• No results found

University of Groningen Neuroendocrine tumors; measures to improve treatment and supportive care de Hosson, Lotte Doortje

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Neuroendocrine tumors; measures to improve treatment and supportive care de Hosson, Lotte Doortje"

Copied!
156
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Neuroendocrine tumors; measures to improve treatment and supportive care

de Hosson, Lotte Doortje

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

de Hosson, L. D. (2019). Neuroendocrine tumors; measures to improve treatment and supportive care.

Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Neuroendocrine tumors;

Measures to improve treatment and supportive care.

(3)

Neuroendocrine tumors; Measures to improve treatment and supportive care Lotte D. de Hosson

PhD dissertation, University of Groningen, the Netherlands

Layout by: XML 2 Publish

Printing by: Gildeprint, Enschede

ISBN: 978-94-034-1443-0

ISBN (electronic version): 978-94-034-1442-3

© Copyright 2019 – Lotte D. de Hosson, Groningen, the Netherlands

All right reserved. No part of this thesis may be reproduced, stored in a retrieval or transmitted in any form or by any means, without prior permission of the author or, when appropriate of the publisher of the published articles.

Printing of this thesis was financially supported by the Stichting Werkgroep Interne Oncologie, Rijksuniversiteit Groningen, Universitair Medisch Centrum Groningen.

(4)

Neuroendocrine tumors; Measures

to improve treatment and

supportive care

Proefschrift

ter verkrijging van de graad van doctor aan de Rijksuniversiteit Groningen

op gezag van de

rector magnificus prof. dr. E. Sterken en volgens besluit van het College voor Promoties.

De openbare verdediging zal plaatsvinden op woensdag 20 februari 2019 om 16.15 uur

door

Lotte Doortje de Hosson

geboren op 15 januari 1985 te Maurik

(5)

Promotor

Prof. dr. E.G.E. de Vries

Copromotor

Dr. A.M.E. Walenkamp

Beoordelingscommissie

Prof. dr. W.W. Herder Prof. dr. H. Hollema Prof. dr. G.A.P. Hospers

Paranimfen

Kim J.H.A. de Hosson Iris I.C. Kok

(6)

Contents

Chapter 1 General introduction and outline of the thesis 6

Chapter 2 Clinical benefit of systemic treatment in patients with advanced

pancreatic and gastro-intestinal neuroendocrine tumors according to ESMO-MCBS and the Net Health Benefit of ASCO revised framework.

16

L.D. de Hosson, L.M. van Veenendaal, Y. Schuller, W.T. Zandee, W.W. de Herder, M.E.T. Tesselaar, H.J. Klümpen, A.M.E. Walenkamp.

Chapter 3 a) Web-based tailored information and support for patients with a

Neuroendocrine tumor; WIN-pilot. 40

G. Bouma, L.D. de Hosson, C. E. van Woerkom, H. van Essen, G.H. de Bock, J.M. Admiraal, A.K. L. Reyners, A. M. E. Walenkamp.

b) Web-based tailored nformation and support for patients with a

Neuroendocrine tumor; WIN-study 58

L.D. de Hosson*, G. Bouma*, J. Stelwagen, H. van Essen, G.H. de Bock, D.J.A. de Groot, E.G.E. de Vries, A.M.E. Walenkamp.

Chapter 4 Towards optimal personalized diet and vitamin supplementation

in NET patients; DIVIT-pilot. 78

L.D. de Hosson, J. Stelwagen, G. Bouma, B. Sijtema, S, Huitema, H.J.R. van Faassen, G.H. de Bock, D.J.A. de Groot, M.J.E. Campmans-Kuijpers, I.P. Kema, E.G.E. de Vries, A.M.E. Walenkamp.

Chapter 5 Heterogeneity in neuroendocrine tumor lesions visible with

[18F]FDOPA PET/CT 88

L.D. de Hosson, A.M. van der Loo- van der Schaaf, R. Boellaard, J. H. van Snick, E.G.E. de Vries, A. H. Brouwers, A.M.E. Walenkamp.

Chapter 6 Reasons for the cold immune microenvironment of

neuroendocrine tumours. 108

L.D. de Hosson*, G. Bouma*, G. Kats-Ugurlu, M. Bulthuis, E.G.E. de Vries, M. van Faassen, I.P. Kema, A.M.E. Walenkamp.

Chapter 7 Summary and future perspectives 126

Appendix Dutch summary, Nederlandse samenvatting 143

(7)
(8)

General introduction and

outline of the thesis

(9)
(10)

1

9

General introduction and outline of the thesis

Background

Neuroendocrine tumors (NETs) are rare tumors with an incidence 3.5/100,000 per year and a prevalence of 35/100,000 (1-3). The tumor arises from secretory cells of the neuroendocrine system. Patients with NET may experience various symptoms from the tumor mass, the output of hormones secreted by the tumor, and treatment accompany-ing side effects (4). Median survival in patients with regional and distant metastatic NET is 105 and 56 months respectively, illustrating the indolent nature of most NETs. Patients with localized and metastatic gastro-intestinal NET who have lived with NET for 5 years have a probability of surviving an additional 5 years of 98.2% and 73.5% respectively (3,5). This underscores the fact that most patients live relatively long. Patients with NET have a lower quality of life (QoL) compared with the general population (6-8). Patients are frequently metastasized at the time of diagnosis. Radical surgery is the only curative treatment for patients presenting with local or regional disease, solitary metastases or resectable liver metastases. Non-curative systemic treatment options have variable, limited success (9-20).

Ideally, in trials investigating novel drugs, the primary aim should be to study clinical relevant endpoints such as overall survival (OS) and QoL (21). However, in gastrointes-tinal and pancreatic NET (GEP-NET) patients, survival analysis can be challenging as the prolonged (natural) course of the disease often results in trials that allow crossover towards the experimental arm, or that allow second line therapies which could influence OS (22-24). Therefore, use of OS as primary endpoint is challenging, and QoL becomes more important to determine meaningful clinical benefit in the treatment of GEP-NET. In cancer survivors adequate information provision is associated with health related QoL and is an essential aspect of supportive care (25,26). For patients with NET it is difficult to find meaningful and understandable information about their diagnosis. Other important factors influencing QoL in patients with NET are gastro-intestinal problems and increased frequency of bowel movements (27). Furthermore somatostatin analogues (SSA) decrease their pancreatic exocrine function and can result in fat-soluble vitamin deficiencies (28-30). In case of high serotonin production, tryptophan deficiency, could

also lead to niacin (vitamin B3) deficiency and lead to symptoms (29,31). Currently,

strik-ingly little is known about how SSA using patients with NET should be best supported and if dietary support and supplementation of vitamins is feasible (32,33).

Clinical symptoms are, among others, associated with reduction in QoL. Symptoms in patients with NET can be caused by the high secretion of neuroendocrine amines. Therefore, local treatment of an active amine producing tumor lesion would be poten-tially meaningful to improve QoL (34). Previously it was demonstrated that tumor burden

measured with total [18F]FDOPA uptake correlates with catecholamine pathway activity

(11)

Chapter 1

10

method in patients with small intestinal NET, this might be of added value in the selec-tion of lesions for local treatment.

Over the last years, immunotherapy with immune checkpoint inhibitors like the pro-grammed death-1 and propro-grammed death ligand-1 (PD1 and PD-L1) antibodies have shown antitumor activity across numerous tumor types by activation of T-cells in the tumor microenvironment (36). Interestingly for NETs, there is also a major interest in the tryptophan-degrading enzymes indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) which are involved in the immune response and are expressed in several tumor types. In serotonin producing NETs IDO and TDO play a special im-portant role since tryptophan is the precursor of the serotonin pathway (37,38). There is, however limited information with regards to the complex interactions of NET tumour cells with their surrounding immune microenvironment. Consequently knowledge about potential targets for immunotherapy in patients with NET is limited.

