Insight in taste alterations during treatment with protein kinase inhibitors

Insight in taste alterations during treatment with protein kinase inhibitors

European Journal of Cancer 86 (2017) 125e134 Available online at www.sciencedirect.com ScienceDirect journal homepage: www.ejcancer.com Review Ins...

750KB Sizes 0 Downloads 55 Views

European Journal of Cancer 86 (2017) 125e134

Available online at www.sciencedirect.com

ScienceDirect journal homepage: www.ejcancer.com

Review

Insight in taste alterations during treatment with protein kinase inhibitors A. van der Werf a,b,1, M. Rovithi a,c,1, J.A.E. Langius b,d, M.A.E. de van der Schueren b,e, H.M.W. Verheul a,* a

Department of Medical Oncology, Cancer Center Amsterdam, VU University Medical Center, P.O. Box 7057, 1081 HV, Amsterdam, The Netherlands b Department of Nutrition and Dietetics, Internal Medicine, VU University Medical Center, P.O. Box 7057, 1081 HV, Amsterdam, The Netherlands c Medical Oncology Unit, Department of Internal Medicine, Agios Nikolaos General Hospital, Const. Paleologos & Knossos, PC 72100, Agios Nikolaos, Crete, Greece d Department of Nutrition and Dietetics, Faculty of Health, Nutrition and Sport, The Hague University of Applied Sciences, P.O. Box 13336, 2501 EH, The Hague, The Netherlands e Faculty of Health and Social Studies, Department of Nutrition and Health, HAN University of Applied Sciences, P.O. Box 6960, 6503 GL, Nijmegen, The Netherlands Received 2 August 2017; accepted 4 September 2017

KEYWORDS Protein kinase inhibitor; Dysgeusia; Dysosmia; Taste; Smell; Oral adverse events; Mucositis

Abstract The role of Protein Kinase Inhibitors (PKI) in the treatment of various types of cancer is increasingly prominent. Their clinical application is accompanied by the development of side effects, among which patient-reported taste alterations. These alterations are missed frequently, but impair nutritional intake, are associated with weight loss and often result in significant morbidity, especially in the context of chronic administration. Accurate reporting of taste alterations is hampered by lack of modules for symptom objectification and inadequate understanding on the underlying mechanisms. In this review we initially describe the physiology of taste and smell and the mechanism of action of PKIs. We proceed to summarize taste related side effects as reported in major clinical trials and describe possible causal factors. Lastly, an in-depth analysis is given on potential molecular pathways responsible for the PKI-

* Corresponding author. E-mail addresses: [email protected] (A. van der Werf), [email protected] (M. Rovithi), [email protected] (J.A.E. Langius), [email protected] (M.A.E. de van der Schueren), [email protected] (H.M.W. Verheul). 1 These authors contributed equally. https://doi.org/10.1016/j.ejca.2017.09.006 0959-8049/ª 2017 Elsevier Ltd. All rights reserved.

126

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

induced taste alterations. Objectification of patient-reported symptoms and universal reporting, along with a better understanding of the underlying mechanisms, will lead to early recognition and optimized treatment, ultimately improving patient adherence and quality of life. ª 2017 Elsevier Ltd. All rights reserved.

1. Introduction/background In the last decade, our increasing insight in the underlying biology of tumorigenesis has revealed distinct pathways that support tumour growth and sustain angiogenesis. Precision blockade of these pathways leads to increased tumour vulnerability and provides the scientific rationale for the development of targeted agents. Protein kinase inhibitors (PKIs) constitute a paradigm category of this class of anticancer agents that act by blocking the intracellular kinase domain of growth receptors or intracellular kinases, thereby inhibiting downstream signalling. Several PKIs have supplanted long-established chemotherapeutic agents and have been approved as standard of care in various treatment lines in the disease course. Clinical application showed their potential to improve patient survival and induce long term disease stabilization in a variety of tumour types. Despite the initial expectations of exclusive tumour cell specificity as opposed to conventional chemotherapeutics, adverse events have been increasingly reported. Chronic administration and subsequent prolongation of survival make the recognition and treatment of these side-effects throughout the treatment period imperative, in order to maintain an appropriate risk/benefit ratio [1e4]. One of the most commonly reported side-effects of PKIs is taste alteration, often referred to as dysgeusia. Taste alterations impair nutritional intake and influences quality of life in patients with cancer [5e8]. Reporting of this side-effect is usually limited to subjective descriptions of alterations; taste function has never been objectively assessed in patients receiving PKIs. The reported prevalence of PKI-induced patientreported taste alterations varies depending on the type of PKI and treatment dose (Table 1). Examples include sunitinib, a Vascular Endothelial Growth Factor Receptor (VEGFR) inhibitor, with a prevalence of patientreported taste alterations varying from 18% [9] to 63% [10] and vismodegib, a Hedgehog signalling pathway inhibitor, with a reported prevalence varying from 19% [11] to 85% [12]. The course of taste alterations has been described in a few studies. Development of oral adverse events was described to develop in 1e15 weeks after introduction of sunitinib or sorafenib [13]. In the majority of patients treated with vismodegib, patient-reported taste alterations developed within the first month after initiation

and reversed within one month after discontinuation of treatment [12]. Although data on prevalence and the course of patient-reported taste alterations have been reported, more detailed data remain limited. The nature of the these alterations is not consistently described and may involve taste loss or taste hypersensitivity, as well as taste alteration, including a metallic, bitter, sour or salty taste [5]. Furthermore, the potential contribution of concurrent smell alterations has been inadequately recognised and recorded. This may be caused by the inconsistency among different trials in recording, reporting and scoring of patient-reported taste alterations while discrepant terminology further smothers the exact prevalence [14]. In addition, there are no consensus recommendations regarding objectification and monitoring of the symptoms. Simultaneously with lack of routine objectification of described symptoms, effort dedicated to unravelling the underlying pathobiology remains limited. Nevertheless, the different manifestations, symptoms, clinical presentations and treatment of oral side-effects caused by targeted agents compared to ‘traditional’ chemotherapy, hint at the possibility of different pathobiology [14].

