2,901
Views
4
CrossRef citations to date
0
Altmetric
Review

A multidisciplinary expert opinion on CINV and RINV, unmet needs and practical real-life approaches

, , , , , , , , & show all
Pages 187-204 | Received 26 Sep 2019, Accepted 29 Jan 2020, Published online: 06 Feb 2020

ABSTRACT

Introduction: A range of combination chemotherapy regimens are currently used in clinical practice. However, international antiemetic guidelines often only categorize the emetogenic potential of single agents rather than the emetogenicity of combination chemotherapy regimens. To manage the nausea and vomiting induced by antineoplastic combinations, guidelines suggest antiemetics that are appropriate for the component drug with the highest emetogenic potential. Furthermore, antiemetic guidelines generally do not consider the influence of other factors, including individual patient characteristics, on the emetic effects of cancer treatments. Similarly, the emetogenic potential of radiotherapy is stratified only according to the site of radiation, while other factors contributing to emetic risk are overlooked.

Areas covered: An Expert Panel was convened to examine unresolved issues and summarize the current clinical research on managing nausea and vomiting associated with combination chemotherapy and radiotherapy.

Expert opinion: The panel identified the incidence of nausea and vomiting induced by multi-drug combination therapies currently used to treat cancer at different anatomic sites and by radiotherapy in the presence of other risk factors. Based on these data and the clinical experience of panel members, several suggestions are made for a practical approach to prevent or manage nausea and vomiting due to chemotherapy regimens and radiation therapy.

1. Introduction

Nausea and vomiting are among the most frequent toxicities associated with antineoplastic agents and radiation treatments used for oncological patients, with an estimated incidence rate of nearly 50% [Citation1Citation3]. These burdensome adverse effects are highly distressing and debilitating for patients, profoundly affecting their quality of life and seriously compromising their compliance with anticancer therapy, with detrimental consequences [Citation4,Citation5].

Not only are nausea and vomiting the strongest concerns for patients and the most feared side effects of antitumoral treatments, but their occurrence also markedly impairs their own and their family’s daily activities [Citation1,Citation5]. Also, health-care providers tend to underestimate the incidence of these symptoms, particularly delayed symptoms that do not develop immediately following the administration of therapy [Citation6].

Nausea and vomiting related to antitumoral therapies are so important and characteristic that they are recognized as specific and well-defined conditions known as CINV (chemotherapy-induced nausea and vomiting) and RINV (radiotherapy-induced nausea and vomiting) for which there are dedicated guidelines by the Multinational Association of Supportive Care in Cancer and the European Society for Medical Oncology (MASCC/ESMO), the American Society of Clinical Oncology (ASCO) and the National Comprehensive Cancer Network (NCCN) [Citation7Citation12].

The main international antiemetic guidelines classify the available single chemotherapeutic agents for anticancer treatment into four groups according to their ‘level of emetogenicity’, that is, the expected frequency of emesis induced in the absence of effective antiemetic prophylaxis [Citation7,Citation13].

Based on solid scientific evidence, the antiemetic guidelines recommend a specific antiemetic approach for each emetogenic level, which limits their applicability because the most common and effective anticancer treatments are combinations of different antineoplastic agents, not the administration of single drugs. With the exception of the doxorubicin and cyclophosphamide (AC) regimen for breast cancer, none of the guidelines has established emetic-risk classes for combination regimens, generally stating that the emetogenic potential of the combination would automatically correspond to the potential of the component drug with the highest emetic risk.

However, as learned through the results of clinical trials and daily clinical practice, the emetogenic potentials of the different agents included in a chemotherapy regimen may be additive (in the case of drugs with a moderate or high emetogenic potential), so that the nausea and vomiting induced by an antitumor drug varies according to its possible combination with other anticancer agents [Citation14].

Moreover, the incidence and severity of CINV (and radiotherapy or chemoradiation as well) is influenced by a wide range of factors, including dosage, duration, schedule, and route of administration of the agents, disease characteristics, and individual patient characteristics, such as age, sex, previous experience of chemotherapy or emesis during pregnancy, motion sickness, history of alcohol use, etc. [Citation9,Citation15]. These important variables are explicitly overlooked by the international antiemetic guidelines, possibly because of a paucity of robust evidence [Citation8,Citation9,Citation16]. As a consequence, the emetic risk classification made by international guidelines can be considered quite arbitrary.

RINV is a common side effect of radiation therapy (RT) that negatively impacts patient quality of life [Citation17]. Importantly, this condition requires expensive supportive care and, in severe cases, can result in treatment delays with a negative effect on tumor control [Citation18].

Both radiotherapy-related and patient-related risk factors contribute to the incidence and severity of RINV [Citation18]. Such factors include the anatomic site of radiation, the volume of the irradiated organs, the radiation dose, and the fractionation schedule (radiotherapy-related factors), along with age, gender, and concurrent or recent chemotherapy (patient-related factors) [Citation18Citation20].

The IGARR (Italian Group for Antiemetic Research in Radiotherapy) studies identified other relevant risk factors for RINV: concomitant radiochemotherapy, previous experience of vomiting caused by chemotherapy, site of irradiation (upper abdomen), and field size (>400 cm2) seem to be significantly correlated with a higher incidence of RINV [Citation19,Citation20].

However, current antiemetic guidelines use only the site of irradiation to categorize the emetogenic potential of RT into four risk categories (high, moderate, low, and minimal) [Citation8,Citation9,Citation16]. The same guidelines state that other known risk factors for the occurrence of RINV, such as radiation dose, fractionation, technique, field size, and concomitant chemotherapy, have not been considered in the stratification of the emetic risk levels of RT, recognizing only previous chemotherapy as a significant patient-related risk factor [Citation8,Citation16,Citation17].

An Italian Expert Panel, involving specialists from a range of fields, was convened to discuss these unresolved issues. They collected data in a non-systematic way from randomized and controlled clinical trials in order to identify the incidence of CINV and RINV, with a particular attention to patient- and disease-related risk factors. The aim also was to highlight any unmet needs in the prevention and control of CINV and RINV, including disease- and patient-specific personalization of CINV risk, the importance of considering the emetogenic risk of combination chemotherapy, possible drug interactions, and the use of different routes of antiemetic drug administration. It should be noted that, while the authors have structured this manuscript by different cancer types, research into antiemetic therapy is often based on the emetogenicity of different antineoplastic regimens, rather than on specific diseases.

Based on the experience gained in clinical practice and reported in the most recent scientific literature available, the authors provide suggestions for the management of CINV and RINV. They describe the antiemetic approaches that they commonly offer to their own patients, under conditions not covered by the guidelines and/or to address limitations of these guidelines.

2. CINV in different pathological conditions

briefly summarizes the epidemiology and key antitumor treatment strategies for the conditions covered in this section.

Table 1. Epidemiology and common antitumor treatment strategies for some of the cancers discussed.

2.1. CINV in head and neck cancer patients

Cisplatin is commonly employed in patients with locally advanced disease (). Since cisplatin is classified as a highly emetogenic chemotherapy, one may expect a high risk of CINV in this disease, which is increased by the concurrent administration of RT. However, the emetic risk in this patient population is influenced by two factors which should be considered when identifying the individual predisposition to this side effect. On one hand, alcohol addiction, which is one of the main risk factors for head and neck cancers (HNC), is actually associated with a diminished risk of emesis. On the other hand, the role of human papillomavirus (HPV) as a causal factor is increasing and patients with HPV infection are much less prone to heavy alcohol intake, so this cancer population could have a higher risk of CINV [Citation61]. The pattern of emetic symptoms has been depicted by Chan and colleagues in an interesting study evaluating different cisplatin schedules () [Citation62]. The incidence of vomiting was shown to be similar with single-day and multiple-day regimens, while the incidence of nausea was higher in patients receiving a single-day regimen than in those receiving a multiple-day regimen.

Table 2. Emetic symptoms associated with different cisplatin schedules in head and neck cancer [Citation62]a.

2.1.1. Unmet needs in the control of nausea and vomiting in this patient population

In HNC, the disease and its treatment can increase the overall risk of nausea and vomiting, making CINV and RINV particularly challenging to manage. Dysgeusia, concomitant infections of the oral cavity, mucositis, sticky saliva, dysphagia, use of a feeding tube, and concurrent use of opioids to alleviate mucositis pain are all linked to an increased risk of emesis in HNC patients.

Overall, we are far from being able to completely control CINV and RINV in HNC patients.

2.2. CINV in lung cancer patients

summarizes reported incidence rates of CINV associated with chemotherapy regimens used for small-cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) [Citation63Citation72].

Table 3. Incidence rates of nausea and vomiting induced by multiple-day chemotherapies commonly used for lung cancer in clinical practice, as reported in the scientific literature.

In a randomized trial conducted in elderly patients with SCLC, as would be expected, the split-dose cisplatin plus etoposide course showed a significantly higher rate of nausea/vomiting toxicity compared with an oxaliplatin plus etoposide regimen: 97.2% vs 65.7%; P = 0.001 () [Citation68].