Aim of the thesis

The aim of this thesis is to measure and improve the clinical benefit of treatment and supportive care provided to patients with NET. We investigated tailored supportive measures for these patients as well as novel treatment approaches.

Outline of the thesis

In chapter 2 an overview of existing literature about trials comparing systemic antitumor treatment for GEP-NET versus a control is presented and their clinical benefit is inter-preted. The electronic databases (PubMed, Embase) were searched for papers report-ing comparative trials, conducted in adult patients with GEP-NET. The clinical benefit of the investigated therapies reported in the selected trials was determined according to the European Society of Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS) and the net health benefit score according to the American Society of Clinical Oncology (ASCO) revised framework by four independent assessors.

QoL is an important factor that contributes to clinical benefit. Factors associated with enhanced QoL include; less clinical symptoms, and adequate information provision (39,40). The internet was found to be a useful source of general information for some

patients with NET (41). In chapter 3A the feasibility of a web based system

consist-ing of self-screenconsist-ing of problems and care needs in patients with NET was analyzed. Newly diagnosed NET patients were randomized between standard care (n=10) or intervention with additional access to the web-based system (n=10) during 12 weeks.

(12)

1

11

General introduction and outline of the thesis

Patients completed questionnaires regarding received information, distress, QoL, and empowerment. The intervention group completed a semi-structured interview to assess patients’ opinion on the web-based system.

Subsequently, in a randomized controlled trial 90 patients with NET were stratified between those newly diagnosed (< 6 months, n=28) or with a longer duration of the disease (n=74) and randomized between standard care (n=49) or intervention with ad-ditional access to the web-based system (n=53) during 12 weeks. Patients completed questionnaires about distress, perception and satisfaction of received information, QoL, and empowerment. The intervention group also completed a questionnaire based on the technical acceptance model regarding their use of and opinion on the web-based

system (chapter 3B).

In chapter 4 we described a feasibility study, in which 15 patients with NET using a SSA for over 6 months were counseled by a dietician for a personalized dietary advice and received supplementation of deficient vitamins A, D, E, K, B12 and vitamin B3 (niacin). Feasibility was assessed by calculation of participation/dropout rate and number of (severe) adverse events related to the intervention. At baseline, after 4 and 18 weeks, vitamins were measured, and QoL, distress, empowerment and nutrition state were assessed.

Part of symptoms in patients with NET are caused by the secretion of neuroendocrine amines and local treatment of a high metabolic active lesion would be another potential

method to improve QoL meaningful in patients with NET (34). Therefore, in chapter 5

we analyzed intertumoral heterogeneity with 18F-dihydroxyphenylalanine (DOPA)

Posi-tron Emission Tomography (PET) scans in patients with small intestinal (SI-) NET and

investigated if tumor lesions with substantially higher [18F]FDOPA uptake than the

ma-jority of the other lesions within a patient could be identified within a patient. [18F]FDOPA

PET scans of 38 patients, of which 35 serotonin producing, were analyzed. For all

tumor lesions the [18F]FDOPA uptake was calculated by dividing the standard uptake

value (SUV) peak of the tumor lesion by the SUV mean of the background organ. The magnitude of heterogeneity between lesions within a patient was calculated by dividing

the lesion with highest [18F]FDOPA uptake by the one with lowest [18F]FDOPA uptake.

In patients (n=20) with more than 10 metastases the lesions with a higher tracer uptake than the upper inner or outer fence (more than 1.5 or 3 times the interquartile range

above the third quartile) were defined as lesions with mild or extreme high [18F]FDOPA

uptake respectively.

In chapter 6 we investigated the immune microenvironment of neuroendocrine tumors,

by pathologic analysis of tumor tissue of 51 patients with serotonin or non-serotonin producing NET grade 1 or 2. Immunohistochemically analyses were performed for PD-L1, T-cells, IDO, TDO, mismatch repair proteins and activated fibroblasts. Serotonin

(13)

Chapter 1

12

production was measured by high performance liquid chromatography fluorometry of 5-hydroxyindolacetic acid (5-HIAA) in 24-h urine and/or serotonin in platelet rich plasma.

Finally the findings of this thesis are summarized in chapter 7 followed by a discussion

on the scientific and clinical implications and direction for future research. Chapter 8 comprises the summary of this thesis in Dutch.

(14)

1

13

General introduction and outline of the thesis

References

1. Lawrence B, Gustafsson BI, Chan A, Svejda B, Kidd M, Modlin IM. The epidemiology of gas-troenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin North Am 2011;40:1-18. 2. Dasari A, Shen C, Halperin D, et al. Trends in the incidence, prevalence, and sur-vival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol 2017;3:1335-42.

3. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 2008;26:3063-72.

4. Oberg KE. The management of neuroendocrine tumours: current and future medical therapy options. Clin Oncol (R Coll Radiol) 2012;24:282-93.

5. Mocellin S, Nitti D. Gastrointestinal carcinoid: epidemiological and survival evidence from a large population-based study (n = 25 531). Ann Oncol 2013;24:3040-4.

6. Beaumont JL, Cella D, Phan AT, Choi S, Liu Z, Yao JC. Comparison of health-related quality of life in patients with neuroendocrine tumors with quality of life in the general US popula-tion. Pancreas 2012;41:461-6.

7. Frojd 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 compara-tive study. Health Qual Life Outcomes 2007;5:18.

8. Haugland T, Vatn MH, Veenstra M, Wahl AK, Natvig GK. Health related quality of life in patients with neuroendocrine tumors compared with the general Norwegian population. Qual Life Res 2009;18:719-26.

9. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011;364:501-13.

10. Faivre S, Niccoli P, Raoul JL, et al. Updated overall survival (OS) analysis from a phase III study of sunitinib vs placebo in patients (PTS) with advanced, unresectable pancreatic neuroendocrine tumor (NET). Ann Oncol 2012;23:ix376.

11. Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospec-tive, 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;27:4656-63.

12. Rinke A, Wittenberg M, Schade-Brittinger C, et al. Placebo-controlled, double-blind, pro-spective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors (PROMID): Results of long-term survival. Neuroendocrinology 2017;104:26-32.

13. Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic enteropancreatic neuroen-docrine tumors. N Engl J Med 2014;371:224-33.

14. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med 2011;364:514-23.

15. Yao JC, Pavel M, Lombard-Bohas C, et al. Everolimus for the treatment of advanced pancreatic neuroendocrine tumors: Overall survival and circulating biomarkers from the randomized, phase III RADIANT-3 study. J Clin Oncol 2016;34:3906-13.

16. Yao JC, Fazio N, Singh S, 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;387:968-77.

(15)

Chapter 1

14

17. Meyer T, Qian W, Caplin ME, et al. Capecitabine and streptozocin +/- cisplatin in advanced gastroenteropancreatic neuroendocrine tumours. Eur J Cancer 2014;50:902-11.

18. Pavel ME, Hainsworth JD, Baudin E, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 2011;378:2005-12. 19. Pavel M, Oberg KE, Hainsworth JD, et al. Everolimus plus octreotide long-acting release

(LAR) for the treatment of advanced neuroendocrine tumors (NET) associated with carci-noid syndrome (RADIANT-2): Updated overall survival results. Eur J Cancer 2013;49:S577. 20. Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of 177Lu-dotatate for midgut

neuro-endocrine tumors. N Engl J Med 2017;376:125-35.