Table 1 Examples of PKIs and reported prevalence of patient-reported taste alteration. PKI

Primary target [30] Daily dose Prevalence taste alterations

Imatinib

ABL, KIT, PDGFR

Dasatinib

ABL, Src

Erlotinib Gefitinib

EGFR EGFR

Osimertinib EGFR Sunitinib KIT, VEGFR, FLT3 Sorafenib VEGFR, BRAF Vismodegib SMO Crizotinib ALK, c-Met, ROS1

400 mg 600 mg 800 mg 100 mg 140 mg 200 mg 150 mg 250 mg 500 mg 80 mg 37.5 mg 50 mg 800 mg 150 mg 500 mg

3e13% [54e56] 14% [54] NR NR NR 3e10% [57] 6% [58] NR 10% [59] NR 20% 18e63% [9,10,60e64] 3e55% [63,65e69] 19e85% [11,12,70e72] 11e26% [73e75]

ABL: ALK: anaplastic lymphoma kinase. EGFR: epidermal growth factor receptor. FLT3: fms-like tyrosine kinase receptor-3. PDGFR: platelet-derived growth factor receptor. SMO: smoothened homologue. VEGFR: vascular endothelial growth factor receptor.

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

127

Inadequate understanding on the underlying pathobiology further restrains the identification of optimal treatment options [5,14,15]. The aim of this article is to provide insight in potential mechanisms of PKI-induced patient-reported taste alterations. First, the physiology of flavour perception is described, as well as the mechanism of action of PKIs, followed by working hypotheses on the molecular pathways underpinning the development of patient-reported taste alterations in patients treated with PKIs.

of flavour is shown in Fig. 1. Because smell is an important contributor to flavour, smell alterations are often reported by patients as taste alterations. Therefore, patient-reported ‘taste alterations’ may be caused by taste and/or smell changes. The physiology of both sensory systems is described below.

2. Taste versus flavour

Perception of taste starts when tastants interact with taste buds. Each taste bud consists of different morphological cells: type I cells are considered to be supporting cells which are wrapped around other cell types and orchestrate reuptake or degradation of neurotransmitters; type II are taste receptor cells for sweet, bitter and umami; type III are taste receptor cells that appear to be sour and possibly salty detectors, although the cells type(s) mediating salty taste remains ambiguous [17,19,20]. Every taste quality is mediated by a specific receptor, while signal transduction cascades are similar for specific cell types. Type II cells contain receptors for sweet, bitter or umami, which are specific G proteincoupled receptors. The intracellular transduction cascade is mediated, among others, by phospholipase Cb2 (PLC-b2) and inositol-trisphosphate (IP3) [16,17]. After depolarisation of the taste receptor cell, the taste

Taste is a chemical sense that evaluates the nutritious content of food and prevents ingestion of potentially toxic substances. The detection of the five different taste qualities (sweet, bitter, salty, sour and umami) by taste bud receptors in the oropharynx and tongue results in the sense of taste [16,17]. However, this sense is frequently confused with ‘flavour’, which refers to the sense produced after integration of both taste and smell in the brain and is related to temperature and tactile information. Thus, whereas taste specifically refers to taste information arising from taste bud receptors, flavour refers to the end result after the convergence of information from multiple senses in the brain [18]. An overview of how the sense of taste and smell are processed and integrated in the brain to produce the sense

3. Physiology of taste 3.1. Signal transduction

Fig. 1. Integration of taste and smell into flavour perception and potential mechanisms in which protein kinase inhibitors may influence flavour processing. Shown in black: Taste and smell are processed via distinct receptors, cranial nerves and brain regions. The orbitofrontal cortex is the first place where taste and smell information converges and, together with temperature and texture, underlie the sense of flavour [18,24]. Shown in grey: Mechanisms in which protein kinase inhibitors may interfere with the sense of taste or the sense of smell, thereby causing an altered flavour perception. cAMP: cyclic adenosine monophosphate. CaMKII: Ca2þ/calmodulin kinase II. DAG: diacylglycerol. EGFR: epidermal growth factor receptor. FGFR: fibroblast growth factor receptor. FLT3: fms-like tyrosine kinase receptor-3. IP3: inositol-trisphosphate. PIP2: phosphatidylinositol-bisphosphate. PLC: phospholipase C. VEGFR: vascular endothelial growth factor receptor.

