/j.bbmt.2018.02.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Elliott, J., Zheleznova, N. N., Wilson, P. D. (2011). c-Src inactivation reduces renal epithelial cell-matrix adhesion, proliferation, and cyst formation. Am. J. Physiol. Cell Physiol. 301 (2), C522–C529. doi: 10.1152/ajpcell.00163.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

Epling-Burnette, P. K., Painter, J. S., Rollison, D. E., Ku, E., Vendron, D., Widen, R., et al. (2007). Prevalence and clinical association of clonal T-cell expansions in myelodysplastic syndrome. Leukemia 21 (4), 659–667. doi: 10.1038/sj.leu.2404590

PubMed Abstract | CrossRef Full Text | Google Scholar

Filgueiras-Rama, D., Rivera Torres, J., Andres, V. (2018). Electrocardiographic abnormalities in patients with Hutchinson–Gilford progeria syndrome. JAMA Cardiol. 3 (10), 1024–1025. doi: 10.1001/jamacardio.2018.2100

PubMed Abstract | CrossRef Full Text | Google Scholar

Fisher, J. P., Heuijerjans, J., Yan, M., Gustafsson, K., Anderson, J. (2014). gammadelta T cells for cancer immunotherapy: a systematic review of clinical trials. Oncoimmunology 3 (1), e27572. doi: 10.4161/onci.27572

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia-Gomez, A., Ocio, E. M., Crusoe, E., Santamaria, C., Hernandez-Campo, P., Blanco, J. F., et al. (2012). Dasatinib as a bone-modifying agent: anabolic and anti-resorptive effects. PLoS One 7 (4), e34914. doi: 10.1371/journal.pone.0034914

PubMed Abstract | CrossRef Full Text | Google Scholar

Geng, L., Liu, Z., Zhang, W., Li, W., Wu, Z., Wang, W., et al. (2018). Chemical screen identifies a geroprotective role of quercetin in premature aging. Protein Cell. 11 (6), 417–435. doi: 10.1007/s13238-018-0567-y

CrossRef Full Text | Google Scholar

Gentles, A. J., Newman, A. M., Liu, C. L., Bratman, S. V., Feng, W., Kim, D., et al. (2015). The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21 (8), 938–945. doi: 10.1038/nm.3909

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalo, S., Kreienkamp, R. (2015). DNA repair defects and genome instability in Hutchinson–Gilford progeria syndrome. Curr. Opin. Cell Biol. 34, 75–83. doi: 10.1016/j.ceb.2015.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Halim, L., Parente-Pereira, A. C., Maher, J. (2017). Prospects for immunotherapy of acute myeloid leukemia using gammadelta T cells. Immunotherapy 9 (2), 111–114. doi: 10.2217/imt-2016-0139

CrossRef Full Text | Google Scholar

Hannon, R. A., Clack, G., Rimmer, M., Swaisland, A., Lockton, J. A., Finkelman, R. D., et al. (2010). Effects of the Src kinase inhibitor saracatinib (AZD0530) on bone turnover in healthy men: a randomized, double-blind, placebo-controlled, multiple-ascending-dose phase I trial. J. Bone Miner. Res. 25 (3), 463–471. doi: 10.1359/jbmr.090830

PubMed Abstract | CrossRef Full Text | Google Scholar

Harmon, B., Campbell, N., Ratner, L. (2010). Role of Abl kinase and the Wave2 signaling complex in HIV-1 entry at a post-hemifusion step. PLoS Pathog. 6 (6), e1000956. doi: 10.1371/journal.ppat.1000956

PubMed Abstract | CrossRef Full Text | Google Scholar

Hollenbaugh, J. A., Gee, P., Baker, J., Daly, M. B., Amie, S. M., Tate, J., et al. (2013). Host factor SAMHD1 restricts DNA viruses in non-dividing myeloid cells. PLoS Pathog. 9 (6), e1003481. doi: 10.1371/journal.ppat.1003481

PubMed Abstract | CrossRef Full Text | Google Scholar

Hughes, A., Clarson, J., Tang, C., Vidovic, L., White, D. L., Hughes, T. P., et al. (2017). CML patients with deep molecular responses to TKI have restored immune effectors and decreased PD-1 and immune suppressors. Blood 129 (9), 1166–1176. doi: 10.1182/blood-2016-10-745992