21. Booth CM, Eisenhauer EA. Progression-free survival: meaningful or simply measurable? J Clin Oncol 2012;30:1030-3.

22. Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treatment of neuroendocrine tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol 2011;29:934-43.

23. Lebwohl D, Kay A, Berg W, Baladi JF, Zheng J. Progression-free survival: gaining on overall survival as a gold standard and accelerating drug development. Cancer J 2009;15:386-94. 24. Riechelmann RP, Rego J. Progression-free survival in neuroendocrine tumors: preferred

end point, but how should it be defined? J Clin Oncol 2011;29:2835-6.

25. Rehnberg G, Absetz P, Aro AR. Women’s satisfaction with information at breast biopsy in breast cancer screening. Patient Educ Couns 2001;42:1-8.

26. Husson O, Mols F, van de Poll-Franse LV. The relation between information provision and health-related quality of life, anxiety and depression among cancer survivors: a systematic review. Ann Oncol 2011;22:761-72.

27. Pearman TP, Beaumont JL, Cella D, Neary MP, Yao J. Health-related quality of life in pa-tients with neuroendocrine tumors: an investigation of treatment type, disease status, and symptom burden. Support Care Cancer 2016;24:3695-703.

28. Fiebrich HB, Van Den Berg G, Kema IP, et al. Deficiencies in fat-soluble vitamins in long-term users of somatostatin analogue. Aliment Pharmacol Ther 2010;32:1398-404. 29. Shah GM, Shah RG, Veillette H, Kirkland JB, Pasieka JL, Warner RR. Biochemical

as-sessment of niacin deficiency among carcinoid cancer patients. Am J Gastroenterol 2005;100:2307-14.

30. Lembcke B, Creutzfeldt W, Schleser S, Ebert R, Shaw C, Koop I. Effect of the somatostatin analogue sandostatin (SMS 201-995) on gastrointestinal, pancreatic and biliary function and hormone release in normal men. Digestion 1987;36:108-24.

31. Bouma G, van Faassen M, Kats-Ugurlu G, de Vries EG, Kema IP, Walenkamp AM. Niacin (Vitamin B3) supplementation in patients with serotonin-producing neuroendocrine tumor. Neuroendocrinology 2016;103:489-94.

32. Maton PN. The carcinoid syndrome. JAMA 1988;260:1602-5.

33. Mohyi D, Tabassi K, Simon J. Differential diagnosis of hot flashes. Maturitas 1997;27:203-14.

34. Pavel M, Baudin E, Couvelard A, et al. ENETS Consensus Guidelines for the manage-ment of patients with liver and other distant metastases from neuroendocrine neoplasms of foregut, midgut, hindgut, and unknown primary. Neuroendocrinology 2012;95:157-76.

(16)

1

15

General introduction and outline of the thesis

35. Fiebrich HB, de Jong JR, Kema IP, et al. Total 18F-dopa PET tumour uptake reflects meta-bolic endocrine tumour activity in patients with a carcinoid tumour. Eur J Nucl Med Mol Imaging 2011;38:1854-61.

36. Guan J, Lim KS, Mekhail T, Chang CC. Programmed death ligand-1 (PD-L1) expression in the programmed death receptor-1 (PD-1)/PD-L1 blockade: A key player against various cancers. Arch Pathol Lab Med 2017;141:851-61.

37. Uyttenhove C, Pilotte L, Theate I, et al. Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 2003;9:1269-74.

38. van Baren N, Van den Eynde BJ. Tryptophan-degrading enzymes in tumoral immune resis-tance. Front Immunol 2015;6:34.

39. Davies AH, Larsson G, Ardill J, et al. Development of a disease-specific Quality of Life questionnaire module for patients with gastrointestinal neuroendocrine tumours. Eur J Cancer 2006;42:477-84.

40. Yadegarfar G, Friend L, Jones L, et al. Validation of the EORTC QLQ-GINET21 question-naire for assessing quality of life of patients with gastrointestinal neuroendocrine tumours. Br J Cancer 2013;108:301-10.

41. Feinberg Y, Law C, Singh S, Wright FC. Patient experiences of having a neuroendocrine tumour: a qualitative study. Eur J Oncol Nurs 2013;17:541-5.

(17)
(18)

Clinical benefit of systemic

treatment in patients with advanced

pancreatic and gastro intestinal

neuroendocrine tumors according to

ESMO-MCBS and ASCO framework

L.D. de Hosson

1

, L.M. van Veenendaal

2

, Y. Schuller

3

, W.T. Zandee

4

,

W.W. de Herder

4

, M.E.T. Tesselaar

2

, H.J. Klümpen

3

, A.M.E. Walenkamp

1

1Department of Medical Oncology, University Medical Center Groningen,

University of Groningen, Groningen, The Netherlands

2Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam,

The Netherlands

3Department of Medical Oncology, Academic Medical Center, Amsterdam,

The Netherlands

(19)

Chapter 2

18

Abstract:

Background

Assessment of clinical benefit of systemic treatments of rare diseases including gastro-enteropancreatic neuroendocrine tumors (GEP-NET) is challenging. Recently several tools have been developed to grade clinical benefit of cancer drugs. European Society for Medical Oncology (ESMO) has developed the ESMO Magnitude of Clinical Benefit Scale (ESMO-MCBS). The American Society of Clinical Oncology (ASCO) developed and revised the ASCO framework consisting of the Net Health Benefit (NHB) score juxtaposed against the costs of the treatment. In this review we graded systemic treat-ments for GEP-NET patients with both frameworks.

Methods

The electronic databases (PubMed, Embase) were searched for papers reporting comparative trials, conducted in adult GEP-NET patients in the English language. Papers were assessed according to the ESMO-MCBS and the NHB part of the ASCO revised Framework (NHB-ASCO-F) by 4 independent assessors, discrepancies were discussed.

Results

The search yielded 32 trials of which 6 trials were eligible for grading with the ESMO-MCBS resulting in scores of 2 or 3. Eight trials were eligible for grading with the NHB-ASCO-F; resulting in scores between 37.6 and 57.4. Trials that were not primary assessable by the tools were analyzed separately. Consensus between assessors was reached in 68% of trials with the ESMO-MCBS and in 23% of trials with the NHB-ASCO-F.

Conclusion

The currently used systemic treatments for GEP-NET patients had low scores accord-ing to the NHB-ASCO-F and none could be graded as meanaccord-ingful clinical beneficial according to the ESMO-MCBS. Despite the low incidence, the heterogeneous patient population and relatively long natural course of NET, future studies on new treatment modalities should aim for high clinical benefit outcomes.

Keywords: ESMO-MCBS, ASCO, pancreatic and gastro-intestinal, neuroendocrine tumors, clinical benefit, value

(20)

2

19

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Introduction

Neuroendocrine tumors are rare malignancies with an incidence of 3.5/100,000 per year and a prevalence of 21.6/100,000 in the last decade (1). Median survival time is 77 months in patients with regional and 24 months in patients with distant metastatic pancreatic NET (pNET) and 105 months and 56 months in case of respectively regional and distant metastatic intestinal NET disease (2). GEP-NET patients are frequently metastasized at the time of initial diagnosis. Surgery is the only curative treatment. Non-curative systemic treatment options include somatostatin analogues (SSA), che-motherapy, targeted agents, and peptide receptor radionuclide therapy (PRRT) with variable, limited success (3-12).

Ideally, in clinical trials investigating novel drugs, the primary aim should be to study endpoints such as the overall survival (OS), quality of life (QoL) and treatment toxicity. However, for trials investigating new agents in GEP-NET patients, survival analysis can be challenging as the prolonged (natural) course of the disease often results in trials al-lowing crossover towards the experimental arm, which influences OS. Crossover to the experimental arm or to second line therapies has impact on OS data (13). Therefore, progression-free survival (PFS) was recommended as a feasible and relevant primary end point for both, phase II and III trials in GEP-NET, by the expert consensus report of the National Cancer Institute Neuroendocrine Tumor Clinical Trials Planning Meeting (13).

