Next Article in Journal
Emerging Canonical and Non-Canonical Roles of Granzyme B in Health and Disease
Previous Article in Journal
Imaging of Oligometastatic Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Health-Related Quality of Life (HRQoL) in Neuroendocrine Tumors: A Systematic Review

1
University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, UPMC Chautauqua Hospital, Jamestown, NY 14701, USA
2
Program in Women’s Oncology, Women and Infants Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
3
Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
4
Department of Gastro Intestinal Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
5
Department of Cancer Prevention & Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(6), 1428; https://doi.org/10.3390/cancers14061428
Submission received: 3 February 2022 / Revised: 8 March 2022 / Accepted: 8 March 2022 / Published: 10 March 2022

Abstract

:

Simple Summary

Neuroendocrine tumors (NETs) are a group of heterogenous neoplasms arising from the diffuse neuroendocrine system. Several therapies have been added to the treatment landscape that have improved long-term outcomes. Despite therapeutic advancements, the symptom burden of the disease remains high, impacting health-related quality of life (HRQoL). In this study, we reviewed the impact of different treatment modalities on HRQoL in NET patients. Through a thorough literature review, 61 out of 2375 publications met the inclusion criteria. All randomized phase III trials leading to drug approvals showed a lack of deterioration of HRQoL, and one showed improved QoL. Capturing and understanding patient-reported outcome data is of vital importance for both patients and physicians to make treatment-related decisions.

Abstract

Therapeutic advancements in neuroendocrine tumors (NETs) have improved survival outcomes. This study aims to review the impact of the current therapeutics on health-related quality of life (HRQoL) in NET patients. A literature review was performed utilizing PubMed, The Cochrane Library, and EMBASE, using the keywords “Carcinoid”, “Neuroendocrine tumor”, “NET”, “Quality of life”, “Chemotherapy”, “Chemoembolization”, “Radiofrequency ablation”, “Peptide receptor radionucleotide therapy”, “PRRT”, “Surgery”, “Everolimus”, “Octreotide”, “Lanreotide”, “Sunitinib”, and “Somatostatin analog”. Letters, editorials, narrative reviews, case reports, and studies not in English were excluded. Out of 2375 publications, 61 studies met our inclusion criteria. The commonly used instruments were EORTC QLQ-C30, FACT G, and EORTC- QLQ GI.NET-21. HRQoL was assessed in all pivotal trials that led to approvals of systemic therapies. All systemic therapies showed no worsening in HRQoL. The NETTER-1 study was the only study to show a statistically significant improvement in HRQoL in several domains. The trial examining sunitinib versus placebo in pancreatic NETs showed no change in QoL, except for worsening of diarrhea. In addition to clinical outcomes, patient-reported outcomes are a key element in making appropriate treatment decisions. HRQoL data should be readily provided to patients to assist in shared decision-making.

1. Introduction

Neuroendocrine tumors (NETs) are a group of heterogeneous malignancies that originate from neuroendocrine cells. Most well-differentiated NETs have an indolent course [1,2]. Multiple new treatments have been approved in recent years, primarily based on the evidence of inhibition of tumor growth and delay of disease progression [3,4,5,6,7,8]. Nevertheless, the symptomatic burden of disease remains high. A recent global survey of patients with NETs, conducted in more than 12 countries, demonstrated the considerable impact of NETs on symptoms, work, activities of daily living, and healthcare resource use [9]. NETs are associated with a wide range of symptoms from excess hormone production to tumor burden, which affect quality of life. The most commonly reported symptoms are fatigue, diarrhea, and flushing [1]. Clinical trials for new treatment options generally focus on disease progression and toxicity rather than quality of life. Moreover, treatment decisions are often based on patient comorbidities and the toxicity profile of the drugs, rather than taking patient preference or symptoms into consideration. Decision-making aimed only at improving overall survival with newer drugs may not achieve a desired symptom control goal or quality of life.
Patient-reported outcomes should play a pivotal role in selecting treatment options in NET, as symptoms can be debilitating. In recent years, health-related quality of life (HRQoL) measurements have gained increasing relevance in clinical trials. There are many available validated tools for evaluating HRQoL, but no clear consensus on the optimal tool for use in trials or clinical practice [10]. Given increasing awareness of the importance of HRQoL in the assessment of cancer treatment, we sought to perform a systematic review of studies measuring patient-reported QoL outcomes in NET patient studies.

2. Materials and Methods

We performed a thorough review of the published literature on health-related quality of life (HRQoL) in the NET population. Our search included the following databases: PubMed, The Cochrane Library, and EMBASE. The systematic research was performed in May 2019. The search combinations used included the Boolean expressions, “AND” and “OR” in combination with the following MeSH and free text terms: “Carcinoid”, “Neuroendocrine tumor”, “NET”, “Quality of life”, “Chemotherapy”, “Chemoembolization”, “Radiofrequency ablation”, “Peptide receptor radionucleotide therapy”, “PRRT”, “Surgery”, “Everolimus”, “Octreotide”, “Lanreotide”, “Sunitinib”, and “Somatostatin analog”. The same search strategy was employed in all 3 databases. Each of the eligible studies were reviewed, and data were extracted independently by two investigators to ensure consistency. Papers addressing HRQoL in NET patients were included in our analysis. Publications that were letters, editorials, narrative reviews, case reports, and studies not in English were excluded. The study is registered in the research registry for systematic analysis (unique identifying number-reviewregistry1315). The study selection process is described in Figure 1.

3. Results

3.1. HRQoL Study Selection & Characteristics

Our literature search revealed a total of 61 studies, which assessed the wellness of patient-reported quality of life (QoL) in NET patients using validated questionnaires. Of these 61 studies, nine were randomized controlled trials (RCTs), 16 were phase II clinical trials, 11 were prospective studies and the remaining 25 were observational studies. The RCTs and phase II clinical trials are listed in Table 1 [4,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34]. The prospective studies and observational studies are listed in Table S1 in Supplementary File [35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71]. In this literature review, we found two review articles focusing on HRQoL in NET patients [72,73]. The first review, by the Spanish NET Group, examined key reported outcomes in HRQoL studies in gastroenteropancreatic (GEP) NET patients. It evaluated different quality assessment tools used in various HRQoL trials and identified appropriate tools to assess HRQoL changes [72]. The second review by Martini et al. evaluated HRQoL studies in GEP NET patients from a methodological standpoint. Of the 48 eligible studies they examined, a range of methodological shortcomings was identified. They concluded that transferring HRQoL into practice is limited not just by the sparsity of studies but also due to the quality of HRQoL data processing [73].
All studies evaluated the quality of life in NET patients using validated QoL questionnaires. The commonly used questionnaires were: European Organization for Research and Treatment of Cancer Quality of Life Core Questionnaire (EORTC QLQ-C30), NET-specific QoL questionnaire (QLQ-GI NET-21), the Functional Assessment of Cancer Therapy-General (FACT-G), and the 12-Item Short Form Health Survey (SF-36).