128

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

signal is transmitted to sensory nerves, in which adenosine triphosphate (ATP) is the key neurotransmitter. Type III cells possess specific ion-channels. Direct entry of the ion generates a positive inward current and additional depolarisation results in signal transduction to sensory nerve fibres, using serotonin (5-HT) and noradrenaline as neurotransmitters [17,19,21]. The signal transduction of each taste quality, including the involved neurotransmitters, is shown in detail in Fig. 2.

progenitor cells (basal keratinocytes) into differentiated keratinocytes, whereas higher levels promote differentiation into taste cell precursors. These cell precursors express sonic hedgehog, which seems to be the key signal that promotes differentiation into either type I, type II or type III cells in response to high, mid or low b-catenin respectively (Fig. 3) [17].

4. Physiology of smell 3.2. Cell renewal 4.1. Signal transduction The average life span of cells within taste buds in rodents is 10e14 days with much variation in subsets of cells. For the subset of taste receptor cells, half-life times vary from 8 days for type II cells to 22 days for type III cells [22]. Consistency in taste perception is ensured by the constant renewal of taste receptor cells, in which the Wnt/b-catenin pathway is a crucial factor. Binding of Wnt to its receptor stimulates the intracellular Wnt/bcatenin pathway, which regulates b-catenin levels [23]. Low b-catenin levels promote differentiation of

Olfactory receptor neurons, the receptor cells for smell, are located in the olfactory epithelium. Every receptor neuron is sensitive to a subset of odorants, dependent on the particular receptor molecules they express. There are believed to be about 400 different odorant receptor proteins, which are G protein-coupled receptors [24]. After receptor stimulation, an intracellular cascade is activated. This cascade includes adenylate cyclase, cyclic adenosine monophosphate (cAMP) and Ca2þ [25,26]. The resulting

Fig. 2. Gustatory signal transduction. Sweet, bitter and umami signals are mediated by type II cells. These cells possess G protein coupled receptors and signal through a common intracellular cascade. Receptor stimulation activates gustducin, a taste specific G protein, which stimulates phospholipase Cb2 (PLC-b2). PLC-b2 hydrolyses phosphatidylinositol-bisphosphate (PIP2) to diacylglycerol (DAG) and inositol-trisphosphate (IP3). This leads to IP3-mediated release of Ca2þ from intracellular stores and Ca2þ influx via opening of Ca2þchannels. The increased intracellular Ca2þ depolarizes the taste receptor cell [16,17,20] and stimulates secretion of adenosine triphosphate (ATP) and subsequent ATP-enhanced ATP release. ATP binds to postsynaptic ATP receptors and excites afferent fibres. Concurrently ATP excites adjacent type III cells to release 5-HT, which inhibits ATP release from receptor cells, thereby forming. The released ATP is degraded by NTPDase2, expressed on type I cells [19,20]. Sour and salty taste may be detected by type III cells via ion-channels by direct entry of Hþ and Naþ, respectively. This generates a positive inward current and activates voltage-gated Naþ-channels. Naþ influx activates voltage-gated Ca2þ-channels and subsequent Ca2þ influx leads to exocytosis of neurotransmitters. Both 5-HT and noradrenaline are released from sour and possibly from salty taste receptor cells [19,21].

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

129

Fig. 3. Differentiation of progenitors cells into taste cells and non-taste epithelium. Low b-catenin levels promote differentiation of progenitor cells (basal keratinocytes) into differentiated keratinocytes, whereas higher levels promote differentiation into taste cell precursors. Taste cell precursors express sonic hedgehog (Shhþ) and further differentiate into either type I, type II or type III cells in response to high, mid or low b-catenin, respectively [17].

action potential is transmitted via axons, synapsed to mitral cells and projected the brain. The signal transduction route is more extensively described in Fig. 4. 4.2. Cell renewal Olfactory receptor neurons have a life span of approximately 30e60 days and are constantly replaced by division of basal stem cells. Horizontal cells, a subtype of basal stem cells expressing epidermal growth factor receptor (EGFR), act as a reserve pool after epithelial damage. They are responsive to EGF, a factor shown to be mitogenic for stem cells in the nervous system. The proliferation rate of these cells is increased after injury, so a relatively stable population of olfactory receptor neurons is maintained [24,27]. 5. Mechanism of action of protein kinase inhibitors While most classic cytostatics interact with DNA and as a consequence target all rapidly dividing cells, comprising both malignant and benign cells, PKIs target specific protein kinases [2,28]. Protein kinases are enzymes that are part of signalling pathways and play a regulatory role in cell proliferation, differentiation and apoptosis, where they catalyse the transfer of phosphate from ATP to the target protein. This changes the shape and function of the target protein and allows it to signal downstream [1,3,4].

In cancer, protein kinase pathways are dysregulated in several ways. Underlying mechanisms of this dysregulation include protein fusions/mutations resulting in protein kinases which are active in the absence of ligand binding and a decrease in factors that limit protein kinase activation. This aberrant activation and consequent downstream signalling can increase survival, proliferation and migration of cells [1,29]. The causative role of protein kinases in tumorigenesis makes them important targets for anti-cancer drugs [3]. These drugs are referred to as small molecule PKIs. PKIs target the ATP binding site on the kinase, preventing phosphorylation of the kinase and thereby inhibiting protein kinase activity [29]. This in turn blocks the downstream pathway and thus the malignant signal. Although originally assumed to comprise a targeted only approach, numerous closely interconnected pathways are involved obscuring the complete understanding of the underlying mechanism [2]. The inhibition of kinases other than the primary target reflects the relatively low selectivity and may cause off-target toxicity by kinase inhibition in normal tissues [28,30]. 6. Potential causes of PKI-induced patient-reported taste alterations Based on the physiology of flavour perception and the mechanism of action, there are several hypotheses that might explain the development of patient-reported taste alterations. These include taste alteration, smell