Recently, among others, ESMO has developed a validated and reproducible tool to assess the magnitude of clinical benefit for drugs for solid tumors, the ESMO-MCBS (14). The ASCO developed the ASCO framework for solid tumors and haematological malignancies consisting of the NHB score juxtaposed against the costs of the treatment (15,16). Both tools have been applied in several solid malignancies (14-18). However, the ASCO framework was assessed only in a few studies. Treatments for GEP-NET are not yet evaluated using the tools. In this review we therefore investigated the value of current systemic antitumor treatments for GEP-NET patients and their eligibility for grading with both ESMO-MCBS and NHB-ASCO-F.

Methods

Search strategy

For detailed description of methods see supplementary data. In brief, trials were

searched between Aug 1st 2015 to Jan 31th 2016, in the databases PubMed and

EM-BASE. The articles that were found, were screened using title and abstract to select trials published in the English language and comparing systemic treatment modalities

(21)

Chapter 2

20

for the treatment of GEP-NET in humans. Furthermore the reference lists of national and international guidelines, included trials and conference abstracts were reviewed for additional relevant articles.

Selection criteria for trials

Comparative trials investigating systemic antitumor treatment for well-differentiated GEP-NET patients were analyzed. Studies in patients with grade 3 neuroendocrine neoplasms were not included. Criteria for assessment are summarized in Table S1. Tri-als were selected if at least 50 % of participants were diagnosed with GEP-NET, and the other participants had a NET of unknown or other origin. Abstracts were not included.

ESMO-MCBS and the NHB-ASCO-F

ESMO-MCBS grades, in the non-curative setting, range from 1-5, with grade 4 and 5 representing meaningful clinical benefit (14). The NHB-ASCO-F ranges from -20 to 180, with a higher score representing a better score, no cut-off value was provided to define clinical benefit (15,16,18). All relevant comparative trials were assessed according to both tools. Relevant trials that did not meet all criteria as mentioned in Table S1 were assessed separately, based on the available data.

Assessment of trials

Four members from all Dutch European Neuroendocrine Tumor Society (ENETS) cen-tres of excellence independently scored the trials according to the ESMO-MCBS and the NHB-ASCO-F. After obtaining scores of these assessors we noticed a wide variation in results. During a consensus meeting additional agreements were defined. Next, trials were assessed again according to the additional agreements. The number of trials to which the same score was awarded between three or four assessors was registered after each of the two scoring sessions. Two phase III trials were published after the consensus meeting and were assessed according our agreements (8,12). Therefore an additional assessment session was not necessary for these trials.

Results

Trial selection and characterization

The primary search strategy yielded 1,942 potentially relevant papers, of which 1,676 remained when duplicates were discarded (Figure S1). After screening of title and abstract, 223 papers were preselected evaluating systemic antitumor treatment for GEP-NET patients.

(22)

2

21

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Thirty nine papers in which 35 comparative trials in GEP-NET patients were described were selected for further analysis. Nine trials fulfilled all criteria as summarized in Table S1 and were assessable according to the ESMO-MCBS and the NHB-ASCO-F (Table 1) (3-12). Only one trial, the RADIANT-4, described in their protocol that crossover was not allowed (10,11). None of the trials investigated adjuvant treatment in GEP-NET or had a curative intent.

The other 26 comparative trials, investigating systemic treatment in GEP-NET patients, did not meet all criteria for assessment with both tools (Table S1), or evaluated partici-pants of which less than 50% had a GEP-NET. Main difficulties for assessment with the tools were summarized in Figure S2. As currently used national and international guidelines are based upon these trials, we analyzed them separately with the available data (Table S2, supplementary data; references).

Assessment according to the ESMO-MCBS

Six trials were assessable with the ESMO-MCBS. The calculated score ranged from 2 to 3 corresponding with a low level of clinical benefit. RADIANT-3 and RADIANT-4 analyzed everolimus versus placebo in pNET and non-functional advanced NET, re-spectively (9-11). Application of ESMO-MCBS for RADIANT-3 and RADIANT-4 resulted in a preliminary score of 3 reflecting a longer PFS. A hazard ratio with a lower limit of the 95% confidence interval ≤ 0.65 for PFS determined the preliminary score. No significant difference in important adverse events as compared to the placebo control arm was detected. QoL of everolimus in pNET patients was reported in a recent abstract which described a single-arm phase IV study, performed in patients who started with evero-limus (19). After 6 months, no improvement in QoL was detected. Because this was a single-arm study and the ESMO-MCBS could only be applied to comparative outcome studies, the preliminary score of the RADIANT-3 will not be downgraded, despite lack of improvement of QoL, when these data are published in full-report. QoL of RADIANT-4 trial was reported in a post-hoc analysis and recently published (11). No improvement of QoL by everolimus versus the control arm was demonstrated. Therefore the preliminary score of the RADIANT-4 was downgraded to a final score 2. The NETTER-1 trial

inves-tigated 177Lu-dotatate with octreotide LAR versus octreotide LAR alone in patients with

metastatic midgut NET (8). The calculated ESMO-MCBS score was 3, which implies a longer PFS in the intervention group as compared with control arm. Furthermore, there was no significant difference in important adverse events as compared to the control arm and no QoL data were documented. In the CLARINET trial GEP-NET patients or patients with NET of unknown origin were treated with lanreotide in the intervention arm versus placebo in the control arm (3). This trial showed an improvement in PFS. Data of QoL did not show an improvement in QoL for the intervention group, resulting in a score of 2. In the PROMID trial patients with a metastasized midgut NET were

(23)

Chapter 2

22

2

23

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Table 1: Clinical benefit of systemic treatment for GEP-NET according to ESMO-MCBS (ESMO-MCBS) and according ASCO revised framework (NHB) ESMO-MCBS

ASCO revised framework

Intervention vs control

Setting and reference

Primary Outcome HR of primary outcome/ RR Intoxication data Improvement QOL (Y es/No) ESMO- MCBS Clinical benefit score

Intoxication

data

Bonus points Net health benefit

Lanreotide vs placebo GEP-NET or NET of unknown origin (3) PFS 0.47 (0.30-0.73) NS No 2 42.4 NC 0 NA

Capecitabine and streptozocin and cisplatin vs capecitabine and streptozocin

Advanced, irresectable, NET

of pancreas, gastroduodenum, or unknown primary (4) RR 16 % vs 12 % NS No NA 0 NC 0 NA

Octreotide and everolimus vs octreotide and placebo

Advanced, progressive NET

with carcinoid syndrome (5) PFS 0.77 (0.59-1.00) NS NA NA 18.4 NC 0 NA Sunitinib vs placebo

Progressive, advanced pancreatic

NET (6) PFS 0.42 (0.26-0.66) NS No 2 46.4 -5 16 57.4

Octreotide LAR vs placebo

First line metastatized midgut

NET (7) PFS 0.34 (0.20-0.59) NS No 2 52.8 NC 16 NA

177Lu-Dotatate vs octreotide LAR

Metastastatic midgut NET

(8) PFS 0.21 (0.13-0.33) NS NA 3 63.8 NC 16 NA Everolimus vs placebo

Advanced pancreatic NET

(9) PFS 0.34 (0.26-0.44) NS NA 3 52.8 -20 16 48.8 Everolimus vs placebo

Advanced, progressive

non-functional lung or GI-NET

(10,1 1) PFS 0.48 (0.35-0.67) NS No 2 41.6 -20 16 37.6

Bevacizumab and octreotide LAR vs interferon alfa-2b and octreotide LAR

Advanced, NET

with progression

or other poor prognostic features

(12) RR 0.90 (0.72-1.12) NS NA NA 8.4 NC 0 8.4

A: Name of first author

. References are reported in reference list.