3.2. QoL Instruments Used

The majority of the studies utilized EORTC QLQ-C30 (41/61 trials), an HRQoL questionnaire that is widely used in oncology trials [74]. The QLQ-C30 questionnaire is a multidimensional tool that has been translated and validated in over 100 languages and used in over 3000 studies worldwide. This questionnaire assesses the quality of life of patients on clinical trials using a series of 30 questions. These questions assess patient well-being based on five functional scales (physical, role, emotional, cognitive, social functioning), and several single- and multi-item symptom subscales (fatigue, nausea/vomiting, pain, dyspnea, insomnia, appetite loss, constipation, diarrhea, financial difficulties) [74]. Responses are translated into a 0–100-point scale to standardize the quality-of-life assessment. A QLQ-C30 score change of 5–10 denotes ‘a little’ change for better or worse on a particular scale (function or symptom), a score change of 10–20 denotes ‘moderate’ change and a change in score greater than 20 corresponds to ‘very much’ change from the baseline [75].
The EORTC has also developed a NET-specific QoL questionnaire, QLQ-GI.NET21. This questionnaire covers issues specific to NET patients that are not covered in the QLQ-C30 questionnaire, such as endocrine or specific gastrointestinal symptoms [64]. Nine of the studies used the QLQ-GI.NET21 questionnaire in conjunction with QLQ-C30. One study used the questionnaire EORTC QLQ Liver Metastases Colorectal (LMC21), in conjunction with QLQ-C30 [76].
The other commonly used questionnaire (n = 3 studies) was the Functional Assessment of Cancer Therapy-General (FACT-G), which comprises 27 items that assess patients’ physical, emotional, functional, and social well-being [77]. Other applied modules of FACT-G, FACT-Anemia, and FACT-hepatobiliary were also utilized by two studies. Another generic questionnaire used by two studies was the Functional Assessment of Chronic Illness Therapy (FACIT), and its modules FACIT-diarrhea and FACIT-fatigue.
Three studies used the Patient-Reported Outcome Measurement Information System 29-item Health Profile (PROMIS-29). This questionnaire assesses the quality of life of patients spanning seven important aspects–depression, anxiety, physical function, pain interference, fatigue, sleep disturbance, and the ability to participate in social roles and activities [78]. In one trial, the psychosocial adjustment to illness scale (PAIS) was used to assess patients’ psychosocial adjustment to their illness. The PAIS covers multiple domains including healthcare orientation, vocational environment, domestic environment, sexual relationships, extended family relationships, social environment, and psychological distress [79].
A total of 16 studies used either their self-made questionnaire, or other tools such as the 36-item Short Form Health Survey (SF-36), the 12-Item Short Form Health Survey (SF-12), the Euroqol-5 Dimension (EQ-5D), the French version of the Nottingham Health Profile (ISPN), and the General Health Questionnaire (GHQ-12). In general, these tools assess a patient’s quality of life from physical, social, and functional aspects.

3.3. HRQoL with Systemic Therapies

Currently approved therapies that are commonly utilized in NET patients include octreotide, lanreotide, everolimus, sunitinib, telotristat, and peptide receptor radionuclide therapy (PRRT) [4,7,13,65,80]. Pivotal randomized phase III trials using these agents evaluated patient quality-of-life while on active treatment or the control arm, utilizing validated HRQoL questionnaires. The study tools used, and results, are summarized in Table 2 and detailed below.
Two randomized phase III studies evaluated the role of somatostatin analogues (SSAs) in NET patients. The PROMID study randomized untreated, well-differentiated, metastatic mid-gut NET patients to receive either octreotide long-acting repeatable (LAR) versus placebo. QoL was assessed as a secondary endpoint using EORTC-QLQ C30 and described as a change from the baseline to after six months of follow-up. The HRQoL was assessed at random visits and at 3-month intervals until tumor progression. The symptoms of carcinoid syndrome were present in 39% of patients at the baseline. Symptomatic response was defined as a reduction of flushing to less than 1 flush/week, <4 bowel movements/week, and the absence of abdominal pain. There was no difference in the baseline mean EORTC-QLQ C30 score between the groups. At the 6-month follow-up mark, the symptomatic responses were higher in the octreotide LAR arm vs. placebo; however, none of these were statistically significant (flushing, 70% vs. 45%, p = 0.08; diarrhea, 33% vs. 19%, p = 0.56, abdominal pain 50% vs. 30%, p = 0.35) [13]. According to the post hoc analysis of the PROMID trial by Rinke et al., a statistically significantly longer time to deterioration (TTD) was observed in the octreotide LAR group than the placebo for fatigue (18.5 months vs. 6.8, p = 0.0006), pain, and insomnia. For diarrhea, there was an improvement in the change from the baseline to week 24 scores in the treatment group, and the score was worsened in the placebo group. Corresponding fatigue scores were stable in the treatment arm and worsened for placebo [82]. The phase III CLARINET study showed the efficacy of another SSA analogue, lanreotide, in comparison to placebo in advanced well/moderately differentiated, non-functioning, grade 1 (G1)/grade 2 (G2) gastroenteropancreatic NETs. The main adverse effect from the lanreotide was reportedly diarrhea, with 26% in the treatment arm having diarrhea when compared to 9% in the placebo arm. The second most common adverse effect was abdominal pain (14% vs. 2% in the treatment and placebo arm, respectively). HRQoL was a secondary endpoint and was assessed using EORTC QLQ-C30 and QLQ-GI.NET21 scores. There were no statistically significant differences in the change from the baseline to post-treatment QoL scores between the two treatment groups [4].
The phase III RADIANT-4 trial evaluated the efficacy of everolimus (mTOR inhibitor) in comparison to placebo in patients with advanced, well-differentiated, G1/G2 gastrointestinal and lung NETs. The Functional Assessment of Cancer Therapy-General (FACT-G) questionnaire was utilized to assess the HRQoL at baseline, every 8 weeks for the first 12 months, and every 12 weeks thereafter. The pre-specified secondary endpoint was the time to definitive deterioration of FACT-G score by at least seven or more points compared to the baseline. The median time to deterioration was similar between both the everolimus (11.3 months) and placebo arms (9.2 months), with no differences between the two groups (HR 0.81, 95% CI 0.55–1.21; p = 0.31) [17]. The most common side effects observed were stomatitis, followed by diarrhea and fatigue. In the treatment arm, 31% of patients had diarrhea with everolimus, compared to 16% observed in the placebo arm. In the everolimus group, grade 3 diarrhea was observed in 6% and grade 4 in 1% of patients, whereas no grade 4 diarrhea was seen in the placebo, and grade 3 was only 2%. Fatigue was also observed more frequently with everolimus when compared to placebo (31% versus 24%) [17,80].
Another targeted agent, sunitinib, a tyrosine kinase inhibitor, was approved in advanced, well-differentiated pancreatic NETs following a prospective trial demonstrating efficacy. In the randomized phase III trial of sunitinib versus placebo, the median progression free survival (PFS) on the sunitinib arm was 11.4 months, and the PFS in the placebo group was 5.5 months, with a hazard ratio (HR) of 0.42 (95% CI 026–0.66; p < 0.001). Functioning tumors were present in 51% of the sunitinib group and in 48% of the placebo group. The frequent adverse effects in the sunitinib group were diarrhea, nausea, vomiting, and fatigue. Diarrhea was seen in 50% of the treatment arm and 39% in the placebo arm. Grade 3 or 4 diarrhea was found in 5% of patients in the treatment arm and 2% in the placebo arm [6]. Quality of life was also a secondary endpoint of the study. EORTC QLQ-C30 scores were assessed at the baseline, day 1 of every cycle, and analyzed for the first 10 cycles. Baseline HRQoL scores were similar in the two arms. Over the first 10 cycles, no differences were noted in the global HRQoL, physical, emotional, cognitive, role, social functioning, or symptom scales, except for diarrhea. There was a clinical and statistically significant worsening of diarrhea in the sunitinib arm with a difference of 21.4 points between the two arms (p < 0.001). Even though the sunitinib arm had a statistically significant worsening of insomnia (p = 0.04), this was not clinically significant as the difference between the groups was 7.8 (between-group clinically significant difference is defined as >10 points) [6,15].
The phase III NETTER-1 trial randomized advanced, progressive, somatostatin-receptor positive, G1/G2 midgut NET patients to 177Lu-Dotatate PRRT plus octreotide LAR vs. octreotide LAR alone. PFS was clinically and statistically higher in the Lu-Dotatate group (65.2% vs. 10.8% at 20 months) with a hazard ratio of 0.18 (95% CI: 0.11, 0.29; p < 0.0001). The overall response rate was also higher in the Lu-Dotatate group (18% vs. 3% p < 0.001). The most common adverse effects in the 177Lu-Dotatate group were nausea (59% vs. 12%) and vomiting (47% vs. 10%), which was attributed to the concurrent amino acid infusion. Fatigue was also higher in the Lu-Dotatate group when compared to the control group (40% vs. 25% p = 0.03). Side effects that may affect the HRQoL, including diarrhea, abdominal pain and flushing, were slightly higher in the Lu-Dotatate group, but this was not statistically significant. The rate of grade 3 and grade 4 side effects was similar in both groups [7]. It was recently found that the median overall survival in Lu-Dotatate group was 48 months and was 36.3 months in the control arm, but this was not statistically significant (HR = 0.84, 95% CI: 0.60, 1.17: p = 0.30) [83]. HRQoL was measured using EORTC QLQ-C30 and GI NET-21 questionnaires every 12 weeks till disease progression. Time to QoL deterioration (TTD) was defined as the time from randomization to first deterioration >/10 points on a 100-point scale for that domain. The TTD was statistically longer in the PRRT arm for a number of domains including global health (HR 0.41, p < 0.001), role functioning (HR = 0.58, p = 0.03), physical functioning (HR 0.52, p = 0.15), disease-related worries (HR 0.57, p = 0.018), body image (HR = 0.43, p = 0.006), diarrhea 0.47 (p = 0.01), pain (HR 0.57, p = 0.025), and fatigue (HR 0.62, p = 0.03). Median TTD was statistically significant in the favor of PRRT for the global health domain (22.7-month difference) and physical functioning domain (13.7-month difference) [18].
The placebo-controlled TELESTAR study studied the role of telotristat ethyl (TE), a tryptophan hydroxylase inhibitor in patients with carcinoid syndrome and >/4 bowel movements (BM) per day despite taking SSAs. Patients were randomized to TE 250 mg three times a day (TID), TE 500 mg TID or placebo (1:1:1) during a double-blind treatment period (DBTP), and all patients received TE 500 mg TID in an open label extension (OLE) to week 48. This study used a unique parameter of durable response defined as a BM frequency reduction of >/30% from baseline for >/50% of the time [3]. An exploratory analysis evaluated the relationship between HRQoL and durable responders (DR)/non-durable responders (NDR). At the end of the 12-week DBTP, 48/135 patients were DR. In the OLE phase, 29/35 DR maintained the DR, and 71 of DBTP-NDRs became OLE_DRs. The analysis showed that DR was associated with better symptom control than NDR in both DBTP and OLE. Similarly, DR had QoL improvements in EORTC QLQ-C30 global health status, nausea, vomiting, pain, diarrhea, and EORTC QLQ-GI.NET21 gastrointestinal symptoms than NDR both over the DBTP and OLE. The EORTC QLQ-C30 diarrhea subscale scores were much improved in the telotristat group. The score improvements for TE 250 mg, 500 mg groups were 19.2 points, 21.6 points, and that of the placebo was only 8.5 points [3,81].