130

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

Fig. 4. Olfactory signal transduction. Olfactory receptor neurons possess G protein coupled receptors. Receptor stimulation activates adenylate cyclase, a catalyser for the conversion of cytosolic ATP to cyclic adenosine monophosphate (cAMP), and leads to opening of cAMP-gated cation-selective channels. The affinity of these cAMP-gated channels for cAMP is reduced by binding of calmodulin, while phoshorlyation of the channel by protein kinase C increases cAMP sensitivity [25].After opening of the cAMP-gated cation channel, Naþ and Ca2þ influx depolarizes the neuron and opens Ca2þ-gated Cl channels. The resulting Cl efflux eventually results in generation of an action potential. The signal is transmitted to glomeruli in the olfactory bulb and synapsed to mitral cells via glutamate [76]. The axons of mitral cells run through the granule cell layer and project to other parts of the brain [24,77]. Olfactory signalling is inhibited by GABAergic and dopaminergic input at the level of the glomerulus [24,76] and granule cell layer. Olfactory signalling is terminated by Ca2þ/calmodulin kinase II, which is activated by the Ca2þ increase in the olfactory receptor neuron. This kinase phosphorylates adenylate cyclase and inhibits its activity [26].

alteration and neurodegeneration. These hypotheses are described below and shown in Fig. 1. 6.1. Taste alterations due to oral toxicity Oral toxicity, such as oral mucositis and xerostomia (dry mouth), occurs frequently [14] and is likely to play an important role in taste alterations. Oral mucositis may be related to taste alterations in several ways. Taste alterations could be the introductory demonstration of off-target effects on the oral mucosa, which could later be clinically apparent by the development of oral mucositis. Therefore, the same reasons that have been implicated in the development of oral mucositis

could mechanistically explain the initial symptom of taste alteration. Oral mucositis will damage the epithelial cell membranes and may also cause formation of compounds causing off-flavour taste perceptions [31]. Nonetheless, patient-reported taste alterations during PKIs may be present in the absence of clinical lesions [14,32], implying that oral mucositis is not the (only) causal factor. Another oral toxicity that probably contributes to taste alterations includes xerostomia, as this hampers the solubilising of food particles and therefore a decreased presentation of tastants to the taste receptors [5]. Further oral factors that may play a role in the development of taste alterations include local disturbance of the immune environment with

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

upregulation of proinflammatory factors and changes in physiologic oral microbiota. These factors have previously been correlated with the development of oral toxicity in the case of other targeted agents, namely mammalian or mechanistic target of rapamycin (mTOR) inhibitors [33e35]. 6.2. Taste alterations by affecting taste receptor cells Taste alterations may be caused by distortion of signal transmission of one or more of the five taste qualities. As described above, sweet, bitter and umami taste signal through a common pathway, with secondary messengers including PLC-b2 and IP3. PKIs interfering with this pathway would be expected to cause taste changes limited to these taste qualities, leaving sour and salty taste as well as smell unaffected. Phosphatidylinositol-bisphosphate (PIP2) is a second messenger within this pathway. PIP2 is also a substrate in downstream signalling of pathways activated by receptor tyrosine kinases. Inhibiting these pathways may result in higher intracellular availability of PIP2. This could be the case for EGFR inhibitors like gefitinib and erlotinib, since a downstream messenger of EGFR is PLC-g, which hydrolyses PIP2 into diacylglycerol (DAG) and IP3 [36,37]. Decreased hydrolysing of PIP2 would lead to higher availability. Another downstream pathway of EGFR is the phosphoinositide 3-kinase (PI3K) pathway, in which PIP2 is also a second messenger. EGFR inhibition of the PI3K pathway may result in less phosphorylation of PIP2 and thereby to higher levels of PIP2 [38,39]. Higher levels of PIP2 could result in increased signal transduction/lower thresholds for specifically sweet, bitter and umami taste. PKIs may also affect taste as a result of a direct stimulatory effect on the taste receptor. In chemotherapy, this has been proposed as a potential mechanism, either by secretion of the drug into the saliva or diffusion from capillaries. This type of taste alteration has been described as a metallic, ‘chemical’ or bitter sensation during chemotherapy [5,6]. Because most PKIs are continuously administered, this would probably cause a continuous chemical drug taste from start of treatment until discontinuation. After drug clearance, taste or smell abnormalities may persist due to damage to the receptor cells or to the afferent neuronal cells. This forms a vicious circle with cancer anorexia that leads to decreased food intake which in turn has been correlated with an increased risk of oral toxicity [40]. Furthermore, PKIs may specifically impair the function of the taste receptors for sweet, bitter and umami, via direct effect on the signalling G protein, gustducin. This protein shows structural and functional similarity to transducin, a G protein crucial for the sense of sight [41,42]. Treatment with targeted agents is often accompanied by ocular toxicities [43,44], hinting at the alikeness of the underlying pathobiology. Another hypothesis to explain PKI-induced taste alterations is by affected renewal of taste receptor cells.