ASCO= American Society of Clinical Oncology , ESMO-MCBS= European Society of Medical Oncology- Magnitude of Clinical Benefit Scale, GEP-NET=gastrointestinal and/ or pancreatic neuro-end ocrine tumor , HR= hazard ratio, NET=neuroendocrine tumor , vs=versus, LAR=long acting reagents, NA= not applicable, NC= no consensus, NS=no significant

(24)

Chapter 2

22

2

23

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

randomized between octreotide LAR and placebo (7). Further enrolment was stopped after inclusion of 85 patients instead of the planned 162 patients, because of observed positive effects of octreotide LAR on tumor growth and a slow recruitment rate. This trial resulted in an ESMO-MCBS score of 2. The trial reported by Raymond et al. analyzed sunitinib versus placebo in patients with progressive, advanced pancreatic NET (6). Ap-plication of ESMO-MCBS resulted in a score of 2. An improvement in PFS was shown. Because reported QoL data did not show improvement, the preliminary score of 3 had to be downgraded one point. This trial had an early closure after randomization of 171 patients, as observed and recommended by the safety monitoring board due to more serious adverse events and a higher frequency of death in the placebo group as well as a difference in PFS favoring sunitinib. The trial analyzing SSA and interferon versus SSA and bevacizumab, in advanced NET patients with progression or other poor prog-nostic features, showed no significant difference in its primary endpoint PFS (12). The RADIANT-2 analyzed everolimus with long acting octreotide in advanced NET tumor patients associated with carcinoid syndrome (5). The PFS with a hazard ratio of 0.77 (0.59-1.00) did not show a statistically significant clinical benefit. The trial analyzing capecitabine and streptozocin with or without cisplatin did also not show a statistically significant clinical benefit (4). Furthermore in these trials no improvement in toxicity, Qol, or OS was seen, and therefore the ESMO-MCBS was not applicable .

Consensus in the ESMO-MCBS score was reached in 2 of 6 trials after the first scor-ing session (3-7,9-11). The variation in awarded scores was related to differences in interpretation of data, like the significance of the primary endpoint, and adjustment of the preliminary score for lack of improvement in QoL. After the second scoring session consensus was reached in 4 of 6 trials (3-7,9-11).

Assessment according to the NHB-ASCO-F

Nine trials were assessable for the NHB-ASCO-F (3-12). The CBS varied from 0 to 63.8. In the trial reported by Raymond et al. and the RADIANT-3 and RADIANT-4 trials the treatment arm experienced more adverse events, resulting in a toxicity score of -5 and -20, respectively (6,9-11). Bonus points were awarded for long term disease control; if, at a time point that was twice the median PFS for the control regimen, the percentage of patients having PFS was at least 50% higher for the intervention arm compared with the control arm, to the trial reported by Raymond et al, PROMID-trial, RADIANT-4, RADIANT-3 and the NETTER-1 (6-11). No consensus was obtained for the score of the trials assessed by the NHB-ASCO-F after 2 scoring sessions. This was generally related to discrepancies in interpretation of toxicity score. Finally, after an additional discussion in 4 trials consensus was obtained for NHB and in 5 trials consensus was reached for the CBS and bonus points.

(25)

Chapter 2

24

Discussion

To define the clinical benefit of systemic antitumor treatment in GEP-NET patients, we systematically applied the ESMO-MCBS and the NHB-ASCO-F to relevant trials. Six out of 35 trials fulfilled all requirements to be assessed with the ESMO-MCBS resulting in an ESMO-MCBS score of 2 or 3, while 9 trials could be assessed with the NHB-ASCO-F and resulted in scores between 37.6 to 57.4. No clear cut-off value to define clinical benefit with the ASCO framework was provided (15,16). None of the trials that were included in our analysis could demonstrate a meaningful clinical benefit according to the employed tools. The ESMO-MCBS scores were generally lower than in other tumor types, like metastasized breast or colorectal cancer (14,17). In more common tumor types, sufficient numbers of patients can be included in trials powered to detect a difference in OS between the intervention and control group. In NET, such large trials are scarce. In addition, OS difference detection is challenging in NET patients. There-fore, PFS is a frequently used, primary endpoint in NET trials. An improvement in PFS has less impact compared to OS in the ESMO-MCBS scores and the NHB-ASCO-F. Furthermore, none of the assessable trials in GEP-NET, showed an improvement in QoL or less toxicity as compared to the control arm. Therefore, these outcomes did not result in an upgrade of the preliminary score in the ESMO-MCBS.

Ideally OS and QoL should be the primary endpoint of all trials with new drugs (20). This is challenging in NET. Reasons for this include the heterogeneous patient popu-lation, the long natural course of the disease and the wide variability of subsequent lines of therapy after progression. The heterogeneity of the patients composing the NET population is important to take into account. This illustrates the importance of well-defined inclusion criteria. The eligibility criteria of the RADIANT-2 trial included patients with; serotonin producing NET and progressive, low- or intermediate-grade and advanced disease (5). Despite these clear criteria still large variation in patient and tumor characteristics between patients exists. Despite the randomized design of the trial, this could have influenced PFS.

Current guidelines for GEP-NET anti-tumor treatment are based on clinical trials and on expert opinion. These guidelines influence our daily clinical practice. Therefore, we also assessed trials that could not be fully assessed by the ESMO-MCBS and ASCO framework (table S2). The control arms of trials analyzing chemotherapy included interferon, dacarbazine, and other kinds of chemotherapy, respectively. Given current knowledge about the effects of chemotherapy, the control arm, nowadays, likely would not have contained chemotherapy. With this analysis we demonstrated, that current guidelines are partially based on studies not powered to determine endpoints that show clinical benefit.

(26)

2

25

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Large trials were conducted by enormous efforts of the international NET community (3-12). This has provided us with important data. Improvements could include trials using a pre-selected patient population. This is already implemented in PRRT, where somatostatin-receptor-positive patients are selected for treatment (8). Furthermore, sequential multiple assignment randomized trials can be used for hypothesis genera-tion followed by a confirmatory randomized controlled trial (RCT) (21). In addigenera-tion, trials should be focused on reporting QoL and be of sufficient follow-up duration. For GEP-NET patients, use of QoL could be of additional importance because OS is generally prolonged in this cohort.

The importance of long follow-up was previously demonstrated in a trial in patients with anaplastic oligodendrogliomas. OS of adjuvant chemoradiotherapy versus radiotherapy was not improved at a median follow-up of 60 months, but was improved in the chemo-radiotherapy arm at a median follow-up of 140 months (22).

International collaboration in clinical trials and investigator-driven trials should be further expanded to increase evidence based data to support treatment decisions and lead to meaningful clinical benefit for GEP-NET patients.

Although not the aim of our analysis, we found some limitations of the tools. The ESMO-MCBS downgrades the preliminary score of a trial if no significant improvement of QoL data was shown (14). If QoL was not reported, the preliminary score did not have to be downgraded (8,9).