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeong, G. U., Park, I. H., Ahn, K., Ahn, B. Y. (2016). Inhibition of hepatitis B virus replication by a dNTPase-dependent function of the host restriction factor SAMHD1. Virology 495, 71–78. doi: 10.1016/j.virol.2016.05.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Jo, T., Noguchi, K., Hayashi, S., Irie, S., Hayase, R., Shioya, H., et al. (2018). Long-lasting memory of cellular immunity in a chronic myeloid leukemia patient maintains molecular response 5 after cessation of dasatinib. Oncol. Lett. 15 (3), 2935–2938. doi: 10.3892/ol.2017.7720

PubMed Abstract | CrossRef Full Text | Google Scholar

Justice, J. N., Nambiar, A. M., Tchkonia, T., LeBrasseur, N. K., Pascual, R., Hashmi, S. K., et al. (2019). Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBio Med. 40, 554–563. doi: 10.1016/j.ebiom.2018.12.052

CrossRef Full Text | Google Scholar

Katsuumi, G., Shimizu, I., Yoshida, Y., Minamino, T. (2018). Vascular senescence in cardiovascular and metabolic diseases. Front. Cardiovasc. Med. 5, 18. doi: 10.3389/fcvm.2018.00018

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, D. H., Kamel-Reid, S., Chang, H., Sutherland, R., Jung, C. W., Kim, H. J., et al. (2009). Natural killer or natural killer/T cell lineage large granular lymphocytosis associated with dasatinib therapy for Philadelphia chromosome positive leukemia. Haematologica 94 (1), 135–139. doi: 10.3324/haematol.13151

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, E. T., White, T. E., Brandariz-Nunez, A., Diaz-Griffero, F., Weitzman, M. D. (2013). SAMHD1 restricts herpes simplex virus 1 in macrophages by limiting DNA replication. J. Virol. 87 (23), 12949–12956. doi: 10.1128/JVI.02291-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Kirkland, J. L., Tchkonia, T. (2015). Clinical strategies and animal models for developing senolytic agents. Exp. Gerontol. 68, 19–25. doi: 10.1016/j.exger.2014.10.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Kirkland, J. L., Tchkonia, T. (2017). Cellular senescence: a translational perspective. EBio Med. 21, 21–28. doi: 10.1016/j.ebiom.2017.04.013

CrossRef Full Text | Google Scholar

Kovacovicova, K., Skolnaja, M., Heinmaa, M., Mistrik, M., Pata, P., Pata, I., et al. (2018). Senolytic cocktail dasatinib+quercetin (D+Q) does not enhance the efficacy of senescence-inducing chemotherapy in liver cancer. Front. Oncol. 8, 459. doi: 10.3389/fonc.2018.00459

PubMed Abstract | CrossRef Full Text | Google Scholar

Kreutzman, A., Juvonen, V., Kairisto, V., Ekblom, M., Stenke, L., Seggewiss, R., et al. (2010). Mono/oligoclonal T and NK cells are common in chronic myeloid leukemia patients at diagnosis and expand during dasatinib therapy. Blood 116 (5), 772–782. doi: 10.1182/blood-2009-12-256800

PubMed Abstract | CrossRef Full Text | Google Scholar

Kreutzman, A., Ladell, K., Koechel, C., Gostick, E., Ekblom, M., Stenke, L., et al. (2011). Expansion of highly differentiated CD8+ T-cells or NK-cells in patients treated with dasatinib is associated with cytomegalovirus reactivation. Leukemia 25 (10), 1587–1597. doi: 10.1038/leu.2011.135

PubMed Abstract | CrossRef Full Text | Google Scholar

Kudlow, B. A., Kennedy, B. K., Monnat, R. J., Jr. (2007). Werner and Hutchinson–Gilford progeria syndromes: mechanistic basis of human progeroid diseases. Nat. Rev. Mol. Cell Biol. 8 (5), 394–404. doi: 10.1038/nrm2161

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. J., Jung, C. W., Kim, D. Y., Lee, K. H., Sohn, S. K., Kwak, J. Y., et al. (2011). Retrospective multicenter study on the development of peripheral lymphocytosis following second-line dasatinib therapy for chronic myeloid leukemia. Am. J. Hematol. 86 (4), 346–350. doi: 10.1002/ajh.21980

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, B., Wang, Z., Zhang, L., Ghosh, S., Zheng, H., Zhou, Z. (2013). Depleting the methyltransferase Suv39h1 improves DNA repair and extends lifespan in a progeria mouse model. Nat. Commun. 4, 1868. doi: 10.1038/ncomms2885