131

Differentiation of progenitor cells into specific receptor cells mainly depends on b-catenin levels [17].b-catenin dynamics may be influenced by several PKIs [45]. For instance, inhibition of fms-like tyrosine kinase receptor3 (FLT3) prevents dissociation of b-catenin from its complex with E-cadherin, thereby reducing b-catenin signalling. FLT3 inhibitor imatinib has been shown to reduce Wnt-b-catenin signalling and this could also be the case for other FLT3 inhibitors, like sorafenib and sunitinib [46]. Reduced b-catenin signalling may also be caused by PKIs targeting EGFR, fibroblast growth factor receptor (FGFR) and VEGFR. The downstream pathway of these receptors includes the PI3K-AKT pathway, which has a stimulatory effect on b-catenin [47,48]. Therefore, inhibition of these receptors, for instance by sunitinib, sorafenib or imatinib, may inhibit b-catenin [49] and influence taste cell renewal. Lower bcatenin levels may lead to less differentiation of progenitor cells into taste cell precursors and/or differentiation into other proportions of type I, II and III taste cells. In case of less differentiation into taste cells, all five taste qualities would be affected, while in case of other proportions of the cell types, sweet, bitter and umami (type II cells) would be more affected that sour and salty taste (type III cells), because for differentiation into type II cells higher levels of b-catenin are needed [17]. Decreased renewal of receptor cells may also be caused by PKIs targeting sonic hedgehog. Sonic hedgehog is expressed by taste cell precursors and plays a role in normal taste cell renewal. Vismodegib inhibits tumour cell proliferation by targeting the sonic hedgehog-pathway [11]. Blockade of this pathway could also inhibit differentiation of taste cell precursors into taste cells and hereby cause taste loss of all five taste qualities [17,50]. 6.3. Smell alteration by affecting olfactory receptor neurons Patient-reported taste alterations may also be a result of smell alteration. Olfactory signal transduction is mediated by protein kinase C. This kinase phosphorylates the cAMP-gated cation-selective channel, increasing its cAMP sensitivity and stimulating olfactory signal transduction. Protein kinase C may be inhibited by PKIs, as has been shown for imatinib [51]. Hypothetically, inhibition of protein kinase C would result in reduced olfactory transduction and/or higher threshold values, which would be likely to occur shortly after initiation of therapy. Another kinase involved in olfactory signal transduction is Ca2þ/calmodulin kinase II (CaMKII), an inhibitor of adenylate cyclase involved in termination of olfactory signalling [26]. Inhibition of CaMKII may reduce termination of olfactory signalling, causing a persistent olfactory signal transduction. There are no PKIs primarily targeting CaMKII, although off target effects have been reported, for instance for sunitinib [30].

132

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

Next to reduced olfactory signal transduction, smell alteration may be caused by reduced renewal of olfactory receptor neurons. The cells that are believed to be stem cells in olfactory epithelium (horizontal cells) are responsive to EGF [27]. Treatment with EGFRinhibitors may lead to blockade of downstream signalling and therefore to less differentiation of these stem cells into olfactory receptor neurons. It can be assumed that a reduced number results in less odorant stimulation of olfactory receptor neurons and therefore to higher smell perception thresholds. 6.4. Neurodegeneration In addition to local, mechanistic causes, taste alterations could be explained by a detrimental effect of PKIs on the neurons and the signal transduction. In vivo data have already demonstrated the degenerative effect of sunitinib on neurons, where impaired VEGFR2 signalling was shown to contribute to increased apoptotic cell death and subsequent neurodegeneration [52]. Abnormal neuronal activity may result in specific taste sensation independent of taste receptor stimulation [5,6,31]. Furthermore, the primary somatic sensory cortex, collecting sensory information from taste buds, is located in the parietal lobe. The reversible posterior leukoencephalopathy syndrome has been reported with some PKIs and it usually affects parietal/occipital lobe [53]. By extrapolation, one could assume that PKI treatment could perhaps result in a more generalised neuronal disturbance directly at the cortex level, ultimately affecting taste perception. 7. Discussion Patient-reported taste alterations are a common sideeffect of treatment with PKIs. To our knowledge, it has never been established whether an objective taste dysfunction develops during PKI treatment, or whether these patient-reported symptoms could have an alternative explanation, for example as a consequence of smell alterations. Fact remains that taste alterations constitute an underrecognised and understudied sideeffect. Especially in the context of chronic drug administration, taste alterations may result in significant morbidity, as they are associated with weight loss and impaired quality of life, all potentially compromising treatment adherence. However, lack of clarity on the underlying pathobiology hampers the search for identifying potential treatment options. We herein described the physiology of taste and smell and the mechanism of action of PKIs, ultimately proposing potential explanations of PKI-induced patientreported taste alterations. The potential explanations described here would lead to specific objective changes. It is important to further explore whether patientreported taste alterations are caused by objective

changes in taste, smell, both or also by other factors. Future studies should describe patient-reported taste alterations in a more detailed manner and also assess taste and smell function objectively, in order to provide a better understanding of the underlying pathobiology. This may help to develop treatment options in order to prevent or overcome these symptoms. Conflict of interest statement HV receives research grants for the institution from Vitromics Healthcare System, Merck, Immunovo B.V., non-financial support from Pfizer. These resources were all received outside the submitted work. The other authors declare no competing interests.

Acknowledgements This work was supported by a grant from Alpe d’HuZes/Dutch Cancer Society (VU 2011e5262).