Furthermore, we noticed during the scoring process, a difference in consensus achieved by assessing trials according to the ESMO-MCBS, compared to trials assessed ac-cording to the NHB-ASCO-F. With the ESMO-MCBS for more trials consensus was achieved. A possible reason for this is the complex toxicity data reporting according to the NHB-ASCO-F. Potentially, a clearer definition of ‘clinical relevant toxicity’ could facilitate the scoring. With the ASCO framework, eight clinicians completed the tool for 11 anticancer drugs. A Cohen’s kappa coefficient of the interrater reliability (ICC) of 0.11 for NHB and 0.06 for toxicity was found, corresponding with ´slightly reliable agreement’ (18).

However in another study where convergent validity and interrater reliability of as-sessment frameworks were analyzed; the ASCO framework had an ICC of 0.80 (23). In another report, 109 RCTs were included and ESMO scores and scores using the concept and revised version of the ASCO framework were determined. Weak to moder-ate correlations were demonstrmoder-ated, suggesting different constructs of clinical benefit measured (24). Other tools, used to define the value and also addressing the costs of drugs, include the National Comprehensive Cancer Network evidence blocks, Institute for Clinical and Economic Review value assessment framework and Memorial Sloan Kettering Drug Abacus (25-27). Every tool has its own aspects. In the future converging

(27)

Chapter 2

26

of tools is expected which will allow to combine the best aspects of these tools to guide policy makers and patient-doctor discussions (28).

Conclusion

The ESMO-MCBS and the NHB of ASCO revised framework could be applied, respec-tively, to only 6 and 9 trials investigating systemic treatments in NET patients. The cur-rently used systemic treatments for GEP-NET patients had low scores according to the NHB-ASCO-F and none could be graded as meaningful clinical beneficial according to the ESMO-MCBS. Despite the low incidence of NET, the heterogeneous patient popula-tion and the relatively long natural course, future studies on new treatment modalities should aim for high clinical benefit outcomes.

(28)

2

27

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

References

1. Mocellin S, Nitti D. Gastrointestinal carcinoid: epidemiological and survival evidence from a large population-based study (n = 25 531). Ann Oncol 2013;24:3040-4.

2. Yao JC, Hassan M, Phan A, et al. One hundred years after “carcinoid”: epidemiology of and prognostic factors for neuroendocrine tumors in 35,825 cases in the United States. J Clin Oncol 2008;26:3063-72.

3. Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic enteropancreatic neuroen-docrine tumors. N Engl J Med 2014;371:224-33.

4. Meyer T, Qian W, Caplin ME, et al. Capecitabine and streptozocin +/- cisplatin in advanced gastroenteropancreatic neuroendocrine tumours. Eur J Cancer 2014;50:902-11.

5. Pavel M, Oberg KE, Hainsworth JD, et al. Everolimus plus octreotide long-acting release (LAR) for the treatment of advanced neuroendocrine tumors (NET) associated with carci-noid syndrome (RADIANT-2): Updated overall survival results. Eur J Cancer 2013;49:S577. 6. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic

neuroendocrine tumors. N Engl J Med 2011;364:501-13.

7. Rinke A, Wittenberg M, Schade-Brittinger C, et al. Placebo-controlled, double-blind, pro-spective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors (PROMID): Results of long-term survival. Neuroendocrinology 2017;104:26-32.

8. Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of 177Lu-dotatate for midgut neuro-endocrine tumors. N Engl J Med 2017;376:125-35.

9. Yao JC, Pavel M, Lombard-Bohas C, et al. Everolimus for the Treatment of Advanced Pancreatic Neuroendocrine Tumors: Overall Survival and Circulating Biomarkers From the Randomized, Phase III RADIANT-3 Study. J Clin Oncol 2016;34:3906-13.

10. Yao JC, Fazio N, Singh S, 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;387:968-77.

11. Pavel ME, Singh S, Strosberg JR, 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;18:1411-22.

12. Yao JC, Guthrie KA, Moran C, et al. Phase III Prospective Randomized Comparison Trial of Depot Octreotide Plus Interferon Alfa-2b Versus Depot Octreotide Plus Bevacizumab in Patients With Advanced Carcinoid Tumors: SWOG S0518. J Clin Oncol 2017;35:1695-703. 13. Kulke MH, Siu LL, Tepper JE, et al. Future directions in the treatment of neuroendocrine

tumors: consensus report of the National Cancer Institute Neuroendocrine Tumor clinical trials planning meeting. J Clin Oncol 2011;29:934-43.

14. Cherny NI, Sullivan R, Dafni U, et al. A standardised, generic, validated approach to stratify the magnitude of clinical benefit that can be anticipated from anti-cancer therapies: the Eu-ropean Society for Medical Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann Oncol 2015;26:1547-73.

15. Schnipper LE, Davidson NE, Wollins DS, et al. American Society of Clinical Oncology Statement: A conceptual framework to assess the value of cancer treatment options. J Clin Oncol 2015;33:2563-77.

(29)

Chapter 2

28

16. Schnipper LE, Davidson NE, Wollins DS, et al. Updating the American Society of Clinical Oncology Value Framework: Revisions and reflections in response to comments received. J Clin Oncol 2016;34:2925-34.

17. Kiesewetter B, Raderer M, Steger GG, et al. The European Society for Medical Oncology Magnitude of Clinical Benefit Scale in daily practice: a single institution, real-life experience at the Medical University of Vienna. ESMO Open 2016;1:e000066.

18. Wilson L, Lin T, Wang L, et al. Evaluation of the ASCO Value Framework for Anticancer Drugs at an Academic Medical Center. J Manag Care Spec Pharm 2017;23:163-9. 19. Ramage J, Punia P, Faluyi O, et al. UK phase IV, observational study to assess quality of

life in patients (pts) with pancreatic neuroendocrine tumours (pNETS) receiving treatment with everolimus: The “real-world” OBLIQUE study. Ann Oncol 2016;27.

20. Booth CM, Eisenhauer EA. Progression-free survival: meaningful or simply measurable? J Clin Oncol 2012;30:1030-3.

21. Moodie EE, Karran JC, Shortreed SM. A case study of SMART attributes: a qualitative as-sessment of generalizability, retention rate, and trial quality. Trials 2016;17:242,016-1368-3. 22. van den Bent MJ, Brandes AA, Taphoorn MJ, et al. Adjuvant procarbazine, lomustine, and

vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951. J Clin Oncol 2013;31:344-50. 23. Bentley TGK, Cohen JT, Elkin EB, et al. Measuring the value of new drugs: Validity and

reliability of 4 value assessment frameworks in the oncology setting. J Manag Care Spec Pharm 2017;23:S34-48.

24. Cheng S, McDonald EJ, Cheung MC, et al. Do the American Society of Clinical Oncol-ogy Value Framework and the European Society of Medical OncolOncol-ogy Magnitude of Clinical Benefit Scale Measure the Same Construct of Clinical Benefit? J Clin Oncol 2017;:JCO2016716894.

25. NCCN Evidence Blocks. National Comprehensive Cancer Network. (at https://www.nccn. org/evidenceblocks/).

26. Drug Abacus. Memorial Sloan Kettering Cancer Center. (2016, at http://www.drugabacus. org/).

27. The ICER approach. Institute for Clinicial and Economic Review. (2017, at https://icer-review.org/methodology/icers-methods/).

28. Schnipper LE, Schilsky RL. Converging on the Value of Value Frameworks. J Clin Oncol 2017;35:2732-2734.

(30)

2

29

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Supplementary Table S1: Eligibility criteria for assessment with ESMO-MCBS and NHB of the revised ASCO framework

ESMO-MCBS NHB part of revised ASCO framework

Design Comparative outcome studies: (one of these) • Randomized design

• Comparative cohort-design • Systematic review

New treatment compared with a prevailing standard of careA in a

well-designed, well-conducted prospective randomized trialB.