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopez-Otin, C., Blasco, M. A., Partridge, L., Serrano, M., Kroemer, G. (2013). The hallmarks of aging. Cell 153 (6), 1194–1217. doi: 10.1016/j.cell.2013.05.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Manning, G., Whyte, D. B., Martinez, R., Hunter, T., Sudarsanam, S. (2002). The protein kinase complement of the human genome. Science 298 (5600), 1912–1934. doi: 10.1126/science.1075762

PubMed Abstract | CrossRef Full Text | Google Scholar

McCarthy, S. D., Sakac, D., Neschadim, A., Branch, D. R. (2016). c-SRC protein tyrosine kinase regulates early HIV-1 infection post-entry. AIDS 30 (6), 849–858. doi: 10.1097/QAD.0000000000001028

PubMed Abstract | CrossRef Full Text | Google Scholar

McHugh, D., Gil, J. (2018). Senescence and aging: causes, consequences, and therapeutic avenues. J. Cell Biol. 217 (1), 65–77. doi: 10.1083/jcb.201708092

PubMed Abstract | CrossRef Full Text | Google Scholar

Mestermann, K., Giavridis, T., Weber, J., Rydzek, J., Frenz, S., Nerreter, T., et al. (2019). The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci. Transl. Med. 11 (499), eaau 5907. doi: 10.1126/scitranslmed.aau5907

CrossRef Full Text | Google Scholar

Mima, A., Abe, H., Nagai, K., Arai, H., Matsubara, T., Araki, M., et al. (2011). Activation of Src mediates PDGF-induced Smad1 phosphorylation and contributes to the progression of glomerulosclerosis in glomerulonephritis. PLoS One 6 (3), e17929. doi: 10.1371/journal.pone.0017929

PubMed Abstract | CrossRef Full Text | Google Scholar

Mustjoki, S., Ekblom, M., Arstila, T. P., Dybedal, I., Epling-Burnette, P. K., Guilhot, F., et al. (2009). Clonal expansion of T/NK-cells during tyrosine kinase inhibitor dasatinib therapy. Leukemia 23 (8), 1398–1405. doi: 10.1038/leu.2009.46

PubMed Abstract | CrossRef Full Text | Google Scholar

Nagata, Y., Ohashi, K., Fukuda, S., Kamata, N., Akiyama, H., Sakamaki, H. (2010). Clinical features of dasatinib-induced large granular lymphocytosis and pleural effusion. Int. J. Hematol. 91 (5), 799–807. doi: 10.1007/s12185-010-0565-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Nomura, S., Ito, T., Satake, A., Ishii, K. (2019). Assessment of soluble cytotoxic T lymphocyte-associated antigen-4, transforming growth factor beta1, and platelet-derived microparticles during dasatinib therapy for patients with chronic myelogenous leukemia. J. Blood Med. 10, 1–8. doi: 10.2147/JBM.S187005

PubMed Abstract | CrossRef Full Text | Google Scholar

Opresko, P. L., von Kobbe, C., Laine, J. P., Harrigan, J., Hickson, I. D., Bohr, V. A. (2002). Telomere-binding protein TRF2 binds to and stimulates the Werner and Bloom syndrome helicases. J. Biol. Chem. 277 (43), 41110–41119. doi: 10.1074/jbc.M205396200

PubMed Abstract | CrossRef Full Text | Google Scholar

Osorio, F. G., Navarro, C. L., Cadinanos, J., Lopez-Mejia, I. C., Quiros, P. M., Bartoli, C., et al. (2011). Splicing-directed therapy in a new mouse model of human accelerated aging. Sci. Transl. Med. 3 (106), 106ra107. doi: 10.1126/scitranslmed.3002847

CrossRef Full Text | Google Scholar

Qiu, Z. Y., Xu, W., Li, J. Y. (2014). Large granular lymphocytosis during dasatinib therapy. Cancer Biol. Ther. 15 (3), 247–255. doi: 10.4161/cbt.27310

PubMed Abstract | CrossRef Full Text | Google Scholar

Raitakari, M., Brown, R. D., Sze, D., Yuen, E., Barrow, L., Nelson, M., et al. (2000). T-cell expansions in patients with multiple myeloma have a phenotype of cytotoxic T cells. Br. J. Haematol. 110 (1), 203–209. doi: 10.1046/j.1365-2141.2000.02131.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Rivera-Torres, J., Acin-Perez, R., Cabezas-Sanchez, P., Osorio, F. G., Gonzalez-Gomez, C., Megias, D., et al. (2013). Identification of mitochondrial dysfunction in Hutchinson–Gilford progeria syndrome through use of stable isotope labeling with amino acids in cell culture. J. Proteomics 91, 466–477. doi: 10.1016/j.jprot.2013.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Rivera-Torres, J., Calvo, C. J., Llach, A., Guzman-Martinez, G., Caballero, R., Gonzalez-Gomez, C., et al. (2016). Cardiac electrical defects in progeroid mice and Hutchinson–Gilford progeria syndrome patients with nuclear lamina alterations. Proc. Natl. Acad. Sci. U. S. A. 113 (46), E7250–E7259. doi: 10.1073/pnas.1603754113