References [1] Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med 2005;353:172e87. [2] Eckstein N, Roper L, Haas B, Potthast H, Hermes U, Unkrig C, et al. Clinical pharmacology of tyrosine kinase inhibitors becoming generic drugs: the regulatory perspective. J Exp Clin Cancer Res 2014;33:15. [3] McDermott U, Settleman J. Personalized cancer therapy with selective kinase inhibitors: an emerging paradigm in medical oncology. J Clin Oncol 2009;27:5650e9. [4] Gotink KJ, Verheul HM. Anti-angiogenic tyrosine kinase inhibitors: what is their mechanism of action? Angiogenesis 2010;13: 1e14. [5] Epstein JB, Barasch A. Taste disorders in cancer patients: pathogenesis, and approach to assessment and management. Oral Oncol 2010;46:77e81. [6] Hovan AJ, Williams PM, Stevenson-Moore P, Wahlin YB, Ohrn KE, Elting LS, et al. A systematic review of dysgeusia induced by cancer therapies. Support Care Cancer 2010;18:1081e7. [7] Bressan V, Stevanin S, Bianchi M, Aleo G, Bagnasco A, Sasso L. The effects of swallowing disorders, dysgeusia, oral mucositis and xerostomia on nutritional status, oral intake and weight loss in head and neck cancer patients: a systematic review. Cancer Treat Rev 2016;45:105e19. [8] Spotten LE, Corish CA, Lorton CM, Ui Dhuibhir PM, O’Donoghue NC, O’Connor B, et al. Subjective and objective taste and smell changes in cancer. Ann Oncol 2017;28:969e84. [9] Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 2006; 368:1329e38. [10] Ferrari V, Sgotti D, Amoroso V, Valcamonico F, Grisanti S, Consoli F, et al. Oral mucositis a side effect in Tyrosin-Kinase Inhibitor Therapy (Sunitinib): the role of assessment of symptoms in evaluation of toxicity. Eur J Cancer 2009:195. [11] Sekulic A, Migden MR, Oro AE, Dirix L, Lewis KD, Hainsworth JD, et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N Engl J Med 2012;366:2171e9. [12] Tang JY, Mackay-Wiggan JM, Aszterbaum M, Yauch RL, Lindgren J, Chang K, et al. Inhibiting the hedgehog pathway in

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

[13]

[14]

[15]

[16] [17] [18]

[19] [20] [21]

[22] [23] [24]

[25]

[26]

[27]

[28]

[29] [30]

[31]

[32]

[33]

[34]

patients with the basal-cell nevus syndrome. N Engl J Med 2012; 366:2180e8. Lee WJ, Lee JL, Chang SE, Lee MW, Kang YK, Choi JH, et al. Cutaneous adverse effects in patients treated with the multitargeted kinase inhibitors sorafenib and sunitinib. Br J Dermatol 2009;161:1045e51. Boers-Doets CB, Epstein JB, Raber-Durlacher JE, Ouwerkerk J, Logan RM, Brakenhoff JA, et al. Oral adverse events associated with tyrosine kinase and mammalian target of rapamycin inhibitors in renal cell carcinoma: a structured literature review. Oncologist 2012;17:135e44. Watters AL, Epstein JB, Agulnik M. Oral complications of targeted cancer therapies: a narrative literature review. Oral Oncol 2011;47:441e8. Chandrashekar J, Hoon MA, Ryba NJ, Zuker CS. The receptors and cells for mammalian taste. Nature 2006;444:288e94. Barlow LA. Progress and renewal in gustation: new insights into taste bud development. Development 2015;142:3620e9. de Araujo IE, Rolls ET, Kringelbach ML, McGlone F, Phillips N. Taste-olfactory convergence, and the representation of the pleasantness of flavour, in the human brain. Eur J Neurosci 2003; 18:2059e68. Chaudhari N. Synaptic communication and signal processing among sensory cells in taste buds. J Physiol 2014;592:3387e92. Roper SD. Signal transduction and information processing in mammalian taste buds. Pflugers Arch 2007;454:759e76. Heath TP, Melichar JK, Nutt DJ, Donaldson LF. Human taste thresholds are modulated by serotonin and noradrenaline. J Neurosci 2006;26:12664e71. Perea-Martinez I, Nagai T, Chaudhari N. Functional cell types in taste buds have distinct longevities. PLoS One 2013;8, e53399. Angers S, Moon RT. Proximal events in Wnt signal transduction. Nat Rev Mol Cell Biol 2009;10:468e77. van Hartevelt TJ, Kringelbach ML. The Olfactory System. The human nervous system. 3rd ed. Academic Press; 2011. p. 1219e38. Muller F, Bonigk W, Sesti F, Frings S. Phosphorylation of mammalian olfactory cyclic nucleotide-gated channels increases ligand sensitivity. J Neurosci 1998;18:164e73. Wei J, Zhao AZ, Chan GC, Baker LP, Impey S, Beavo JA, et al. Phosphorylation and inhibition of olfactory adenylyl cyclase by CaM kinase II in neurons: a mechanism for attenuation of olfactory signals. Neuron 1998;21:495e504. Beites CL, Kawauchi S, Crocker CE, Calof AL. Identification and molecular regulation of neural stem cells in the olfactory epithelium. Exp Cell Res 2005;306:309e16. Gharwan H, Groninger H. Kinase inhibitors and monoclonal antibodies in oncology: clinical implications. Nat Rev Clin Oncol 2016;13:209e27. Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer 2009;9:28e39. Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE, Campbell BT, et al. A quantitative analysis of kinase inhibitor selectivity. Nat Biotechnol 2008;26:127e32. Hong JH, Omur-Ozbek P, Stanek BT, Dietrich AM, Duncan SE, Lee YW, et al. Taste and odor abnormalities in cancer patients. J Support Oncol 2009;7:58e65. Epstein JB, Thariat J, Bensadoun RJ, Barasch A, Murphy BA, Kolnick L, et al. Oral complications of cancer and cancer therapy: from cancer treatment to survivorship. CA Cancer J Clin 2012;62: 400e22. Bankvall M, Sjoberg F, Gale G, Wold A, Jontell M, Ostman S. The oral microbiota of patients with recurrent aphthous stomatitis. J Oral Microbiol 2014;6:25739. Hijazi K, Lowe T, Meharg C, Berry SH, Foley J, Hold GL. Mucosal microbiome in patients with recurrent aphthous stomatitis. J Dent Res 2015;94:87Se94S.

