Skip to main content
Top

01-12-2016 | Nausea and vomiting | Book chapter | Article

3. First-Generation 5-HT3 Receptor Antagonists

Authors: Roy Chen, MD, Kathy Deng, MD, Harry Raftopoulos, MD

Publisher: Springer International Publishing

Abstract

Chemotherapy-induced nausea and vomiting (CINV) is a significant side effect of cancer therapy and can lead to poor compliance with therapy, treatment delays, dehydration, hospitalization, and a marked decrement in patient quality of life. With appropriate CINV control, safe outpatient administration of chemotherapy can be accomplished with no change in patients’ pre-therapy quality of life. Over the last 30 years, developments have improved in the control of CINV, including the advent of 5-hydroxytryptamine-3 (5HT-3) receptor antagonists which are an integral ingredient in regimens used today. With the variety of chemotherapy regimens today and the ongoing development of new combinations in addition to targeted therapy, there have been corresponding dynamic goals for control of not only CINV in general but in differentiating control of nausea over and above that of vomiting. In fact, current antiemetic therapy conceptually fulfills the true definition of “targeted therapy” as there is significant understanding of pathways involved in emesis as well as specific targeted antagonists to these pathways. This chapter will focus on the 5HT-3 pathway and the specific receptor antagonists to this pathway.

3.1 Introduction

Chemotherapy-induced nausea and vomiting (CINV) is a significant side effect of cancer therapy and can lead to poor compliance with therapy, treatment delays, dehydration, hospitalization, and a marked decrement in patient quality of life. With appropriate CINV control, safe outpatient administration of chemotherapy can be accomplished with no change in patients’ pre-therapy quality of life. Over the last 30 years, developments have improved in the control of CINV, including the advent of 5-hydroxytryptamine-3 (5-HT3) receptor antagonists which are an integral ingredient in regimens used today. With the variety of chemotherapy regimens today and the ongoing development of new combinations in addition to targeted therapy, there have been corresponding dynamic goals for control of not only CINV in general but in differentiating control of nausea over and above that of vomiting. In fact, current antiemetic therapy conceptually fulfills the true definition of “targeted therapy” as there is significant understanding of pathways involved in emesis as well as specific targeted antagonists to these pathways. This chapter will focus on the 5-HT3 pathway and the specific receptor antagonists to this pathway.

3.2 Physiology of CINV

The intrinsic emetogenicity of a given chemotherapeutic agent is the key determinant of the probability of clinical emesis, although chemotherapy dose, as well as patient factors such as female gender and young age may increase the probability of emesis [1]. Guideline groups divide chemotherapeutic agents into four emetogenic groups: high, moderate, low, and minimum [2] (Table 3.1). The clinical phases of emesis have been defined largely from observations using cisplatin where typically in the absence of antiemetics all patients will experience nausea and vomiting 1–2 h post administration. At 18–24 h the CINV abates, only to resurface and peak again at 48–72 h after the cisplatin administration [3]. As a result, acute emesis is defined as occurring within the first 24 h and delayed after 24 h (usually up to 120 h) [2]. Other agents may also cause delayed emesis, although not usually with the same biphasic pattern as cisplatin.
Table 3.1
Emetogenic potential of antineoplastic agents
Highly emetogenic (IV)
AC (doxorubicin or epirubicin with cyclophosphamide)
Dacarbazine
Ifosfamide >2 g/m2per dose
Carmustine >250 mg/m2
Doxorubicin >60 mg/m2
Mechlorethamine
Cisplatin
Epirubicin >90 mg/m2
Streptozocin
Cyclophosphamide >1,500 mg/m2
  
Moderately emetogenic (IV)
Aldesleukin >12–15 million IU/m2
Clofarabine
Ifosfamide <2 g/m2per dose
Amifostine >300 mg/m2
Cyclophosphamide <1,500 mg/m2
Interferon alfa >10 million IU/m2
Arsenic trioxide
Cytarabine >200 mg/m2
Irinotecan
Azacitidine
Dactinomycin
Melphalan
Bendamustine
Daunorubicin
Methotrexate >250mg/m2
Busulfan
Doxorubicin <60 mg/m2
Oxaliplatin
Carboplatin
Epirubicin <90 mg/m2
Temozolomide
Carmustine < 250 mg/m2
Idarubicin
 
Low emetic risk (IV)
Ado-trastuzumab emtansine
Etoposide
Paclitaxel
Amifostine <300 mg
5-fluorouracil
Paclitaxel-albumin
Aldesleukin <12 million IU/m2
Floxuridine
Pemetrexed
Brentuximab vedotin
Interferon alfa >5 <10 million IU/m2
Pentostatin
Cabazitaxel
Ixabepilone
Pralatrexate
Carfilzomib
Methotrexate >50mg/m2 <250 mg/m2
Romidepsin
Cytarabine 100–200mg/m2
Mitomycin
Thiotepa
Docetaxel
Mitoxantrone
Topotecan
Doxorubicin (liposomal)
Omacetaxine
Ziv-aflibercept
Eribulin
  
Minimal emetic risk (IV)
Alemtuzumab
Dexrazoxane
Pertuzumab
Asparaginase
Fludarabine
Rituximab
Bevacizumab
Interferon alfa <5 million IU/m2
Temsirolimus
Bleomycin
Ipilimumab
Trastuzumab
Bortezomib
Methotrexate <50 mg/m2
Valrubicin
Cetuximab
Nelarabine
Vinblastine
Cladribine
Ofatumumab
Vincristine
Cytarabine <100 mg/m2
Panitumumab
Vincristine (liposomal)
Decitabine
Pegaspargase
Vinorelbine
Denileukin diftitox
Peginterferon
 
Moderate to high emetic risk (oral)
Altretamine
Estramustine
Procarbazine
Busulfan >4 mg/day
Etoposide
Temozolomide >75mg/m2/day
Crizotinib
Lomustine (single day)
Vismodegib
Cyclophosphamide >100 mg/m2/day
Mitotane
 
Minimal to low emetic risk (oral)
Axitinib
Gefitinib
Ruxolitinib
Bexarotene
Hydroxyurea
Sorafenib
Bosutinib
Imatinib
Sunitinib
Busulfan <4 mg/day
Lapatinib
Temozolomide <75mg/m2/day
Cabozantinib
Lenalidomide
Thalidomide
Capecitabine
Melphalan
Thioguanine
Chlorambucil
Mercaptopurine
Topotecan
Cyclophosphamide <100 mg/m2/day
Methotrexate
Trametinib
Dasatinib
Nilotinib
Tretinoin
Dabrafenib
Pazopanib
Vandetanib
Erlotinib
Pomalidomide
Vemurafenib
Everolimus
Ponatinib
Vorinostat
Fludarabine
Regorafenib
 
