Skip to main content
Top
Published in: BMC Cancer 1/2019

Open Access 01-12-2019 | Neuroendocrine Tumor | Research article

Tumor growth rate as a metric of progression, response, and prognosis in pancreatic and intestinal neuroendocrine tumors

Authors: Clarisse Dromain, Marianne E. Pavel, Philippe Ruszniewski, Alison Langley, Christine Massien, Eric Baudin, Martyn E. Caplin, on behalf of the CLARINET Study Group

Published in: BMC Cancer | Issue 1/2019

Login to get access

Abstract

Background

Lanreotide depot/autogel antitumor activity in intestinal/pancreatic neuroendocrine tumors (NETs) was demonstrated in the phase-3 CLARINET study (NCT00353496), based on significantly prolonged progression-free survival (PFS) versus placebo.

Methods

During CLARINET, patients with metastatic intestinal/pancreatic NETs received lanreotide depot/autogel 120 mg or placebo every 4 weeks for 96 weeks. Imaging data (response evaluation criteria in solid tumors [RECIST] v1.0, centrally reviewed) were re-evaluated in this post hoc analysis of tumor growth rate (TGR) in NETs. TGR (%/month) was calculated from two imaging scans during relevant periods: pre-treatment (TGR0); 12–24 weeks before randomization versus baseline; each treatment visit versus baseline (TGRTx-0); between consecutive treatment visits (TGRTx-Tx). To assess TGR as a measure of prognosis, PFS was compared for TGR0 subgroups stratified by optimum TGR0 cut-off; a multivariate analysis was conducted to identify prognostic factors for PFS.

Results

TGR0 revealed tumors growing during pre-treatment (median [interquartile range] TGR0: lanreotide 2.1%/month [0.2; 6.1]; placebo 2.7%/month [0.15; 6.8]), contrary to RECIST status. TGR was significantly reduced by 12 weeks with lanreotide versus placebo (difference in least-square mean TGR0–12 of − 2.9 [− 5.1, − 0.8], p = 0.008), a difference that was maintained at most subsequent visits. TGR0 > 4%/month had greater risk of progression/death than ≤4%/month (hazard ratio 4.1; [95% CI 2.5–6.5]; p < 0.001); multivariate analysis revealed lanreotide treatment, progression at baseline, TGR0, hepatic tumor load, and primary tumor type were independently associated with PFS.

Conclusions

TGR provides valuable information on tumor activity and prognosis in patients with metastatic intestinal/pancreatic NETs, and identifies early lanreotide depot/autogel antitumor activity.