133

[35] Murtaza B, Hichami A, Khan AS, Ghiringhelli F, Khan NA. Alteration in taste perception in cancer: causes and strategies of treatment. Front Physiol 2017;8:134. [36] Longo DL. Cancer cell biology and angiogenesis. In: Longo DL, Kasper DL, Jameson JL, Fauci AS, Hauser SL, Loscalzo J, editors. Harrison’s hematology and oncology. 2nd ed. New York: McGraw-Hill Education; 2013. p. 312e32. [37] Nyati MK, Morgan MA, Feng FY, Lawrence TS. Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer 2006; 6:876e85. [38] Yuan TL, Cantley LC. PI3K pathway alterations in cancer: variations on a theme. Oncogene 2008;27:5497e510. [39] Zhao ZQ, Yu ZY, Li J, Ouyang XN. Gefitinib induces lung cancer cell autophagy and apoptosis via blockade of the PI3K/AKT/mTOR pathway. Oncol Lett 2016;12:63e8. [40] Peterson DE, O’Shaughnessy JA, Rugo HS, Elad S, Schubert MM, Viet CT, et al. Oral mucosal injury caused by mammalian target of rapamycin inhibitors: emerging perspectives on pathobiology and impact on clinical practice. Cancer Med 2016;5:1897e907. [41] He W, Yasumatsu K, Varadarajan V, Yamada A, Lem J, Ninomiya Y, et al. Umami taste responses are mediated by alphatransducin and alpha-gustducin. J Neurosci 2004;24:7674e80. [42] McLaughlin SK, McKinnon PJ, Margolskee RF. Gustducin is a taste-cell-specific G protein closely related to the transducins. Nature 1992;357:563e9. [43] Leisy H, Ahmad M, Smith RT. Photopsias during systemic bevacizumab therapy. Case Rep Ophthalmol Med 2016;2016: 1926178. [44] Huillard O, Bakalian S, Levy C, Desjardins L, Lumbroso-Le Rouic L, Pop S, et al. Ocular adverse events of molecularly targeted agents approved in solid tumours: a systematic review. Eur J Cancer 2014;50:638e48. [45] Kahn M. Can we safely target the WNT pathway? Nat Rev Drug Discov 2014;13:513e32. [46] Zhou L, An N, Haydon RC, Zhou Q, Cheng H, Peng Y, et al. Tyrosine kinase inhibitor STI-571/Gleevec down-regulates the beta-catenin signaling activity. Cancer Lett 2003;193:161e70. [47] Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol 2014; 15:178e96. [48] Zhang W, Zhang H, Wang N, Zhao C, Zhang H, Deng F, et al. Modulation of beta-catenin signaling by the inhibitors of MAP kinase, tyrosine kinase, and PI3-kinase pathways. Int J Med Sci 2013;10:1888e98. [49] Ott PA, Adams S. Small-molecule protein kinase inhibitors and their effects on the immune system: implications for cancer treatment. Immunotherapy 2011;3:213e27. [50] Miura H, Kusakabe Y, Sugiyama C, Kawamatsu M, Ninomiya Y, Motoyama J, et al. Shh and Ptc are associated with taste bud maintenance in the adult mouse. Mech Dev 2001;106: 143e5. [51] Deininger MW, Druker BJ. Specific targeted therapy of chronic myelogenous leukemia with imatinib. Pharmacol Rev 2003;55: 401e23. [52] Abdel-Aziz AK, Mantawy EM, Said RS, Helwa R. The tyrosine kinase inhibitor, sunitinib malate, induces cognitive impairment in vivo via dysregulating VEGFR signaling, apoptotic and autophagic machineries. Exp Neurol 2016;283:129e41. [53] Hadj JO, Braven RD, Tillier C, Schrijver HM, Verheul HM, HJ VDV. Reversible posterior leukoencephalopathy syndrome during sunitinib therapy for metastatic renal cell carcinoma. Oncol Lett 2012;3:1293e6. [54] Demetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts PJ, et al. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. N Engl J Med 2002;347:472e80.