PubMed Abstract | CrossRef Full Text | Google Scholar

Rix, U., Hantschel, O., Durnberger, G., Remsing Rix, L. L., Planyavsky, M., Fernbach, N. V., et al. (2007). Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 110 (12), 4055–4063. doi: 10.1182/blood-2007-07-102061

PubMed Abstract | CrossRef Full Text | Google Scholar

Roos, C. M., Zhang, B., Palmer, A. K., Ogrodnik, M. B., Pirtskhalava, T., Thalji, N. M., et al. (2016). Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 15 (5), 973–977. doi: 10.1111/acel.12458

PubMed Abstract | CrossRef Full Text | Google Scholar

Roskoski, R., Jr. (2015). Src protein–tyrosine kinase structure, mechanism, and small molecule inhibitors. Pharmacol. Res. 94, 9–25. doi: 10.1016/j.phrs.2015.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Scheper, W., Grunder, C., Kuball, J. (2013). Multifunctional gammadelta T cells and their receptors for targeted anticancer immunotherapy. Oncoimmunology 2 (5), e23974. doi: 10.4161/onci.23974

PubMed Abstract | CrossRef Full Text | Google Scholar

Sillaber, C., Herrmann, H., Bennett, K., Rix, U., Baumgartner, C., Bohm, A., et al. (2009). Immunosuppression and atypical infections in CML patients treated with dasatinib at 140 mg daily. Eur. J. Clin. Invest. 39 (12), 1098–1109. doi: 10.1111/j.1365-2362.2009.02206.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sommer, A. F., Riviere, L., Qu, B., Schott, K., Riess, M., Ni, Y., et al. (2016). Restrictive influence of SAMHD1 on hepatitis B virus life cycle. Sci. Rep. 6, 26616. doi: 10.1038/srep26616

PubMed Abstract | CrossRef Full Text | Google Scholar

Steegmann, J. L., Baccarani, M., Breccia, M., Casado, L. F., Garcia-Gutierrez, V., Hochhaus, A., et al. (2016). European LeukemiaNet recommendations for the management and avoidance of adverse events of treatment in chronic myeloid leukaemia. Leukemia 30 (8), 1648–1671. doi: 10.1038/leu.2016.104

PubMed Abstract | CrossRef Full Text | Google Scholar

Sweeney, W. E., Jr., von Vigier, R. O., Frost, P., Avner, E. D. (2008). Src inhibition ameliorates polycystic kidney disease. J. Am. Soc. Nephrol. 19 (7), 1331–1341. doi: 10.1681/ASN.2007060665

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanaka, H., Nakashima, S., Usuda, M. (2012). Rapid and sustained increase of large granular lymphocytes and rare cytomegalovirus reactivation during dasatinib treatment in chronic myelogenous leukemia patients. Int. J. Hematol. 96 (3), 308–319. doi: 10.1007/s12185-012-1132-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Taniguchi, K., Xia, L., Goldberg, H. J., Lee, K. W., Shah, A., Stavar, L., et al. (2013). Inhibition of Src kinase blocks high glucose–induced EGFR transactivation and collagen synthesis in mesangial cells and prevents diabetic nephropathy in mice. Diabetes 62 (11), 3874–3886. doi: 10.2337/db12-1010

PubMed Abstract | CrossRef Full Text | Google Scholar

Trigueros-Motos, L., Gonzalez, J. M., Rivera, J., Andres, V. (2011). Hutchinson–Gilford progeria syndrome, cardiovascular disease and oxidative stress. Front. Biosci. (Schol. Ed.) 3, 1285–1297. doi: 10.2741/226

PubMed Abstract | CrossRef Full Text | Google Scholar

Varela, I., Cadinanos, J., Pendas, A. M., Gutierrez-Fernandez, A., Folgueras, A. R., Sanchez, L. M., et al. (2005). Accelerated ageing in mice deficient in Zmpste24 protease is linked to p53 signalling activation. Nature 437 (7058), 564–568. doi: 10.1038/nature04019