Although carboplatin is considered to be moderately emetogenic, in one trial a carboplatin plus etoposide regimen gave a very low incidence of grade 3–4 nausea and vomiting in patients with SCLC (0.2% and 0.7% respectively; ) [Citation69].

In the Hoosier Oncology Group study in patients with SCLC, the combination of split-dose cisplatin + ifosfamide + etoposide was associated with grade 3–4 nausea/vomiting toxicity in 13.0% of patients (grade 4 = 4.0%), which was higher than 8% rate in patients receiving a regimen without ifosfamide (grade 4 = 0.0%) () [Citation66].

Etoposide administered intravenously for 5 days or given per os for 21 days to patients with SCLC gives a low incidence of nausea/vomiting [Citation73].

The epidermal growth factor receptor (EGFR) inhibitors (e.g. afatinib, erlotinib, gefitinib), used in patients with NSCLC and EGFR mutations, are characterized by a low rate of vomiting toxicity; osimertinib, which is selective for both EGFR-tyrosine kinase inhibitor sensitizing and T790M resistant mutations, rarely causes grade 3 nausea/vomiting (1.0%) [Citation74].

In patients with metastatic NSCLC, the next-generation anaplastic lymphoma kinase inhibitors have demonstrated incidences of grade 3–4 nausea and vomiting of 1.1% (alectinib), 10.9% (ceritinib), and 1.4% (brigatinib), but a not negligible rate of all-grade CINV [Citation75].

2.2.1. Unmet needs in the control of nausea and vomiting in this patient population

Nausea and vomiting are frequent adverse effects of many lung cancer treatments, including chemotherapy regimens and some targeted therapies, although the emetogenic risk differs between agents. A comprehensive intervention is needed to effectively manage and relieve these symptoms which further impact the quality of life of lung cancer patients already very debilitated by their pathology and cancer treatments. In particular, attention should be given to CINV management in patients receiving certain treatments, such as multiple-day schedules with cisplatin or treatment with some oral agents (including ceritinib or crizotinib) on a daily continuous schedule, where long-term low-grade nausea or vomiting may severely impact the patient’s quality of life.

2.3. CINV in breast cancer patients

presents an overview of reported incidence rates of CINV caused by multiple-day chemotherapy regimens commonly administered for different types of breast cancer [Citation76Citation81].

Table 4. Incidence rates of nausea and vomiting induced by multiple-day chemotherapies commonly used for breast cancer in clinical practice, as reported in the scientific literature.

The oral version of cyclophosphamide, methotrexate and 5-fluorouracil with a concomitant 14-day administration of cyclophosphamide has shown moderate potential for causing nausea/vomiting [Citation82].

Numerous other regimens were associated with a low emetogenic potential: oral hormonal therapy (tamoxifen, aromatase inhibitors, and megestrol) [Citation83,Citation84], targeted therapy with lapatinib or everolimus [Citation85,Citation86], palbociclib or ribociclib plus letrozole (as first-line therapy in metastatic hormone receptor-positive patients) [Citation87,Citation88], and abemaciclib plus letrozole/anastrazole [Citation89].

2.3.1. Unmet needs in the control of nausea and vomiting in this patient population

CINV is still a critical issue in breast cancer patients. As outlined herein, numerous chemotherapeutic agents are commonly used for breast cancer, including new agents, and they have emetogenic potential, particularly when administered in combination regimens. The high risk for emesis in this group may be due not only to chemotherapy but also to patient-specific risk factors (female, often aged <50 years old, and anxious about their diagnosis and disease prognosis). Although guidelines recommend a triple antiemetic combination (consisting of a neurokinin 1 receptor antagonist (NK1 RA), 5HT3 RA and dexamethasone ± olanzapine) in patients receiving an AC-based regimen, evidence-based guidelines lack recommendations that address CINV related to other antineoplastic schemes, highlighting the need for specific antiemetic strategies to control this unpleasant symptom.

2.4. CINV in upper gastrointestinal tract cancer patients

2.4.1. Gastric cancer

In the ToGA trial, the incidences of nausea and vomiting were similar in patients receiving trastuzumab plus chemotherapy and those receiving chemotherapy alone () [Citation90], demonstrating the minimal emetic potential of trastuzumab.

Table 5. Incidence rates of nausea and vomiting induced by multiple-day chemotherapies commonly used for gastric cancer in clinical practice, as reported in the scientific literature.

Vascular endothelial growth factor receptor 2 (VEGFR-2) plays a role in gastric cancer pathogenesis and progression. The RAINBOW study explored the role of ramucirumab, a monoclonal antibody directed against VEGFR-2, with or without paclitaxel. Overall survival was longer in the exploratory arm than the control arm, and the regimen was associated with a low rate of gastrointestinal toxicity, especially in terms of CINV (grade 3 nausea: 2.0%; ) [Citation91].

In addition to CINV, patients with this disease may also have an increased risk of nausea and vomiting due to the risk of esophageal-gastric obstruction.

2.4.2. Pancreatic cancer

In a trial in patients with pancreatic cancer, the rates of vomiting (grade 3–4) were 14.5% with FOLFOXIRI (irinotecan, oxaliplatin, 5-fluorouracil, and leucovorin) and 8.3% with gemcitabine [Citation92]. In another trial, there was no grade 3–4 nausea and vomiting in patients receiving nanoparticle albumin-bound (nab)-paclitaxel in combination with gemcitabine; this result is unsurprising, given the low emetogenic potential of these agents [Citation93].

In patients with pancreatic cancer, disease-related factors such as bowel obstruction and cachexia can influence the incidence and severity of CINV.

2.4.3. Biliary tract cancer

In one trial, when patients were randomly assigned to receive cisplatin (25 mg/m2) plus gemcitabine (1,000 mg/m2) on day 1 and 8 every 3 weeks or gemcitabine alone, rates of nausea and vomiting were very low in both arms (about 4.0–5.0%) [Citation94]; given that cisplatin is considered to be highly emetogenic, regardless of dose, this result is surprising.

2.4.4. Unmet needs in the control of nausea and vomiting in the upper gastrointestinal tract cancer population

Overall, the most common combination regimens used for upper gastrointestinal malignancies are characterized by a moderate gastrointestinal toxicity profile, especially when oxaliplatin is included instead of cisplatin [Citation95].

However, patients suffering from upper gastrointestinal cancer may be particularly susceptible to complications arising from poorly controlled CINV, such as dehydration, nutrient depletion, metabolic imbalances, and performance status deterioration, all of which may impair their quality of life. These conditions sometimes interfere with chemotherapy by causing dose reductions, delays, or discontinuations [Citation96]. Therefore, CINV can exacerbate concurrent symptoms caused by the disease itself, and is more likely to develop when combination chemotherapy regimens are used.

2.5. CINV in colorectal cancer patients

According to the 2016 MASCC/ESMO guideline, the agents used in the treatment of metastatic colorectal cancer (CRC) are classified as moderate risk (irinotecan and oxaliplatin), low risk (5-fluorouracil), and minimal risk (bevacizumab and cetuximab) for CINV causation [Citation16]. The FOLFOXIRI regimen may be more appropriately considered as moderate/high risk; however, being a combination chemotherapy regimen, the emetogenic potential of this combination is not specified in the antiemetic guidelines.

The proportion of patients who experienced nausea and vomiting while receiving multiple-day regimens in clinical studies are shown in [Citation97Citation105]. Other possible risk factors for CINV in colorectal cancer patients are bowel obstruction or electrolyte imbalances.

Table 6. Incidence rates of nausea and vomiting induced by multiple-day chemotherapies commonly used for colorectal cancer in clinical practice, as reported in the scientific literature.

2.5.1. Unmet needs in the control of nausea and vomiting in this patient population

International guidelines recommend using 5-HT3 RA (day 1) in combination with dexamethasone (days 1–3) to prevent CINV associated with chemotherapy with moderate emetic risk, with the exception of carboplatin, for which guidelines recommend using a 3-drug antiemetic regimen, including an NK1 RA [Citation8,Citation16]. The most recent guidelines from the NCCN recommend that NK1 RAs should be given selectively to patients with additional risk factors or to those who experienced CINV during previous therapy while receiving two-drug antiemetic medication [Citation9].

There is a paucity of evidence from the literature on the management of delayed nausea and vomiting in patients treated with oxaliplatin- or irinotecan-based chemotherapy regimens.

In a prospective study, oxaliplatin-induced delayed nausea in 10% of patients. The use of a 5-HT3 RA and dexamethasone prior to oxaliplatin resulted in excellent control of nausea and vomiting during the 24 h after chemotherapy (complete response in 90% of patients). However, without further antiemetic treatments, only 54% had a complete response during the delayed phase [Citation106]. Therefore, routine antiemetic prophylaxis for delayed nausea and vomiting following oxaliplatin-based chemotherapy should be recommended. The use of dexamethasone on days 2 to 3, granisetron transdermal patch 24 h before chemotherapy or palonosetron on day 1 of therapy can be considered [Citation16,Citation107].