134

A. van der Werf et al. / European Journal of Cancer 86 (2017) 125e134

[55] Cohen MH, Cortazar P, Justice R, Pazdur R. Approval summary: imatinib mesylate in the adjuvant treatment of malignant gastrointestinal stromal tumors. Oncologist 2010;15:300e7. [56] Morishima Y, Ogura M, Nishimura M, Yazaki F, Bessho M, Mizoguchi H, et al. Efficacy and safety of imatinib mesylate for patients in the first chronic phase of chronic myeloid leukemia: results of a Japanese phase II clinical study. Int J Hematol 2004;80:261e6. [57] Lassman AB, Pugh SL, Gilbert MR, Aldape KD, Geinoz S, Beumer JH, et al. Phase 2 trial of dasatinib in target-selected patients with recurrent glioblastoma (RTOG 0627). Neuro Oncol 2015;17:992e8. [58] Minemura H, Yokouchi H, Azuma K, Hirai K, Sekine S, Oshima K, et al. A phase II trial of erlotinib monotherapy for pretreated elderly patients with advanced EGFR wild-type nonsmall cell lung cancer. BMC Res Notes 2015;8:220. [59] Dawson NA, Guo C, Zak R, Dorsey B, Smoot J, Wong J, et al. A phase II trial of gefitinib (Iressa, ZD1839) in stage IV and recurrent renal cell carcinoma. Clin Cancer Res 2004;10:7812e9. [60] Uemura H, Shinohara N, Yuasa T, Tomita Y, Fujimoto H, Niwakawa M, et al. A phase II study of sunitinib in Japanese patients with metastatic renal cell carcinoma: insights into the treatment, efficacy and safety. Jpn J Clin Oncol 2010;40:194e202. [61] Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:115e24. [62] Kollmannsberger C, Soulieres D, Wong R, Scalera A, Gaspo R, Bjarnason G. Sunitinib therapy for metastatic renal cell carcinoma: recommendations for management of side effects. Can Urol Assoc J 2007;1:S41e54. [63] Zhao J, Zhu Y, Zhang C, Wang X, He H, Wang H, et al. Sorafenib or sunitinib as postoperative adjuvant therapy for Chinese patients with locally advanced clear cell renal cell carcinoma at high risk for disease recurrence. Urol Oncol 2013;31:1800e5. [64] Kulke MH, Lenz HJ, Meropol NJ, Posey J, Ryan DP, Picus J, et al. Activity of sunitinib in patients with advanced neuroendocrine tumors. J Clin Oncol 2008;26:3403e10. [65] Kloos RT, Ringel MD, Knopp MV, Hall NC, King M, Stevens R, et al. Phase II trial of sorafenib in metastatic thyroid cancer. J Clin Oncol 2009;27:1675e84. [66] Lam ET, Ringel MD, Kloos RT, Prior TW, Knopp MV, Liang J, et al. Phase II clinical trial of sorafenib in metastatic medullary thyroid cancer. J Clin Oncol 2010;28:2323e30. [67] Locati LD, Perrone F, Cortelazzi B, Bergamini C, Bossi P, Civelli E, et al. A phase II study of sorafenib in recurrent and/or

[68]

[69]

[70]

[71]

[72]

[73]

[74]

[75]

[76]

[77]

metastatic salivary gland carcinomas: translational analyses and clinical impact. Eur J Cancer 2016;69:158e65. Motzer RJ, Escudier B, Tomczak P, Hutson TE, Michaelson MD, Negrier S, et al. Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol 2013;14:552e62. Motzer RJ, Porta C, Vogelzang NJ, Sternberg CN, Szczylik C, Zolnierek J, et al. Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial. Lancet Oncol 2014;15: 286e96. Sofen H, Gross KG, Goldberg LH, Sharata H, Hamilton TK, Egbert B, et al. A phase II, multicenter, open-label, 3-cohort trial evaluating the efficacy and safety of vismodegib in operable basal cell carcinoma. J Am Acad Dermatol 2015;73:99e105e1. LoRusso PM, Rudin CM, Reddy JC, Tibes R, Weiss GJ, Borad MJ, et al. Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors. Clin Cancer Res 2011;17:2502e11. Kaye SB, Fehrenbacher L, Holloway R, Amit A, Karlan B, Slomovitz B, et al. A phase II, randomized, placebo-controlled study of vismodegib as maintenance therapy in patients with ovarian cancer in second or third complete remission. Clin Cancer Res 2012;18:6509e18. Camidge DR, Theodoro M, Maxson DA, Skokan M, O’Brien T, Lu X, et al. Correlations between the percentage of tumor cells showing an anaplastic lymphoma kinase (ALK) gene rearrangement, ALK signal copy number, and response to crizotinib therapy in ALK fluorescence in situ hybridization-positive nonsmall cell lung cancer. Cancer 2012;118:4486e94. Shaw AT, Kim DW, Nakagawa K, Seto T, Crino L, Ahn MJ, et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N Engl J Med 2013;368:2385e94. Kim DW, Ahn MJ, Shi MM, DePas TM, Yang PC, Riely GJ, et al. Results of a global phase II study with crizotinib in advanced ALK-positive non-small cell lung cancer. J Clin Oncol 2012;30 (suppl; abstr 7533). Aroniadou-Anderjaska V, Ennis M, Shipley MT. Glomerular synaptic responses to olfactory nerve input in rat olfactory bulb slices. Neuroscience 1997;79:425e34. Chen WR, Xiong W, Shepherd GM. Analysis of relations between NMDA receptors and GABA release at olfactory bulb reciprocal synapses. Neuron 2000;25:625e33.