Animal studies have defined significant neuroanatomic components of the emetic reflex. Initially, Thumas proposed a single vomiting center (dorsal vagal nucleus) in 1891 based on canine studies [4]; Wang and Borison further refined the concept to include a “sensor” area in the area postrema (often called the chemoreceptor trigger zone) and an “effector” area, the vomiting center, in the medulla [5]. More recent studies suggest two areas of afferent input in the dorsal vagal nucleus, the area postrema and the nucleus tractus solitarius, and rather than a discrete “vomiting center,” several neuronal areas loosely organized to effect the emetic reflex, termed the “central pattern generator” [6].
The neurotransmitters involved in the emetic reflex related to chemotherapy are underpinned by 5-hydroxytryptamine (5-HT). Chemotherapy administration leads to local release of mediators by enterochromaffin cells in the proximal small intestine. These mediators include 5-HT, substance P, and cholecystokinin, which then bind to respective receptors (5-HT3, neurokinin-1, and cholecystokinin-1) located on terminal ends of abdominal vagal efferents [7]. This binding leads to a signal conducted by these vagal fibers which terminates in the nucleus tractus solitarius and leads to activation of the central pattern generator. The local release of 5-HT in the gastrointestinal tract and signal transduction by the vagal afferents is thought to be the chief mechanism whereby chemotherapeutic agents cause emesis [7]. Direct stimulation of the chemoreceptor trigger zone as described by Wang and Borison is a further mechanism, albeit thought to be a less important mechanism with CINV [8].

3.3 Development of First-Generation 5-HT3 Antagonists

In 1985, studies showed that high-dose metoclopramide (a dopamine-2 receptor antagonist) combined with dexamethasone provided meaningful protection from cisplatin-induced emesis at the expense of significant extrapyramidal side effects [9]. Curiously, at that time, results could not be replicated with other dopamine-2 receptor antagonists, suggesting the antiemetic effect was mediated by interaction with another neurotransmitter receptor. The 5-HT3 receptor emerged as a likely mediator of the antiemetic effect, and the pharmaceutical industry employed various strategies to develop selective 5-HT3 receptor antagonists. These included:
(a)
Screening indole analogues, leading to the development of ondansetron [10]
 
(b)
Structure-activity relationships around cocaine resulting in the development of dolasetron [11]
 
(c)
Using serotonin as a basis, yielding tropisetron [12]
 
(d)
Structure-activity relationships around tropisetron developed granisetron [13]
 
The structures of the four first-generation 5-HT3 antagonists are shown in Fig. 3.1 [1417]. Since ondansetron and granisetron are the most used agents with dolasetron no longer indicated for CINV and tropisetron in limited use, a greater focus will be devoted to ondansetron and granisetron. The role of 5-HT3 receptor antagonists in postoperative nausea and vomiting is beyond the scope of this text.

3.3.1 Ondansetron

Ondansetron is an indole derivative, selective 5-HT3 receptor antagonist with weak affinity for other 5-HT receptors and dopamine receptors. Oral formulations are rapidly absorbed with an approximate 60 % bioavailability [18]; ondansetron is extensively metabolized primarily by hydroxylation of the indole ring with subsequent conjugation [19]. The elimination half-life is approximately 4 h (Table 3.2); metabolites are not significant contributors to activity. The elderly and patients with hepatic impairment show reduced clearance; however, this is not clinically meaningful [19].
Table 3.2
5-HT3 receptor antagonist characteristics
5-HT3 receptor antagonist
Half-life (hours)
Oral bioavailability (%)
QTc prolongation
First generation
Ondansetron
4
59
Yes
Dolasetron (hydrodolasetron)
7.3
76
Yes
Tropisetron
8
60
Yes
Granisetron
9
60
Yes
Second generation
Palonosetron
42
97
No
Adverse events with ondansetron are largely confined to headache and constipation. An EKG study conducted in 2012 demonstrated significant QTc prolongation with a 32 mg dose of ondansetron prompting the FDA to limit the dose administered intravenously to 16 mg [20].

3.3.2 Dolasetron

Dolasetron is a selective 5-HT3 receptor antagonist derived after extensive chemical substitutions to the cocaine molecule. Dolasetron is rapidly metabolized to the active metabolite hydroxydolasetron, which is predominantly excreted in the urine and has an elimination half-life of 7.3 h (Table 3.2) [21]. Renal impairment increases the elimination time, but is once again not clinically relevant.
As with ondansetron, headache and constipation are the most significant adverse effects. QTc prolongation is also evident at higher doses, and in 2010, due to the QTc concerns, the approval for intravenous dolasetron in CINV was withdrawn.

3.3.3 Tropisetron

Tropisetron is another selective competitive antagonist of the 5-HT3 receptor, derived by chemical modification of serotonin (5-HT) and thus is also an indole derivative. Oral absorption is rapid with approximately 60 % bioavailability; metabolism is similar to ondansetron (hydroxylation followed by conjugation), with metabolites being inactive [22]. In most patients, the elimination half-life is approximately 8 h (Table 3.2), although in some poor metabolizers, the half-life may be up to 45 h [23]. Tropisetron is not available in the USA, but is used in the East and Australia.

3.3.4 Granisetron

Granisetron is a selective 5-HT3 receptor antagonist derived by making chemical alterations to tropisetron. Oral bioavailability is similar to other agents and metabolism is the primary means of elimination. The elimination half-life is approximately 9 h (Table 3.2), with no significant changes noted in elderly and those with renal or hepatic impairment [24]. In addition to the intravenous and oral formulations, a transdermal delivery system (patch) is available in the USA, delivering granisetron directly through intact skin by passive diffusion with levels peaking at 48 h and sustained for further 5 days. Adverse events once again include headache and constipation and reports of QTc prolongation.

3.4 Development of Palonosetron

Chemical strategies that examined conformational alterations in 5-HT3 receptor antagonists led to compounds with significantly increased affinity for the receptor and further alterations led to the development of palonosetron, deemed a second-generation 5-HT3 antagonist for its enhanced receptor binding affinity and prolonged half-life (approximately 42 h) (Table 3.2) [25]. The structure of palonosetron is shown in Fig. 3.1 [26]. In addition, distinguishing palonosetron from first-generation agents, it does not appear to have a meaningful effect on QTc [27]. Oral bioavailability is excellent at 97 % [28].