PubMed Abstract | CrossRef Full Text | Google Scholar

Villa-Bellosta, R., Rivera-Torres, J., Osorio, F. G., Acin-Perez, R., Enriquez, J. A., Lopez-Otin, C., et al. (2013). Defective extracellular pyrophosphate metabolism promotes vascular calcification in a mouse model of Hutchinson–Gilford progeria syndrome that is ameliorated on pyrophosphate treatment. Circulation 127 (24), 2442–2451. doi: 10.1161/CIRCULATIONAHA.112.000571

PubMed Abstract | CrossRef Full Text | Google Scholar

von Muhlinen, N., Horikawa, I., Alam, F., Isogaya, K., Lissa, D., Vojtesek, B., et al. (2018). p53 isoforms regulate premature aging in human cells. Oncogene 37 (18), 2379–2393. doi: 10.1038/s41388-017-0101-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Zhuang, S. (2017). Src family kinases in chronic kidney disease. Am. J. Physiol. Renal. Physiol. 313 (3), F721–F728. doi: 10.1152/ajprenal.00141.2017

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, C., Li, L., Menon, M. C., Zhang, W., Fu, J., Kidd, B., et al. (2017). Genomic analysis of kidney allograft injury identifies hematopoietic cell kinase as a key driver of renal fibrosis. J. Am. Soc. Nephrol. 28 (5), 1385–1393. doi: 10.1681/ASN.2016020238

PubMed Abstract | CrossRef Full Text | Google Scholar

Weichsel, R., Dix, C., Wooldridge, L., Clement, M., Fenton-May, A., Sewell, A. K., et al. (2008). Profound inhibition of antigen-specific T-cell effector functions by dasatinib. Clin. Cancer Res. 14 (8), 2484–2491. doi: 10.1158/1078-0432.CCR-07-4393

PubMed Abstract | CrossRef Full Text | Google Scholar

Weisberg, E., Manley, P. W., Cowan-Jacob, S. W., Hochhaus, A., Griffin, J. D. (2007). Second generation inhibitors of BCR-ABL for the treatment of imatinib-resistant chronic myeloid leukaemia. Nat. Rev. Cancer 7 (5), 345–356. doi: 10.1038/nrc2126

PubMed Abstract | CrossRef Full Text | Google Scholar

Wheaton, K., Campuzano, D., Ma, W., Sheinis, M., Ho, B., Brown, G. W., et al. (2017). Progerin-induced replication stress facilitates premature senescence in Hutchinson–Gilford progeria syndrome. Mol. Cell Biol. 37 (14), e00659–16. doi: 10.1128/MCB.00659-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, H., Shi, Y., Deng, X., Su, Y., Du, C., Wei, J., et al. (2015). Inhibition of c-Src/p38 MAPK pathway ameliorates renal tubular epithelial cells apoptosis in db/db mice. Mol. Cell Endocrinol. 417, 27–35. doi: 10.1016/j.mce.2015.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, F., Fofana, I., Thumann, C., Mailly, L., Alles, R., Robinet, E., et al. (2015). Synergy of entry inhibitors with direct-acting antivirals uncovers novel combinations for prevention and treatment of hepatitis C. Gut 64 (3), 483–494. doi: 10.1136/gutjnl-2013-306155

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, M., Pirtskhalava, T., Farr, J. N., Weigand, B. M., Palmer, A. K., Weivoda, M. M., et al. (2018). Senolytics improve physical function and increase lifespan in old age. Nat. Med. 24 (8), 1246–1256. doi: 10.1038/s41591-018-0092-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Y., Tchkonia, T., Pirtskhalava, T., Gower, A. C., Ding, H., Giorgadze, N., et al. (2015). The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14 (4), 644–658. doi: 10.1111/acel.12344

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Src tyrosine kinase inhibitors, dasatinib, immunotherapy, LGLs, γδ T cells, senescence, senolytics, progeria

Citation: Rivera-Torres J and San José E (2019) Src Tyrosine Kinase Inhibitors: New Perspectives on Their Immune, Antiviral, and Senotherapeutic Potential. Front. Pharmacol. 10:1011. doi: 10.3389/fphar.2019.01011

Received: 18 March 2019; Accepted: 08 August 2019;
Published: 18 September 2019.

Edited by:

Olivier Feron, Catholic University of Louvain, Belgium

Reviewed by:

Andaleeb Sajid, National Institutes of Health (NIH), United States
Thomas E. Smithgall, University of Pittsburgh, United States

Copyright © 2019 Rivera-Torres and San José. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: José Rivera Torres, jose.rivera@universidadeuropea.es; Esther San José Martínez, esther.sanjose@universidadeuropea.es

These authors have contributed equally to this work and share senior authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.