4. Discussion

Patient-centered outcomes have become an important focus of research, and few physicians have had formal training in this aspect of patient evaluation. This form of research assesses the benefits and harms of preventive, diagnostic, therapeutic, palliative, and other interventions for shared decision-making. This knowledge further helps in tailoring the treatments based on the meaningful endpoint of quality of life and highlights outcomes that are relevant to patients and caretakers which eventually lead to treatment satisfaction. Most clinical trials and retrospective studies focus on the survival benefits and toxicity profiles of the treatments rather than impact on quality of life. In a patient-centric treatment model, treatment discussions should also include data on HRQoL outcomes. Decisions informed by HRQoL can help to improve the patients’ physical and mental health and decrease the burden on health care by minimizing frequent hospital admissions and health care utilization [84].
HRQoL is a particularly important consideration for patients with NETs, who can develop both tumor- and hormone-related symptoms during their disease course [9]. The common symptoms affecting the HRQoL of NET patients are listed in Table 3. Patients with relatively slow-growing tumors may be prescribed local and systemic treatments for prolonged periods lasting a decade or more, and many hope to remain functional throughout their journey. Further, although the incidence of NET in patients 65 years or above is highest, it is also diagnosed in younger adults, whose health goals may be different [2].
HRQoL studies assessing locoregional modalities of surgery or interventional radiology (IR) procedures are limited by size, absence of a formal HRQoL analysis, and/or lack of prospective analysis. Therefore, a definitive impact of these procedures in the HRQoL of NET patients is largely unknown. There are multiple studies on the effect of surgeries and IR techniques for treating hepatic metastasis such as embolization, radiofrequency ablation, and cryoablation in patients with NET [25,26,44,46,55]. Very few surgical or IR studies have studied the impact of the procedures on the patient’s quality of life. Most of them lack a formal HRQoL analysis.
Validated tools and uniform definitions are key for assessing HRQoL and allowing comparison between studies of approved therapy options to aid treatment selection. There is a wide variety of tools and patient surveys for assessing HRQoL of cancer patients [10] in trials and in clinical practice. As summarized here, all the key phase 3 studies in NET patients used validated tools to assess HRQoL. Most of the studies used the EORTC QLQ-C30 questionnaire, which is a general oncology questionnaire. As fatigue and diarrhea are common in NET patients at the baseline, some trials used FACIT-fatigue and FACIT-D scoring while some studies used QLQ-GI.NET21, which is a NET-specific questionnaire. Cross-study comparisons will be easier with uniform adoption of HRQoL tools for future use, and facilitate treatment-choice discussions in clinical practice. One limitation, even with these validated tools, is reliance on recall when patients and caregivers fill out the HRQoL. A majority are done during office visits, and do not always have journals to track changes in the HRQoL events between visits. Besides journals to assist in recall, the availability of mobile phone applications, which can collect real-time data, may increase patient compliance and confidence in data capture from self-reporting [85].
While using a global score and a prior definition for improved QoL are important, there are also some limitations with looking only at overall scores. In the multiple practice-changing clinical trials leading to the approvals of octreotide LAR, lanreotide auto gel, everolimus, and sunitinib, no decline of QoL global scores was seen compared to the control arms, suggesting that therapy did not worsen the quality of life [4,6,13,17]. However, the trial examining sunitinib versus placebo in pancreatic NETs showed that, despite no change in QoL in all aspects, the diarrhea scores reflected that sunitinib worsened this quality measure. In addition, in a retrospective analysis of the PROMID trial, Rinke et al. observed a significantly longer time to deterioration in quality of life in the octreotide LAR group compared to the placebo in clinically relevant NET symptoms such as fatigue, diarrhea, pain, and insomnia, suggesting maintained or improved HRQoL with octreotide LAR [82]. Similarly, everolimus was also associated with more diarrhea and fatigue than the placebo group [80].
The NETTER-1 trial was the only study to have favorably impacted decline in HRQoL compared to control in key domains including global health, physical functioning, role functioning, fatigue, pain, and diarrhea, providing more granular insight on the impact of therapy on life. Therapy discussions should include these outcomes along with discussion of survival benefits [18].
Most trial publications focus on grade 3 and 4 events that result in dose reduction or discontinuation of therapy. However, chronic low-grade side effects such as fatigue can have a major impact on HRQoL. In the global health survey by Singh et al., the impact of the illness on employment was captured in those still working and 49% of patients reported taking days off work, 27% asked for accommodations, and 24% reduced work hours. In addition, among 82% of those who were unemployed, the reason for not working was disease-related symptoms [9]. Assessing financial impact is only partially included in most QoL assessments and is an important gap in knowledge for NET patients that future studies should aim to address. The EORTC QLQ-30 has clinical thresholds for physical functioning (<80), emotional functioning (<70), fatigue (>39), pain (>25), and role function (<90) scales based on literature [74]. These threshold scores provide valuable insight into clinically important problems that should be addressed and may impact not just treatment choices but also bring to light the need for timely referrals to supportive care services.

5. Conclusions

Health-related quality of life should be integrated into tailoring treatment selection in neuroendocrine cancer patients as patient-reported outcomes are a key element of shared decision-making and patient-centered care. In NET patients, the impact on QoL of all currently approved therapies compared to their control arms has been studied using well-validated tools, and should be provided to patients during their pretherapy counseling along with toxicity information. Toxicity assessment of specific treatments should not be a surrogate measure for quality of life of patients, and careful assessment of global QoL scores, as well as individual symptom scores, may provide key insights into the impact of the disease on quality of life and functioning. Real-life studies on the impact of therapy, where most of the care delivery occurs, and HRQoL studies during periods between active treatment need to be conducted to identify the true impact of therapy choices on patients living with NETs.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14061428/s1, Table S1: Prospective and Observational Studies in NET Patients with Reporting of HRQoL Outcome Measures.