3.5 Clinical Studies

Over the last 30 years, numerous clinical studies have been conducted in the CINV space that have established the benefits of first-generation 5-HT3 receptor antagonists as well as have helped to refine dosing and steroid combinations. During the same period, standards and definitions as far as end points and study design have also evolved, such that initial studies had more variability in terms of measured end points and use of patient-reported outcomes. Currently, important definitions include the acute phase (0–24 h post emetogenic chemotherapy), delayed phase (24–120 h post), complete response (no emesis and no use of rescue medication), and complete control (no emesis and no more than minimal nausea) [1]. While not perfect, these standard definitions allow studies to be interpreted in context and determine whether any differences seen are meaningful. In addition, standardization has assisted in the development of meaningful guidelines to help translate clinical trial benefits to global benefits.

3.5.1 Initial Studies

3.5.1.1 Placebo-Controlled Studies

Two studies compared the efficacy of ondansetron and granisetron to placebo in preventing cisplatin-induced emesis. Cubeddu [29] randomized 28 chemotherapy-naive patients about to receive cisplatin to ondansetron or placebo as antiemetic prophylaxis. The control of emesis was significantly improved in the active treatment arm in terms of number of emetic episodes, time to emesis, and need for rescue medication. Cupissol [30] similarly randomized 28 chemotherapy-naive patients to receive granisetron or placebo with their first dose of cisplatin. Once again, the active therapy group had significant control of emesis – 13 of 14 granisetron patients had no acute nausea or vomiting whereas only 1 of 14 placebo patients was free of nausea or vomiting in the first 24 h. In patients receiving cyclophosphamide (at that time deemed a moderately emetogenic agent irrespective of the combination), Cubeddu randomized 20 patients to ondansetron or placebo [31] and predictably 70 % of ondansetron-treated patients experienced no emesis, compared to 0 % in the placebo arm. Two further studies in Japan used a placebo design to confirm the activity of ondansetron and tropisetron [32, 33]. The continued use of placebo controls was strongly discouraged except for studies involving low emetogenic risk agents, and further studies needed to compare new treatments with the best available existing therapy [34]. An individual patient data meta-analysis [35] reinforced the dismal outcome in terms of emesis control in placebo-treated patients.

3.5.1.2 Dose-Finding Studies

Principles of cancer supportive care, unlike primary therapy, dictate that the lowest effective dose of an agent can be used rather than the maximally tolerated dose: many initial studies sought to define optimal dosing of the first-generation 5-HT3 receptor antagonists. In patients receiving cisplatin, a double-blind trial of three different ondansetron doses (0.015, 0.15, and 0.30 mg/kg, each given three times 4 h apart) demonstrated the 0.15 mg/kg dose was superior to the lower dose, and no added improvement with the higher dose was noted [36]. Granisetron was also evaluated in patients receiving cisplatin in a double-blind study [37]. Doses evaluated were 2, 10, and 40 μg/kg with the 10 and 40 μg/kg doses being superior to the lower dose in terms of preventing cisplatin-induced CINV. There was no difference observed between the 10 and 40 μg/kg doses. A second study examined granisetron doses of 40 μg/kg or 160 μg/kg and found no differences in efficacy [38]. Similarly studies with tropisetron and dolasetron [39, 40] confirmed intermediate doses as effective in preventing cisplatin-induced CINV with no added benefit to dose escalation. Importantly, though, even at much higher doses than required for maximum efficacy, adverse events were seldom significantly increased.

3.5.1.3 Comparison Studies with Older Agents

Ondansetron (0.15-mg/kg × 3 doses) was compared with high-dose metoclopramide (2 mg/kg × 6 doses) in a single-blind trial in 307 patients receiving high-dose cisplatin [41]. Ondansetron was found to be superior to metoclopramide and produced fewer adverse events; in particular no extrapyramidal effects were noted with ondansetron. Similar findings were seen with two earlier smaller studies; however, the schedule of ondansetron administration was not standard (continuous infusion) [42, 43].
Granisetron trials provided clues that helped the understanding of the neurophysiology of emesis and the pharmacology of the antiemetics: granisetron alone was superior to either chlorpromazine or prochlorpromazine given together with dexamethasone for moderately emetogenic chemotherapy [44, 45]; yet when granisetron alone was compared to high-dose metoclopramide with dexamethasone for high-dose cisplatin, no differences in antiemetic control could be discerned [45, 46].
These observations validated the original findings by Gralla using metoclopramide and confirming the role of the 5-HT3 receptor in emesis. In addition clues about the utility of corticosteroids emerged.

3.5.2 The Role of Corticosteroids

Corticosteroids, in particular dexamethasone, have been used as antiemetics for CINV and have demonstrated efficacy, but no clear mechanism of action since their protective effects appear much sooner than conventional corticosteroid mechanism would allow. With the advent of 5-HT3 receptor antagonists, numerous studies examined the question of single-agent 5-HT3 receptor antagonist compared to corticosteroid combinations. The studies are summarized by Jantunen [47] and combining 11 studies in a meta-analysis demonstrated the odds ratio of acute vomiting to be 0.42, strongly in favor of the combination arms. The Italian Group for Antiemetic Research went further to define the optimal dosing of dexamethasone with 5-HT3 receptor antagonists both for highly emetogenic [48] and moderately emetogenic therapy [49].

3.5.3 First-Generation 5-HT3 Antagonists Compared

The first study to compare ondansetron to granisetron randomized 496 patients to receive either ondansetron 8 mg, ondansetron 32 mg, or granisetron 3 mg prior to cisplatin-based therapy [50]. No significant difference was seen between any of the groups related to emesis, nausea, or adverse events. Multiple other studies compared various doses or schedules of ondansetron with granisetron both as prophylaxis for highly emetogenic chemotherapy and for moderately emetogenic chemotherapy [5156]. Other studies compared tropisetron [57, 58] as well as dolasetron [5961]. Despite numerous studies, no significant differences emerged. Even large meta-analyses did not show any appreciable differences in clinical efficacy and adverse events [62, 63], although in the larger analysis, tropisetron was not as effective as granisetron [63].