Outcome Relative benefit of treatments; (one of these, if given as primary endpoint)

• Survival • QoL

• Surrogate outcomes for survival or QoL, • Treatment toxicities

Benefit: (one of these) • If available -> OS

• If OS data is not available ->PFS • If OS and PFS data are not

available ->RR

Toxicity Toxicity data

(Can up- or down grade the final score) Toxicity data graded in accordance with WHO criteria in grade 1,2 and grade 3,4. (Can up- or down grade the final score)

Disease Solid cancers Advanced disease or potentially curable clinical presentationsB in

medical oncology settingA. Same clinical

question evaluated in more studies

Results derived from well-powered registration

trial should be given priority. Only the highest quality evidence availableA.

Pre-planned

subgroups These can be scored with a maximum of three subgroups, scoring should be done separately when for more than one subgroup statistically significant results are reported.

NA

Post-hoc subgroup

analysis These are not graded, except for studies that incorporate collection of tissue samples to enable re-stratification based on new genetic or other biomarkers.

NA

In addition

(Only if data are available)

QoL

(Can up- or down grade the final score) Bonus: • Tail of the curve • Palliation • QoL

• Treatment free intervalB

(Can upgrade the final score) ASCO= American Society of Clinical Oncology, ESMO-MCBS= European Society of Medical Oncology- Magnitude of Clinical Benefit Scale, NA= not applicable, NHB= net health benefit, OS= overall survival, PFS=progression free survival, QoL=quality of life, RR=response rate

A) In conceptual framework B) In revised framework

(31)

Chapter 2

30

2

31

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Supplementary T

able S2: Clinical benefit of systemic treatment for GEP-NET according to ESMO-MCBS (ESMO-MCBS) and according

ASCO revised framework (NHB)

Intervention vs control

ESMO-MCBS

ASCO revised framework

Setting Primary Outcome HR of primary outcome/ RR Intoxication data Improvement QOL (Y es/No) ESMO- MCBS Clinical benefit score

Intoxication

data

Bonus points

NHB

References

Everolimus versus placebo

Previous SSA

use

before participating in RADIANT_2 trial

PFS NS NS NA NA 0 NC 0 NA Anthony (1)

Everolimus versus placebo

SSA-naive before participating in RADIANT_2 trial

PFS NS NS NA NA 0 NC 0 NA Anthony (1)

Octreotide vs no or other treatment

Intestinal neuro- endocrine tumor

PFS NS NA NA NA 0 NA 0 NA Arauyo (2)

Octreotide and interferon vs octreotide

Progressive. metastatic GEP-NET OS (and TTF) NS NS No NA 0 NC 0 NA Arnold (3)

Lanreotide autogel vs lanreotide microparticels

Sporadic NET

.

new diagnosis or progression

RR NS NS NA NA 0 NC 0 NA Bajetta (4)

Streptozocin and 5FU vs interferon

Unresectable malignant carcinoid tumor PFS NS NS No NA 0 NC 0 NA Dahan (5)

Streptozocin and 5FU vs doxorubicin

Unresectable.

progressive metastatic

carcinoid.

OS and response rate

NS NS NA NA 0 NC 0 NA Engstrom (6) Interferon vs lanreotide Progressive.

metastatic. treatment- naive GEP

NET RR NS NS NA NA 0 NC 0 NA Faiss (7)

Lanreotide vs interferon and lanreotide

Progressive.

metastatic. treatment- naive GEP

NET RR NS NS NA NA 0 NC 10 NA Faiss (7) Octreotide vs placebo Gastro-intestinal NET

Number of symptom episodes and 5-HIAA

NA NA Yes. in 2/5 domains of P AIS 3 0 NA 10 NA Jacobsen (8)

(32)

Chapter 2

30

2

31

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Supplementary T

able S2: Clinical benefit of systemic treatment for GEP-NET according to ESMO-MCBS (ESMO-MCBS) and according

ASCO revised framework (NHB)

(continued

)

Intervention vs control

ESMO-MCBS

ASCO revised framework

Setting Primary Outcome HR of primary outcome/ RR Intoxication data Improvement QOL (Y es/No) ESMO- MCBS Clinical benefit score

Intoxication

data

Bonus points

NHB

References

Interferon alfa. streptozocin and doxorubicin vs interferon

Carcinoid tumor RR NS NS NA NA 0 NC 0 NA Janson (9)

Octreotide and interferon vs octreotide

Carcinoid tumor of midgut with liver

metastases Risk of tumor progression 0.28 (0.16-0.45) NS NA NA NC NA 0 NA Kölby (10) B

Telotristat Ethyl and SSA

vs placebo and

SSA

Metastatic NET

with

carcinoid syndrome and ≥ 4 BM a day BM freq reduction 44 vs 20% NS NA NA NA NA 10 NA Kulke (1 1)

Telotristat Ethyl and SSA

vs placebo and

SSA

Metastatic NET

with

carcinoid syndrome and ≥ 4 BM a day BM freq reduction 42 vs 20% NS NA NA NA NA 10 NA Kulke (1 1)

Chemotherapy before everolimus or placebo

Chemo-naive

participants of the RADIANT

-3 trial PFS 0.32 (0.21-0.48) NA NA 3 44 -20 0 24 Lombard- Bohas (12)

Chemotherapy before everolimus or placebo

Participants with prior chemotherapy of the RADIANT

-3 trial PFS 0.45 (0.29-0.70) NA NA 3 54.4 -20 16 50.4 Lombard- Bohas (12)

Streptozocin and Cyclophosphamide vs Streptozocin and 5FU

Unresecatble metastastic carcinoid tumor RR 26% vs 33 % No NA NA 0 0 0 0 Moertel ’79 (13)

Streptozocin and 5FU vs streptozocin

Advanced islet cell

carcinoma RR 63 vs 36% No NA 2 44 NC 0 NA Moertel ’80 (14)

Chlorozotocin vs streptozocin and FU vs Streptozocin and doxorubicin

Unresectable or metastatic islet cell carcinoma.

RR. but also OS

is given

1.4yr vs 2.2 yr Strep/FU vs Strep/dox

NS NA 4 151.9 NC 16 NA Moertel ’92 (15) C

(33)

Chapter 2

32

2

33

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Supplementary T

able S2: Clinical benefit of systemic treatment for GEP-NET according to ESMO-MCBS (ESMO-MCBS) and according

ASCO revised framework (NHB)

(continued

)

Intervention vs control

ESMO-MCBS

ASCO revised framework

Setting Primary Outcome HR of primary outcome/ RR Intoxication data Improvement QOL (Y es/No) ESMO- MCBS Clinical benefit score

Intoxication

data

Bonus points

NHB

References

Interferon vs Streptozocin and 5FU

Carcinoid tumor RR or stable disease 0 vs 20 % NS NA 2 14 NC 0 NA Oberg ’89 (16)

Streptozocin and 5FU vs streptozocin and 5FU followed by interferon

Carcinoid tumor with liver metastases.

RR. but also OS is given 8 vs72 m p<0.001 NS NA 4 NC NC 20 NA Oberg ‘91 (17) D

Streptozocin and 5FU vs first-line interferon

Carcinoid tumor with liver metastases.

RR. but also OS is given 8 vs 56 m NS NA 4 NC NA 20 NA Oberg ‘91 (17) D Lanreotide vs octreotide

Carcinoid tumor with carcinoid syndrome.

QoL disappearance of flushes NS NS No NA 0 NA 0 NA O’ T oole (18)

Octreotide LAR vs no or other treatment

Metastasized NET

patients of ≥ 65 years registered in the SEER database

OS 0.61 (0.47- 0.79) NA NA 4 39 NA 0 NA Shen (19)

Octreotide LAR vs no treatment with octreotide LAR

NET

patients ≥ 65

year registered in the SEER database. with loco-regional disease.

OS NS NA NA NA 0 NA 0 NA Shen (19)

Octreotide LAR with previous SSA vs Octreotide LAR without previous SSA

Foregut. midgut or hindgut NET

-patients

included in the

placebo arm of the RADIANT

-2 trial. OS 33.5 m vs 50.6 m NA NA NA NA NA NA NA Strosberg (20) E

Streptozocin /5FU vs doxorubicin/5FU followed by DTIC if PD

Unresectable. advanced carcinoid tumors RR or OS 15.7m vs 24.3m OS p=0.0267 NS NA 4 54.8 NC 0 NA Sun (21)

(34)

Chapter 2

32

2

33

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Supplementary T

able S2: Clinical benefit of systemic treatment for GEP-NET according to ESMO-MCBS (ESMO-MCBS) and according

ASCO revised framework (NHB)

(continued

)

Intervention vs control

ESMO-MCBS

ASCO revised framework

Setting Primary Outcome HR of primary outcome/ RR Intoxication data Improvement QOL (Y es/No) ESMO- MCBS Clinical benefit score

Intoxication data Bonus points NHB References Y-DOT AT OC vs Y-DOT AT OC and LU-DOT AT OC Progressive. metastatic neuroendocrine tumor . OS 0.64 (0.47- 0.88) NS NA 4 36 NC 0 NA Villard (22) Lanreotide vs placebo NET patients with carcinoid syndrome

Symptom control (days of octreotide)

NS NS NS NA NA NA 0 NA Vinik (23)

Pasireotide LAR vs octreotide LAR

Carcinoid tumor of digestive tract with inadequately controlled diarrhea and/or flushing. Symptom control NS NS NA 3 43.2 NC 0 NA W olin (24)

Octreotide and bevacizumab vs PEG-interferon and Octreotide

Metastatic carcinoid tumor Response (and PFS) NS NS NA NA 0 NC 0 NA Yao ’08 (25)

Everolimus with octreotide LAR vs everolimus without octreotide LAR

Advanced pancreatic NET with progressive disease after chemotherapy

RR NA NS NA NA 0 NC 0 NA Yao ’10 (26) F ASCO= American Society of Clinical Oncology . BM= bowel movements. BM freq reduction= reponse. defined as a bm frequency reduction ≥ 30% from baseline for ≥ 50% of the treatment period. ESMO-MCBS= Europea n Society of Medical Oncology- Magnitud e of Clinical Benefit Scale. GEP-NET=gastrointestinal and/ or pancreatic neuro-end ocrine tumor . GI=gastro-intestinal. HR= hazard ratio. NET=neuroendocrine tumor . vs=versus. LAR=long acting reagents. NA= not applicable NC. no consensus. ND= no improvement or no im -portant difference. NS=no significant difference. OS=overall survival. PFS=progression free survival. QoL=quality of life. RR=response rate. SSA=somatostatin analogue vs= versus. A First author

. References are reported in the supplementary reference list.

B Although the risk of tumor progression is significant different. this endpoint can not be used in the ESMO-MCBS. nor in the ASCO Framework. Some centers used this endpoint as

being a HR of PFS. or used instead of this endpoint the response rate (although no p-value or significance level was given with the response rate). C Although

the time to tumor progression is significant different this endpoint can not be used in the ESMO-MCBS and ASCO Framework. In ASCO framework overall survival is

not permitted because cross over

.

D Patients were not randomized. but assigned to a treatment.

This probably have led to bias.

E Patients were divided based on the treatment the participant had before inclusion in the RADIANT

-2 trial. No statistical analysis is performed between the two groups.

(35)

Chapter 2

34

Supplementary Figure S1. Inclusion process for the literature analysis 1942 articles identified by database search:-1442 Embase -500 PubMed 1676 articles 266 Duplicates excluded

1453 ‘Clearly not relevant’ articles excluded based on title and abstract

39 articles remaining for analysis including 35 trials 223 articles

189 articles excluded based on:

The clinical question of 43 articles, was published later in a better article or with a better trial design. In 83 articles the main subject was not the comparison of intervention

8 Articles showed only a multivariate analysis 1 Article was not only about NET

20 Articles were post-hoc analyses, without comparison of intervention

21 Articles did not have a cohort design, a randomized trial design, or a design of a meta-analysis.

7 Articles only showed baseline results or the planned study

1 Article used an outcome that was not eligible for scoring (i.e. blood flow in liver metastases) 5 Only abstract available

Nog precies natellen

5 articles with publication date after literature search included later,

4 Phase III trials,

1 post-hoc analysis of predefined quality of life data

(36)

2

35

Clinical benefit of systemic treatment in patients with advanced pancreatic and gastro intestinal neuroendocrine tumors according to ESMO-MCBS and ASCO framework

Figure 2. Eligibility criteria for a valid evaluation with the tools

Refe ren ce Bo th th oo ls Bo th to ols Bo th to ols ASCO ASCO ASCO ES M O-M CB S ES M O-M CB S

A B C D E F G H Eligibility criteria that should

Caplin (1) be met for a valid score:

Meyer (2) Eligibility criteria are

Pavel (3,4) not met.

Raymond (5) Eligibility criteria are

Rinke (6,7) partly met.

Strosberg (8) All eligibility criteria

Yao’11, Yao‘16 (9,10) are met.

Yao’16 (11) Yao’17 (12) Wolin (34) Faiss (19) Arnold (15) Dahan (17) Moertel ’92 (26) Jacobsen (20) Janson (21) Bajetta (16) Kölby (22) Moertel ’79 (24) Sun (32) Moertel ’80 (25) Oberg ’89 (27) Yao ’10 (35) O’ Toole (29) Shen (30) Villard (33) Lombard-Bohas (23) Arauyo (14) Anthony (13) Strosberg (31) Oberg ‘91 (28) Engstrom (18)

A; More than 50% of participants has another NET than a gastro-intestinal or pancreatic NET B; Sample size of pre-planned power analysis not reached

C; Same clinical question evaluated in more studies D; Toxicity data are not available and graded E; Outcome data available

F; No prospective, randomized design

G; No randomized, comparative cohort design, or meta-analysis H; No toxicity data available

Referenties

GERELATEERDE DOCUMENTEN

Patients were randomized 1:1 to the control group, receiving standard care, or the intervention group, which received standard care with additional access to WINS. Randomization

This is the first prospective feasibility study examining whether a personalized dietary intervention with supplementation of deficient vitamins is feasible in SSA-using patients

background activity with PET scans and investigated if tumor lesions with substantially higher [ 18 F]FDOPA uptake could be identified in patients with SI-NET.... Materials

In patients with metastatic urothelial carcinoma, lack of response to immune checkpoint blockade is associated with increased TGF-β signalling in fibroblasts in the

Potentially, in patients with carcinoid syndrome [ 18 F]FDOPA PET scans can be used to select lesions with substantially higher [ 18 F]FDOPA uptake for local treatment like

Daarnaast hebben we gekeken of we op 18Fluorine-L-dihydroxyphenylalanine (18F-DOPA) PET scans van patiënten met een NET laesies met hoge opname konden worden geïdentificeerd,

Lieve vriendinnen, lieve Marlies, hartelijk dank voor alle keren dat je heerlijk voor me hebt gekookt, een rondje mee ging hardlopen en de kopjes (of bierpul met) thee waarbij

Bij patiënten met gemetastaseerde neuro-endocriene tumoren van de dunne darm is er heterogene 18 F-DOPA tracer opname tussen tumor laesies binnen een patiënt. Met 18 F-DOPA