A 2011 study demonstrated that irinotecan has a modest tendency to cause delayed nausea and vomiting. Without any further antiemetic treatment, most patients (82%) do not experience delayed emesis or require rescue antiemetics. Routine prophylaxis for delayed emesis following irinotecan does not appear to be warranted. As observed in other studies, patients not achieving a complete response during the first 24 h after chemotherapy may have a higher risk of delayed emesis [Citation108]. Therefore, patients not achieving a complete response during the first 24 h should receive prophylaxis for delayed nausea and vomiting similar to that used for patients treated with oxaliplatin-based regimes.

2.6. CINV in gynecological cancer patients

Many of these patients are treated with highly emetogenic (cisplatin based) and moderately emetogenic (carboplatin based) chemotherapy. Additionally, factors that can increase their risk of emesis include that these patients are female, and many of them have abdominal disease (e.g. ovarian or cervical cancer) and are at increased risk for bowel obstruction. Pain management medications such as opioid analgesics, which are often required in these patients, can also worsen nausea and vomiting.

In a post-hoc subset analysis of two trials, the fixed-combination antiemetic netupitant/palonosetron plus dexamethasone was effective at preventing CINV in patients with gynecological cancers who were receiving cisplatin- or carboplatin-based chemotherapy; the proportion of patients experiencing no significant nausea was >90% in the acute phase and >80% in the delayed phase [Citation109].

2.7. CINV in soft-tissue sarcoma patients

In our clinical experience, the emetogenic potential of chemotherapy for soft-tissue sarcoma (STS) varies depending on whether they are used as single agents (e.g. moderate for anthracyclines, ifosfamide, and trabectedin, low for paclitaxel and pazopanib) or as combination regimens: high for ifosfamide + epirubicin, etoposide + ifosfamide, and doxorubicin + dacarbazine; moderate for ifosfamide + epirubicin and gemcitabine + docetaxel. The addition of olaratumab to doxorubicin or its use as maintenance monotherapy does not require a modification of the antiemetic prophylaxis, since olaratumab is characterized by a low emetogenic potential [Citation9].

According to the ASCO, MASCC/ESMO and NCCN guidelines, the recommended antiemetic prophylaxis for a highly emetogenic chemotherapy includes the use of three drugs (dexamethasone, single dose of 5-HT3 RA and NK1 RA) in the acute phase and, in the late phase, of dexamethasone (from day 2 to day 4) + aprepitant, depending on the NK1 RA used in the acute phase [Citation8,Citation9,Citation16].

Alternatively, in patients receiving moderate emetic risk chemotherapy, the combination of 5-HT3 RA + dexamethasone is used as CINV prophylaxis during the acute phase and dexamethasone alone is administered in the late phase (days 2 and 3) [Citation8,Citation9,Citation16].

2.7.1. Unmet needs in the control of nausea and vomiting in this patient population

Sometimes, STS patients are treated with alternating combination chemotherapies, resulting in the frequent occurrence of intercycle nausea. The issue of how best to minimize intercycle nausea is still a matter of debate, as is the optimal approach to rescue therapy in order to maximize adherence to chemotherapy. Furthermore, STS patients are often young adults; their young age and possible high anxiety regarding prognosis and treatment outcomes can increase their risk of CINV.

2.8. CINV in melanoma patients

Adjuvant therapy is historically based on interferon for which, depending on the dosage, antiemetic prophylaxis can be avoided [Citation110]. Similarly, no specific prophylaxis is required in patients receiving newer immunotherapy agents (ipilimumab, pembrolizumab, nivolumab, and their combinations) [Citation8,Citation9,Citation16].

Targeted treatments, such as BRAF inhibitor monotherapy (dabrafenib, vemurafenib) are deemed to have a low or minimal emetogenic potential and therefore require the administration of a single antiemetic drug for acute phase prophylaxis (dexamethasone, 5-HT3 RA, or metoclopramide) and no therapy in the delayed phase [Citation8,Citation9,Citation16]. The mitogen-activated protein kinase inhibitors cobimetinib and trametinib are not covered in the current guidelines; however, as they also have low-to-minimal emetogenic risk, the same anti-emetic approach may be used.

In light of recent innovations, the role of chemotherapy in melanoma is more marginal and the drugs used, such as dacarbazine (high emetic risk), fotemustine and temozolomide (moderate) and taxanes (low), should be administered with appropriate prophylaxis according to the emetogenic risk as described in the existing guidelines [Citation8,Citation9,Citation16].

2.8.1. Unmet needs in the control of nausea and vomiting in this patient population

The impact of therapeutic innovation on the management of CINV has been much lower in melanoma than in other cancers, given the limited emetic effect of the new drugs used.

2.9. CINV in myelodysplastic syndrome patients

Myelodysplastic syndrome (MDS) is a heterogeneous group of hematologic diseases resulting from a clonal disorder of pluripotent hematopoietic stem cells [Citation111,Citation112]. MDS is classified as either primary (de novo) or secondary; in the latter, it is possible to identify a previous hemopathy or exposure to myelotoxic substances.

Drugs used in low-risk/intermediate-1 risk MDS and immunomodulatory agents such as lenalidomide () are not emetogenic. Demethylating agents () are characterized by a moderate emetic risk. Nausea induced by these drugs may be managed with a prophylactic regimen of oral antiemetic agents (ondansetron/granisetron/metoclopramide) before starting on the 7 days of subcutaneous chemotherapy administration.

2.9.1. Unmet needs in the control of nausea and vomiting in this patient population

Many patients with myelodysplasia are elderly and already have concomitant conditions that negatively impact their quality of life and require daily administration of multiple medications. For these patients, offering antiemetic prophylaxis via an alternative route to oral therapy may be convenient and advantageous.

2.10. CINV in acute myeloid leukemia patients

The number of studies on nausea and vomiting in patients with acute myeloid leukemia (AML) is limited () [Citation113]. It has been observed that more than 70% of patients have no nausea in the first 24 h of chemotherapy and about 50% experience emesis in the following days [Citation114]. Mattiuzzi and colleagues compared the effect of ondansetron and palonosetron in patients treated with high doses of cytarabine, and found no significant difference in the incidence of acute nausea between the two treatment groups; however, patients treated with palonosetron had significantly less delayed nausea compared with the group receiving ondansetron [Citation114].

Table 7. Response rates to antiemetic regimens in patients with hematologic malignancies (including acute myeloid leukemia) receiving multiple-day chemotherapy, as reported in the scientific literature.

According to recent guidelines, in patients receiving moderately or highly emetogenic multiday chemotherapies (fludarabine in combination with cytosine arabinoside and idarubicin at intermediate/high doses), it is advisable to use an NK1 RA in conjunction with a 5-HT3 RA and dexamethasone [Citation8,Citation9,Citation16]. However, prolonged use of dexamethasone should be avoided because of the well-established side effects (insomnia, agitation, etc.) and increased susceptibility to infections [Citation115].

In older patients treated with demethylating agents, nausea is managed with prophylactic antiemetic treatment using ondansetron, granisetron, or metoclopramide administered intravenously or orally before and during the course of chemotherapy.

2.10.1. Unmet needs in the control of nausea and vomiting in this patient population

Because of the advanced age of most AML patients, they frequently have concomitant diseases requiring multiple oral medications, which can negatively affect their quality of life. For these patients, the availability of an alternative route to oral therapy would therefore be useful.

Breakthrough emesis is still difficult to manage and is often irreversible. In such cases, antiemetic administration via the oral route is not feasible, so intravenous, transdermal, or rectal administration is required.

2.11. CINV in acute lymphoblastic leukemia patients

In patients with acute lymphoblastic leukemia (ALL), the intensive therapeutic approach consists of active drugs with different mechanisms of action and administered at their maximum dose, to achieve a rapid reduction in the tumor mass and avoid inducing resistance.

There is limited information on nausea/vomiting in hematologic diseases, particularly concerning CINV in ALL. Nausea is managed with prophylactic antiemetics (ondansetron, granisetron, or metoclopramide) administered intravenously or orally before and during chemotherapy [Citation10,Citation116,Citation117].

The ondansetron and aprepitant combination appears to be effective at preventing nausea and vomiting in patients undergoing multiday moderately/highly emetogenic chemotherapy [Citation10,Citation116,Citation117].

3. RINV in different pathological conditions

3.1. RINV in patients with brain, head and neck, thorax and upper trunk cancers

3.1.1. Overview

RINV is a common and troublesome side effect experienced by patients undergoing RT. It is commonly believed to be an exclusive or, at least, prevalent symptom of abdomen RT, but a significant percentage of patients undergoing RT in other parts of the body, like head and neck and thorax, also experience RINV. In this context, at the beginning of the 1990s, Scarantino et al. suggested that serotonin could play a role in mediating RINV induced by RT in the upper body: they documented an increase in urinary levels of the serotonin active metabolite 5-hyroxyindoleacetic acid following emetogenic upper and mid hemibody irradiation, and the efficacy of 5-HT3 RAs in the prevention of RINV [Citation118,Citation119].