Author Contributions

Conceptualization, R.I.; methodology, R.G. and M.G.; validation, R.G., M.G. and A.M.R.; formal analysis, R.G., M.G. and A.M.R.; data curation, R.G. and M.G.; writing—original draft preparation, R.G., M.G. and A.M.R.; writing—review and editing, J.S., K.M.G. and R.I.; visualization, R.I.; supervision, R.I. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by the Roswell Park Comprehensive Cancer Center and National Cancer Institute (NCI) grant P30CA016056.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gosain, R.; Mukherjee, S.; Yendamuri, S.S.; Iyer, R. Management of Typical and Atypical Pulmonary Carcinoids Based on Different Established Guidelines. Cancers 2018, 10, 510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Dasari, A.; Shen, C.; Halperin, D.M.; Zhao, B.; Zhou, S.; Xu, Y.; Shih, T.; Yao, J.C. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States. JAMA Oncol. 2017, 3, 1335–1342. [Google Scholar] [CrossRef] [PubMed]
  3. Kulke, M.H.; Hörsch, D.; Caplin, M.E.; Anthony, L.B.; Bergsland, E.; Öberg, K.; Welin, S.; Warner, R.R.; Lombard-Bohas, C.; Kunz, P.L.; et al. Telotristat Ethyl, a Tryptophan Hydroxylase Inhibitor for the Treatment of Carcinoid Syndrome. J. Clin. Oncol. 2017, 35, 14–23. [Google Scholar] [CrossRef] [PubMed]
  4. Caplin, M.E.; Pavel, M.; Ćwikła, J.B.; Phan, A.T.; Raderer, M.; Sedláčková, E.; Cadiot, G.; Wolin, E.M.; Capdevila, J.; Wall, L.; et al. Lanreotide in Metastatic Enteropancreatic Neuroendocrine Tumors. N. Engl. J. Med. 2014, 371, 224–233. [Google Scholar] [CrossRef]
  5. Öberg, K.; Lamberts, S.W.J. Somatostatin analogues in acromegaly and gastroenteropancreatic neuroendocrine tumours: Past, present and future. Endocr.-Relat. Cancer 2016, 23, R551–R566. [Google Scholar] [CrossRef] [Green Version]
  6. Raymond, E.; Dahan, L.; Raoul, J.-L.; Bang, Y.-J.; Borbath, I.; Lombard-Bohas, C.; Valle, J.; Metrakos, P.; Smith, D.; Vinik, A.; et al. Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N. Engl. J. Med. 2011, 364, 501–513. [Google Scholar] [CrossRef] [Green Version]
  7. Strosberg, J.; El-Haddad, G.; Wolin, E.; Hendifar, A.; Yao, J.; Chasen, B.; Mittra, E.; Kunz, P.L.; Kulke, M.H.; Jacene, H.; et al. Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine Tumors. N. Engl. J. Med. 2017, 376, 125–135. [Google Scholar] [CrossRef]
  8. Yao, J.C.; Shah, M.H.; Ito, T.; Bohas, C.L.; Wolin, E.M.; Van Cutsem, E.; Hobday, T.J.; Okusaka, T.; Capdevila, J.; de Vries, E.; et al. Everolimus for Advanced Pancreatic Neuroendocrine Tumors. N. Engl. J. Med. 2011, 364, 514–523. [Google Scholar] [CrossRef] [Green Version]
  9. Singh, S.; Granberg, D.; Wolin, E.; Warner, R.; Sissons, M.; Kolarova, T.; Goldstein, G.; Pavel, M.; Öberg, K.; Leyden, J. Patient-Reported Burden of a Neuroendocrine Tumor (NET) Diagnosis: Results from the First Global Survey of Patients with NETs. J. Glob. Oncol. 2017, 3, 43–53. [Google Scholar] [CrossRef]
  10. White, B.E.; Druce, M.R.; Grozinsky-Glasberg, S.; Srirajaskanthan, R.; Gamper, E.M.; Gray, D.; Mujica-Mota, R.; Ramage, J. Health-related quality of life in neuroendocrine neoplasms: A critical review. Endocr.-Relat. Cancer 2020, 27, R267–R280. [Google Scholar] [CrossRef]
  11. Arnold, R.; Rinke, A.; Klose, K.-J.; Müller, H.-H.; Wied, M.; Zamzow, K.; Schmidt, C.; Schade-Brittinger, C.; Barth, P.; Moll, R.; et al. Octreotide versus Octreotide Plus Interferon-Alpha in Endocrine Gastroenteropancreatic Tumors: A Randomized Trial. Clin. Gastroenterol. Hepatol. 2005, 3, 761–771. [Google Scholar] [CrossRef]
  12. Bajetta, E.; Procopio, G.; Catena, L.; Dsc, A.M.; De Dosso, S.; Ricci, S.; Lecchi, A.S.; Boscani, P.F.; Iacobelli, S.; Carteni, G.; et al. Lanreotide autogel every 6 weeks compared with Lanreotide microparticles every 3 weeks in patients with well differentiated neuroendocrine tumors: A Phase III Study. Cancer 2006, 107, 2474–2481. [Google Scholar] [CrossRef]
  13. Rinke, A.; Müller, H.-H.; Schade-Brittinger, C.; Klose, K.-J.; Barth, P.; Wied, M.; Mayer, C.; Aminossadati, B.; Pape, U.-F.; Bläker, M.; et al. Placebo-Controlled, Double-Blind, Prospective, Randomized Study on the Effect of Octreotide LAR in the Control of Tumor Growth in Patients with Metastatic Neuroendocrine Midgut Tumors: A Report from the PROMID Study Group. J. Clin. Oncol. 2009, 27, 4656–4663. [Google Scholar] [CrossRef]
  14. Meyer, T.; Qian, W.; Caplin, M.E.; Armstrong, G.; Lao-Sirieix, S.-H.; Hardy, R.; Valle, J.; Talbot, D.C.; Cunningham, D.; Reed, N.; et al. Capecitabine and streptozocin±cisplatin in advanced gastroenteropancreatic neuroendocrine tumours. Eur. J. Cancer 2014, 50, 902–911. [Google Scholar] [CrossRef]
  15. Vinik, A.; Bottomley, A.; Korytowsky, B.; Bang, Y.-J.; Raoul, J.-L.; Valle, J.W.; Metrakos, P.; Hörsch, D.; Mundayat, R.; Reisman, A.; et al. Patient-Reported Outcomes and Quality of Life with Sunitinib versus Placebo for Pancreatic Neuroendocrine Tumors: Results from an International Phase III Trial. Target. Oncol. 2016, 11, 815–824. [Google Scholar] [CrossRef]
  16. Vinik, A.I.; Wolin, E.M.; Liyanage, N.; Gomez-Panzani, E.; Fisher, G.A. Evaluation of Lanreotide Depot/Autogel Efficacy and Safety as a Carcinoid Syndrome Treatment (Elect): A Randomized, Double-Blind, Placebo-Controlled Trial. Endocr. Pract. 2016, 22, 1068–1080. [Google Scholar] [CrossRef]
  17. E Pavel, M.; Singh, S.; Strosberg, J.R.; Bubuteishvili-Pacaud, L.; Degtyarev, E.; Neary, M.P.; Carnaghi, C.; Tomasek, J.; Wolin, E.; Raderer, M.; et al. Health-related quality of life for everolimus versus placebo in patients with advanced, non-functional, well-differentiated gastrointestinal or lung neuroendocrine tumours (RADIANT-4): A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017, 18, 1411–1422. [Google Scholar] [CrossRef]
  18. Strosberg, J.; Wolin, E.; Chasen, B.; Kulke, M.; Bushnell, D.; Caplin, M.; Baum, R.P.; Kunz, P.; Hobday, T.; Hendifar, A.; et al. Health-Related Quality of Life in Patients with Progressive Midgut Neuroendocrine Tumors Treated with 177Lu-Dotatate in the Phase III NETTER-1 Trial. J. Clin. Oncol. 2018, 36, 2578–2584. [Google Scholar] [CrossRef]
  19. Wymenga, A.; Eriksson, B.; Salmela, P.; Jacobsen, M.; Van Cutsem, E.; Fiasse, R.; Välimäki, M.; Renstrup, J.; De Vries, E.; Öberg, K. Efficacy and Safety of Prolonged-Release Lanreotide in Patients with Gastrointestinal Neuroendocrine Tumors and Hormone-Related Symptoms. J. Clin. Oncol. 1999, 17, 1111. [Google Scholar] [CrossRef]