3.5.4 Intravenous Compared to Oral Therapy

Oral forms of 5-HT3 receptor antagonists were developed shortly after the intravenous forms and theoretically provide more convenient dosing. The agents are well absorbed and undergo some first-pass metabolism although they are generally conserved; the bioavailability of oral ondansetron is 59 % [18], granisetron 60 % [64], tropisetron 60 % [22], and dolasetron (hydrodolasetron) 76 % [65].
Initial studies of oral 5-HT3 receptor antagonists examined ranges of oral doses and efficacy was significantly superior to historical placebo controls for ondansetron [66], dolasetron [67], and granisetron [68]. A direct systematic comparison of the same agent, comparing intravenous and oral forms in large randomized studies was only performed in a single study. The Ondansetron Acute Emesis Study Group randomized 530 patients to receive a single dose of either oral ondansetron 24 mg or intravenous ondansetron 8 mg together with dexamethasone prior to cisplatin therapy [69]. The acute (<24 h) complete response rates (no emesis and no rescue medication) were 85 % and 83 % for the oral and intravenous groups, respectively. Further randomized studies compared different formulations and agents: oral granisetron was compared to intravenous ondansetron prior to highly emetogenic therapy [70] and prior to moderately emetogenic therapy [71]; oral ondansetron was compared to intravenous granisetron prior to cisplatin therapy [72]. All three studies showed no difference in emesis control between randomized arms.
Oral 5-HT3 receptor antagonists allowed the opportunity to explore the use of these agents beyond the first day in an attempt to lessen delayed emesis. Some studies showed reduced delayed emesis with the use of 5-HT3 receptor antagonists beyond 24 h [73], while others failed to show any benefit [74]. A meta-analysis of ten randomized studies, including five that used dexamethasone in the delayed setting and five that did not, determined the reduction in emesis risk with longer treatment [75]. The absolute emesis risk reduction for monotherapy was 8.2 % (95 % CI, 3.0–13.4 %), whereas the reduction was only 2.6 % (95 % CI, −0.6–5.8 %) with the use of dexamethasone.
Oral therapy also allowed the evaluation of these agents in other oncology settings, particularly in preventing radiation-induced emesis. In patients receiving fractionated upper abdominal radiation, a randomized study of 260 patients demonstrated significant control of radiation-induced emesis with granisetron 2 mg daily compared with placebo [76]. Spitzer et al. [77] compared oral granisetron or ondansetron prior to total body irradiation to historical control patients. Emesis rates were significantly reduced in patients receiving either 5-HT3 receptor antagonist compared to the historical controls.

3.5.5 Summary of Characteristics of First-Generation 5-HT3 Receptor Antagonists

The four first-generation 5-HT3 receptor antagonists discussed here were derived from different processes but essentially exhibit more similarities than differences. These characteristics are listed below and are used as principles to formulate guideline recommendations for CINV prophylaxis:
(i)
They all contain an indole ring and are highly selective antagonists of the 5-HT3 receptor.
 
(ii)
The elimination half-life is approximately 4–8 h.
 
(iii)
Oral formulations are well absorbed with an approximate 60 % bioavailability and are equivalent to intravenous formulations.
 
(iv)
The lowest effective dose has been determined.
 
(v)
Antiemetic efficacy is vastly superior to placebo for both highly and moderately emetogenic chemotherapy.
 
(vi)
Antiemetic efficacy is superior to older antiemetics except for high-dose metoclopramide.
 
(vii)
No significant differences in antiemetic efficacy are discernible between the four agents in individual studies.
 
(viii)
Antiemetic efficacy is superior when combined with corticosteroids.
 
(ix)
Side effects are largely confined to constipation and headache, with no appreciable increase in adverse events, even at escalated doses, although QTc prolongation has become a regulatory concern.
 

3.6 Palonosetron: A Second-Generation 5-HT3 Antagonist

Unlike the first-generation 5-HT3 antagonists, palonosetron is not based on an indole moiety; rather it contains a fused tricyclic ring and a quinuclidine moiety and has a half-life of approximately 40 h [28]. At the receptor level, it appears to bind to the 5-HT3 receptor more avidly than first-generation agents as well as exhibiting allosteric binding in contrast to the pure competitive binding seen with first-generation agents [78]. Further, it has been noted to cause receptor internalization [79], resulting in additional prolongation of duration of action. These unique properties are thought to account for some of the clinical efficacy differences seen in comparison to first-generation 5-HT3 receptor antagonists.
In the moderately emetogenic setting, palonosetron was compared to ondansetron [80] and dolasetron [81], respectively, in two double-blind randomized phase III trials. Both studies included three arms with only a single dose: palonosetron 0.25 mg and 0.75 mg and first-generation 5-HT3 receptor antagonist; no dexamethasone was administered as part of the protocol. Results from both studies demonstrated that a single dose of palonosetron (0.25 mg) was as effective as a single dose of a first-generation 5-HT3 receptor antagonist in preventing acute CINV and superior in preventing delayed CINV. In a randomized study conducted in the highly emetogenic setting, palonosetron was as effective as ondansetron and in a subset of patients also treated with dexamethasone appeared more effective [82].
Since the palonosetron registration studies were conducted without mandated corticosteroid therapy, the magnitude of benefit of palonosetron over the first-generation 5-HT3 receptor antagonists remained in question. A randomized study comparing palonosetron with dexamethasone to granisetron with dexamethasone in patients receiving highly or moderately emetogenic therapy helped to define the benefit [83]. There was no difference in the acute phase, but in the delayed phase, approximately 12 % more patients were emesis-free in the palonosetron arm as compared to the granisetron arm for both highly and moderately emetogenic therapy. Similar to first-generation 5-HT3 receptor antagonists, oral palonosetron has high bioavailability and similar efficacy to intravenous palonosetron with 0.5 mg being the preferred oral dose [84].
The improved efficacy observed with palonosetron has been attributed to the receptor-binding effects discussed above rather than the longer half-life, since administration of 5-HT3 receptor antagonists beyond day 1 results in negligible benefit in emesis control [75].

3.7 Novel Delivery Methods

Alternative routes of administration of first-generation 5-HT3 receptor antagonists have been explored, including subcutaneous, intramuscular, rectal, transdermal, and nasal/buccal sprays. In general, bioavailability of any route has been high with comparable efficacy where studied [85, 86]. Two delivery methods warrant further discussion: transdermal granisetron and polymer encapsulated slow-release granisetron.

3.7.1 Transdermal Granisetron

A transdermal formulation of granisetron is approved in the USA for the prevention of nausea and vomiting in patients receiving highly or moderately emetogenic chemotherapy. This granisetron transdermal delivery system is a 52 cm2 patch containing 34.3 mg of granisetron, which is delivered transdermally as 3.1 mg/24 h and essentially achieves a similar exposure to that of a 2 mg oral dose providing continuous delivery of granisetron over 6 days [87]. A randomized, double-blind study included 641 patients receiving chemotherapy and demonstrated that the transdermal delivery system was non-inferior to oral granisetron [88]. Although balanced between treatment arms, the patient population studied was heterogeneous in terms of emetogenicity of chemotherapy (high or moderate), prior chemotherapy exposure and the use of corticosteroids. In addition, a strong limitation on the utility of this delivery system is the requirement to place patch 24 h before scheduled chemotherapy. Finally, since the benefit of first-generation 5-HT3 receptor antagonists beyond the first day of chemotherapy is limited [75], protracted delivery would appear to hold little advantage. Intuitively, such delivery systems may be more useful for preventing emesis from radiation or oral agents: no such studies have been conducted. Nevertheless, the transdermal system is approved in the USA and remains a potential choice for prevention of chemotherapy-induced nausea and vomiting.