3.1.2. Radiation therapy and incidence rates of RINV

According to the MASCC Consensus Conference on Antiemetic Therapy, the emetogenic potential of radiation alone is classified as ‘low’ (30–60%) for RT of the brain, head and neck, and thorax, and as ‘minimal’ for RT of the breast (<30%) [Citation11,Citation12]. However, these guidelines were prepared before the implementation of intensity-modulated radiation therapy (IMRT) and have not been significantly updated since 2016 [Citation16].

In the IGARR study, a prospective observational multicenter trial of 1020 patients treated with RT (with or without chemotherapy) at 45 Italian radiation oncology centers, nausea, and vomiting were most common in patients treated for brain cancer, followed by those treated for cancer of the thorax, head and neck, and breast () [Citation19].

Table 8. Incidence rates of nausea and vomiting induced by radiation therapy in patients with brain, head and neck, thorax, and upper trunk cancers, as reported in the scientific literature.

The pathophysiological mechanism of RINV in patients with brain cancer or HNC is probably linked to a release of serotonin from the chemoreceptor trigger zone in the brainstem (the area postrema and dorsal vagal complex) when it is included in the radiation portals [Citation120Citation122]. In Rosenthal’s experience [Citation120], the incidence of nausea and vomiting is associated with irradiation of the area postrema at a dose >36 Gy for patients treated using IMRT [Citation120]. This observation was confirmed by Ciura et al. [Citation122], who suggested that RINV developed around the second week of treatment, with a possible correlation between dose and toxicity in the range of 15 Gy to 25 Gy. In both these experiences, the incidence of RINV in patients with HNC was higher when IMRT was combined with emetogenic chemotherapy compared with the incidence typically reported for the same chemotherapy used in combination with 3D conformational RT or compared with IMRT alone (). Kocak-Uzel and colleagues [Citation123] have also shown that, in patients with HNC receiving definitive IMRT, the development of RINV was related to the dose of radiation delivered to specific CNS non-target structures, including the area postrema and dorsal vagal complex [Citation123]. Practical contouring guidelines of brainstem structures involved in the occurrence of RINV (according to a magnetic resonance-based atlas) have been recently published [Citation124].

Unlike what we know about the causes of RINV in brain and head and neck RT, the cause and mechanisms of RINV are still unclear in patients undergoing chest RT when the upper abdomen is not included in the treatment volume. The irradiated volume is one determinant of risk for RINV, in addition to irradiation site, gender, previous or concurrent chemotherapy. IMRT and volumetric modulated arc therapy (VMAT), which are increasingly used for cancer treatment, can reduce acute toxicity by decreasing the radiation doses to uninvolved healthy tissue near the tumor targets; however, previously unaffected tissues, such as the celiac plexus and gastroesophageal junction (GEJCP) for breast or thorax cancers, may receive clinically significant doses that lead to side effects such as nausea and vomiting [Citation125].

After having observed unanticipated RINV in some breast cancer patients treated with postoperative upper trunk IMRT-VMAT, an Italian study group hypothesized that these symptoms could be attributed to an unduly dispensed dose to the upper abdominal anatomical structures underlying the planning target volume [Citation125]. In a retrospective analysis of standard weekly management visit forms, these researchers found >60% of patients with breast cancer experienced acute RINV (). Using the original planning computed tomography (CT) scans, the researchers retrospectively contoured a volume containing the anatomical structures relevant to the emesis-related vagal parasympathetic afferent pathways, such as the GEJCP. RINV was significantly related to a maximal dose >10 Gy and a mean dose <3 Gy to the GEJCP (P < 0.001). The development of RINV was weakly correlated with irradiation of the left breast (P < 0.01) and a planned treatment volume >700 cm3 (P < 0.03), but no correlation was observed with age, previous systemic therapy, or nodal irradiation [Citation125].

Palliative hypofractionated radiotherapy (8 Gy/1 fraction or 20 Gy/5 fractions) of the spine has been classified as a moderately emetogenic treatment schedule. Dexamethasone monotherapy given on the day of irradiation is not sufficient to control delayed nausea and vomiting; a prospective pilot study suggested that the combination of aprepitant and granisetron may provide more effective prophylaxis [Citation126].

3.1.3. Unmet needs in the control of nausea and vomiting in this patient population

The literature on RINV related to the new radiation techniques, such as IMRT and VMAT, is still not extensive. The VMAT modality has become an increasingly implemented radiation technique for cancer, but the paths of the arc beams often cross healthy structures that would not be directly irradiated if using 3D-conformational RT techniques. This effect is responsible for a new acute morbidity profile not seen before the IMRT era [Citation125]. Well-designed prospective randomized trials are needed to investigate (1) how modern radiotherapy modalities induce these symptoms, (2) the maximum doses' targets can receive to avoid these side effects, (3) suitable supportive therapy to prevent or minimize discomfort in patients. Such studies will help to identify treatment planning solutions that can be integrated into future IMRT VMAT treatments.

3.2. RINV in gastrointestinal cancer patients

To date, the largest observational studies on RINV have been conducted by the IGARR group and evaluated a total of 1,934 patients [Citation19,Citation20]. Within these studies, 29% of the patients who received RT to the upper abdomen reported vomiting and 56% experienced nausea. In a smaller observational series, 63% of patients treated with abdominal or pelvic RT experienced nausea [Citation127]. In a prospective study including 45 patients with gastrointestinal cancers undergoing abdominal and/or pelvic RT with curative or palliative intent, alone or with concomitant chemoradiotherapy, nausea was reported in 83% of patients and emesis in 54% [Citation128].

3.2.1. Unmet needs in the control of nausea and vomiting in this patient population

The neurotransmitter serotonin is thought to be the most important chemical mediator of RINV. The gastrointestinal tract houses approximatively 95% of the body’s serotonin and is an important anatomic region related to RINV [Citation129]. According to antiemetic practice guidelines, radiation treatments to the upper abdomen are considered moderately emetogenic, with an estimated risk of 60–90% [Citation12,Citation16]. In patients receiving radiotherapy with moderate emetogenic risk, antiemetic prophylaxis using a 5-HT3 RA is recommended, with the option of combining this with a short course (day 1–5) of dexamethasone [Citation12,Citation16].

As stated in the guidelines [Citation12,Citation16], although all chemotherapy drugs associated with RT for the treatment of cancer of this body region belong to the intermediate-low risk class, the antiemetic prophylaxis strategy should take into account the intermediate risk of RT itself [Citation12,Citation16].

Over the last two decades, the most extensively used antiemetics for RINV in clinical practice have been the 5-HT3 RAs. Many trials have reported that, in patients receiving upper abdominal irradiation, 5-HT3 RAs provided significantly greater protection against RINV than metoclopramide, phenothiazines, or placebo [Citation130,Citation131].

The side effects of 5-HT3 RAs are generally mild and mainly consist of headache, constipation, diarrhea, and weakness. Furthermore, it has been suggested that these agents may reduce the frequency of diarrhea, a debilitating adverse effect of RT for acute gastrointestinal cancers [Citation132].

The efficacy of antiemetic drugs in the treatment of RINV has been studied in a few randomized clinical trials, and it seems clear that the prevention of RINV must be preferred to a rescue intervention [Citation132].

However, the appropriate duration of antiemetic prophylaxis for patients receiving fractionated RT has not been determined, since there are no randomized trials comparing a five-day course of 5-HT3 RA treatment with a more prolonged period [Citation132]. Nonetheless, according to a systematic review including 25 randomized and non-randomized trials, 5-HT3 RAs appear to be most commonly administered for the entire phase of radiotherapy [Citation132]. In short, the duration and the timing of therapy with this class of antiemetics have not yet been defined and are additional unmet needs in this field of research [Citation132].

4. Suggestions for a practical approach to CINV and RINV

The therapeutic armamentarium offers several options and strategies to relieve CINV and RINV for particular cases and conditions commonly observed in clinical practice, basing on innovative drugs or on new formulations of known compounds. For what concerns the latter case, we focused our attention on transdermal granisetron as an example of possible advantages given by an ‘old’ drug given in a different way, applied in different diseases. Based on the authors’ experience in managing CINV and RINV, several suggestions are provided below.

We recommend that antiemetic therapy should be targeted to the patient’s needs, based on the emetogenic potential of their antineoplastic regimen, as defined by the antiemetic guidelines, along with the characteristics of the disease and the patient’s risk factors.

4.1. HNC treated concurrently with chemotherapy and RT

Patients with HNC often experience swallowing difficulties related to tumor location and treatment consequences, which may require a feeding tube in the last weeks of treatment. In such circumstances, oral formulations of antiemetic drugs are often difficult to administer, affecting compliance. Using an alternative route of antiemetic administration could overcome this problem, and deserves further clinical trials.

The transdermal formulation of the 5-HT3 RA granisetron avoids the oral route of administration and could improve the management of CINV in patients with HNC, by improving compliance in patients with swallowing difficulties, allowing them to receive optimal antiemetic dose intensity and protection from symptoms. This form of antiemetic therapy could also benefit patients who are experiencing symptoms related to RT (sticky saliva, mucositis, tube feeding, etc.) by providing sustained protection against nausea and vomiting without the need to swallow a tablet or capsule. Further studies are needed to better position the use of transdermal granisetron in the prevention and treatment of CINV in patients with HNC, including evaluating its effects on quality of life.