  20. Ruszniewski, P.; Ish-Shalom, S.; Wymenga, M.; O’Toole, D.; Arnold, R.; Tomassetti, P.; Bax, N.; Caplin, M.; Eriksson, B.; Glaser, B.; et al. Rapid and Sustained Relief from the Symptoms of Carcinoid Syndrome: Results from an Open 6-Month Study of the 28-Day Prolonged-Release Formulation of Lanreotide. Neuroendocrinology 2004, 80, 244–251. [Google Scholar] [CrossRef]
  21. Zuetenhorst, J.M.; Olmos, R.A.V.; Muller, M.; A Hoefnagel, C.; Taal, B.G. Interferon and meta-iodobenzylguanidin combinations in the treatment of metastatic carcinoid tumours. Endocr.-Relat. Cancer 2004, 11, 553–561. [Google Scholar] [CrossRef]
  22. Frilling, A.; Weber, F.; Saner, F.; Bockisch, A.; Hofmann, M.; Mueller-Brand, J.; Broelsch, C.E. Treatment with 90Y- and 177Lu-DOTATOC in patients with metastatic neuroendocrine tumors. Surgery 2006, 140, 968–976; discussion 976–977. [Google Scholar] [CrossRef]
  23. Kulke, M.H.; Lenz, H.-J.; Meropol, N.J.; Posey, J.; Ryan, D.P.; Picus, J.; Bergsland, E.; Stuart, K.; Tye, L.; Huang, X.; et al. Activity of Sunitinib in Patients With Advanced Neuroendocrine Tumors. J. Clin. Oncol. 2008, 26, 3403–3410. [Google Scholar] [CrossRef]
  24. Korse, C.M.; Bonfrer, J.M.; Aaronson, N.K.; Hart, A.A.; Taal, B.G. Chromogranin A as an Alternative to 5-Hydroxyindoleacetic Acid in the Evaluation of Symptoms during Treatment of Patients with Neuroendocrine Tumors. Neuroendocrinology 2009, 89, 296–301. [Google Scholar] [CrossRef]
  25. Bushnell, D.L., Jr.; O’Dorisio, T.M.; O’Dorisio, M.S.; Menda, Y.; Hicks, R.J.; Van Cutsem, E.; Baulieu, J.-L.; Borson-Chazot, F.; Anthony, L.; Benson, A.B.; et al. 90Y-Edotreotide for Metastatic Carcinoid Refractory to Octreotide. J. Clin. Oncol. 2010, 28, 1652–1659. [Google Scholar] [CrossRef] [Green Version]
  26. Cwikla, J.B.; Sankowski, A.; Seklecka, N.; Buscombe, J.R.; Nasierowska-Guttmejer, A.; Jeziorski, K.; Mikolajczak, R.; Pawlak, D.; Stepien, K.; Walecki, J. Efficacy of radionuclide treatment DOTATATE Y-90 in patients with progressive metastatic gastroenteropancreatic neuroendocrine carcinomas (GEP-NETs): A phase II study. Ann. Oncol. 2010, 21, 787–794. [Google Scholar] [CrossRef]
  27. Bodei, L.; Cremonesi, M.; Grana, C.M.; Fazio, N.; Iodice, S.; Baio, S.M.; Bartolomei, M.; Lombardo, D.; Ferrari, M.E.; Sansovini, M.; et al. Peptide receptor radionuclide therapy with 177Lu-DOTATATE: The IEO phase I-II study. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 2125–2135. [Google Scholar] [CrossRef]
  28. Claringbold, P.G.; Brayshaw, P.A.; Price, R.A.; Turner, J.H. Phase II study of radiopeptide 177Lu-octreotate and capecitabine therapy of progressive disseminated neuroendocrine tumours. Eur. J. Nucl. Med. Mol. Imaging 2011, 38, 302–311. [Google Scholar] [CrossRef]
  29. Khan, S.; Krenning, E.P.; van Essen, M.; Kam, B.L.; Teunissen, J.J.; Kwekkeboom, D.J. Quality of Life in 265 Patients with Gastroenteropancreatic or Bronchial Neuroendocrine Tumors Treated with [177Lu-DOTA0,Tyr3]Octreotate. J. Nucl. Med. 2011, 52, 1361–1368. [Google Scholar] [CrossRef] [Green Version]
  30. Kvols, L.K.; E Oberg, K.; O’Dorisio, T.M.; Mohideen, P.; De Herder, W.W.; Arnold, R.; Hu, K.; Zhang, Y.; Hughes, G.; Anthony, L.; et al. Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: Results from a phase II study. Endocr.-Relat. Cancer 2012, 19, 657–666. [Google Scholar] [CrossRef] [Green Version]
  31. Martín-Richard, M.; Massutí, B.; Pineda, E.; Alonso, V.; Mármol, M.; Castellano, D.; Fonseca, E.; Galán, A.; Llanos, M.; Sala, M.A.; et al. Antiproliferative effects of lanreotide autogel in patients with progressive, well-differentiated neuroendocrine tumours: A Spanish, multicentre, open-label, single arm phase II study. BMC Cancer 2013, 13, 427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Delpassand, E.S.; Samarghandi, A.; Zamanian, S.; Wolin, E.M.; Hamiditabar, M.; Espenan, G.D.; Erion, J.L.; O’Dorisio, T.M.; Kvols, L.K.; Simon, J.; et al. Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE for Patients with Somatostatin Receptor–Expressing Neuroendocrine Tumors: The first US phase 2 experience. Pancreas 2014, 43, 518–525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Ducreux, M.; Dahan, L.; Smith, D.; O’Toole, D.; Lepère, C.; Dromain, C.; Vilgrain, V.; Baudin, E.; Lombard-Bohas, C.; Scoazec, J.-Y.; et al. Bevacizumab combined with 5-FU/streptozocin in patients with progressive metastatic well-differentiated pancreatic endocrine tumours (BETTER trial)—A phase II non-randomised trial. Eur. J. Cancer 2014, 50, 3098–3106. [Google Scholar] [CrossRef] [PubMed]
  34. Mitry, E.; Walter, T.; Baudin, E.; Kurtz, J.-E.; Ruszniewski, P.; Dominguez-Tinajero, S.; Bengrine-Lefevre, L.; Cadiot, G.; Dromain, C.; Farace, F.; et al. Bevacizumab plus capecitabine in patients with progressive advanced well-differentiated neuroendocrine tumors of the gastro-intestinal (GI-NETs) tract (BETTER trial)—A phase II non-randomised trial. Eur. J. Cancer 2014, 50, 3107–3115. [Google Scholar] [CrossRef]
  35. O’Toole, D.; Ducreux, M.; Bommelaer, G.; Wemeau, J.L.; Bouche, O.; Catus, F.; Blumberg, J.; Ruszniewski, P. Treatment of carcinoid syndrome: A prospective crossover evaluation of lanreotide versus octreotide in terms of efficacy, patient accepta-bility, and tolerance. Cancer 2000, 88, 770–776. [Google Scholar] [CrossRef]
  36. Jacobsen, M.B.; Hanssen, L.E. Clinical effects of octreotide compared to placebo in patients with gastrointestinal neuroendocrine tumours Report on a double-blind, randomized trial. J. Intern. Med. 1995, 237, 269–275. [Google Scholar] [CrossRef]
  37. Larsson, G.; Sjoden, P.O.; Oberg, K.; Eriksson, B.; von Essen, L. Health-related Quality of Life, Anxiety and Depression in Patients with Midgut Carcinoid Tumours. Acta Oncol. 2001, 40, 825–831. [Google Scholar] [CrossRef]
  38. Kwekkeboom, D.J.; Bakker, W.H.; Kam, B.L.; Teunissen, J.J.M.; Kooij, P.P.M.; De Herder, W.W.; Feelders, R.A.; Van Eijck, C.H.J.; De Jong, M.; Srinivasan, A.; et al. Treatment of patients with gastro-entero-pancreatic (GEP) tumours with the novel radiolabelled somatostatin analogue [177Lu-DOTA0,Tyr3]octreotate. Eur. J. Nucl. Med. Mol. Imaging 2003, 30, 417–422. [Google Scholar] [CrossRef] [Green Version]
  39. Pasieka, J.L.; McEwan, A.J.; Rorstad, O. The palliative role of 131I-MIBG and 111In-octreotide therapy in patients with metastatic progressive neuroendocrine neoplasms. Surgery 2004, 136, 1218–1226. [Google Scholar] [CrossRef]
  40. Teunissen, J.J.; Kwekkeboom, D.J.; Krenning, E.P. Quality of Life in Patients With Gastroenteropancreatic Tumors Treated with [177Lu-DOTA(0),Tyr3]octreotate. J. Clin. Oncol. 2004, 22, 2724–2729. [Google Scholar] [CrossRef]