3.7.2 Sustained-Release Subcutaneous Granisetron

APF530 is a subcutaneously administered polymeric formulation of granisetron that provides slow, controlled, and sustained release of granisetron [89]. APF530 comprises 2 % granisetron and a polymer that is designed to undergo controlled hydrolysis, imparting the drug release characteristics. In a phase 3 non-inferiority trial, the clinical efficacy of APF530 250 mg subcutaneously and 500 mg subcutaneously (containing granisetron 5 and 10 mg, respectively) was compared with 0.25 mg palonosetron intravenously in patients receiving moderately or highly emetogenic chemotherapy [90]. Patients were stratified according to emetogenicity of chemotherapy and received study drug together with appropriate placebo (subcutaneous saline for palonosetron group; intravenous saline for APF530 group). All patients received guideline-appropriate doses of dexamethasone. The study demonstrated non-inferiority of the 500 mg dose of APF530 compared with palonosetron in preventing CINV both in the acute and delayed setting. Since the classification of emetogenicity in the initial study design was based on the older Hesketh algorithm [91], a reanalysis of the study was undertaken, using the latest American Society of Clinical Oncology emetogenic classification [92]. The reanalysis confirmed the initial findings of non-inferiority of APF530 [93].
The observations from the single study of APF530 question the mechanism used to explain the superiority of palonosetron over first-generation 5-HT3 receptor antagonists: that receptor binding was more important than the extended half-life. A clear explanation to unify these observations will require further study.

3.8 Conclusion

First-generation 5-HT3 receptor antagonists dramatically altered the delivery of cytotoxic chemotherapy, changing intolerable regimens to tolerable ones, shifting many chemotherapy regimens to the ambulatory setting, and improving quality of life for many patients. Further understandings of the mechanisms of emesis and clinical trial observations have allowed refinements in their use; 5-HT3 receptor antagonists form the backbone of most antiemetic regimens. Improvements have also been seen with palonosetron and with newer agents such as neurokinin-1 receptor inhibitors; however, nausea remains a persistent problem and will require further refinements in the use of multiple agents, together with a better understanding of the mechanisms of chemotherapy-induced nausea to improve overall CINV control.
Literature
1.
Markman M (2002) Progress in preventing chemotherapy-induced nausea and vomiting. Cleve Clin J Med 69:609–610, 612, 615–7CrossRefPubMed
2.
Roila F, Herrstedt J, Aapro M et al (2010) Guideline update for MASCC and ESMO in the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting: results of the Perugia consensus conference. Ann Oncol 21(Suppl 5):v232–v243CrossRefPubMed
3.
Kris MG, Gralla RJ, Clark RA et al (1985) Incidence, course, and severity of delayed nausea and vomiting following the administration of high-dose cisplatin. J Clin Oncol 3:1379–1384PubMed
4.
Thumas L (1891) Ueber das Brechcentrum und über die Wirkung einiger pharmakologischer Mittel auf dasselbe. Virchows Arch 123:44–69CrossRef
5.
Wang SC, Borison HL (1950) The vomiting center; a critical experimental analysis. Arch Neurol Psychiatry 63:928–941CrossRefPubMed
6.
Koga T, Fukuda H (1992) Neurons in the nucleus of the solitary tract mediating inputs from emetic vagal afferents and the area postrema to the pattern generator for the emetic act in dogs. Neurosci Res 14:166–179CrossRefPubMed
7.
Andrews PL, Davis CJ, Bingham S et al (1990) The abdominal visceral innervation and the emetic reflex: pathways, pharmacology, and plasticity. Can J Physiol Pharmacol 68:325–345CrossRefPubMed
8.
Leslie RA, Shah Y, Thejomayen M et al (1990) The neuropharmacology of emesis: the role of receptors in neuromodulation of nausea and vomiting. Can J Physiol Pharmacol 68:279–288CrossRefPubMed
9.
Kris MG, Gralla RJ, Tyson LB et al (1985) Improved control of cisplatin-induced emesis with high-dose metoclopramide and with combinations of metoclopramide, dexamethasone, and diphenhydramine. Results of consecutive trials in 255 patients. Cancer 55:527–534CrossRefPubMed
10.
Butler A, Hill JM, Ireland SJ et al (1988) Pharmacological properties of GR38032F, a novel antagonist at 5-HT3 receptors. Br J Pharmacol 94:397–412PubMedCentralCrossRefPubMed
11.
Fozard JR, Mobarok Ali AT, Newgrosh G (1979) Blockade of serotonin receptors on autonomic neurones by (-)-cocaine and some related compounds. Eur J Pharmacol 59:195–210CrossRefPubMed
12.
Buchheit KH, Costall B, Engel G et al (1985) 5-Hydroxytryptamine receptor antagonism by metoclopramide and ICS 205–930 in the guinea-pig leads to enhancement of contractions of stomach muscle strips induced by electrical field stimulation and facilitation of gastric emptying in-vivo. J Pharm Pharmacol 37:664–667CrossRefPubMed
13.
Yan D, Schulte MK, Bloom KE et al (1999) Structural features of the ligand-binding domain of the serotonin 5-HT3 receptor. J Biol Chem 274:5537–5541
14.
National Center for Biotechnology Information. PubChem Compound Database; CID = 4595. http://​pubchem.​ncbi.​nlm.​nih.​gov/​compound/​ondansetron#section=​Top. Accessed 11 Feb 2015
15.
National Center for Biotechnology Information (2015) PubChem Compound Database; CID = 3148. http://​pubchem.​ncbi.​nlm.​nih.​gov/​compound/​3148. Accessed 11 Feb 2015
16.
National Center for Biotechnology Information (2015) PubChem Compound Database; CID = 656665. http://​pubchem.​ncbi.​nlm.​nih.​gov/​compound/​tropisetron. Accessed 11 Feb 2015
17.
National Center for Biotechnology Information (2015) PubChem Compound Database; CID = 5284566. http://​pubchem.​ncbi.​nlm.​nih.​gov/​compound/​granisetron. Accessed 11 Feb 2015
18.
Blackwell CP, Harding SM (1989) The clinical pharmacology of ondansetron. Eur J Cancer Clin Oncol 25(Suppl 1):S21–S24, discussion S25–7PubMed
19.
Pritchard JF, Bryson JC, Kernodle AE et al (1992) Age and gender effects on ondansetron pharmacokinetics: evaluation of healthy aged volunteers. Clin Pharmacol Ther 51:51–55CrossRefPubMed
20.
Zuo P, Haberer LJ, Fang L et al (2014) Integration of modeling and simulation to support changes to ondansetron dosing following a randomized, double-blind, placebo-, and active-controlled thorough QT study. J Clin Pharmacol 54:1221–1229CrossRefPubMed
21.
Boxenbaum H, Gillespie T, Heck K et al (1992) Human dolasetron pharmacokinetics: I. Disposition following single-dose intravenous administration to normal male subjects. Biopharm Drug Dispos 13:693–701CrossRefPubMed
22.
Kees F, Farber L, Bucher M et al (2001) Pharmacokinetics of therapeutic doses of tropisetron in healthy volunteers. Br J Clin Pharmacol 52:705–707PubMedCentralCrossRefPubMed
23.
Kim MK, Cho JY, Lim HS et al (2003) Effect of the CYP2D6 genotype on the pharmacokinetics of tropisetron in healthy Korean subjects. Eur J Clin Pharmacol 59:111–116PubMed
24.
Carmichael J, Cantwell BM, Edwards CM et al (1989) A pharmacokinetic study of granisetron (BRL 43694A), a selective 5-HT3 receptor antagonist: correlation with anti-emetic response. Cancer Chemother Pharmacol 24:45–49PubMed
25.
Clark RD, Miller AB, Berger J et al (1993) 2-(Quinuclidin-3-yl)pyrido[4,3-b]indol-1-ones and isoquinolin-1-ones. Potent conformationally restricted 5-HT3 receptor antagonists. J Med Chem 36:2645–2657CrossRefPubMed
26.
National Center for Biotechnology Information (2015) PubChem Compound Database; CID = 6337614. http://​pubchem.​ncbi.​nlm.​nih.​gov/​compound/​Palonosetron. Accessed 11 Feb 2015
27.
Yavas C, Dogan U, Yavas G et al (2012) Acute effect of palonosetron on electrocardiographic parameters in cancer patients: a prospective study. Support Care Cancer 20:2343–2347CrossRefPubMed
28.
Yang LP, Scott LJ (2009) Palonosetron: in the prevention of nausea and vomiting. Drugs 69:2257–2278CrossRefPubMed
29.
Cubeddu LX, Hoffmann IS, Fuenmayor NT et al (1990) Efficacy of ondansetron (GR 38032F) and the role of serotonin in cisplatin-induced nausea and vomiting. N Engl J Med 322:810–816CrossRefPubMed
30.
Cupissol DR, Serrou B, Caubel M (1990) The efficacy of granisetron as a prophylactic anti-emetic and intervention agent in high-dose cisplatin-induced emesis. Eur J Cancer 26(Suppl 1):S23–S27PubMed
31.
Cubeddu LX, Hoffman IS, Fuenmayor NT et al (1990) Antagonism of serotonin S3 receptors with ondansetron prevents nausea and emesis induced by cyclophosphamide-containing chemotherapy regimens. J Clin Oncol 8:1721–1727PubMed
32.
Ikeda M, Taguchi T, Ota K et al (1992) Evaluation of SN-307 (ondansetron), given intravenously in the treatment of nausea and vomiting caused by anticancer drugs including cisplatin – a placebo-controlled, double-blind comparative study. Gan To Kagaku Ryoho 19:2071–2084PubMed
33.
Kondo M, Furue H, Taguchi T et al (1995) Clinical phase III study of tropisetron capsule in the treatment of nausea and vomiting induced by anti-cancer drug; a placebo-controlled, multicenter, double-blind comparative study. Gan To Kagaku Ryoho 22:1223–1234PubMed