4.2. Upper gastrointestinal malignancies

For the most used RT regimens in these conditions, the use of 5-HT3 RAs and dexamethasone seems to be adequate to control RINV. For schedules with extended exposure to chemotherapy, there may be potential for 5-HT3 RAs characterized by a prolonged action, such as the granisetron transdermal patch, to be used. However, studies are needed to explore this potential.

4.3. STS

The granisetron transdermal patch may be considered appropriate in patients with sarcoma undergoing highly emetogenic polychemotherapy administered over several days, particularly in patients who have difficulty taking oral medications or have a previous experience of nausea and vomiting not adequately controlled by the prescribed antiemetic prophylaxis.

4.4. MDS

The transdermal formulation of granisetron may be an alternative route to oral therapy. It could be applied on the day before demethylating therapy commences and kept in place for up to 7 days.

This formulation does not require any special recommendations and is extremely useful for elderly patients with swallowing dysfunction, mucositis, gastropathy, and anorexia, and may help to improve the quality of life of patients who are already taking multiple oral medications.

4.5. Brain, head and neck, thorax and upper trunk cancers treated with RT

In everyday practice, patients treated exclusively with RT on the brain usually receive prophylactic dexamethasone.

RINV prophylaxis is not usually prescribed to patients irradiated for breast or chest neoplasms (lung, thymus, and upper esophagus); these patients usually receive antiemetics only at the onset of symptoms. When chemotherapy is associated with radiotherapy, RINV prophylaxis should follow the guidelines provided for the most emetogenic treatment. In these cases, the administration of transdermal granisetron is preferred in patients with swallowing difficulties.

4.6. Gastrointestinal cancer treated with RT

The AVERT study, a multicenter phase II trial, assessed the effectiveness, safety, and tolerability of protracted dual NK1 RA and 5-HT3 RA as RINV prophylaxis in patients receiving RT to the upper abdomen and radiosensitizing chemotherapy. The authors concluded that RINV still represents a serious morbidity in patients receiving RT to the upper abdomen and that aprepitant plus ondansetron, as dosed in this trial, were not superior to standard ondansetron monotherapy. The authors noted that other potential prophylactic options may be considered for future investigations, such as the granisetron transdermal patch, intravenous fosaprepitant, olanzapine, and alternative NK1 RA formulations (e.g. the combination of netupitant and palonosetron and rolapitant as a single agent) [Citation133].

4.7. Quadruple antiemetic combinations

Several studies have explored the potential benefit of combining four antiemetic agents to alleviate CINV in patients undergoing highly emetogenic chemotherapy (HEC; ). In a randomized, double-blind, phase III trial in 380 chemotherapy-naïve patients with malignancies receiving HEC, the addition of olanzapine significantly improved the antiemetic efficacy of the three-drug regimen of a 5-HT3 RA, an NK1 RA and dexamethasone () [Citation134]. Based on the improvements achieved with this new quadruple regimen, the ASCO and NCCN antiemetic guidelines included this therapeutic option in their recommendations [Citation8,Citation9].

Table 9. Efficacy of quadruple versus triple antiemetic therapy in patients receiving highly emetogenic chemotherapy.

In a single-blind, randomized trial in 97 chemotherapy-naïve patients with breast cancer undergoing HEC, the addition of metoclopramide to antiemetic prophylaxis with palonosetron, aprepitant, and dexamethasone significantly improved the complete response rate () [Citation135].

The efficacy of multi-drug combinations in managing CINV probably arises from their ability to inhibit the emetic response at multiple sites [Citation136].

5. Conclusions

Many cancer patients are affected by CINV and RINV, and these conditions are two of the major barriers to patient acceptance of antineoplastic treatments. Nonetheless, continuous efforts and scientific advances are being made to further develop novel therapeutic options, formulations, and combination strategies of antiemetic therapies. These will certainly contribute to extend the availability of effective antiemetic treatments in the near future, with the ultimate objective of providing promising solutions tailored to suit the condition and the needs of each individual patient.

6. Expert opinion

CINV and RINV are two of the most troublesome issues that patients experience during anticancer treatment, with a high impact on quality of life. Despite the great efforts and improvements made in recent years, many areas of uncertainty and unsolved issues still exist.

First of all, research should be tailored to identify more precise treatment-, patient-, and disease-specific risk factors for the development of nausea and vomiting. Until now, only limited clinical predictive factors have been identified, but these have not yet been included in the guidelines to differentiate preventive strategies. In this regard, prospective studies aimed at personalizing antiemetic therapies could be the next step in the landscape of a tailored approach to supportive care in cancer. Moreover, the impact of genomic predisposition to CINV and RINV should be explored, by defining categories of risk and identifying possible new targets for improving treatment of these symptoms.

Secondly, the cluster of symptoms linked to nausea and vomiting should be better explored, in order to provide new information about the associated patterns of toxicity. It is plausible that a better control of CINV and RINV may positively impact other symptoms, such as taste and swallowing, fatigue, anxiety, and depression. An analysis of the patient-reported outcome (PRO) measures, listing each symptom as perceived by the patients themselves, would allow for a comprehensive assessment. Moreover, the analysis of adverse events should not be limited to intensity or frequency, but should also evaluate the duration of symptoms of toxicity. For CINV and RINV, one would assume that low-grade toxicities lasting several days or weeks would deeply affect the quality of life of patients, however this analysis is often lacking in many studies.

Thirdly, greater attention should be devoted to nausea and vomiting with targeted agents, particularly when administered orally for a long time. Some agents are associated with long-lasting nausea, which bothers patients. The impact of nausea with targeted therapy should be studied more broadly; in fact, while there are data on the greater impact of nausea on patient quality of life compared with the impact of vomiting, this information is lacking for targeted agents [Citation137]. Greater efforts are needed to understand how to prevent and treat nausea in such cases, as these treatments are often used for periods lasting months or years.

Finally, the studies of new drug compounds or combinations should proceed in parallel with the evaluation of different schedules (intensified or de-intensified) of antiemetics and of alternative ways of drug administration. Moreover, the study of non-pharmacological therapies should be encouraged, with the aim of reducing CINV/RINV symptoms. Non-pharmacological approaches equally deserve to be studied in a rigorous manner, using well-designed trials, to precisely define the role of such strategies in controlling nausea and vomiting.

Article highlights

  • The international antiemetic guidelines only partially consider the emetogenic risk of combination chemotherapy regimens.

  • Guidelines for treatment of RINV have not yet been updated to consider the use of modern technologies, taking into account different risks and the corresponding preventive approaches.

  • The prevention of nausea and vomiting in the continuous administration of oral chemotherapy or targeted agents is another unmet need.

  • Antiemetic therapy should be targeted to individual patients’ needs, based on the emetogenic potential of their antineoplastic regimen, as well as patient- and disease-specific risk factors.

  • New approaches in prevention and treatment of CINV and RINV may derive from new drugs, new combinations or the use of different formulations.

This box summarizes key points contained in the article.

Author contributions

P Bossi was involved with the conception, design of the study, and in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. M Airoldi was involved with the conception, design of the study and in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. MA Aloe Spiriti was involved with the conception, design of the study and in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. A Antonuzzo was involved with the conception, design of the study and in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. G Bonciarelli was involved in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. A Campagna was involved in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. A Cassano was involved in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. R Murialdo was involved in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. D Musio was involved in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission. G Silvano was involved in the analysis and interpretation of the data, drafting, and critically revising the manuscript and final approval of the manuscript before submission.

Declaration of interest

P Bossi has disclosed that they have had a consulting or advisory role for Angelini, AstraZeneca, Bristol-Myers Squibb, Kyowa Kirin, Merck Serono, MSD Oncology, Roche and Sanofi/Regeneron. M Airoldi has disclosed that they have had a consulting or advisory role for Angelini, Bristol-Myers Squibb, Elsevier, Merck Serono and Novartis. A Antonuzzo has disclosed that they have had a consulting or advisory role for Kyowa Kirin. D Musio has disclosed that they have had a consulting or advisory role for Kyowa Kirin. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgments

The authors thank Brunilde Iovene, an independent medical writer, who provided editorial assistance during manuscript preparation on behalf of Springer Healthcare Communications. They also thank Catherine Rees, an independent medical writer on behalf of Springer Healthcare Communications, who styled the manuscript for submission. Support for this assistance was funded by Kyowa Kirin Srl (Milan, Italy).

Additional information

Funding

This paper was not funded.