  41. Fröjd, C.; Lampic, C.; Larsson, G.; Von Essen, L. Is satisfaction with doctors’ care related to health-related quality of life, anxiety and depression among patients with carcinoid tumours? A longitudinal report. Scand. J. Caring Sci. 2009, 23, 107–116. [Google Scholar] [CrossRef]
  42. Fröjd, C.; Larsson, G.; Lampic, C.; Von Essen, L. Health related quality of life and psychosocial function among patients with carcinoid tumours. A longitudinal, prospective, and comparative study. Health Qual. Life Outcomes 2007, 5, 18. [Google Scholar] [CrossRef] [Green Version]
  43. Larsson, G.; Janson, E. Anemia in patients with midgut carcinoid, treated with alpha interferon: Effects by erythropoietin treatment on the perceived quality of life. Eur. J. Cancer Care 2008, 17, 200–204. [Google Scholar] [CrossRef]
  44. Kalinowski, M.; Dressler, M.; König, A.; El-Sheik, M.; Rinke, A.; Höffken, H.; Gress, T.M.; Arnold, R.; Klose, K.-J.; Wagner, H.-J. Selective Internal Radiotherapy with Yttrium-90 Microspheres for Hepatic Metastatic Neuroendocrine Tumors: A Prospective Single Center Study. Digestion 2009, 79, 137–142. [Google Scholar] [CrossRef]
  45. Haugland, T.; Veenstra, M.; Vatn, M.H.; Wahl, A.K. Improvement in Stress, General Self-Efficacy, and Health Related Quality of Life following Patient Education for Patients with Neuroendocrine Tumors: A Pilot Study. Nurs. Res. Pract. 2013, 2013, 695820. [Google Scholar] [CrossRef]
  46. Spolverato, G.; Bagante, F.; Wagner, D.; Buettner, S.; Gupta, R.; Kim, Y.; Maqsood, H.; Pawlik, T.M. Quality of life after treatment of neuroendocrine liver metastasis. J. Surg. Res. 2015, 198, 155–164. [Google Scholar] [CrossRef]
  47. Larsson, G.; Von Essen, L.; Sjödén, P.O. Quality of life in patients with endocrine tumors of the gastrointestinal tract: Patient and staff perceptions. Cancer Nurs. 1998, 21, 411–420. [Google Scholar] [CrossRef]
  48. Larsson, G.; Sjödén, P.O.; Öberg, K.; Von Essen, L. Importance-satisfaction discrepancies are associated with health-related quality of life in five-year survivors of endocrine gastrointestinal tumours. Ann. Oncol. 1999, 10, 1321–1327. [Google Scholar] [CrossRef]
  49. Larsson, G.; Von Essen, L.; Sjödén, P.-O. Health-related Quality of Life in Patients with Endocrine Tumours of the Gastrointestinal Tract. Acta Oncol. 1999, 38, 481–490. [Google Scholar] [CrossRef]
  50. Von Essen, L.; Larsson, G.; Öberg, K.; Sjödén, P. ‘Satisfaction with care’: Associations with health-related quality of life and psychosocial function among Swedish patients with endocrine gastrointestinal tumours. Eur. J. Cancer Care 2002, 11, 91–99. [Google Scholar] [CrossRef]
  51. Berglund, G.; Lidén, A.; Hansson, M.G.; Öberg, K.; Sjöden, P.O.; Nordin, K. Quality of life in patients with multiple endocrine neoplasia type 1 (MEN 1). Fam. Cancer 2003, 2, 27–33. [Google Scholar] [CrossRef]
  52. Larsson, G.; Haglund, K.; Von Essen, L. Distress, quality of life and strategies to ‘keep a good mood’ in patients with carcinoid tumours: Patient and staff perceptions. Eur. J. Cancer Care 2003, 12, 46–57. [Google Scholar] [CrossRef]
  53. Davies, A.; Larsson, G.; Ardill, J.; Friend, E.; Jones, L.; Falconi, M.; Bettini, R.; Koller, M.; Sezer, O.; Fleissner, C.; et al. Development of a disease-specific quality of life questionnaire module for patients with gastrointestinal neuroendocrine tumours. Eur. J. Cancer 2006, 42, 477–484. [Google Scholar] [CrossRef]
  54. Larsson, G.; Von Essen, L.; Sjödén, P.-O. Are importance–satisfaction discrepancies with regard to ratings of specific health-related quality-of-life aspects valid indicators of disease- and treatment-related distress among patients with endocrine gastrointestinal tumours? Eur. J. Cancer Care 2007, 16, 493–499. [Google Scholar] [CrossRef]
  55. You, Y.N.; Thompson, G.B.; Young, W.F.; Larson, D.; Farley, D.R.; Richards, M.; Grant, C.S. Pancreatoduodenal surgery in patients with multiple endocrine neoplasia type 1: Operative outcomes, long-term function, and quality of life. Surgery 2007, 142, 829–836; discussion 836.e821. [Google Scholar] [CrossRef]
  56. Haugland, T.; Vatn, M.H.; Veenstra, M.; Wahl, A.K.; Natvig, G.K. Health related quality of life in patients with neuroendocrine tumors compared with the general Norwegian population. Qual. Life Res. 2009, 18, 719–726. [Google Scholar] [CrossRef] [Green Version]
  57. Pezzilli, R.; Campana, D.; Morselli-Labate, A.M.; Fabbri, M.C.; Brocchi, E.; Tomassetti, P. Patient-reported outcomes in subjects with neuroendocrine tumors of the pancreas. World J. Gastroenterol. 2009, 15, 5067–5073. [Google Scholar] [CrossRef]
  58. Van Der Horst-Schrivers, A.N.; Van Ieperen, E.; Wymenga, A.M.; Weijmar-Schultz, W.C.; Kema, I.P.; Willemse, P.H.; De Vries, E.G.; Boezen, H.M.; Meijer, W.G.; De Herder, W.W.; et al. Sexual Function in Patients with Metastatic Midgut Carcinoid Tumours. Neuroendocrinology 2008, 89, 231–236. [Google Scholar] [CrossRef]
  59. Pezzilli, R.; Campana, D.; Morselli-Labate, A.M.; Galassi, E.; Brocchi, E.; Nori, F.; Cipollini, M.L.; Tomassetti, P. Patient-reported outcomes in patients with endocrine tumors of the ileum. Eur. J. Gastroenterol. Hepatol. 2009, 22, 689–694. [Google Scholar] [CrossRef]
  60. Vinik, E.; Silva, M.P.; Vinik, A.I. Measuring the Relationship of Quality of Life and Health Status, Including Tumor Burden, Symptoms, and Biochemical Measures in Patients with Neuroendocrine Tumors. Endocrinol. Metab. Clin. N. Am. 2011, 40, 97–109. [Google Scholar] [CrossRef]
  61. Beaumont, J.L.; Cella, D.; Phan, A.T.; Choi, S.; Liu, Z.; Yao, J.C. Comparison of Health-Related Quality of Life in Patients with Neuroendocrine Tumors with Quality of Life in the General US Population. Pancreas 2012, 41, 461–466. [Google Scholar] [CrossRef] [PubMed]
  62. Petzel, M.Q.B.; Parker, N.H.; Valentine, A.D.; Simard, S.; Nogueras-Gonzalez, G.M.; Lee, J.E.; Pisters, P.W.T.; Vauthey, J.-N.; Fleming, J.B.; Katz, M.H.G. Fear of Cancer Recurrence after Curative Pancreatectomy: A Cross-sectional Study in Survivors of Pancreatic and Periampullary Tumors. Ann. Surg. Oncol. 2012, 19, 4078–4084. [Google Scholar] [CrossRef] [PubMed]
  63. Pavel, M.; Unger, N.; Borbath, I.; Ricci, S.; Hwang, T.-L.; Brechenmacher, T.; Park, J.; Herbst, F.; Beaumont, J.L.; Bechter, O. Safety and QOL in Patients with Advanced NET in a Phase 3b Expanded Access Study of Everolimus. Target. Oncol. 2016, 11, 667–675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Yadegarfar, G.; Friend, L.; Jones, L.; Plum, L.M.; Ardill, J.; Taal, B.; Larsson, G.; Jeziorski, K.; Kwekkeboom, D.; Ramage, J.K.; et al. Validation of the EORTC QLQ-GINET21 questionnaire for assessing quality of life of patients with gastrointestinal neuroendocrine tumours. Br. J. Cancer 2013, 108, 301–310. [Google Scholar] [CrossRef] [Green Version]