34.
McVie JG, de Bruijn KM (1992) Methodology of antiemetic trials. Drugs 43(Suppl 3):1–5CrossRefPubMed
35.
Kris MG, Cubeddu LX, Gralla RJ et al (1996) Are more antiemetic trials with a placebo necessary? Report of patient data from randomized trials of placebo antiemetics with cisplatin. Cancer 78:2193–2198CrossRefPubMed
36.
Grunberg SM, Lane M, Lester EP et al (1993) Randomized double-blind comparison of three dose levels of intravenous ondansetron in the prevention of cisplatin-induced emesis. Cancer Chemother Pharmacol 32:268–272CrossRefPubMed
37.
Riviere A (1994) Dose finding study of granisetron in patients receiving high-dose cisplatin chemotherapy. The Granisetron Study Group. Br J Cancer 69:967–971PubMedCentralCrossRefPubMed
38.
Soukop M (1994) A dose-finding study of granisetron, a novel antiemetic, in patients receiving high-dose cisplatin. Granisetron Study Group. Support Care Cancer 2:177–183CrossRefPubMed
39.
Van Belle SJ, Stamatakis L, Bleiberg H et al (1994) Dose-finding study of tropisetron in cisplatin-induced nausea and vomiting. Ann Oncol 5:821–825PubMed
40.
Kris MG, Grunberg SM, Gralla RJ et al (1994) Dose-ranging evaluation of the serotonin antagonist dolasetron mesylate in patients receiving high-dose cisplatin. J Clin Oncol 12:1045–1049PubMed
41.
Hainsworth J, Harvey W, Pendergrass K et al (1991) A single-blind comparison of intravenous ondansetron, a selective serotonin antagonist, with intravenous metoclopramide in the prevention of nausea and vomiting associated with high-dose cisplatin chemotherapy. J Clin Oncol 9:721–728PubMed
42.
De Mulder PH, Seynaeve C, Vermorken JB et al (1990) Ondansetron compared with high-dose metoclopramide in prophylaxis of acute and delayed cisplatin-induced nausea and vomiting. A multicenter, randomized, double-blind, crossover study. Ann Intern Med 113:834–840CrossRefPubMed
43.
Marty M, Pouillart P, Scholl S et al (1990) Comparison of the 5-hydroxytryptamine3 (serotonin) antagonist ondansetron (GR 38032F) with high-dose metoclopramide in the control of cisplatin-induced emesis. N Engl J Med 322:816–821CrossRefPubMed
44.
Warr D, Willan A, Fine S et al (1991) Superiority of granisetron to dexamethasone plus prochlorperazine in the prevention of chemotherapy-induced emesis. J Natl Cancer Inst 83:1169–1173CrossRefPubMed
45.
Marty M (1992) A comparison of granisetron as a single agent with conventional combination antiemetic therapies in the treatment of cytostatic-induced emesis. The Granisetron Study Group. Eur J Cancer 28A(Suppl 1):S12–S16CrossRefPubMed
46.
Warr D, Wilan A, Venner P et al (1992) A randomised, double-blind comparison of granisetron with high-dose metoclopramide, dexamethasone and diphenhydramine for cisplatin-induced emesis. An NCI Canada Clinical Trials Group Phase III Trial. Eur J Cancer 29A:33–36PubMed
47.
Jantunen IT, Kataja VV, Muhonen TT (1997) An overview of randomised studies comparing 5-HT3 receptor antagonists to conventional anti-emetics in the prophylaxis of acute chemotherapy-induced vomiting. Eur J Cancer 33:66–74CrossRefPubMed
48.
Double-blind, dose-finding study of four intravenous doses of dexamethasone in the prevention of cisplatin-induced acute emesis. Italian Group for Antiemetic Research (1998) J Clin Oncol 16:2937–2942
49.
Randomized, double-blind, dose-finding study of dexamethasone in preventing acute emesis induced by anthracyclines, carboplatin, or cyclophosphamide (2004) J Clin Oncol 22:725–729
50.
Ruff P, Paska W, Goedhals L et al (1994) Ondansetron compared with granisetron in the prophylaxis of cisplatin-induced acute emesis: a multicentre double-blind, randomised, parallel-group study. The Ondansetron and Granisetron Emesis Study Group. Oncology 51:113–118CrossRefPubMed
51.
Noble A, Bremer K, Goedhals L et al (1994) A double-blind, randomised, crossover comparison of granisetron and ondansetron in 5-day fractionated chemotherapy: assessment of efficacy, safety and patient preference. The Granisetron Study Group. Eur J Cancer 30A:1083–1088CrossRefPubMed
52.
Ondansetron versus granisetron, both combined with dexamethasone, in the prevention of cisplatin-induced emesis. Italian Group of Antiemetic Research (1995) Ann Oncol 6:805–810
53.
Bonneterre J, Hecquet B (1995) Granisetron (IV) compared with ondansetron (IV plus oral) in the prevention of nausea and vomiting induced by moderately-emetogenic chemotherapy. A cross-over study. Bull Cancer 82:1038–1043PubMed
54.
Navari R, Gandara D, Hesketh P et al (1995) Comparative clinical trial of granisetron and ondansetron in the prophylaxis of cisplatin-induced emesis. The Granisetron Study Group. J Clin Oncol 13:1242–1248PubMed
55.
Stewart A, McQuade B, Cronje JD et al (1995) Ondansetron compared with granisetron in the prophylaxis of cyclophosphamide-induced emesis in out-patients: a multicentre, double-blind, double-dummy, randomised, parallel-group study. Emesis Study Group for Ondansetron and Granisetron in Breast Cancer Patients. Oncology 52:202–210CrossRefPubMed
56.
Martoni A, Angelelli B, Guaraldi M et al (1996) An open randomised cross-over study on granisetron versus ondansetron in the prevention of acute emesis induced by moderate dose cisplatin-containing regimens. Eur J Cancer 32A:82–85CrossRefPubMed
57.
Chua DT, Sham JS, Kwong DL et al (2000) Comparative efficacy of three 5-HT3 antagonists (granisetron, ondansetron, and tropisetron) plus dexamethasone for the prevention of cisplatin-induced acute emesis: a randomized crossover study. Am J Clin Oncol 23:185–191CrossRefPubMed
58.
Oge A, Alkis N, Oge O et al (2000) Comparison of granisetron, ondansetron and tropisetron for control of vomiting and nausea induced by cisplatin. J Chemother 12:105–108PubMed
59.
Audhuy B, Cappelaere P, Martin M et al (1996) A double-blind, randomised comparison of the anti-emetic efficacy of two intravenous doses of dolasetron mesylate and granisetron in patients receiving high dose cisplatin chemotherapy. Eur J Cancer 32A:807–813PubMed
60.
Fauser AA, Duclos B, Chemaissani A et al (1996) Therapeutic equivalence of single oral doses of dolasetron mesylate and multiple doses of ondansetron for the prevention of emesis after moderately emetogenic chemotherapy. European Dolasetron Comparative Study Group. Eur J Cancer 32A:1523–1529CrossRefPubMed
61.
Lofters WS, Pater JL, Zee B et al (1997) Phase III double-blind comparison of dolasetron mesylate and ondansetron and an evaluation of the additive role of dexamethasone in the prevention of acute and delayed nausea and vomiting due to moderately emetogenic chemotherapy. J Clin Oncol 15:2966–2973PubMed
62.
Hesketh PJ (2000) Comparative review of 5-HT3 receptor antagonists in the treatment of acute chemotherapy-induced nausea and vomiting. Cancer Investig 18:163–173CrossRef
63.
Jordan K, Hinke A, Grothey A et al (2007) A meta-analysis comparing the efficacy of four 5-HT3-receptor antagonists for acute chemotherapy-induced emesis. Support Care Cancer 15:1023–1033CrossRefPubMed
64.
Clarke SE, Austin NE, Bloomer JC et al (1994) Metabolism and disposition of 14C-granisetron in rat, dog and man after intravenous and oral dosing. Xenobiotica 24:1119–1131CrossRefPubMed
65.