References

  • Barbour SY. Management of patients with chemotherapy-induced nausea and vomiting. J Adv Pract Oncol. 2017;8:303–308.
  • The Antiemetic Subcommittee of the Multinational Association of Supportive Care in Cancer (MASCC). Prevention of chemotherapy- and radiotherapy-induced emesis: results of Perugia consensus conference. Ann Oncol. 1998;9:811–819.
  • Vidall C, Fernández-Ortega P, Cortinovis D, et al. Impact and management of chemotherapy/radiotherapy-induced nausea and vomiting and the perceptual gap between oncologists/oncology nurses and patients: a cross-sectional multinational survey. Support Care Cancer. 2015;23:3297–3305.
  • Feyer P, Jordan K. Update and new trends in antiemetic therapy: the continuing need for novel therapies. Ann Oncol. 2011;22:30–38.
  • Sasaki H, Tamura K, Naito Y, et al. Patient perceptions of symptoms and concerns during cancer chemotherapy: ‘affects my family’ is the most important. Int J Clin Oncol. 2017;22:793–800.
  • Grunberg SM, Deuson RR, Mavros P, et al. Incidence of chemotherapy-induced nausea and emesis after modern antiemetics. Cancer. 2004;100:2261–2268.
  • Razvi Y, Chan S, McFarlane T, et al. ASCO, NCCN, MASCC/ESMO: a comparison of antiemetic guidelines for the treatment of chemotherapy-induced nausea and vomiting in adult patients. Support Care Cancer. 2019;27:87–95.
  • Hesketh PJ, Kris MG, Basch E, et al. Antiemetics: American society of clinical oncology clinical practice guideline update. J Clin Oncol. 2017;35:3240–3261.
  • National Comprehensive Cancer Network (NCCN). NCCN (National Comprehensive Cancer Network®) clinical practice guidelines in oncology (NCCN guidelines®). Antiemesis. Version 1.2019. [updated 2019 Feb 28; cited 2019 Mar 3]. Available from: http://www.nccn.org/professionals/physician_gls/pdf/antiemesis.pdf
  • Patel P, Robinson PD, Thackray J, et al. Guideline for the prevention of acute chemotherapy-induced nausea and vomiting in pediatric cancer patients: a focused update. Pediatr Blood Cancer. 2017;64:e26542.
  • Maranzano E, Feyer P, Molassiotis A, et al. Evidence-based recommendations for the use of antiemetics in radiotherapy. Radiother Oncol. 2005;76:227–233.
  • Feyer PC, Maranzano E, Molassiotis A, et al. Radiotherapy-induced nausea and vomiting (RINV): MASCC/ESMO guideline for antiemetics in radiotherapy: update 2009. Support Care Cancer. 2011;19(Suppl 1):S5–14.
  • Grunberg SM, Osoba D, Hesketh PJ, et al. Evaluation of new antiemetic agents and definition of antineoplastic agent emetogenicity–an update. Support Care Cancer. 2005;13:80–84.
  • Navari RM. Pathogenesis-based treatment of chemotherapy-induced nausea and vomiting–two new agents. J Support Oncol. 2003;1:89–103.
  • Dranitsaris G, Molassiotis A, Clemons M, et al. The development of a prediction tool to identify cancer patients at high risk for chemotherapy-induced nausea and vomiting. Ann Oncol. 2017;28:1260–1267.
  • Roila F, Molassiotis A, Herrstedt J, et al. 2016 MASCC and ESMO guideline update for the prevention of chemotherapy- and radiotherapy-induced nausea and vomiting and of nausea and vomiting in advanced cancer patients. Ann Oncol. 2016;27:v119–v33.
  • Naeim A, Dy SM, Lorenz KA, et al. Evidence-based recommendations for cancer nausea and vomiting. J Clin Oncol. 2008;26:3903–3910.
  • Poon M, Hwang J, Dennis K, et al. A novel prospective descriptive analysis of nausea and vomiting among patients receiving gastrointestinal radiation therapy. Support Care Cancer. 2016;24:1545–1561.
  • Maranzano E, De Angelis V, Pergolizzi S, et al. A prospective observational trial on emesis in radiotherapy: analysis of 1020 patients recruited in 45 Italian radiation oncology centres. Radiother Oncol. 2010;94:36–41.
  • The Italian Group for Antiemetic Research in Radiotherapy. Radiation-induced emesis: a prospective observational multicenter Italian trial. The Italian group for antiemetic research in radiotherapy. Int J Radiat Oncol Biol Phys. 1999;44:619–625.
  • Huber RM, Tufman A. Update on small cell lung cancer management. Breathe. 2012;8:315–330.
  • National Cancer Institute. Small cell lung cancer treatment (PDQ®). Health Professional Version. [cited 2019 Feb 24]. Available from: http://www.cancer.gov/types/lung/hp/small-cell-lung-treatment-pdq
  • Wallace E. Small cell lung cancer: staging, treatment and prognosis. WIN. 2012;20:43–44.
  • Bhoo-Pathy N, Verkooijen HM, Tan EY, et al. Trends in presentation, management and survival of patients with de novo metastatic breast cancer in a Southeast Asian setting. Sci Rep. 2015;5:16252.
  • Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med. 2018;379:111–121.
  • Hu B, El Hajj N, Sittler S, et al. Gastric cancer: classification, histology and application of molecular pathology. J Gastrointest Oncol. 2012;3:251–261.
  • Office for National Statistics. Cancer survival in England: adults diagnosed in 2009 to 2013, followed up to 2014 2015. [cited 2019 Mar 3]. Available from: http://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/stomach-cancer/survival#heading-One
  • American Cancer Society. Cancer facts & figures. 2019 [cited 2019 Mar 3]. Available from: http://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2019/cancer-facts-and-figures-2019.pdf
  • Lee J, Bass AJ, Ajani JA. Gastric adenocarcinoma: an update on genomics, immune system modulations, and targeted therapy. Am Soc Clin Oncol Educ Book. 2016;35:104–111.
  • Inadomi K, Kusaba H, Matsushita Y, et al. Efficacy and safety analysis of oxaliplatin-based chemotherapy for advanced gastric cancer. Anticancer Res. 2017;37:2663–2671.
  • Cunningham D, Starling N, Rao S, et al. Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 2008;358:36–46.
  • Kang YK, Kang WK, Shin DB, et al. Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: a randomised phase III noninferiority trial. Ann Oncol. 2009;20:666–673.
  • Weledji EP, Enoworock G, Mokake M, et al. How grim is pancreatic cancer? Oncol Rev. 2016;10:294.
  • Burris HA 3rd, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol. 1997;15:2403–2413.
  • Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817–1825.
  • Kim MJ, Oh DY, Lee SH, et al. Gemcitabine-based versus fluoropyrimidine-based chemotherapy with or without platinum in unresectable biliary tract cancer: a retrospective study. BMC Cancer. 2008;8:374.
  • Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.
  • Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. 2016;27:1386–1422.
  • Wibmer C, Leithner A, Zielonke N, et al. Increasing incidence rates of soft tissue sarcomas? A population-based epidemiologic study and literature review. Ann Oncol. 2010;21:1106–1111.
  • Wortman JR, Tirumani SH, Jagannathan JP, et al. Radiation therapy for soft-tissue sarcomas: a primer for radiologists. Radiographics. 2016;36:554–572.
  • Tap WD, Jones RL, Van Tine BA, et al. Olaratumab and doxorubicin versus doxorubicin alone for treatment of soft-tissue sarcoma: an open-label phase 1b and randomised phase 2 trial. Lancet. 2016;388:488–497.
  • Rastrelli M, Tropea S, Rossi CR, et al. Melanoma: epidemiology, risk factors, pathogenesis, diagnosis and classification. In Vivo. 2014;28:1005–1011.
  • Associazione Italiana di Oncologia Medica (AIOM). Linee guida: melanoma. Edizione. 2018 [cited 2019 Feb 24]. Available from: https://www.aiom.it/wp-content/uploads/2018/11/2018_LG_AIOM_Melanoma.pdf
  • Weber J, Mandala M, Del Vecchio M, et al. Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med. 2017;377:1824–1835.
  • Eggermont AMM, Blank CU, Mandala M, et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N Engl J Med. 2018;378:1789–1801.
  • Cheng L, Lopez-Beltran A, Massari F, et al. Molecular testing for BRAF mutations to inform melanoma treatment decisions: a move toward precision medicine. Mod Pathol. 2018;31:24–38.
  • Aul C, Bowen DT, Yoshida Y. Pathogenesis, etiology and epidemiology of myelodysplastic syndromes. Haematologica. 1998;83:71–86.
  • Williamson PJ, Kruger AR, Reynolds PJ, et al. Establishing the incidence of myelodysplastic syndrome. Br J Haematol. 1994;87:743–745.
  • Thein MS, Ershler WB, Jemal A, et al. Outcome of older patients with acute myeloid leukemia: an analysis of SEER data over 3 decades. Cancer. 2013;119:2720–2727.
  • Raut LS. Novel formulation of cytarabine and daunorubicin: a new hope in AML treatment. South Asian J Cancer. 2015;4:38–40.