  65. Gelhorn, H.L.; Kulke, M.H.; O’Dorisio, T.; Yang, Q.M.; Jackson, J.; Jackson, S.; Boehm, K.A.; Law, L.; Kostelec, J.; Auguste, P.; et al. Patient-reported Symptom Experiences in Patients With Carcinoid Syndrome After Participation in a Study of Telotristat Etiprate: A Qualitative Interview Approach. Clin. Ther. 2016, 38, 759–768. [Google Scholar] [CrossRef] [Green Version]
  66. Haugland, T.; Wahl, A.K.; Hofoss, D.; Devon, H.A. Association between general self-efficacy, social support, cancer-related stress and physical health-related quality of life: A path model study in patients with neuroendocrine tumors. Health Qual. Life Outcomes 2016, 14, 11. [Google Scholar] [CrossRef] [Green Version]
  67. Pearman, T.P.; Beaumont, J.L.; Cella, D.; Neary, M.P.; Yao, J. Health-related quality of life in patients with neuroendocrine tumors: An investigation of treatment type, disease status, and symptom burden. Support. Care Cancer 2016, 24, 3695–3703. [Google Scholar] [CrossRef]
  68. Ruszniewski, P.; Valle, J.W.; Lombard-Bohas, C.; Cuthbertson, D.; Perros, P.; Holubec, L.; Delle Fave, G.; Smith, D.; Niccoli, P.; Maisonobe, P.; et al. Patient-reported outcomes with lanreotide Autogel/Depot for carcinoid syndrome: An international observational study. Dig. Liver Dis. 2016, 48, 552–558. [Google Scholar] [CrossRef] [Green Version]
  69. Halperin, D.M.; Huynh, L.; Beaumont, J.L.; Cai, B.; Totev, T.; Bhak, R.H.; Duh, M.S.; Neary, M.P.; Cella, D. Impact of carcinoid syndrome symptoms and long-term use of somatostatin analogs on quality of life in patients with carcinoid syndrome: A survey study. Medicine 2018, 97, e13390. [Google Scholar] [CrossRef]
  70. Marinova, M.; Mücke, M.; Mahlberg, L.; Essler, M.; Cuhls, H.; Radbruch, L.; Conrad, R.; Ahmadzadehfar, H. Improving quality of life in patients with pancreatic neuroendocrine tumor following peptide receptor radionuclide therapy assessed by EORTC QLQ-C30. Eur. J. Nucl. Med. Mol. Imaging 2017, 45, 38–46. [Google Scholar] [CrossRef]
  71. Milanetto, A.C.; Nordenström, E.; Sundlöv, A.; Almquist, M. Health-Related Quality of Life after Surgery for Small Intestinal Neuroendocrine Tumours. World J. Surg. 2018, 42, 3231–3239. [Google Scholar] [CrossRef]
  72. Jiménez-Fonseca, P.; Carmona-Bayonas, A.; Martín-Pérez, E.; Crespo, G.; Serrano, R.; Llanos, M.; Villabona, C.; García-Carbonero, R.; Aller, J.; Capdevila, J.; et al. Health-related quality of life in well-differentiated metastatic gastroenteropancreatic neuroendocrine tumors. Cancer Metastasis Rev. 2015, 34, 381–400. [Google Scholar] [CrossRef]
  73. Martini, C.; Gamper, E.-M.; Wintner, L.; Nilica, B.; Sperner-Unterweger, B.; Holzner, B.; Virgolini, I. Systematic review reveals lack of quality in reporting health-related quality of life in patients with gastroenteropancreatic neuroendocrine tumours. Health Qual. Life Outcomes 2016, 14, 127. [Google Scholar] [CrossRef] [Green Version]
  74. Aaronson, N.K.; Ahmedzai, S.; Bergman, B.; Bullinger, M.; Cull, A.; Duez, N.J.; Filiberti, A.; Flechtner, H.; Fleishman, S.B.; De Haes, J.C.J.M.; et al. The European Organization for Research and Treatment of Cancer QLQ-C30: A Quality-of-Life Instrument for Use in International Clinical Trials in Oncology. J. Natl. Cancer Inst. 1993, 85, 365–376. [Google Scholar] [CrossRef]
  75. Fayers, P.; Bottomley, A. Quality of life research within the EORTC—The EORTC QLQ-C30. Eur. J. Cancer 2002, 38, 125–133. [Google Scholar] [CrossRef]
  76. Kavadas, V.; Blazeby, J.; Conroy, T.; Sezer, O.; Holzner, B.; Koller, M.; Buckels, J. Development of an EORTC disease-specific quality of life questionnaire for use in patients with liver metastases from colorectal cancer. Eur. J. Cancer 2003, 39, 1259–1263. [Google Scholar] [CrossRef]
  77. Cella, D.F.; Tulsky, D.S.; Gray, G.; Sarafian, B.; Linn, E.; Bonomi, A.; Silberman, M.; Yellen, S.B.; Winicour, P.; Brannon, J.; et al. The Functional Assessment of Cancer Therapy scale: Development and validation of the general measure. J. Clin. Oncol. 1993, 11, 570–579. [Google Scholar] [CrossRef]
  78. Cella, D.; Riley, W.; Stone, A.; Rothrock, N.; Reeve, B.; Yount, S.; Amtmann, D.; Bode, R.; Buysse, D.; Choi, S.; et al. The Patient-Reported Outcomes Measurement Information System (PROMIS) developed and tested its first wave of adult self-reported health outcome item banks: 2005–2008. J. Clin. Epidemiol. 2010, 63, 1179–1194. [Google Scholar] [CrossRef] [Green Version]
  79. Derogatis, L.R. The psychosocial adjustment to illness scale (PAIS). J. Psychosom. Res. 1986, 30, 77–91. [Google Scholar] [CrossRef]
  80. Yao, J.C.; Fazio, N.; Singh, S.; Buzzoni, R.; Carnaghi, C.; Wolin, E.; Tomasek, J.; Raderer, M.; Lahner, H.; Voi, M.; et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): A randomised, placebo-controlled, phase 3 study. Lancet 2016, 387, 968–977. [Google Scholar] [CrossRef]
  81. Cella, D.; Beaumont, J.; Hudgens, S.; Marteau, F.; Feuilly, M.; Houchard, A.; Lapuerta, P.; Ramage, J.; Pavel, M.; Hörsch, D.; et al. Relationship between Symptoms and Health-related Quality-of-life Benefits in Patients with Carcinoid Syndrome: Post Hoc Analyses from TELESTAR. Clin. Ther. 2018, 40, 2006–2020.e2002. [Google Scholar] [CrossRef] [Green Version]
  82. Rinke, A.; Neary, M.P.; Eriksson, J.; Hunger, M.; Doan, T.; Karli, D.; Arnold, R. Health-Related Quality of Life for Long-Acting Octreotide versus Placebo in Patients with Metastatic Midgut Neuroendocrine Tumors in the Phase 3 PROMID Trial. Neuro-endocrinology 2019, 109, 141–151. [Google Scholar] [CrossRef]
  83. Virgolini, I. Overall survival results from the NETTER-1 trial in neuroendocrine tumours: An important milestone. Lancet Oncol. 2021, 22, 1645–1646. [Google Scholar] [CrossRef]
  84. Basch, E.; Deal, A.M.; Kris, M.G.; Scher, H.I.; Hudis, C.A.; Sabbatini, P.; Rogak, L.; Bennett, A.V.; Dueck, A.C.; Atkinson, T.M.; et al. Symptom Monitoring with Patient-Reported Outcomes during Routine Cancer Treatment: A Randomized Controlled Trial. J. Clin. Oncol. 2016, 34, 557–565. [Google Scholar] [CrossRef]
  85. Rincon, E.; Monteiro-Guerra, F.; Rivera-Romero, O.; Dorronzoro-Zubiete, E.; Sanchez-Bocanegra, C.L.; Gabarron, E. Mobile Phone Apps for Quality of Life and Well-Being Assessment in Breast and Prostate Cancer Patients: Systematic Review. JMIR mHealth uHealth 2017, 5, e187. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow chart describing systematic research search and study selection process.