Dimmitt DC, Choo YS, Martin LA et al (1999) Intravenous pharmacokinetics and absolute oral bioavailability of dolasetron in healthy volunteers: part 1. Biopharm Drug Dispos 20:29–39CrossRefPubMed
66.
Needles B, Miranda E, Garcia Rodriguez FM et al (1999) A multicenter, double-blind, randomized comparison of oral ondansetron 8 mg b.i.d., 24 mg q.d., and 32 mg q.d. in the prevention of nausea and vomiting associated with highly emetogenic chemotherapy. S3AA3012 Study Group. Support Care Cancer 7:347–353CrossRefPubMed
67.
Rubenstein EB, Gralla RJ, Hainsworth JD et al (1997) Randomized, double blind, dose-response trial across four oral doses of dolasetron for the prevention of acute emesis after moderately emetogenic chemotherapy. Oral Dolasetron Dose-Response Study Group. Cancer 79:1216–1224CrossRefPubMed
68.
Bleiberg HH, Spielmann M, Falkson G et al (1995) Antiemetic treatment with oral granisetron in patients receiving moderately emetogenic chemotherapy: a dose-ranging study. Clin Ther 17:38–51CrossRefPubMed
69.
Krzakowski M, Graham E, Goedhals L et al (1998) A multicenter, double-blind comparison of i.v. and oral administration of ondansetron plus dexamethasone for acute cisplatin-induced emesis. Ondansetron Acute Emesis Study Group. Anticancer Drugs 9:593–598CrossRefPubMed
70.
Gralla RJ, Navari RM, Hesketh PJ et al (1998) Single-dose oral granisetron has equivalent antiemetic efficacy to intravenous ondansetron for highly emetogenic cisplatin-based chemotherapy. J Clin Oncol 16:1568–1573PubMed
71.
Perez EA, Hesketh P, Sandbach J et al (1998) Comparison of single-dose oral granisetron versus intravenous ondansetron in the prevention of nausea and vomiting induced by moderately emetogenic chemotherapy: a multicenter, double-blind, randomized parallel study. J Clin Oncol 16:754–760PubMed
72.
Spector JI, Lester EP, Chevlen EM et al (1998) A comparison of oral ondansetron and intravenous granisetron for the prevention of nausea and emesis associated with cisplatin-based chemotherapy. Oncologist 3:432–438PubMed
73.
Navari RM, Madajewicz S, Anderson N et al (1995) Oral ondansetron for the control of cisplatin-induced delayed emesis: a large, multicenter, double-blind, randomized comparative trial of ondansetron versus placebo. J Clin Oncol 13:2408–2416PubMed
74.
Olver I, Paska W, Depierre A et al (1996) A multicentre, double-blind study comparing placebo, ondansetron and ondansetron plus dexamethasone for the control of cisplatin-induced delayed emesis. Ondansetron Delayed Emesis Study Group. Ann Oncol 7:945–952CrossRefPubMed
75.
Geling O, Eichler HG (2005) Should 5-hydroxytryptamine-3 receptor antagonists be administered beyond 24 hours after chemotherapy to prevent delayed emesis? Systematic re-evaluation of clinical evidence and drug cost implications. J Clin Oncol 23:1289–1294CrossRefPubMed
76.
Lanciano R, Sherman DM, Michalski J et al (2001) The efficacy and safety of once-daily Kytril (granisetron hydrochloride) tablets in the prophylaxis of nausea and emesis following fractionated upper abdominal radiotherapy. Cancer Invest 19:763–772CrossRefPubMed
77.
Spitzer TR, Friedman CJ, Bushnell W et al (2000) Double-blind, randomized, parallel-group study on the efficacy and safety of oral granisetron and oral ondansetron in the prophylaxis of nausea and vomiting in patients receiving hyperfractionated total body irradiation. Bone Marrow Transplant 26:203–210CrossRefPubMed
78.
Rojas C, Stathis M, Thomas AG et al (2008) Palonosetron exhibits unique molecular interactions with the 5-HT3 receptor. Anesth Analg 107:469–478CrossRefPubMed
79.
Rojas C, Thomas AG, Alt J et al (2010) Palonosetron triggers 5-HT(3) receptor internalization and causes prolonged inhibition of receptor function. Eur J Pharmacol 626:193–199CrossRefPubMed
80.
Gralla R, Lichinitser M, Van Der Vegt S et al (2003) Palonosetron improves prevention of chemotherapy-induced nausea and vomiting following moderately emetogenic chemotherapy: results of a double-blind randomized phase III trial comparing single doses of palonosetron with ondansetron. Ann Oncol 14:1570–1577CrossRefPubMed
81.
Eisenberg P, Figueroa-Vadillo J, Zamora R et al (2003) Improved prevention of moderately emetogenic chemotherapy-induced nausea and vomiting with palonosetron, a pharmacologically novel 5-HT3 receptor antagonist: results of a phase III, single-dose trial versus dolasetron. Cancer 98:2473–2482CrossRefPubMed
82.
Aapro MS, Grunberg SM, Manikhas GM et al (2006) A phase III, double-blind, randomized trial of palonosetron compared with ondansetron in preventing chemotherapy-induced nausea and vomiting following highly emetogenic chemotherapy. Ann Oncol 17:1441–1449CrossRefPubMed
83.
Saito M, Aogi K, Sekine I et al (2009) Palonosetron plus dexamethasone versus granisetron plus dexamethasone for prevention of nausea and vomiting during chemotherapy: a double-blind, double-dummy, randomised, comparative phase III trial. Lancet Oncol 10:115–124CrossRefPubMed
84.
Boccia R, Grunberg S, Franco-Gonzales E et al (2013) Efficacy of oral palonosetron compared to intravenous palonosetron for the prevention of chemotherapy-induced nausea and vomiting associated with moderately emetogenic chemotherapy: a phase 3 trial. Support Care Cancer 21:1453–1460PubMedCentralCrossRefPubMed
85.
Gurpide A, Sadaba B, Martin-Algarra S et al (2007) Randomized crossover pharmacokinetic evaluation of subcutaneous versus intravenous granisetron in cancer patients treated with platinum-based chemotherapy. Oncologist 12:1151–1155CrossRefPubMed
86.
Roila F, Del Favero A (1995) Ondansetron clinical pharmacokinetics. Clin Pharmacokinet 29:95–109CrossRefPubMed
87.
Howell J, Smeets J, Drenth HJ et al (2009) Pharmacokinetics of a granisetron transdermal system for the treatment of chemotherapy-induced nausea and vomiting. J Oncol Pharm Pract 15:223–231CrossRefPubMed
88.
Boccia RV, Gordan LN, Clark G et al (2011) Efficacy and tolerability of transdermal granisetron for the control of chemotherapy-induced nausea and vomiting associated with moderately and highly emetogenic multi-day chemotherapy: a randomized, double-blind, phase III study. Support Care Cancer 19:1609–1617PubMedCentralCrossRefPubMed
89.
Heller J, Barr J (2005) Biochronomer technology. Expert Opin Drug Deliv 2:169–183CrossRefPubMed
90.
Raftopoulos H, Cooper W, O’Boyle E et al (2015) Comparison of an extended-release formulation of granisetron (APF530) versus palonosetron for the prevention of chemotherapy-induced nausea and vomiting associated with moderately or highly emetogenic chemotherapy: results of a prospective, randomized, double-blind, noninferiority phase 3 trial. Support Care Cancer 23:723–732PubMedCentralCrossRefPubMed
91.
Hesketh PJ, Kris MG, Grunberg SM et al (1997) Proposal for classifying the acute emetogenicity of cancer chemotherapy. J Clin Oncol 15:103–109PubMed
92.
Basch E, Prestrud AA, Hesketh PJ et al (2011) Antiemetics: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol 29:4189–4198CrossRefPubMed
93.
Raftopoulos H, Boccia RV, Cooper W et al (2014) A prospective, randomized, double-blind phase 3 trial of extended-release granisetron (APF530) versus palonosetron (PALO) for preventing chemotherapy-induced nausea and vomiting (CINV) associated with moderately (MEC) or highly (HEC) emetogenic chemotherapy: does a reanalysis using newer ASCO emetogenicity criteria affect study conclusions? ASCO Meet Abstr 32:9648