  • Ferrara F, Pinto A. Acute myeloid leukemia in the elderly: current therapeutic results and perspectives for clinical research. Rev Recent Clin Trials. 2007;2:33–41.
  • Nanah R, McCullough K, Hogan W, et al. Outcome of elderly patients after failure to hypomethylating agents given as frontline therapy for acute myeloid leukemia: single institution experience. Am J Hematol. 2017;92:866–871.
  • Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. J Clin Oncol. 2012;30:2670–2677.
  • Inaba H, Greaves M, Mullighan CG. Acute lymphoblastic leukaemia. Lancet. 2013;381:1943–1955.
  • Sultan S, Irfan SM, Parveen S, et al. Acute lymphoblastic leukemia in adults - an analysis of 51 cases from a tertiary care center in Pakistan. Asian Pac J Cancer Prev. 2016;17:2307–2309.
  • Takeuchi J, Kyo T, Naito K, et al. Induction therapy by frequent administration of doxorubicin with four other drugs, followed by intensive consolidation and maintenance therapy for adult acute lymphoblastic leukemia: the JALSG-ALL93 study. Leukemia. 2002;16:1259–1266.
  • Kantarjian H, Thomas D, O’Brien S, et al. Long-term follow-up results of hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone (Hyper-CVAD), a dose-intensive regimen, in adult acute lymphocytic leukemia. Cancer. 2004;101:2788–2801.
  • Barry E, DeAngelo DJ, Neuberg D, et al. Favorable outcome for adolescents with acute lymphoblastic leukemia treated on Dana-Farber cancer institute acute lymphoblastic leukemia consortium protocols. J Clin Oncol. 2007;25:813–819.
  • Vignetti M, Fazi P, Cimino G, et al. Imatinib plus steroids induces complete remissions and prolonged survival in elderly Philadelphia chromosome-positive patients with acute lymphoblastic leukemia without additional chemotherapy: results of the Gruppo Italiano Malattie Ematologiche dell’Adulto (GIMEMA) LAL0201-B protocol. Blood. 2007;109:3676–3678.
  • Lazarus HM, Richards SM, Chopra R, et al. Central nervous system involvement in adult acute lymphoblastic leukemia at diagnosis: results from the international ALL trial MRC UKALL XII/ECOG E2993. Blood. 2006;108:465–472.
  • Chaturvedi AK, Engels EA, Pfeiffer RM, et al. Human papillomavirus and rising oropharyngeal cancer incidence in the United States. J Clin Oncol. 2011;29:4294–4301.
  • Chan A, Shwe M, Gan Y, et al. Trajectory and risk factors for chemotherapy-induced nausea and vomiting in Asian patients with head and neck cancer. Head Neck. 2015;37:1349–1357.
  • Camidge DR, Bang YJ, Kwak EL, et al. Activity and safety of crizotinib in patients with ALK-positive non-small-cell lung cancer: updated results from a phase 1 study. Lancet Oncol. 2012;13:1011–1019.
  • Gregor A, Drings P, Burghouts J, et al. Randomized trial of alternating versus sequential radiotherapy/chemotherapy in limited-disease patients with small-cell lung cancer: a European organization for research and treatment of cancer lung cancer cooperative group study. J Clin Oncol. 1997;15:2840–2849.
  • Lara PN Jr., Natale R, Crowley J, et al. Phase III trial of irinotecan/cisplatin compared with etoposide/cisplatin in extensive-stage small-cell lung cancer: clinical and pharmacogenomic results from SWOG S0124. J Clin Oncol. 2009;27:2530–2535.
  • Loehrer PJ Sr., Ansari R, Gonin R, et al. Cisplatin plus etoposide with and without ifosfamide in extensive small-cell lung cancer: a Hoosier oncology group study. J Clin Oncol. 1995;13:2594–2599.
  • Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol. 2017;18:1307–1316.
  • Pu D, Hou M, Li Z, et al. A randomized controlled study of chemotherapy: etoposide combined with oxaliplatin or cisplatin regimens in the treatment of extensive-stage small cell lung cancer in elderly patients. Zhongguo Fei Ai Za Zhi. 2013;16:20–24.
  • Socinski MA, Smit EF, Lorigan P, et al. Phase III study of pemetrexed plus carboplatin compared with etoposide plus carboplatin in chemotherapy-naive patients with extensive-stage small-cell lung cancer. J Clin Oncol. 2009;27:4787–4792.
  • Sun Y, Cheng Y, Hao X, et al. Randomized phase III trial of amrubicin/cisplatin versus etoposide/cisplatin as first-line treatment for extensive small-cell lung cancer. BMC Cancer. 2016;16:265.
  • Tiseo M, Boni L, Ambrosio F, et al. Italian, multicenter, phase III, randomized study of cisplatin plus etoposide with or without bevacizumab as first-line treatment in extensive-disease small-cell lung cancer: the GOIRC-AIFA FARM6PMFJM trial. J Clin Oncol. 2017;35:1281–1287.
  • Treat J, Huang CH, Lane SR, et al. Topotecan in the treatment of relapsed small cell lung cancer patients with poor performance status. Oncologist. 2004;9:173–181.
  • de Jong RS, Mulder NH, Dijksterhuis D, et al. Review of current clinical experience with prolonged (oral) etoposide in cancer treatment. Anticancer Res. 1995;15:2319–2330.
  • Mok TS, Wu YL, Ahn MJ, et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med. 2017;376:629–640.
  • Costa RB, Costa RLB, Talamantes SM, et al. Systematic review and meta-analysis of selected toxicities of approved ALK inhibitors in metastatic non-small cell lung cancer. Oncotarget. 2018;9:22137–22146.
  • Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal hormone-receptor-positive advanced breast cancer. N Engl J Med. 2012;366:520–529.
  • Burnell M, Levine MN, Chapman JA, et al. Cyclophosphamide, epirubicin, and fluorouracil versus dose-dense epirubicin and cyclophosphamide followed by paclitaxel versus doxorubicin and cyclophosphamide followed by paclitaxel in node-positive or high-risk node-negative breast cancer. J Clin Oncol. 2010;28:77–82.
  • Cazzaniga ME, Cortesi L, Ferzi A, et al. Metronomic chemotherapy with oral vinorelbine (mVNR) and capecitabine (mCAPE) in advanced HER2-negative breast cancer patients: is it a way to optimize disease control? Final results of the VICTOR-2 study. Breast Cancer Res Treat. 2016;160:501–509.
  • Harbeck N, Saupe S, Jäger E, et al. A randomized phase III study evaluating pegylated liposomal doxorubicin versus capecitabine as first-line therapy for metastatic breast cancer: results of the PELICAN study. Breast Cancer Res Treat. 2017;161:63–72.
  • Pivot X, Manikhas A, Żurawski B, et al. CEREBEL (EGF111438): a phase III, randomized, open-label study of lapatinib plus capecitabine versus trastuzumab plus capecitabine in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. J Clin Oncol. 2015;33:1564–1573.
  • Tawfik H, Rostom Y, Elghazaly H. All-oral combination of vinorelbine and capecitabine as first-line treatment in HER2/Neu-negative metastatic breast cancer. Cancer Chemother Pharmacol. 2013;71:913–919.
  • Bonadonna G, Rossi A, Valagussa P, et al. The CMF program for operable breast cancer with positive axillary nodes. Updated analysis on the disease-free interval, site of relapse and drug tolerance. Cancer. 1977;39:2904–2915.
  • Rimawi MF, Osborne CK. Chapter 43: adjuvant systemic therapy: endocrine therapy. In: editors, Harris JR, Lippman ME, Morrow M, et al. Diseases of the breast. 5th ed. Philadelphia: Lippincott Williams and Wilkins; 2014.
  • Bines J, Dienstmann R, Obadia RM, et al. Activity of megestrol acetate in postmenopausal women with advanced breast cancer after nonsteroidal aromatase inhibitor failure: a phase II trial. Ann Oncol. 2014;25:831–836.
  • Blackwell KL, Burstein HJ, Storniolo AM, et al. Overall survival benefit with lapatinib in combination with trastuzumab for patients with human epidermal growth factor receptor 2-positive metastatic breast cancer: final results from the EGF104900 Study. J Clin Oncol. 2012;30:2585–2592.
  • Yardley DA, Noguchi S, Pritchard KI, et al. Everolimus plus exemestane in postmenopausal patients with HR+ breast cancer: BOLERO-2 final progression-free survival analysis. Adv Ther. 2013;30:870–884.
  • Rugo HS, Finn RS, Diéras V, et al. Palbociclib plus letrozole as first-line therapy in estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer with extended follow-up. Breast Cancer Res Treat. 2019;174:719–729.
  • Hortobagyi GN, Stemmer SM, Burris HA, et al. Updated results from MONALEESA-2, a phase III trial of first-line ribociclib plus letrozole versus placebo plus letrozole in hormone receptor-positive, HER2-negative advanced breast cancer. Ann Oncol. 2018;29:1541–1547.
  • Goetz MP, Toi M, Campone M, et al. MONARCH 3: abemaciclib as initial therapy for advanced breast cancer. J Clin Oncol. 2017;35:3638–3646.
  • Bang YJ, Van Cutsem E, Feyereislova A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010;376:687–697.
  • Wilke H, Muro K, Van Cutsem E, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol. 2014;15:1224–1235.
  • Li X, Ma T, Zhang Q, et al. Modified-FOLFIRINOX in metastatic pancreatic cancer: a prospective study in Chinese population. Cancer Lett. 2017;406:22–26.
  • Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369:1691–1703.
  • Valle J, Wasan H, Palmer DH, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N Engl J Med. 2010;362:1273–1281.
  • Montagnani F, Turrisi G, Marinozzi C, et al. Effectiveness and safety of oxaliplatin compared to cisplatin for advanced, unresectable gastric cancer: a systematic review and meta-analysis. Gastric Cancer. 2011;14:50–55.
  • Oyama K, Fushida S, Kaji M, et al. Evaluation of the efficacy of palonosetron for prevention of chemotherapy-induced nausea and vomiting in patients with gastric cancer treated with S-1 plus cisplatin. Int J Clin Oncol. 2016;21:483–490.
  • André T, Boni C, Mounedji-Boudiaf L, et al. Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. N Engl J Med. 2004;350:2343–2351.
  • Buroker TR, O’Connell MJ, Wieand HS, et al. Randomized comparison of two schedules of fluorouracil and leucovorin in the treatment of advanced colorectal cancer. J Clin Oncol. 1994;12:14–20.
  • de Gramont A, Bosset JF, Milan C, et al. Randomized trial comparing monthly low-dose leucovorin and fluorouracil bolus with bimonthly high-dose leucovorin and fluorouracil bolus plus continuous infusion for advanced colorectal cancer: a French intergroup study. J Clin Oncol. 1997;15:808–815.
  • Falcone A, Ricci S, Brunetti I, et al. Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol. 2007;25:1670–1676.
  • Fuchs CS, Marshall J, Mitchell E, et al. Randomized, controlled trial of irinotecan plus infusional, bolus, or oral fluoropyrimidines in first-line treatment of metastatic colorectal cancer: results from the BICC-C Study. J Clin Oncol. 2007;25:4779–4786.
  • Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern cooperative oncology group study E3200. J Clin Oncol. 2007;25:1539–1544.
  • Tournigand C, André T, Achille E, et al. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol. 2004;22:229–237.
  • Van Cutsem E, Köhne CH, Hitre E, et al. Cetuximab and chemotherapy as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;360:1408–1417.
  • Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol. 2012;30:3499–3506.
  • Hesketh PJ, Sanz-Altamira P, Bushey J, et al. Prospective evaluation of the incidence of delayed nausea and vomiting in patients with colorectal cancer receiving oxaliplatin-based chemotherapy. Support Care Cancer. 2012;20:1043–1047.
  • Boccia RV, Gordan LN, Clark G, et al. 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. 2011;19:1609–1617.
  • Hesketh PJ, Bosnjak SM, Nikolic V, et al. Incidence of delayed nausea and vomiting in patients with colorectal cancer receiving irinotecan-based chemotherapy. Support Care Cancer. 2011;19:2063–2066.
  • Bosnjak SM, Stamatovic L, Borroni ME, et al. Efficacy and safety of oral NEPA (netupitant/palonosetron), the first fixed-combination antiemetic, in patients with gynecological cancers receiving platinum-based chemotherapy. Int J Gynecol Cancer. 2018;28:1153–1161.
  • Sanlorenzo M, Vujic I, Carnevale-Schianca F, et al. Role of interferon in melanoma: old hopes and new perspectives. Expert Opin Biol Ther. 2017;17:475–483.
  • Heaney ML, Golde DW. Myelodysplasia. N Engl J Med. 1999;340:1649–1660.
  • List AF, Doll DC. The myelodysplastic syndromes. Wintrobe’s clinical hematology. 10th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 1999. p. 2320.
  • Musso M, Scalone R, Bonanno V, et al. Palonosetron (Aloxi) and dexamethasone for the prevention of acute and delayed nausea and vomiting in patients receiving multiple-day chemotherapy. Support Care Cancer. 2009;17:205–209.
  • Mattiuzzi GN, Cortes JE, Blamble DA, et al. Daily palonosetron is superior to ondansetron in the prevention of delayed chemotherapy-induced nausea and vomiting in patients with acute myelogenous leukemia. Cancer. 2010;116:5659–5666.
  • Vardy J, Chiew KS, Galica J, et al. Side effects associated with the use of dexamethasone for prophylaxis of delayed emesis after moderately emetogenic chemotherapy. Br J Cancer. 2006;94:1011–1015.
  • Totadri S. Prophylaxis and management of antineoplastic drug induced nausea and vomiting in children with cancer. Pediatr Hematol Oncol. 2016;1:50–55.
  • Felix-Ukwu F, Reichert K, Bernhardt MB, et al. Evaluation of aprepitant for acute chemotherapy-induced nausea and vomiting in children and adolescents with acute lymphoblastic leukemia receiving high-dose methotrexate. Pediatr Blood Cancer. 2018;65:e26857.
  • Scarantino CW, Ornitz RD, Hoffman LG, et al. On the mechanism of radiation-induced emesis: the role of serotonin. Int J Radiat Oncol Biol Phys. 1994;30:825–830.
  • Scarantino CW, Ornitz RD, Hoffman LG, et al. Radiation-induced emesis: effects of ondansetron. Semin Oncol. 1992;19:38–43.
  • Rosenthal DI, Chambers MS, Fuller CD, et al. Beam path toxicities to non-target structures during intensity-modulated radiation therapy for head and neck cancer. Int J Radiat Oncol Biol Phys. 2008;72:747–755.
  • Urba S. Radiation-induced nausea and vomiting. J Natl Compr Canc Netw. 2007;5:60–65.
  • Ciura K, McBurney M, Nguyen B, et al. Effect of brain stem and dorsal vagus complex dosimetry on nausea and vomiting in head and neck intensity-modulated radiation therapy. Med Dosim. 2011;36:41–45.
  • Kocak-Uzel E, Gunn GB, Colen RR, et al. Beam path toxicity in candidate organs-at-risk: assessment of radiation emetogenesis for patients receiving head and neck intensity modulated radiotherapy. Radiother Oncol. 2014;111:281–288.
  • Beddok A, Faivre JC, Coutte A, et al. Practical contouring guidelines with an MR-based atlas of brainstem structures involved in radiation-induced nausea and vomiting. Radiother Oncol. 2019;130:113–120.
  • Lazzari G, Terlizzi A, Leo MG, et al. VMAT radiation-induced nausea and vomiting in adjuvant breast cancer radiotherapy: the incidental effect of low-dose bath exposure. Clin Transl Radiat Oncol. 2017;7:43–48.
  • Dennis K, De Angelis C, Jon F, et al. Aprepitant and granisetron for the prophylaxis of radiotherapy-induced nausea and vomiting after moderately emetogenic radiotherapy for bone metastases: a prospective pilot study. Curr Oncol. 2014;21:e760–7.
  • Enblom A, Bergius Axelsson B, Steineck G, et al. One third of patients with radiotherapy-induced nausea consider their antiemetic treatment insufficient. Support Care Cancer. 2009;17:23–32.
  • Poon M, Dennis K, DeAngelis C, et al. A prospective study of gastrointestinal radiation therapy-induced nausea and vomiting. Support Care Cancer. 2014;22:1493–1507.
  • Berger M, Gray JA, Roth BL. The expanded biology of serotonin. Annu Rev Med. 2009;60:355–366.
  • Lanciano R, Sherman DM, Michalski J, et al. 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. 2001;19:763–772.
  • Salvo N, Doble B, Khan L, et al. Prophylaxis of radiation-induced nausea and vomiting using 5-hydroxytryptamine-3 serotonin receptor antagonists: a systematic review of randomized trials. Int J Radiat Oncol Biol Phys. 2012;82:408–417.
  • Feyer P, Jahn F, Jordan K. Prophylactic management of radiation-induced nausea and vomiting. Biomed Res Int. 2015;2015:893013.
  • Ades S, Halyard M, Wilson K, et al. Effectiveness of aprepitant in addition to ondansetron in the prevention of nausea and vomiting caused by fractionated radiotherapy to the upper abdomen (AVERT). Support Care Cancer. 2017;25:1503–1510.
  • Navari RM, Qin R, Ruddy KJ, et al. Olanzapine for the prevention of chemotherapy-induced nausea and vomiting. N Engl J Med. 2016;375:134–142.
  • Oflazoglu U, Varol U, Alacacioglu A, et al. A prospective randomized controlled trial of metoclopramide combined with triple antiemetic therapy to prevent anthracycline-based chemotherapy-induced nausea and vomiting in patients with breast cancer. Ann Oncol. 2018;29:1704P.
  • Kessler JF, Alberts DS, Plezia PM, et al. An effective five-drug antiemetic combination for prevention of chemotherapy-related nausea and vomiting. Experience in eighty-four patients. Cancer Chemother Pharmacol. 1986;16:282–286.
  • Pessi MA, Necchi A, Bossi P, et al. Nausea and vomiting during the first 3 intercycle periods in chemo-naive cancer patients receiving moderately/highly emetogenic therapy. Tumori. 2015;101:692–696.