Figure 1. Flow chart describing systematic research search and study selection process.
Cancers 14 01428 g001
Table 1. RCTs and phase II clinical trials in NET patients with reporting of HRQoL outcome measures.
Table 1. RCTs and phase II clinical trials in NET patients with reporting of HRQoL outcome measures.
StudyType of StudySample SizeTreatmentTumor SubtypeHRQoL Measure
Arnold et al., 2005 [11]RCT109octreotide vs. octreotide + IFNGI NETsQLQ-C30
Bajetta et al., 2006 [12]RCT60lanreotide autogel vs. lanreotide microparticleGI NETsQLQ-C30
Rinke et al., 2009 [13]RCT85octreotide LAR vs. placeboGI NETsQLQ-C30
Caplin et al., 2014 [4]RCT204lanreotide autogel vs. placeboGI NETsQLQ-C30, QLQ-GI. NET21
Meyer et al., 2014 [14]RCT86capecitabine + streptozocin + cisplatin vs.
capecitabine + streptozocin
GI NETsQLQ-C30
Vinik et al., 2016 [15]RCT171sunitinib vs. placeboGI NETsQLQ-C30
Vinik et al., 2016 [16]RCT115lanreotide depot/autogel vs. placeboGI NETsQLQ-C30, QLQ-GI.NET-21
Pavel et al., 2017 [17]RCT302everolimus vs. placeboNETsFACT-G
Strosberg et al., 2018 [18]RCT231177Lu-DOTATATE vs. octreotideGI NETsQLQ-C30, QLQ-GI.NET-21
Wymenga et al., 1999 [19]Phase II55lanreotide prolonged-releaseNETsQLQ-C30
Ruszniewski et al., 2004 [20]Phase II71lanreotide prolonged-releaseGI NETsQLQ-C30
Zuetenhorst et al., 2004 [21]Phase II26Interferon followed by meta- iodbenzylguanidinNETsQLQ-C30
Frilling et al., 2006 [22]Phase II1890Y-DOTATOC; 177Lu-DOTATATENETsSF-36
Kulke et al., 2008 [23]Phase II107SunitinibNETsEQ-5D, FACIT-Fatigue scale
Korse et al., 2009 [24]Phase II39octreotide LARGI NETsQLQ-C30
Bushnell et al., 2010 [25]Phase II9090Y-DOTADOCNETsEQ-5D
Cwikla et al., 2010 [26]Phase II6090Y-DOTATATEGI NETsQLQ-C30, QLQ-GI.NET-21
Bodei et al., 2011 [27]Phase II51177Lu-DOTATATENETsQLQ-C30
Claringbold et al., 2011 [28]Phase II33177Lu-DOTATATENETsQLQ-C30
Khan et al., 2011 [29]Phase II256177Lu-DOTATATENETsQLQ-C30
Kvols et al., 2012 [30]Phase II45PasireotideGI NETsFACIT-D
Martin-Richard et al., 2013 [31]Phase II30lanreotide autogelNETsQLQ-C30
Delpassand et al., 2014 [32]Phase II37177Lu-DOTATATEGI NETsQLQ-C30
Ducreux et al., 2014 [33]Phase II34bevacizumab + 5FU/streptozocinGI NETsQLQ-C30
Mitry et al., 2014 [34]Phase II49bevacizumab + capecitabineGI NETsQLQ-C30
Abbreviations: NET = neuroendocrine tumors, HRQoL = health-related quality of life, RCT = randomized controlled trial; GI = gastrointestinal, vs. = versus, IFN = interferon alpha; LAR = long-acting release, 5FU = 5-Flurouracil, SSA = Somatostatin Analogue, PRRT = peptide receptor radionuclide therapy, EORTC QLQ-30 = European Organization for Research and Treatment of Cancer Quality of Life Core Questionnaire, EORTC QLQ GI.NET21 = NET specific EORTC QoL questionnaire, FACT-G = Functional Assessment of Cancer Therapy-General, GHQ = general health questionnaire, SF-36 = 36-Item Short Form Health Survey, FACIT = The Functional Assessment of Chronic Illness Therapy.
Table 2. Health-related quality of life in phase III neuroendocrine tumor studies.
Table 2. Health-related quality of life in phase III neuroendocrine tumor studies.
Clinical TrialHRQoL Tool UsedPatient Population StudiedHRQoL in Comparison to Control Arm
Octreotide vs. Placebo (PROMID) [13]EORTC QLQ-C30GI and unknown primary NETsAt 6-month follow up mark, no statistically significant difference from baseline was observed between two arms.
Lanreotide vs. Placebo (CLARINET) [4]EORTC QLQ-C30, EORTC QLQ-GI.NET21GI, pancreatic NETs, and unknown primaryNo statistically significant difference in the change from baseline to post treatment QoL scores between the two arms.
Everolimus vs. Placebo (RADIANT 4) [17,80]FACT-GGI and lung NETsThe median time to definitive deterioration in FACT-G score was similar between both arms with no significant difference between both arms.
Sunitinib vs. Placebo [6,15]EORTC QLQ-C30Pancreatic NETsOver the first 10 cycles, no differences were observed in the global HRQoL, physical, emotional, cognitive, role, social functioning, or symptom scales except for diarrhea. Statistically significant worsening of diarrhea in the sunitinib arm with a difference of 21.4 points between the two arms was observed.
PRRT vs. Octreotide (NETTER 1) [7]EORTC QLQ-C30, QLQ-GI.NET21Midgut NETsTime to QoL deterioration was statistically longer in the PRRT arm for multiple domains including global health, role functioning, physical functioning, disease-related worries, body image, diarrhea, pain and fatigue.
Telotristat vs. Placebo (TELESTAR) [3,81]EORTC QLQ-C30
EORTC QLQ-GI.NET21
Carcinoid syndrome with diarrheaDurable responders had QoL improvements in EORTC QLQ-C30 global health status, nausea and vomiting, pain, diarrhea, and EORTC QLQ-GI.NET21 gastrointestinal symptoms than non-durable responders both over the DBPT and OLE period *.
* Durable response defined as a BM frequency reduction of >/30% from baseline for >/50% of the time Abbreviations: NET = neuroendocrine tumor, GI = gastrointestinal, HRQoL = health-related quality of life, EORTC QLQ-30 = European Organization for Research and Treatment of Cancer Quality of Life Core Questionnaire, EORTC QLQ GI.NET21 = NET specific EORTC QoL questionnaire, FACT-G = Functional Assessment of Cancer Therapy-General, PRRT = peptide receptor radionuclide therapy, DBTP = double blind treatment period, OLE = open label extension.
Table 3. Common symptoms affecting HRQoL of NET patients.
Table 3. Common symptoms affecting HRQoL of NET patients.
Hormone-related symptomsDiarrhea, flushing, fatigue, loss of appetite, dyspnea, palpitation, loss of weight
Tumor burden-related symptomsAbdominal pain, abdominal distension, ascites, jaundice, compression of adjacent organs
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gosain, R.; Gupta, M.; Roy, A.M.; Strosberg, J.; Glaser, K.M.; Iyer, R. Health-Related Quality of Life (HRQoL) in Neuroendocrine Tumors: A Systematic Review. Cancers 2022, 14, 1428. https://doi.org/10.3390/cancers14061428

AMA Style

Gosain R, Gupta M, Roy AM, Strosberg J, Glaser KM, Iyer R. Health-Related Quality of Life (HRQoL) in Neuroendocrine Tumors: A Systematic Review. Cancers. 2022; 14(6):1428. https://doi.org/10.3390/cancers14061428

Chicago/Turabian Style

Gosain, Rohit, Medhavi Gupta, Arya Mariam Roy, Jonathan Strosberg, Kathryn M. Glaser, and Renuka Iyer. 2022. "Health-Related Quality of Life (HRQoL) in Neuroendocrine Tumors: A Systematic Review" Cancers 14, no. 6: 1428. https://doi.org/10.3390/cancers14061428

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop