Impact of liver tumour burden, alkaline phosphatase elevation, and target lesion size on treatment outcomes with 177Lu-Dotatate: an analysis of the NETTER-1 study

Jonathan Strosberg, Pamela L Kunz, Andrew Hendifar, James Yao, David Bushnell, Matthew H Kulke, Richard P Baum, Martyn Caplin, Philippe Ruszniewski, Ebrahim Delpassand, Timothy Hobday, Chris Verslype, Al Benson, Rajaventhan Srirajaskanthan, Marianne Pavel, Jaume Mora, Jordan Berlin, Enrique Grande, Nicholas Reed, Ettore Seregni, Giovanni Paganelli, Stefano Severi, Michael Morse, David C Metz, Catherine Ansquer, Frédéric Courbon, Adil Al-Nahhas, Eric Baudin, Francesco Giammarile, David Taïeb, Erik Mittra, Edward Wolin, Thomas M O'Dorisio, Rachida Lebtahi, Christophe M Deroose, Chiara M Grana, Lisa Bodei, Kjell Öberg, Berna Degirmenci Polack, Beilei He, Maurizio F Mariani, Germo Gericke, Paola Santoro, Jack L Erion, Laura Ravasi, Eric Krenning, NETTER-1 study group, Jonathan Strosberg, Pamela L Kunz, Andrew Hendifar, James Yao, David Bushnell, Matthew H Kulke, Richard P Baum, Martyn Caplin, Philippe Ruszniewski, Ebrahim Delpassand, Timothy Hobday, Chris Verslype, Al Benson, Rajaventhan Srirajaskanthan, Marianne Pavel, Jaume Mora, Jordan Berlin, Enrique Grande, Nicholas Reed, Ettore Seregni, Giovanni Paganelli, Stefano Severi, Michael Morse, David C Metz, Catherine Ansquer, Frédéric Courbon, Adil Al-Nahhas, Eric Baudin, Francesco Giammarile, David Taïeb, Erik Mittra, Edward Wolin, Thomas M O'Dorisio, Rachida Lebtahi, Christophe M Deroose, Chiara M Grana, Lisa Bodei, Kjell Öberg, Berna Degirmenci Polack, Beilei He, Maurizio F Mariani, Germo Gericke, Paola Santoro, Jack L Erion, Laura Ravasi, Eric Krenning, NETTER-1 study group

Abstract

Purpose: To assess the impact of baseline liver tumour burden, alkaline phosphatase (ALP) elevation, and target lesion size on treatment outcomes with 177Lu-Dotatate.

Methods: In the phase 3 NETTER-1 trial, patients with advanced, progressive midgut neuroendocrine tumours (NET) were randomised to 177Lu-Dotatate (every 8 weeks, four cycles) plus octreotide long-acting release (LAR) or to octreotide LAR 60 mg. Primary endpoint was progression-free survival (PFS). Analyses of PFS by baseline factors, including liver tumour burden, ALP elevation, and target lesion size, were performed using Kaplan-Meier estimates; hazard ratios (HRs) with corresponding 95% CIs were estimated using Cox regression.

Results: Significantly prolonged median PFS occurred with 177Lu-Dotatate versus octreotide LAR 60 mg in patients with low (< 25%), moderate (25-50%), and high (> 50%) liver tumour burden (HR 0.187, 0.216, 0.145), and normal or elevated ALP (HR 0.153, 0.177), and in the presence or absence of a large target lesion (diameter > 30 mm; HR, 0.213, 0.063). Within the 177Lu-Dotatate arm, no significant difference in PFS was observed amongst patients with low/moderate/high liver tumour burden (P = 0.7225) or with normal/elevated baseline ALP (P = 0.3532), but absence of a large target lesion was associated with improved PFS (P = 0.0222). Grade 3 and 4 liver function abnormalities were rare and did not appear to be associated with high baseline liver tumour burden.

Conclusions: 177Lu-Dotatate demonstrated significant prolongation in PFS versus high-dose octreotide LAR in patients with advanced, progressive midgut NET, regardless of baseline liver tumour burden, elevated ALP, or the presence of a large target lesion. Clinicaltrials.gov : NCT01578239, EudraCT: 2011-005049-11.

Keywords: 177Lu-Dotatate; Liver tumour burden; NETTER-1; Neuroendocrine tumour; Octreotide.

Conflict of interest statement

J. Strosberg reports fees for consulting or advisory roles with Novartis; participation in speakers’ bureaus with Ipsen and Lexicon; and research funding from Merck and Novartis.

P. L. Kunz reports fees for consulting or advisory roles with Advanced Accelerator Applications, Ipsen, Lexicon, and Novartis; research funding from Advanced Accelerator Applications, Ipsen, Lexicon, Xencor, and Brahams; and is a stockholder with Guardant Health.

A. Hendifar reports fees for consulting or advisory roles with Novartis and Ipsen; and research funding from Halo, Ipsen, Novartis, Merck, Xencor, AbbVie.

J. Yao reports fees for consulting or advisory roles with Novartis, Ipsen, Hutchison Medi Pharma, and Tarveda.

D. Bushnell reports honoraria from Novartis, Advanced Accelerator Applications; consulting or advisory roles with Novartis, Advanced Accelerator Applications; and research funding from Novartis, Advanced Accelerator Applications.

M. H. Kulke reports fees for consulting or advisory roles with Novartis, Lexicon, Ipsen, Tarveda; and research funding from Lexicon and Ipsen; and providing expert testimony on behalf of Novartis.

R. P. Baum reports fees for consulting or advisory roles with ITG; and is a stockholder with Advanced Accelerator Applications and Endocyte.

M. Caplin reports honoraria from Advanced Accelerator Applications, Novartis, Ipsen, and Pfizer; consulting or advisory roles with Advanced Accelerator Applications, Novartis, Ipsen, and Pfizer; participation in speakers’ bureaus with Advanced Accelerator Applications, Novartis, Ipsen, and Pfizer; research funding from Advanced Accelerator Applications and Ipsen; and travel, accommodations, or expenses from Advanced Accelerator Applications and Ipsen.

P. Ruszniewski reports honoraria from Ipsen, Novartis, Advanced Accelerator Applications, ITN, and Keocyt; fees for consulting or advisory roles with Ipsen, Novartis, and Advanced Accelerator Applications; travel, accommodations, or expenses from Ipsen; and research funding from Novartis; and fees for providing expert testimony on behalf of Advanced Accelerator Applications.

E. Delpassand reports honoraria from Advanced Accelerator Applications and Endocyte; fees for consulting or advisory roles with Endocyte; participation in speakers’ bureaus with Advanced Accelerator Applications; patents, royalties or other intellectual property with Radiomedix, Inc.; travel, accommodations, or expenses from Endocyte, Advanced Accelerator Applications ITG/ITM GmbH; and is a stockholder with Radiomedix, Inc., Excel Diagnostics, Westchase Imaging, Endocyte, and GE.

C. Verslype reports fees for consulting or advisory roles with Ipsen, Novartis, Bayer, Sirtex; participation in speakers’ bureaus with Bayer; and research funding from Ipsen and Bayer;

A. Benson reports fees for consulting or advisory roles with Bristol-Myers Squibb, Guardant Health, Eli Lilly & Company, Exelixis, Purdue Pharma, inVentive Health Inc., Axio, Genentech, Bayer, Merck, Rafael Pharmaceuticals, Astellas, Terumo, Taiho, Thera Bionic, LSK, Axio, and Incyte Corporation; and research funding from Acerta, Celegene, Advanced Accelerator Applications, Novartis, Infinity Pharmaceuticals, Merck Sharp and Dohme, Taiho, Bristol-Myers Squibb, MedImmune/AstraZeneca, Xencor, PreECOG, Astellas, Amgen, and ECOG-ACRIN.

R. Srirajaskanthan reports honoraria from Novartis, Ipsen, and Mylan; fees for participation in speakers’ bureaus with Mylan; and travel, accommodations, or expenses from Ipsen.

M. Pavel reports honoraria from Novartis, Ipsen, Pfizer, and Lexicon; fees for consulting or advisory roles with Novartis, Ipsen, Pfizer, and Lexicon; and research funding from Novartis, Ipsen, Pfizer, and Lexicon.

J. Berlin reports fees for consulting or advisory roles with Rafael, Celgene, Taiho, FivePrime, EMD Serono, Arno, Gritstone, Erytech, Astra Zeneca, Eisai, LSK Pharmaceuticals; Bayer, Seattle Genetics; research funding from Novartis (Array), AbbVie, Immunomedics, Taiho, Genentech/Roche, Bayer, Lilly, Incyte, Pharmacyclics, FivePrime, Loxo, EMD Serono, Bayer, Boston Biomedical, PsiOxus, Macrogenics, Boston Biomedical, Symphogen; fees for participation in speakers’ bureaus with Nestle; travel, accommodations, or expenses from NCI; and DSMB from Astrazeneca.

E. Grande reports receiving honoraria for speaking and expert testimony for Pfizer, Ipsen, BMS, Eisai, Roche, MSD, Sanofi-Genzyme, Adacap, Novartis, EUSA Pharma, Pierre Fabre, and Lexicon; expert testimony for Celgene; research funding from Astra Zeneca, Pfizer, Ipsen, MTEM/Threshold, and Lexicon; medical educational grants from MSD and Roche; and has had leadership roles with ENETS, GETNE, and GETHI.

N. Reed reports fees for consulting or advisory roles with Novartis, Advanced Accelerator Applications, Ipsen, and Eisai; and participation in speakers’ bureaus with Novartis, Advanced Accelerator Applications, Ipsen, and Eisai.

S. Severi reports travel, accommodations, or expenses from Novartis.

M. Morse reports honoraria from Genetech, Bayer, Exelixis, Eisai, Lexicon, Novartis, Advanced Accelerator Applications, and Taiho; fees for participation in speakers’ bureaus with Genetech, Bayer, Exelixis, Eisai, Lexicon, Novartis, Advanced Accelerator Applications, and Taiho; and research funding from BMS, Medimmune/AstraZeneca, and Eisai; and has held a patent with Duke University for targeting HER3.

D. C. Metz reports honoraria from Advanced Accelerator Applications; fees for consulting or advisory roles with Takeda and Lexicon; research funding from Lexicon, Wren Laboratories, and Advanced Accelerator Applications; providing expert testimony on behalf of Mylan; research funding from Lexicon, Wren Laboratories, and Advanced Accelerator Applications; and has held a patent or has intellectual property interests with Capital Academics for a GI board review syllabus.

C. Ansquer reports honoraria from Ipsen, Novartis, and Advanced Accelerator Applications; fees for consulting or advisory roles with Ipsen, Novartis, and Advanced Accelerator Applications; and travel, accommodations, or expenses from Novartis, Advanced Accelerator Applications, and Eisai.

F. Courbon reports honoraria from Novartis, Bayer, GEHC, Ipsen, and Norgine; fees for consulting or advisory roles with Novartis, Bayer, Ipsen, Advanced Accelerator Applications, and Norgine; participation in speakers’ bureaus with Novartis, Bayer, GEHC, Ipsen, Norgine, and Advanced Accelerator Applications; expert testimony on behalf of Novartis, Bayer, GEHC, Ipsen, Norgine, and Advanced Accelerator Applications; research funding from GEHC, Curium, and Advanced Accelerator Applications; and travel, accommodations, or expenses with Novartis, Bayer, GEHC, Ipsen, Norgine, and Advanced Accelerator Applications.

E. Baudin reports honoraria from Advanced Accelerator Applications; fees for consulting or advisory roles with Advanced Accelerator Applications; and research funding from Advanced Accelerator Applications.

E. Mittra reports honoraria from Advanced Accelerator Applications/Novartis; fees for consulting or advisory roles with Novartis, Curium, and Ipsen; and research funding from Endocyte/Novartis.

E. Wolin reports fees for consulting or advisory roles with Advanced Accelerator Applications, Lexicon, and Ipsen.

R. Lebtahi reports honoraria from Advanced Accelerator Applications; fees for consulting or advisory roles with Advanced Accelerator Applications; and travel, accommodations, or expenses with Advanced Accelerator Applications.

C. M. Deroose reports fees for consulting or advisory roles with Ipsen, Novartis, Terumo, and Advanced Accelerator Applications; participation in speakers’ bureaus with Terumo and Advanced Accelerator Applications; and travel, accommodations, or expenses with General Electric and Terumo.

C. M. Grana reports fees for consulting or advisory roles with Norgine and Ipsen; and travel, accommodations, or expenses with Iason, Ipsen - IBA.

L. Bodei reports honoraria from Advanced Accelerator Applications and Ipsen; fees for consulting or advisory roles with Advanced Accelerator Applications and Ipsen; participation in speakers’ bureaus with Advanced Accelerator Applications and Ipsen; and travel, accommodations, or expenses from Advanced Accelerator Applications.

K. Öberg reports fees for consulting or advisory roles with Advanced Accelerator Applications.

B. Degirmenci Polack is an employee of, has had leadership roles with, and is a stockholder with Advanced Accelerator Applications.

B. He is an employee of Advanced Accelerator Applications, a Novartis company, and is a stockholder with Novartis.

M. F. Mariani reports honoraria from Norgine, Italy, and GE Healthcare, Italy.

G. Gericke reports travel, accommodations, or expenses from Novartis AG, CH; is a stockholder with Novartis AG, CH; has held patents, royalties, or other intellectual property from Novartis AG, CH.

P. Santoro is an employee of and a stockholder with Advanced Accelerator Applications.

J. L. Erion reports travel, accommodations, or expenses from Advanced Accelerator Applications; and is an employee of, has held leadership roles at, has held patents, royalties, or other intellectual property from, and is a stockholder with Advanced Accelerator Applications, Inc.

L. Ravasi is an employee of and a stockholder with Advanced Accelerator Applications.

E. Krenning reports travel, accommodations, or expenses from Advanced Accelerator Applications; and has held patents, royalties, or other intellectual property from, and is a stockholder with Advanced Accelerator Applications.

T. Hobday, E. Seregni, A. Al-Nahhas, F. Giammarile, J. Mora, G. Paganelli, D. Taïeb, and T. M. O’Dorisio have no disclosures to report.

Figures

Fig. 1
Fig. 1
Kaplan-Meier analysis of progression-free survival by treatment arm (patients randomised to four cycles of peptide receptor radionuclide therapy with 177Lu-Dotatate + octreotide LAR 30 mg or octreotide LAR 60 mg) and baseline extent of liver tumour burden (low [< 25%], moderate [25–50%], or high [> 50%]). Liver tumour burden is calculated according to liver tumour volume divided by total liver volume by computed tomography or magnetic resonance imaging. Data cutoff: 24 July 2015. HRs with corresponding 95% CIs and P-values were estimated using a Cox regression model with randomised treatment, liver tumour burden at baseline, and liver tumour burden × randomised treatment interaction term as covariates. Log-rank test used for within-treatment arm comparisons of PFS. CI: confidence interval, HR: hazard ratio, LAR: long-acting release, NR: not reached, PFS: progression-free survival
Fig. 2
Fig. 2
Kaplan-Meier analysis of progression-free survival by treatment arm (patients randomised to four cycles of peptide receptor radionuclide therapy with 177Lu-Dotatate + octreotide LAR 30 mg or octreotide LAR 60 mg) and baseline normal (≤ ULN) or elevated (> ULN) alkaline phosphatase levels (based on institutional ULN). Data cutoff: 24 July 2015. One-hundred twelve patients in either treatment arm had evaluable baseline ALP levels and were included in this analysis. HRs with corresponding 95% CIs and P-values were estimated using a Cox regression model with randomised treatment, alkaline phosphatase level, and alkaline phosphatase level × randomised treatment interaction term as covariates. Log-rank test was used for within-treatment arm comparisons of PFS. ALP: alkaline phosphatase, CI: confidence interval, HR: hazard ratio, LAR: long-acting release, NR: not reached, PFS: progression-free survival, ULN: upper limit of normal
Fig. 3
Fig. 3
Kaplan-Meier analysis of progression-free survival by treatment arm (patients randomised to four cycles of peptide receptor radionuclide therapy with 177Lu-Dotatate + octreotide LAR 30 mg or octreotide LAR 60 mg) and presence or absence of at least one large (> 30 mm diameter) target lesion at any site of the body at baseline imaging with computed tomography or magnetic resonance imaging. Data cutoff: 24 July 2015. HRs with corresponding 95% CIs and P-values were estimated using a Cox regression model with randomised treatment, presence/absence of large target lesion, and presence/absence of large target lesion × randomised treatment interaction term as covariates. Log-rank test was used for within–treatment arm comparisons of PFS. CI: confidence interval, HR: hazard ratio, LAR: long-acting release, NR: not reached, PFS: progression-free survival
Fig. 4
Fig.  4
Least squares mean percentage change from baseline in the size of liver lesions at each study visit in the 177Lu-Dotatate arm, stratified by baseline liver lesion size. Data cutoff: 30 June 2016. A lesion-based mixed model repeated measures analysis included study visit, baseline target liver lesion size (≤ 30 mm or > 30 mm), and baseline target liver lesion size × study visit interaction as fixed effects

References

    1. Riihimaki M, Hemminki A, Sundquist K, et al. The epidemiology of metastases in neuroendocrine tumors. Int J Cancer. 2016;139:2679–2686. doi: 10.1002/ijc.30400.
    1. Rinke A, Müller HH, Schade-Brittinger C, 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. doi: 10.1200/JCO.2009.22.8510.
    1. Rinke A, Wittenberg M, Schade-Brittinger C, 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 (PROMID): results on long term survival. Neuroendocrinology. 2017;104:26–32. doi: 10.1159/000443612.
    1. Yalchin M, Oliveira A, Theocharidou E, et al. The impact of radiological response to peptide receptor radionuclide therapy on overall survival in patients with metastatic midgut neuroendocrine tumors. Clin Nucl Med. 2017;42:e135–e141. doi: 10.1097/RLU.0000000000001457.
    1. Ezziddin S, Khalaf F, Vanezi M, et al. Outcome of peptide receptor radionuclide therapy with 177Lu-octreotate in advanced grade 1/2 pancreatic neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2014;41:925–933. doi: 10.1007/s00259-013-2677-3.
    1. Ezziddin S, Attassi M, Yong-Hing CJ, et al. Predictors of long-term outcome in patients with well-differentiated gastroenteropancreatic neuroendocrine tumors after peptide receptor radionuclide therapy with 177Lu-octreotate. J Nucl Med. 2014;55:183–190. doi: 10.2967/jnumed.113.125336.
    1. Panzuto F, Merola E, Pavel ME, et al. Stage IV gastro-entero-pancreatic neuroendocrine neoplasms: a risk score to predict clinical outcome. Oncologist. 2017;22:409–415. doi: 10.1634/theoncologist.2016-0351.
    1. Foulfoin M, Graillot E, Adham M, et al. Treatment of metastatic pancreatic neuroendocrine tumors: relevance of ENETS 2016 guidelines. Endocr Relat Cancer. 2017;24:71–81. doi: 10.1530/ERC-16-0464.
    1. Brabander T, van der Zwan WA, Teunissen JJM et al. Long-Term Efficacy, Survival, and Safety of [(177)Lu-DOTA(0),Tyr(3)]octreotate in Patients with Gastroenteropancreatic and Bronchial Neuroendocrine Tumors. Clin Cancer Res 2017; 23: 4617–4624.
    1. Andriantsoa M, Hoibian S, Autret A, et al. An elevated serum alkaline phosphatase level in hepatic metastases of grade 1 and 2 gastrointestinal neuroendocrine tumors is unusual and of prognostic value. PLoS One. 2017;12:e0177971. doi: 10.1371/journal.pone.0177971.
    1. Fairweather M, Swanson R, Wang J, et al. Management of neuroendocrine tumor liver metastases: long-term outcomes and prognostic factors from a large prospective database. Ann Surg Oncol. 2017;24:2319–2325. doi: 10.1245/s10434-017-5839-x.
    1. Jimenez-Fonseca P, Krug S, Tamagno G, et al. Identifying prognostic factors for well-differentiated metastatic pancreatic neuroendocrine tumours (pNETs): a retrospective international multicenter cohort study. Neuroendocrinology. 2018. 10.1159/000492223[Epub ahead of print].
    1. Clancy TE, Sengupta TP, Paulus J, et al. Alkaline phosphatase predicts survival in patients with metastatic neuroendocrine tumors. Dig Dis Sci. 2006;51:877–884. doi: 10.1007/s10620-006-9345-4.
    1. Putzer D, Gabriel M, Henninger B, et al. Bone metastases in patients with neuroendocrine tumor: 68Ga-DOTA-Tyr3-octreotide PET in comparison to CT and bone scintigraphy. J Nucl Med. 2009;50:1214–1221. doi: 10.2967/jnumed.108.060236.
    1. Bodei L, Cremonesi M, Grana CM, et al. Yttrium-labelled peptides for therapy of NET. Eur J Nucl Med Mol Imaging. 2012;39(Suppl 1):S93–102. doi: 10.1007/s00259-011-2002-y.
    1. Bodei L, Mueller-Brand J, Baum RP, et al. The joint IAEA, EANM, and SNMMI practical guidance on peptide receptor radionuclide therapy (PRRNT) in neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2013;40:800–816. doi: 10.1007/s00259-012-2330-6.
    1. Hicks RJ, Kwekkeboom DJ, Krenning E, et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasia: peptide receptor radionuclide therapy with radiolabeled somatostatin analogues. Neuroendocrinology. 2017;105:295–309. doi: 10.1159/000475526.
    1. de Jong M, Breeman WA, Valkema R, et al. Combination radionuclide therapy using 177Lu- and 90Y-labeled somatostatin analogs. J Nucl Med. 2005;46(Suppl 1):13S–17S.
    1. Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of 177Lu-Ddotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376:125–135. doi: 10.1056/NEJMoa1607427.
    1. Strosberg J, Wolin E, Chasen B, 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. doi: 10.1200/JCO.2018.78.5865.
    1. Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371:224–233. doi: 10.1056/NEJMoa1316158.
    1. de Jong M, Breeman WA, Bernard BF, et al. Tumor response after [(90)Y-DOTA(0),Tyr(3)]octreotide radionuclide therapy in a transplantable rat tumor model is dependent on tumor size. J Nucl Med. 2001;42:1841–1846.

Source: PubMed

3
Abonner