Trial registration

Retrospective registration, 18 July 2006; EudraCT: 2005–004904-35; ClinicalTrials.gov: NCT00353496.
Appendix
Available only for authorised users
Literature
1.
go back to reference Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92(3):205–16.PubMedCrossRef Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, Verweij J, Van Glabbeke M, van Oosterom AT, Christian MC, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000;92(3):205–16.PubMedCrossRef
2.
go back to reference Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.PubMedCrossRef Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, Dancey J, Arbuck S, Gwyther S, Mooney M, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–47.PubMedCrossRef
3.
go back to reference Sharma MR, Maitland ML, Ratain MJ. RECIST: no longer the sharpest tool in the oncology clinical trials toolbox---point. Cancer Res. 2012;72(20):5145–9 discussion 5150.PubMedCrossRef Sharma MR, Maitland ML, Ratain MJ. RECIST: no longer the sharpest tool in the oncology clinical trials toolbox---point. Cancer Res. 2012;72(20):5145–9 discussion 5150.PubMedCrossRef
4.
go back to reference Abramson RG, McGhee CR, Lakomkin N, Arteaga CL. Pitfalls in RECIST data extraction for clinical trials: Beyond the Basics. Acad Radiol. 2015;22(6):779–86.PubMedPubMedCentralCrossRef Abramson RG, McGhee CR, Lakomkin N, Arteaga CL. Pitfalls in RECIST data extraction for clinical trials: Beyond the Basics. Acad Radiol. 2015;22(6):779–86.PubMedPubMedCentralCrossRef
5.
go back to reference Sundin A, Rockall A. Therapeutic monitoring of gastroenteropancreatic neuroendocrine tumors: the challenges ahead. Neuroendocrinology. 2012;96(4):261–71.PubMedCrossRef Sundin A, Rockall A. Therapeutic monitoring of gastroenteropancreatic neuroendocrine tumors: the challenges ahead. Neuroendocrinology. 2012;96(4):261–71.PubMedCrossRef
6.
go back to reference Kappeler C, Reichardt P, Kang Y-K, Blay J-Y, Joensuu H, Schaefer K, Chung J, Wagner A, Casali PG, Demetri G. O-016 exploratory analysis of tumor growth rate in patients with advanced gastrointestinal stromal tumors (GIST) treated with regorafenib in the GRID phase 3 trial. Ann Oncol. 2015;26(suppl 4):iv114.CrossRef Kappeler C, Reichardt P, Kang Y-K, Blay J-Y, Joensuu H, Schaefer K, Chung J, Wagner A, Casali PG, Demetri G. O-016 exploratory analysis of tumor growth rate in patients with advanced gastrointestinal stromal tumors (GIST) treated with regorafenib in the GRID phase 3 trial. Ann Oncol. 2015;26(suppl 4):iv114.CrossRef
7.
go back to reference Kappeler C, Healy DP, Baumer C, Meinhardt G, Elisei R, Schlumberger M, Brose MS. Analysis of tumor growth rate for radioiodine (RAI)-refractory differentiated thyroid cancer patients receiving placebo and/or sorafenib in the phase III DECISION study. J Clin Oncol. 2015;33(suppl):abst 6015. Kappeler C, Healy DP, Baumer C, Meinhardt G, Elisei R, Schlumberger M, Brose MS. Analysis of tumor growth rate for radioiodine (RAI)-refractory differentiated thyroid cancer patients receiving placebo and/or sorafenib in the phase III DECISION study. J Clin Oncol. 2015;33(suppl):abst 6015.
8.
go back to reference Madeira I, Terris B, Voss M, Denys A, Sauvanet A, Flejou JF, Vilgrain V, Belghiti J, Bernades P, Ruszniewski P. Prognostic factors in patients with endocrine tumours of the duodenopancreatic area. Gut. 1998;43(3):422–7.PubMedPubMedCentralCrossRef Madeira I, Terris B, Voss M, Denys A, Sauvanet A, Flejou JF, Vilgrain V, Belghiti J, Bernades P, Ruszniewski P. Prognostic factors in patients with endocrine tumours of the duodenopancreatic area. Gut. 1998;43(3):422–7.PubMedPubMedCentralCrossRef
9.
go back to reference Palazzo M, Lombard-Bohas C, Cadiot G, Matysiak-Budnik T, Rebours V, Vullierme MP, Couvelard A, Hentic O, Ruszniewski P. Ki67 proliferation index, hepatic tumor load, and pretreatment tumor growth predict the antitumoral efficacy of lanreotide in patients with malignant digestive neuroendocrine tumors. Eur J Gastroenterol Hepatol. 2013;25(2):232–8.PubMedCrossRef Palazzo M, Lombard-Bohas C, Cadiot G, Matysiak-Budnik T, Rebours V, Vullierme MP, Couvelard A, Hentic O, Ruszniewski P. Ki67 proliferation index, hepatic tumor load, and pretreatment tumor growth predict the antitumoral efficacy of lanreotide in patients with malignant digestive neuroendocrine tumors. Eur J Gastroenterol Hepatol. 2013;25(2):232–8.PubMedCrossRef
10.
go back to reference Durante C, Boukheris H, Dromain C, Duvillard P, Leboulleux S, Elias D, de Baere T, Malka D, Lumbroso J, Guigay J, et al. Prognostic factors influencing survival from metastatic (stage IV) gastroenteropancreatic well-differentiated endocrine carcinoma. Endocr Relat Cancer. 2009;16(2):585–97.PubMedCrossRef Durante C, Boukheris H, Dromain C, Duvillard P, Leboulleux S, Elias D, de Baere T, Malka D, Lumbroso J, Guigay J, et al. Prognostic factors influencing survival from metastatic (stage IV) gastroenteropancreatic well-differentiated endocrine carcinoma. Endocr Relat Cancer. 2009;16(2):585–97.PubMedCrossRef
11.
go back to reference Ferte C, Fernandez M, Hollebecque A, Koscielny S, Levy A, Massard C, Balheda R, Bot B, Gomez-Roca C, Dromain C, et al. Tumor growth rate is an early indicator of antitumor drug activity in phase I clinical trials. Clin Cancer Res. 2014;20(1):246–52.PubMedCrossRef Ferte C, Fernandez M, Hollebecque A, Koscielny S, Levy A, Massard C, Balheda R, Bot B, Gomez-Roca C, Dromain C, et al. Tumor growth rate is an early indicator of antitumor drug activity in phase I clinical trials. Clin Cancer Res. 2014;20(1):246–52.PubMedCrossRef
12.
go back to reference Ferte C, Koscielny S, Albiges L, Rocher L, Soria JC, Iacovelli R, Loriot Y, Fizazi K, Escudier B. Tumor growth rate provides useful information to evaluate sorafenib and everolimus treatment in metastatic renal cell carcinoma patients: an integrated analysis of the TARGET and RECORD phase 3 trial data. Eur Urol. 2014;65(4):713–20.PubMedCrossRef Ferte C, Koscielny S, Albiges L, Rocher L, Soria JC, Iacovelli R, Loriot Y, Fizazi K, Escudier B. Tumor growth rate provides useful information to evaluate sorafenib and everolimus treatment in metastatic renal cell carcinoma patients: an integrated analysis of the TARGET and RECORD phase 3 trial data. Eur Urol. 2014;65(4):713–20.PubMedCrossRef
13.
go back to reference Ito T, Honma Y, Hijioka S, Kudo A, Fukutomi A, Nozaki A, Kimura Y, Motoi F, Isayama H, Komoto I, et al. Phase II study of lanreotide autogel in Japanese patients with unresectable or metastatic well-differentiated neuroendocrine tumors. Investig New Drugs. 2017;35(4):499–508.CrossRef Ito T, Honma Y, Hijioka S, Kudo A, Fukutomi A, Nozaki A, Kimura Y, Motoi F, Isayama H, Komoto I, et al. Phase II study of lanreotide autogel in Japanese patients with unresectable or metastatic well-differentiated neuroendocrine tumors. Investig New Drugs. 2017;35(4):499–508.CrossRef
14.
go back to reference Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–33.PubMedCrossRef Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371(3):224–33.PubMedCrossRef
15.
go back to reference Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al. Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocr Relat Cancer. 2016;23(3):191–9.PubMedPubMedCentralCrossRef Caplin ME, Pavel M, Cwikla JB, Phan AT, Raderer M, Sedlackova E, Cadiot G, Wolin EM, Capdevila J, Wall L, et al. Anti-tumour effects of lanreotide for pancreatic and intestinal neuroendocrine tumours: the CLARINET open-label extension study. Endocr Relat Cancer. 2016;23(3):191–9.PubMedPubMedCentralCrossRef
16.
go back to reference Gomez-Roca C, Koscielny S, Ribrag V, Dromain C, Marzouk I, Bidault F, Bahleda R, Ferte C, Massard C, Soria JC. Tumour growth rates and RECIST criteria in early drug development. Eur J Cancer. 2011;47(17):2512–6.PubMedCrossRef Gomez-Roca C, Koscielny S, Ribrag V, Dromain C, Marzouk I, Bidault F, Bahleda R, Ferte C, Massard C, Soria JC. Tumour growth rates and RECIST criteria in early drug development. Eur J Cancer. 2011;47(17):2512–6.PubMedCrossRef
17.
go back to reference Stein WD, Gulley JL, Schlom J, Madan RA, Dahut W, Figg WD, Ning YM, Arlen PM, Price D, Bates SE, et al. Tumor regression and growth rates determined in five intramural NCI prostate cancer trials: the growth rate constant as an indicator of therapeutic efficacy. Clin Cancer Res. 2011;17(4):907–17.PubMedCrossRef Stein WD, Gulley JL, Schlom J, Madan RA, Dahut W, Figg WD, Ning YM, Arlen PM, Price D, Bates SE, et al. Tumor regression and growth rates determined in five intramural NCI prostate cancer trials: the growth rate constant as an indicator of therapeutic efficacy. Clin Cancer Res. 2011;17(4):907–17.PubMedCrossRef
18.
go back to reference Stein WD, Wilkerson J, Kim ST, Huang X, Motzer RJ, Fojo AT, Bates SE. Analyzing the pivotal trial that compared sunitinib and IFN-alpha in renal cell carcinoma, using a method that assesses tumor regression and growth. Clin Cancer Res. 2012;18(8):2374–81.PubMedPubMedCentralCrossRef Stein WD, Wilkerson J, Kim ST, Huang X, Motzer RJ, Fojo AT, Bates SE. Analyzing the pivotal trial that compared sunitinib and IFN-alpha in renal cell carcinoma, using a method that assesses tumor regression and growth. Clin Cancer Res. 2012;18(8):2374–81.PubMedPubMedCentralCrossRef
19.
go back to reference Le Tourneau C, Servois V, Dieras V, Ollivier L, Tresca P, Paoletti X. Tumour growth kinetics assessment: added value to RECIST in cancer patients treated with molecularly targeted agents. Br J Cancer. 2012;106(5):854–7.PubMedPubMedCentralCrossRef Le Tourneau C, Servois V, Dieras V, Ollivier L, Tresca P, Paoletti X. Tumour growth kinetics assessment: added value to RECIST in cancer patients treated with molecularly targeted agents. Br J Cancer. 2012;106(5):854–7.PubMedPubMedCentralCrossRef
20.
go back to reference Liu Y, Litiere S, de Vries EG, Sargent D, Shankar L, Bogaerts J, Seymour L. The role of response evaluation criteria in solid tumour in anticancer treatment evaluation: results of a survey in the oncology community. Eur J Cancer. 2014;50(2):260–6.PubMedCrossRef Liu Y, Litiere S, de Vries EG, Sargent D, Shankar L, Bogaerts J, Seymour L. The role of response evaluation criteria in solid tumour in anticancer treatment evaluation: results of a survey in the oncology community. Eur J Cancer. 2014;50(2):260–6.PubMedCrossRef
21.
go back to reference Neal ML, Trister AD, Cloke T, Sodt R, Ahn S, Baldock AL, Bridge CA, Lai A, Cloughesy TF, Mrugala MM, et al. Discriminating survival outcomes in patients with glioblastoma using a simulation-based, patient-specific response metric. PLoS One. 2013;8(1):e51951.PubMedPubMedCentralCrossRef Neal ML, Trister AD, Cloke T, Sodt R, Ahn S, Baldock AL, Bridge CA, Lai A, Cloughesy TF, Mrugala MM, et al. Discriminating survival outcomes in patients with glioblastoma using a simulation-based, patient-specific response metric. PLoS One. 2013;8(1):e51951.PubMedPubMedCentralCrossRef
22.
go back to reference Neal ML, Trister AD, Ahn S, Baldock A, Bridge CA, Guyman L, Lange J, Sodt R, Cloke T, Lai A, et al. Response classification based on a minimal model of glioblastoma growth is prognostic for clinical outcomes and distinguishes progression from pseudoprogression. Cancer Res. 2013;73(10):2976–86.PubMedPubMedCentralCrossRef Neal ML, Trister AD, Ahn S, Baldock A, Bridge CA, Guyman L, Lange J, Sodt R, Cloke T, Lai A, et al. Response classification based on a minimal model of glioblastoma growth is prognostic for clinical outcomes and distinguishes progression from pseudoprogression. Cancer Res. 2013;73(10):2976–86.PubMedPubMedCentralCrossRef
23.
go back to reference Iacovelli R, Massari F, Albiges L, Loriot Y, Massard C, Fizazi K, Escudier B. Evidence and clinical relevance of tumor flare in patients who discontinue tyrosine kinase inhibitors for treatment of metastatic renal cell carcinoma. Eur Urol. 2015;68(1):154–60.PubMedCrossRef Iacovelli R, Massari F, Albiges L, Loriot Y, Massard C, Fizazi K, Escudier B. Evidence and clinical relevance of tumor flare in patients who discontinue tyrosine kinase inhibitors for treatment of metastatic renal cell carcinoma. Eur Urol. 2015;68(1):154–60.PubMedCrossRef
24.
go back to reference Barnes J, Johnson SJ, French JJ. Correlation of Ki-67 indices from biopsy and resection specimens of neuroendocrine tumours. Ann R Coll Surg Engl. 2017;99(3):193–7.PubMedCrossRef Barnes J, Johnson SJ, French JJ. Correlation of Ki-67 indices from biopsy and resection specimens of neuroendocrine tumours. Ann R Coll Surg Engl. 2017;99(3):193–7.PubMedCrossRef
25.
go back to reference Kloppel G, La Rosa S. Ki67 labeling index: assessment and prognostic role in gastroenteropancreatic neuroendocrine neoplasms. Virchows Arch. 2018;472(3):341–9.PubMedCrossRef Kloppel G, La Rosa S. Ki67 labeling index: assessment and prognostic role in gastroenteropancreatic neuroendocrine neoplasms. Virchows Arch. 2018;472(3):341–9.PubMedCrossRef
26.
go back to reference Richards-Taylor S, Ewings SM, Jaynes E, Tilley C, Ellis SG, Armstrong T, Pearce N, Cave J. The assessment of Ki-67 as a prognostic marker in neuroendocrine tumours: a systematic review and meta-analysis. J Clin Pathol. 2016;69(7):612–8.PubMedCrossRef Richards-Taylor S, Ewings SM, Jaynes E, Tilley C, Ellis SG, Armstrong T, Pearce N, Cave J. The assessment of Ki-67 as a prognostic marker in neuroendocrine tumours: a systematic review and meta-analysis. J Clin Pathol. 2016;69(7):612–8.PubMedCrossRef
27.
go back to reference Wolin EM, Caplin ME, Pavel ME, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, Cadiot G, Capdevila J, Wall L, et al. Multivariate analysis of progression free survival in the CLARINET study of lanreotide autogel/depot vs placebo identifies prognostic factors in neuroendocrine tumors. Pancreas. 2016;45:486. Wolin EM, Caplin ME, Pavel ME, Ćwikła JB, Phan AT, Raderer M, Sedláčková E, Cadiot G, Capdevila J, Wall L, et al. Multivariate analysis of progression free survival in the CLARINET study of lanreotide autogel/depot vs placebo identifies prognostic factors in neuroendocrine tumors. Pancreas. 2016;45:486.
Metadata
Title
Tumor growth rate as a metric of progression, response, and prognosis in pancreatic and intestinal neuroendocrine tumors
Authors
Clarisse Dromain
Marianne E. Pavel
Philippe Ruszniewski
Alison Langley
Christine Massien
Eric Baudin
Martyn E. Caplin
on behalf of the CLARINET Study Group
Publication date
01-12-2019
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2019
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-5257-x

Other articles of this Issue 1/2019

BMC Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine