esmo e-learning overview of management of well ...€¦ · recently, it has become possible to use...

59
OVERVIEW OF MANAGEMENT OF WELL-DIFFERENTIATED PANCREATIC NEUROENDOCRINE TUMOURS (PanNETs) Current State-of-the-Art and Future Steps Angela Lamarca The Christie NHS Foundation Trust; University of Manchester Manchester, United Kingdom

Upload: others

Post on 23-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • OVERVIEW OF MANAGEMENT OF WELL-DIFFERENTIATED PANCREATIC NEUROENDOCRINE TUMOURS (PanNETs)Current State-of-the-Art and Future Steps

    Angela Lamarca

    The Christie NHS Foundation Trust; University of Manchester

    Manchester, United Kingdom

  • INTRODUCTION TOPANCREATIC NETsEpidemiology

    Clinical presentation

    Grading

    Staging

  • EPIDEMIOLOGY

    NETs are rare malignancies but incidence is increasing

    Incidence of NETs is increasing

    ▪ Most dramatic rise in incidence in

    patients 65 years or older (8-fold rise to

    25.3 per 100,000 persons)

    Diagnosis of localised stages and

    grade 1 tumours is increasing:

    better diagnosis?

    Dasari A, et al. JAMA Oncol 2017;3(10):1335–42.

    p

  • EPIDEMIOLOGY

    PanNETs are rare (low incidence)

    Increase in incidence of NETs

    from 1973 to 2012 across all

    sites, stages, and grades

    ▪ 15-fold increase in the stomach

    ▪ 2-fold increase in the cecum

    PanNETs represent

  • EPIDEMIOLOGY

    Most PanNETs are sporadic

    Most PanNETs are sporadic (non-hereditary); risk factors could include diabetes, smoking, chronic pancreatitis

    Hereditary syndromes include:

    ◆ MEN1 (multiple endocrine neoplasia 1)

    ◆ VHL (von Hippel Lindau disease)

    ◆ NF1 (von Recklinghausen’s syndrome; neurofibromatosis 1)

    ◆ TS (tuberous sclerosis)

    Halfdanarson TR, et al. Pancreas 2014;43(8):1219–22; Capurso G, et al. Am J Gastroenterol 2009;104:2175–81; Falconi M, et al. Neuroendocrinology 2016;103(2):153–71

    Patients who develop PanNETs in the context of an hereditary syndrome are expected to be associated with

    a more indolent course; consider separately for management / prognosis

  • CLINICAL PRESENTATION

    Functioning vs. Non-Functioning

    Most (60–90%) of PanNETs are

    non-functioning

    ◆ 10–40% are expected to be

    functioning

    ◆ Specific GUT hormones may be

    assessed at diagnosis if clinical

    suspicion of specific syndrome

    (+CgA)

    Falconi M, et al. Neuroendocrinology 2016;103(2):153–71 CgA: Chromogranin A; WDHA, watery diarrhoea, hypokalaemia, achlorhydria

    Most frequent functioning PanNETs

    Name

    Biologically

    active

    peptide(s)

    secreted

    Incidence (new cases/106

    population/year) Tumour location

    Malignant,

    %

    Associated

    with

    MEN1, % Main symptoms/signs

    The most common F-P-NET syndromes

    Insulinoma Insulin 1–32 Pancreas (>99%) 100 cases)

    VIPoma (Verner-

    Morrison

    syndrome,

    pancreatic

    cholera, WDHA)

    Vasoactive

    intestinal

    peptide

    0.05–0.2 Pancreas (90%,

    adult)

    Other (10%,

    neural, adrenal,

    periganglionic)

    40–70 6 Diarrhoea (90–100%)

    Hypokalaemia (80–100%)

    Dehydration (83%)

    Glucagonoma Glucagon 0.01–0.1 Pancreas (100%) 50–80 1–20 Rash (67–90%)

    Glucose intolerance (38–87%)

    Weight loss (66–96%)

  • CLINICAL PRESENTATION

    Functioning vs. Non-Functioning

    Falconi M, et al. Neuroendocrinology 2016;

    103(2):153–71

    Less frequent functioning PanNETs

    Among functioning

    PanNETs, hormone

    secretion may

    drive treatment

    strategy and needs

    to be taken into

    account

    Name

    Biologically active

    peptide(s) secreted

    Incidence(new cases/106

    population/year) Tumour location Malignant, %

    Associated with

    MEN1, % Main symptoms/signs

    SSoma Somatostatin Rare Pancreas (55%)

    Duodenum/jejunum (44%)

    >70 45 Diabetes mellitus (63–90%)

    Cholelithiases (65–90%)

    Diarrhoea (35–90%)

    GRHoma Growth hormone-

    releasing hormone

    Unknown Pancreas (30%)

    Lung (54%)

    Jejunum (7%)

    Other (13%)

    >60 16 Acromegaly (100%)

    ACTHoma ACTH Rare Pancreas (4–16% all

    ectopic Cushing’s

    syndrome)

    >95 Rare Cushing’s syndrome (100%)

    P-NET causing carcinoid

    syndrome

    Serotonin

    ? Tachykinins

    Rare (43 cases) Pancreas (

  • GRADING

    WHO classification update 2017: G3-NET

    Classification relies on grade (Ki-67 / mitotic index) and tumour morphology

    ◆ NEN: neuroendocrine neoplasms (NET + NEC; regardless of morphology / grade)

    ◆ NET: neuroendocrine tumours (well-differentiated morphology)

    ◆ NEC: neuroendocrine carcinoma (poorly-differentiated morphology)

    Lloyd RV, et al. WHO Classification of Tumours of Neuroendocrine Organs 4th Ed 2017

    Classification / grade Ki-67 proliferation index (%) Mitotic index

    Well-differentiated PanNENs

    PanNET G1 20

    Poorly-differentiated PanNENs

    PanNEC G3 >20 >20

  • GRADING

    G3-NET vs. G3-NEC

    Coriat R, et al. The Oncologist 2016;21(19):1191–9;

    The Oncologist by Society for Translational Oncology. Reproduced with permission of JOHN WILEY & SONS - JOURNALS in the format Use in an e-coursepack via Copyright Clearance Center

    Differential immunolabeling and molecular alterations of pancreatic NET and NECs

  • GRADING: G3-NEC, A SEPARATE ENTITY

    Principles of management: chemotherapy is the cornerstone of treatment for G3-NEC

    ◆ Affects approximately 7% of patients with PanNENs

    ◆ Presents with locally advanced (20%) or metastatic (65%) disease

    ◆ Nordic NEC study:

    ◆ Patients with Ki67 ≥55% had greater response rate (42% vs. 15%, p

  • STAGING

    AJCC vs. ENETS

    classification

    Current classifications

    Very similar: except

    T4, M1 status

    Klöppel G, et al. Virchows Arch 2010;456(6):595–7; Rindi G, et al. J Natl Cancer Inst 2012;104(10):764–77;

    Amin M., Edge S., Greene F., Byrd D. R., Brookland R. K., Washington M. K., et al. 2017. AJCC cancer staging manual, 8th ed. Springer, New York, NY.

    ENETS TNM (2010) AJCC 8th Edition (2017)

    T Stage

    T1 Confined for pancreas 4cm, or invasion of

    duodenum or bile duct

    Tumour limited to pancreas, more than 4 cm in greatest

    dimension or tumour invading duodenum or bile duct

    T4 Invasion of adjacent organs or major vessels Tumour perforates visceral peritoneum (serosa) or invades

    other organs or adjacent structures

    N Stage

    NX Regional lymph nodes cannot be assessed Regional lymph nodes cannot be assessed

    N0 No regional lymph node metastasis No regional lymph node metastasis

    N1 Regional lymph node metastasis Regional lymph node metastasis

    M Stage

    M0 No distant metastasis No distant metastasis

    M1 Distant metastasis Distant metastasis

    M1a / M1b / M1c n/a Hepatic only / Extrahepatic only / Both

    TNM Stage groups

    Stage I T1, N0, M0 T1, N0, MO

    Stage IIa T2, N0, M0 T2, N0, M0

    Stage IIb T3, N0, MO T3, N0, MO

    Stage IIIa T4, N0, MO T4, N0, MO

    Stage IIIb Any T, N1, M0 Any T, N1, M0

    Stage IV Any T, Any N, M1 Any T, Any N, M1

    ENETS: European Neuroendocrine Tumour Society; AJCC: American Joint Committee on Cancer .

  • STAGING

    AJCC vs. ENETS classification

    Multiple versions of the AJCC

    TNM classification

    Klöppel G, et al. Virchows Arch 2010;456(6):595–7; Teo RYA, et al. Surgery 2019;165(4):672–85

    ENETS 2010 AJCC v.7 (2010)

    Retrospective analysis:

    ◆ 1072 PanNEN patients

    ◆ ENETS vs. AJCCv.7 (2010)

    ◆ ENETS classification seemed

    superior?

    Rindi G, et al. TNM staging of neoplasms of the endocrine pancreas: results from a large international cohort study, J Natl Cancer Inst. 2012;104(10):764–77, by permission of Oxford University Press.

  • STAGING

    AJCC vs. ENETS classification

    Cross-sectional imaging +/- Somatostatin receptor scintigraphy (SRS; OctreoScan®)

    Recently, it has become possible to use somatostatin receptor PET/CT (i.e. 68Ga-PET)

    instead, which might improve diagnostic quality:

    ◆ Systematic review and metanalysis (22 studies)

    ◆ DTA of 68Ga-PET in NETs: Se 93%, Sp 90%

    Exception: insulinomas (Se 25%)

    ◆ Glucagon-Like Peptide-1 Receptor and 68Ga-NOTA-Exendin-4 PET/CT may have a role

    (currently on development)

    Falconi M, et al. Neuroendocrinology 2016;103(2):153–71; Sharma P, et al. Q J Nucl Med Mol Imaging 2016;60(1):69–76

    68Ga-PET is the method of choice to fully stage disease in patients

    with PanNETs; expected change of management (surgical,

    medical, staging) in 20–55% of patients

    Reprinted by permission from Springer Nature: Geijer Eur J Nucl Med Mol Imaging, Somatostatin receptor PET/CT in neuroendocrine tumours: update on systematic review and meta-analysis, Geijer H, et al. Copyright 2013;

  • LOCALISED STAGE: MANAGEMENTSurgical management

    Adjuvant treatment

    Risk stratification

    Post-surgical follow-up

  • SURGICAL MANAGEMENT

    Localised disease; 2 cm size cut-off

    In selected patients with

    tumours 2 cm – surgery

    Surveillance depending on final

    pathology

    Tumour >2 cm

    Surgeryb

    Limited resection only if conditions

    favourable to preserve organ

    function (otherwise oncological

    resection)

    See section on treatment for

    advanced disease

  • ADJUVANT TREATMENT

    There is NO evidence to support adjuvant treatment

    Clinical trials not performed in this setting, mainly due to lack of definition of “population at risk of relapse”

    ◆ Overall, relapse rate is low in the completely resected population

    ◆ Predicting clinical behaviour in PanNETs has been difficult due to lack of data

    Stratification for risk of relapse is crucial for the development of adjuvant strategies

    Falconi M, et al. Neuroendocrinology 2016;103(2):153–71

  • RISK STRATIFICATION

    Risk factors for relapse (PanNETs)

    Gao H, et al. Cancer Lett. 2018;412:188–93; Sho S, et al. J Gastroint Surg 2018 Oct 23 [Epub ahead of print]; Genç CG, et al. Ann Surg 2018;267(6):1148–54; Ausania F, Pancreatology 2019;19(2):367–71; Marchegiani G, et

    al. Neuroendocrinology 2018 Nov 27 [Epub ahead of print]

    Relapse Rate Risk factors Site of recurrence

    Gao 2018Relapse rate 129/505 (25.5%).

    Median disease-free survival of 19 months (range 6–96 months) T3, T4, N1, Ki67 >2%, functional Not reported

    Sho 2018

    Relapse rate 23/140 (16.3%).

    5- and 10-year relapse-free survival were 84.6% and 67.1%,

    respectively

    Size >5 cm, N1, Ki67 >20%All recurrences were distant

    (liver, peritoneal and bone)

    Genç 2018

    Relapse rate 35/211 (17%).

    The 5- and 10-year disease-specific/overall survival was 98%/91%

    and 84%/68%, respectively.

    Median timeto recurrence was 43 months (IQR 23–62)

    Grade 2, N1, perineural invasion

    Pancreatic remnant (69%),

    distant (14%), 1 patient had

    lymph node metastasis

    Ausania 2019Relapse rate 19/137 (13.9%).

    Median DFS was 55 months

    Tumour size >2 cm, N1, Ki67>5%

    or mitotic index >2Not reported

    Marchegiani 2018

    Recurrence rate 12.3%.

    Recurrence occurred either during the first year of follow-up (n=9),

    or after ten years (n=4)

    >21 mm size, G3, N1,

    vascular infiltration

    Liver (11.1%), local recurrence

    (2.3%), lymph node (2.1%),

    other organs (1.6%)

  • RISK STRATIFICATION

    Risk factors for relapse (PanNETs)

    Gao H, et al. Cancer Lett. 2018;412:188–93; Sho S, et al. J Gastroint Surg 2018 Oct 23 [Epub ahead of print]; Genç CG, et al. Ann Surg 2018;267(6):1148–54; Ausania F, Pancreatology 2019;19(2):367–71; Marchegiani G, et

    al. Neuroendocrinology 2018 Nov 27 [Epub ahead of print]

    Relapse Rate Risk factors Site of recurrence

    Gao 2018Relapse rate 129/505 (25.5%).

    Median disease-free survival of 19 months (range 6–96 months)T3, T4, N1, Ki67 >2%, functional Not reported

    Sho 2018

    Relapse rate 23/140 (16.3%).

    5- and 10-year Relapse-free survival were 84.6% and 67.1%,

    respectively

    Size >5 cm, N1, Ki67 >20%All recurrences were distant

    (liver, peritoneal and bone)

    Genç 2018

    Relapse rate 35/211 (17%).

    The 5- and 10-year disease-specific/overall survival was 98%/91%

    and 84%/68%, respectively.

    Median time to recurrence was 43 months (IQR 23–62)

    Grade 2, N1, perineural invasion

    Pancreatic remnant (69%),

    distant (14%), 1 patients had

    lymph node metastasis

    Ausania 2019Relapse rate 19/137 (13.9%).

    Median DFS was 55 months

    Tumour size >2 cm, N1, Ki67>5%

    or mitotic index >2Not reported

    Marchegiani 2018

    Relapse rate (12.3%).

    Recurrence occurred either during the first year of follow-up (n=9),

    or after ten years (n= 4)

    >21 mm size, G3, N1,

    vascular infiltration

    Liver (11.1%), local recurrence

    (2.3%), lymph node (2.1%),

    other organs (1.6%)

    Lesson 1: Relapse rate 12–25%

  • RISK STRATIFICATION

    Risk factors for relapse (PanNETs)

    Gao H, et al. Cancer Lett. 2018;412:188–93; Sho S, et al. J Gastroint Surg 2018 Oct 23 [Epub ahead of print]; Genç CG, et al. Ann Surg 2018;267(6):1148–54; Ausania F, Pancreatology 2019;19(2):367–71; Marchegiani G, et

    al. Neuroendocrinology 2018 Nov 27 [Epub ahead of print]

    Relapse Rate Risk factors Site of recurrence

    Gao 2018Relapse rate 129/505 (25.5%).

    Median disease-free survival of 19 months (range 6–96 months) T3, T4, N1, Ki67 >2%, functional Not reported

    Sho 2018

    Relapse rate 23/140 (16.3%).

    5- and 10-year Relapse-free survival were 84.6% and 67.1%,

    respectively

    Size >5 cm, N1, Ki67 >20%All recurrence were distant

    (liver, peritoneal and bone)

    Genç 2018

    Relapse rate 35/211 (17%).

    The 5- and 10-year disease-specific/overall survival was 98%/91%

    and 84%/68%, respectively.

    Median time to recurrence was 43 months (IQR 23–62)

    Grade 2, N1, perineural invasion

    Pancreatic remnant (69%),

    distant (14%), 1 patients had

    lymph node metastasis

    Ausania 2019Relapse rate 19/137 (13.9%).

    Median DFS was 55 months

    Tumour size >2 cm, N1, Ki67>5%

    or mitotic index >2 Not reported

    Marchegiani 2018

    Recurrence rate 12.3%.

    Recurrence occurred either during the first year of follow-up (n=9),

    or after ten years (n=4)

    >21 mm size, G3, N1,

    vascular infiltration

    Liver (11.1%), local recurrence

    (2.3%), lymph node (2.1%),

    other organs (1.6%)

    Lesson 1: Relapse rate 12–25%

    Lesson 2: Late relapses DO exist (>5/10 years)

  • RISK STRATIFICATION

    Risk factors for relapse (PanNETs)

    Gao H, et al. Cancer Lett. 2018;412:188–93; Sho S, et al. J Gastroint Surg 2018 Oct 23 [Epub ahead of print]; Genç CG, et al. Ann Surg 2018;267(6):1148–54; Ausania F, Pancreatology 2019;19(2):367–71; Marchegiani G, et

    al. Neuroendocrinology 2018 Nov 27 [Epub ahead of print]

    Relapse Rate Risk factors Site of recurrence

    Gao 2018Relapse rate 129/505 (25.5%).

    Median disease-free survival of 19 months (range 6–96 months). T3, T4, N1, Ki67 >2%, functional Not reported

    Sho 2018

    Relapse rate 23/140 (16.3%).

    5- and 10-year Relapse-free survival were 84.6% and 67.1%,

    respectively

    Size >5 cm, N1, Ki67 >20%All recurrences were distant

    (liver, peritoneal and bone)

    Genç 2018

    Relapse rate 35/211 (17%).

    The 5- and 10-year disease-specific/overall survival was 98%/91%

    and 84%/68%, respectively.

    Median time to recurrence was 43 months (IQR 23 – 62)

    Grade 2, N1, perineural invasion

    Pancreatic remnant (69%),

    distant (14%), 1 patients had

    lymph node metastasis

    Ausania 2019Relapse rate 19/137 (13.9%).

    Median DFS was 55 months

    Tumour size >2 cm, N1,

    Ki67>5% or mitotic index >2 Not reported

    Marchegiani 2018

    Recurrence rate 12.3%.

    Recurrence occurred either during the first year of follow-up (n= 9),

    or after ten years (n= 4)

    >21 mm size, G3, N1,

    vascular infiltration

    Liver (11.1%), local recurrence

    (2.3%), lymph node (2.1%),

    other organs (1.6%)

    Lesson 1: Relapse rate 12–25%

    Lesson 2: Late relapses DO exist (>5/10 years)

    Lesson 3: Size/T, N, Ki67/grade are repetitive prognostic factors

  • RISK STRATIFICATION

    Risk factors for relapse (PanNETs)

    Gao H, et al. Cancer Lett. 2018;412:188–93; Sho S, et al. J Gastroint Surg 2018 Oct 23 [Epub ahead of print]; Genç CG, et al. Ann Surg 2018;267(6):1148–54; Ausania F, Pancreatology 2019;19(2):367–71; Marchegiani G, et

    al. Neuroendocrinology 2018 Nov 27 [Epub ahead of print]

    Relapse Rate Risk factors Site of recurrence

    Gao 2018Relapse rate 129/505 (25.5%).

    Median disease-free survival of 19 months (range 6–96 months)T3, T4, N1, Ki67 >2%, functional Not reported

    Sho 2018

    Relapse rate 23/140 (16.3%).

    5- and 10-year Relapse-free survival were 84.6% and 67.1%,

    respectively

    Size >5 cm, N1, Ki67 >20%All recurrences were distant

    (liver, peritoneal and bone)

    Genç 2018

    Relapse rate 35/211 (17%).

    The 5- and 10-year disease-specific/overall survival was 98%/91%

    and 84%/68%, respectively.

    Median time to recurrence was 43 months (IQR 23–62)

    Grade 2, N1, perineural

    invasion

    Pancreatic remnant (69%),

    distant (14%), 1 patients had

    lymph node metastasis

    Ausania 2019Relapse rate 19/137 (13.9%).

    Median DFS was 55 months

    Tumour size >2 cm, N1, Ki67>5%

    or mitotic index >2 Not reported

    Marchegiani 2018

    Recurrence rate 12.3%.

    Recurrence occurred either during the first year of follow-up (n=9),

    or after ten years (n=4)

    >21 mm size, G3, N1,

    vascular infiltration

    Liver (11.1%), local recurrence

    (2.3%), lymph node (2.1%),

    other organs (1.6%)

    Lesson 1: Relapse rate 12–25%

    Lesson 2: Late relapses DO exist (>5/10 years)

    Lesson 3: Size/T, N, Ki67/grade are repetitive prognostic factors

    Lesson 4: Other factors may require further confirmation

  • RISK STRATIFICATION

    Risk factors for relapse (PanNETs)

    Gao H, et al. Cancer Lett. 2018;412:188–93; Sho S, et al. J Gastroint Surg 2018 Oct 23 [Epub ahead of print]; Genç CG, et al. Ann Surg 2018;267(6):1148–54; Ausania F, Pancreatology 2019;19(2):367–71; Marchegiani G, et

    al. Neuroendocrinology 2018 Nov 27 [Epub ahead of print]

    Relapse Rate Risk factors Site of recurrence

    Gao 2018Relapse rate 129/505 (25.5%).

    Median disease-free survival of 19 months (range 6–96 months) T3, T4, N1, Ki67 >2%, functional Not reported

    Sho 2018

    Relapse rate 23/140 (16.3%).

    5- and 10-year Relapse-free survival were 84.6% and 67.1%,

    respectively

    Size >5 cm, N1, Ki67 >20%All recurrences were distant

    (liver, peritoneal and bone)

    Genç 2018

    Relapse rate 35/211 (17%).

    The 5- and 10-year disease-specific/overall survival was 98%/91%

    and 84%/68%, respectively. Median time

    to recurrence was 43 months (IQR 23–62)

    Grade 2, N1, perineural invasion

    Pancreatic remnant (69%),

    distant (14%), 1 patients had

    lymph node metastasis

    Ausania 2019Relapse rate 19/137 (13.9%).

    Median DFS was 55 months.

    Tumour size >2 cm, N1, Ki67>5%

    or mitotic index >2 Not reported

    Marchegiani 2018

    Recurrence rate 12.3%.

    Recurrence occurred either during the first year of follow-up (n=9),

    or after ten years (n=4)

    >21 mm size, G3, N1,

    vascular infiltration

    Liver (11.1%), local recurrence

    (2.3%), lymph node (2.1%),

    other organs (1.6%)

    Lesson 1: Relapse rate 12–25%

    Lesson 2: Late relapses DO exist (>5/10 years)

    Lesson 3: Size/T, N, Ki67/grade are repetitive prognostic factors

    Lesson 4: Other factors may require further confirmation

    Lesson 5: Site of recurrence distal (liver) > local

  • RISK STRATIFICATION

    Towards development of adjuvant strategies for selected resected PanNETs

    Clinical trials not performed in this setting, mainly due to lack of definition of “population at risk of relapse”

    ◆ Overall, relapse rate is

  • POST-SURGICAL FOLLOW-UP

    Recommendations

    Frequency of review / radiology investigations should be adjusted to presence of risk factors of relapse

    T1N0, G1 and N0 insulinomas may require a less intensive follow-up

    NCCN v3.2018 guidelines

    3–12 months post-resection:

    ◆ Patient history and physical examination

    ◆ Biochemistry follow-up as clinically required based on previous findings (CgA vs. GUT hormones)

    ◆ Cross-sectional imaging (CT/MRI)

    ◆ OctreoScan®/68Ga-Pet (if not previously performed)

    After 1st year and until 10 years post-resection

    6–12 monthly: history/physical examination; biochemistry; radiological follow-up (cross-sectional imaging)

  • POST-SURGICAL FOLLOW-UP

    Current practice is variable between centres

    Data from a US/Canada survey:

    ◆ Clinicians aware of guidelines but there was

    still very significant variability between sites

    Chan DL, et al. Neuroendocrinology 2017;107(1):32–41

    The current guidelines are not widely adopted,

    potentially due to a lack of high-quality evidence to

    inform follow-up for this heterogeneous disease

    We should work towards improved

    standardisation of follow-up

    Follow-up for Resected Gastroenteropancreatic

    Neuroendocrine Tumours: A Practice Survey of the

    Commonwealth Neuroendocrine Tumour Collaboration

    (CommNETS) and the North American Neuroendocrine

    Tumour Society (NANETS)

  • ADVANCED STAGE: MANAGEMENTHow to select the most adequate treatment

    Liver-directed therapies

    Somatostatin analogues

    Targeted therapies

    Chemotherapy

    PRRT

  • HOW TO SELECT THE MOST ADEQUATE TREATMENT

    ENETS guidelines

    ◆ Surgery can have a role in

    metastatic disease

    ◆ Liver-directed therapy plays a role

    in selected patients

    ◆ Multiple systemic therapy options

    Importance of individualised

    treatment and planification of a

    treatment strategy by an

    experienced MDT

    Pavel M, et al. Neuroendocrinology 2016;103(2):172–85

    Resection of primary

    Two-step surgery

    1.Minor resection

    ± RFA, RPVE,

    RPVL

    2.Sequential

    major liver

    resection

    Resection (minor

    or anatomical)

    Surgery

    contraindicated

    Selected

    cases (

  • HOW TO SELECT THE MOST ADEQUATE TREATMENT

    ENETS guidelines

    ◆ Surgery can have a role in

    metastatic disease

    ◆ Liver-directed therapy plays a role

    in selected patients

    ◆ Multiple systemic therapy options

    Importance of individualised

    treatment and planification of a

    treatment strategy by an

    experienced MDT

    Resection of primary

    Two-step surgery

    1.Minor resection

    ± RFA, RPVE,

    RPVL

    2.Sequential

    major liver

    resection

    Resection (minor

    or anatomical)

    Surgery

    contraindicated

    Selected

    cases (

  • LIVER-DIRECTED THERAPIES

    A very significant proportion of patients diagnosed with

    PanNET will have liver metastases

    Reprinted from The Lancet Oncol 2014, 15(1), Frilling A, et al. Recommendations for management of patients with neuroendocrine liver metastases, e8-e21, Copyright (2014), with permission from Elsevier.

    Resection of liver metastases from PanNETs

    Resection of liver metastases (if resectable) seems to

    improve survival (no prospective randomised data available)

  • HOW TO SELECT THE MOST ADEQUATE TREATMENT

    ENETS guidelines

    ◆ Surgery can have a role in

    metastatic disease

    ◆ Liver-directed therapy plays a role

    in selected patients

    ◆ Multiple options of systemic therapy

    Importance of individualised

    treatment and planification of a

    treatment strategy by an

    experienced MDT

    Resection of primary

    Two-step surgery

    1.Minor resection

    ± RFA, RPVE,

    RPVL

    2.Sequential

    major liver

    resection

    Resection (minor

    or anatomical)

    Surgery

    contraindicated

    Selected

    cases (

  • LIVER-DIRECTED THERAPIES

    Overall survival worse than after resection

    (likely to reflect unresectable (more extensive

    disease)

    Main role in patients with functioning

    tumours and with liver-predominant

    disease

    Data for NETs (no specific data in PanNETs):

    • symptomatic response 53–100%

    • morphological response 35–74%

    • progression-free survival 18 months

    • 5-year survival of 40–83%

    Reprinted from The Lancet Oncol 2014, 15(1), Frilling A, et al. Recommendations for management of patients with neuroendocrine liver metastases , e8-e21, Copyright (2014), with permission from Elsevier.

    Liver embolisation (TAE, TACE) in PanNETs

  • HOW TO SELECT THE MOST ADEQUATE TREATMENT

    ENETS guidelines

    ◆ Surgery can have a role in

    metastatic disease

    ◆ Liver-directed therapy plays a role

    in selected patients

    ◆ Multiple options of systemic

    therapy

    Importance of individualised

    treatment and planification of a

    treatment strategy by an

    experienced MDT

    Resection of primary

    Two-step surgery

    1.Minor resection

    ± RFA, RPVE,

    RPVL

    2.Sequential

    major liver

    resection

    Resection (minor

    or anatomical)

    Surgery

    contraindicated

    Selected

    cases (

  • HOW TO SELECT THE MOST ADEQUATE (SYSTEMIC) TREATMENT

    Understanding the effect of different systemic treatments

    Chemotherapy (20–50%)

    Targeted (5-10%)

    SSA (0%)

    PRRT (18%)May be higher in

    PanNETs (58%)

    SSA: somatostatin analogue; PRRT: Peptide Receptor Radionuclide Therapy Adapted from Lamarca A, et al. J Oncopathol 2014;2(1):15–25;

    Caplin ME, et al. N Engl Med 2014;371: 224–33; Strosberg J, et al. N Engl J Med 2017;376:125–35; Ramage J, et al. Semin Oncol. 2018;45(4):236–48

  • Principles for treatment selection:

    1. Targeted therapies are effective in treatment-naïve as well as

    chemotherapy pre-treated patients

    2. Chemotherapy is associated with a higher response rate

    3. Treatment decision is based on the aims of therapy (disease

    response versus time to progression)

    4. Decision may depend on expected toxicities

    5. Concept of ''mitotically-active'' disease

    6. Patients usually live long enough to receive multiple therapies

    7. Need to identify sub-groups of patients (through research) who

    benefit most from each therapy

    8. One-size does not fit all (importance of MDTs)

    Adapted from Lamarca A, et al. J Oncopathol 2014;2(1):15–25

    Decision based on tumour burden, Ki-67 and rate of progression (tumour kinetics)

    HOW TO SELECT THE MOST ADEQUATE (SYSTEMIC) TREATMENT

    or SSA

    or PRRT

  • CAP, Capecitabine; TEM, temozolomide. *≤6-12 months. †Cisplatin can be replaced by carboplatin.Pavel M, et al. Neuroendocrinology 2016;103(2):172–85.

    Tailoring systemic treatment to patient/tumour characteristics

    HOW TO SELECT THE MOST ADEQUATE (SYSTEMIC) TREATMENT

    Therapeutic options and conditions for preferential use as first-line therapy in advanced NEN

    Drug Functionality Grading Primary site SSTR status Special considerations

    Octreotide +/- G1 Midgut + Low tumour burden

    Lanreotide +/- G1/G2 (-10%) Midgut, pancreas + Low and high (>25%) liver tumor burden

    IFN-α 2b +/- G1/G2 Midgut If SSTR negative

    STZ/5-FU +/- G1/G2 Pancreas Progressive in short-term* or high tumour burden or

    symptomatic

    TEM/CAP +/- G2 Pancreas Progressive in short-term* or high tumour burden or

    symptomatic; if STZ is contraindicated or not available

    Everolimus +/- G1/G2 Lung

    Pancreas

    Midgut

    Atypical carcinoid and/or SSTR negative

    Insulinoma or contraindication for CTX

    If SSTR negative

    Sunitinib +/- G1/G2 Pancreas Contraindication for CTX

    PRRT +/- G1/G2 Midgut + (required) Extended disease; extrahepatic disease, e.g. bone

    metastasis

    Cisplatin†/etoposide +/- G3 Any All poorly differentiated NEC

  • SOMATOSTATIN ANALOGUES

    Rinke A, et al. J Clin Oncol 2009;27(28):4656–63

    Octreotide (PROMID study)

    ◆ G1 metastatic or locally advanced well diff,

    functioning* or non-functioning midgut NETs

    (no PanNETs included)

    ◆ Randomisation 1:1 (n=85 patients)

    R

    Octreotide LAR 30 mg

    4-weekly

    Placebo

    Primary endpoint: Progression free survival

    *Only patients tolerating flushing without intervention or responding to treatment with loperamide or cholestyramine in case of

    diarrhoea were included

  • SOMATOSTATIN ANALOGUES

    Caplin ME, et al. N Engl J Med 2014;371: 224–33

    Lanreotide (CLARINET study)

    ◆ G1 or G2 (Ki-67

  • SOMATOSTATIN ANALOGUES

    Rinke A, et al. J Clin Oncol. 2009;27(28):4656–63; Caplin ME, et al. N Engl J Med 2014;371: 224–33.

    Octreotide vs. Lanreotide (role beyond anti-hormone function)

    Octreotide LAR

    30 mg, im 4-weekly

    (PROMID study; vs. placebo)

    Lanreotide Autogel

    120 mg; deep sc, 4-weekly

    (CLARINET study; vs. placebo)

    Population of patients

    Advanced, functional or non-functional midgut

    primary tumour or tumour of unknown.

    PanNETs were excluded.

    Advanced, non-functioning, somatostatin

    receptor-positive, grade 1 or 2 (Ki-67

  • TARGETED THERAPIES

    Two main pathways to target:

    ◆ mTOR (everolimus)

    ◆ Angiogenesis (sunitinib)

    Adapted from Lamarca A, et al. J Oncopathol 2014;2(1):15–25

    Potential pathways to target

    Molecular biology in pNETs

  • TARGETED THERAPIES

    Sunitinib

    ◆ G1 or G2 metastatic or locally advanced well

    diff, functioning or non-functioning PanNETs

    ◆ Progressed within previous 12 m

    ◆ Randomisation 1:1 (n=171 patients

    randomised*)

    R

    Sunitinib 37.5 mg PO OD

    Placebo

    Primary endpoint: Progression-free survival

    *Enrolment completed in the first interim analysis (therefor recruitment not fully completed)

    Raymond E, et al. N Engl J Med 2011;364:501–13

  • TARGETED THERAPIES

    Everolimus

    ◆ G1 or G2 metastatic or locally advanced well

    diff, functioning or non-functioning PanNETs

    ◆ Progressed within previous 12 m

    ◆ Randomisation 1:1 (n=410 patients)

    R

    Everolimus 10 mg PO OD

    Placebo

    Primary endpoint: Progression-free survival

    Yao JC, et al. N Engl J Med 2011; 364:514-523

  • TARGETED THERAPIES

    Sunitinib and Everolimus

    Sunitinib

    37.5 mg once daily

    (Phase 3 vs. placebo)

    Everolimus

    10 mg once daily

    (Phase 3 vs. placebo)

    Population of patientsUnresectable or metastatic, well- or moderately-

    differentiated PanNETs

    Unresectable or metastatic, well- or moderately-

    differentiated PanNETs

    Documented disease

    progression at study entryYes Yes

    Objective response rate 9.3% vs. 0% 5% vs. 2%

    Median PFS (experiment vs.

    placebo) (months)

    11.4 vs. 5.5

    HR 0.42 (95% CI 0.26, 0.66); p

  • TARGETED THERAPIES

    Sunitinib and Everolimus: Toxicity profile

    Sunitinib Everolimus

    Yao JC, et al. N Engl J Med 2011;364:514–23; Raymond E, et al. N Engl J Med 2011;364:501–13; Lombard-Bohas C, et al. Pancreas 2015;44(2):181–9.

  • TARGETED THERAPIES

    Sunitinib and Everolimus: Toxicity profile

    Sunitinib Everolimus

    Selection between Sunitinib or Everolimus usually relies on comorbidities due to different toxicity profile

    Yao JC, et al. N Engl J Med 2011;364:514–23; Raymond E, et al. N Engl J Med 2011;364:501–13; Lombard-Bohas C, et al. Pancreas 2015;44(2):181–9.

  • CHEMOTHERAPY

    Systematic review and meta-analysis: Among well-differentiated NETs, PanNETs seem to benefit more from

    chemotherapy (increased response rate) than other NETs (i.e. small intestinal NETs)

    Benefit from chemotherapy in PanNETs: Objective response

    Non-PanNETs

    Response Rate

    9.5%

    PanNETs

    Response Rate

    26.3%

    Vs.

    Reprinted from Cancer Treat Rev 2014, 44(16), Lamarca A, et al. Chemotherapy for advanced non-pancreatic well-differentiated neuroendocrine tumours of the gastrointestinal tract, a systematic review and meta-analysis: A

    lost cause?, 26-41, Copyright 2014, with permission from Elsevier.

  • CHEMOTHERAPY OPTIONS OF CHEMOTHERAPY

    Multiple different chemotherapy

    combinations have been tested

    over the years, most of them

    in retrospective series of small

    prospective studies

    Treatment Type of Trial Patients’

    characteristics

    Number of pNET

    patients

    Response rate (%) Disease

    control

    rate (%)

    Median PFS

    (months)

    Median overall

    survival (months)

    Year of publication

    (References)

    Chemotherapy

    Chlorozotocin vs.

    Streptozocin and

    fluorouracil vs. Doxorubicin

    and streptozocin

    Phase III Previous chemo

    allowed (no data)

    33

    33

    36

    30

    45

    69

    No data 17

    14

    18

    18

    16.8

    26.4

    1992 (12)

    Doxorubicin and

    streptozocin

    Retrospective

    analysis

    24% previous

    chemotherapy

    45 36 60 16 2-year survival rate:

    50.2%

    2004 (13)

    Streptozocin, doxorubicin

    and fluorouracil

    Retrospective

    analysis

    5% second line 84 39 89 18 37 2004 (14)

    5-fluororuracil, cisplatin and

    streptozocin

    Retrospective

    analysis

    Chemo naïve

    patients

    82 (49 pNETs) 38 86 9.1 31.4 2010 (15)

    Capecitabine and

    streptozocin +/-cisplatin

    Randomised

    Phase II

    Previous chemo

    allowed (no data)

    86 (48% pNETs) 14/8 78/82 9.7/10.2 34.7 2012 (16)

    Decarbazine Phase II 44% second line 50 34 No data No data 19.3 2001 (18)

    Temozolomide and

    thalidomine

    Phase II 45% second line 29 (38%

    pancreatic)

    45 93 No data 2-year survival rate

    61%

    2006 (19)

    Temozolomide and

    capecitabine

    Retrospective

    analysis

    Chemo naïve

    patients

    30 70 97 18 2-year survival rate

    92%

    2011 (21)

    Temozolomide and

    bevacizumab

    Phase II 44% second line 35 (44%

    pancreatic)

    33.3 87 14.3 41.7 2012 (20)

    Temozolomide, everolimus Phase I/II 33% second line 43 40 93 15.4 No data 2013 (37)

    Capecitabine and

    oxaliplatin

    Retrospective

    analysis

    Second line 27 well-

    differentiated NETs

    included

    27 well-

    differentiated NETs

    (unknown number

    of pNETs)

    30 78 20 40 2007 (23)

    5-fluorouracil, oxaliplatin

    and bevacizumab

    Phase II No 6 of 13 patients

    included

    20 100 No data No data 2008 (24)

    Capecitabine, oxaliplatin

    and bevacizumab

    Phase II No data 20 30 94 13.7 No data 2011 (25)

    Chemotherapy and Targeted Therapy Studies for Well-differentiated Pancreatic NETs

    Adapted from Lamarca A, et al.

    J Oncopathol 2014;2(1):15–25

  • CHEMOTHERAPY

    ◆ Capecitabine (750 mg/m2 twice

    daily day 1–14) + temozolomide

    (200 mg/m2 once daily day

    10–14); 28-day cycle

    ◆ Median PFS 18 months

    ◆ Radiological response rate:

    70%

    ◆ Toxicity: myelosuppression

    Strosberg JR, et al. Cancer 2011;117(2); 268–75. Courtesy of John Wiley and Sons. Copyright © 2010 American Cancer Society

    Temozolomide + Capecitabine

    % c

    han

    ge

    Best radiographic response | 70% of patients achieved PR (RECIST)

  • CHEMOTHERAPY

    Randomised phase II study in progressive PanNETS

    Kunz P, et al. J Clin Oncol 36, 2018 (suppl; abstr 4004). By permission of Dr Pamela Kunz

    Temozolomide Capecitabine vs. Capecitabine: E2211 clinical trial

    Progressive,

    G1 / G2, metastatic

    pancreatic NETsR

    Arm A:

    Temozolomide 200 mg/m2 po QD days 1–5

    Arm B:

    Capecitabine 750 mg/m2 po BID days 1–14

    Temozolomide 200 mg/m2 po QD days 10–14Stratified by:

    ◆ Prior everolimus

    ◆ Prior sunitinib

    ◆ Concurrent octreotide

    Primary endpoint:

    PFS (local review)

    Secondary endpoint:

    RR, OS, toxicity

    Correlative endpoints:

    MGMT by IHC, MGMT by

    promoter methylation

    1:1 n=72

    n=72

    Cycle length = 28 days; max 13 cycles.

    Imaging performed every 12 weeks (RECIST 1.1)

  • CHEMOTHERAPY

    Randomised Phase 2 study in progressive PanNETS

    Kunz P, et al. J Clin Oncol 36, 2018 (suppl; abstr 4004) 2018. By permission of Dr Pamela Kunz.

    Temozolomide Capecitabine vs. Temozolomide: E2211 clinical trial

    Tem TemCap Comments

    Grade 1

    Grade 2

    45.1%

    54.9%

    68.1%

    31.9%

    PFS (median) (months) 14.4 22.7HR 0.58 (95% CI 0.36, 0.93); p=0.023

    If adjusted for grade results are

    unchanged (HR 0.61 p=0.042)

    OS (median) (months) 38.0 Not reachedHR 0.41 (95% CI 0.21, 0.82); p=0.012

    If adjusted for grade results are

    unchanged (HR 0.46 p=0.033)

    Complete Response 2.8% 0%

    Partial Response 25.0% 33.3%

    Response duration (months) 9.7 12.1

  • PEPTIDE RECEPTOR RADIONUCLIDE THERAPY (PRRT)

    ◆ Radiolabelled octreotide: selection of patients based on somatostatin receptor positive imaging

    ◆ Metastatic midgut neuroendocrine tumours (excluding PanNETs) with progressive disease to SSA

    ◆ Benefit in terms of survival (PFS: HR 0.21 [95%CI 0.13, 0.33]) → approved for its use in GEP-NETs

    (including PanNETs)

    From The New England Journal of Medicine, Strosberg J, et al.,

    Phase 3 Trial of 177Lu-Dotatate for Midgut Neuroendocrine

    Tumors, 376(2), 125–35. Copyright © 2017 Massachusetts

    Medical Society. Reprinted with permission from

    Massachusetts Medical Society

    NETTER-1 clinical trial: PRRT vs. Octreotide 60 mg

  • PEPTIDE RECEPTOR RADIONUCLIDE THERAPY (PRRT)

    ◆ Review of the literature; multiple retrospective studies

    ◆ Among these studies, the median disease control rate was 83% (range 50%–94%) and the median

    objective response rate was 58% (range 13%–73%)

    ◆ Reported median progression-free survival for the overall PanNET population ranged from 25 to 34 months;

    the median overall survival ranged from 42 to 71 months

    Ramage J, et al. Semin Oncol 2018;45(4):236–48

    Experience of PRRT in PanNETs

    “The effect may be at least as great as in midgut NET”

  • THE FUTURE OF MANAGEMENT OF PANCREATIC NETSNew targeted therapies

    Challenges for treatment sequencing

    Immunotherapy

  • NEW TARGETED THERAPIES

    Role of Lenvatinib: TALENT study

    ◆ Cohort A: Lenvatinib in PanNETs (pre-treated); n=55

    ◆ Objective response rate (RECIST): 40.4%

    ◆ Median PFS: 14.2 months (95% CI 11.4, not reached)

    ◆ Dose reduction required: 88%

    ◆ AEs: G3 8.6%; G4 0.5%; G5 0.1%

    ◆ Good response rate in PanNETs. Confirmatory trials

    awaited

    Capdevila J, et al. Ann Oncol 2018;29(Suppl 8): Abstract 1307O (presented at ESMO 2018); By permission of Dr Jaume Capdevila.

    Chan ASCO-GI 2017; Xu ENETS 2017

    Cabozantinib, Sulfatinib

    ◆ Phase 3 trials ongoing in view of promising Phase 2 results

  • CHALLENGES FOR TREATMENT SEQUENCING

    Targeted or PRRT: COMPETE study (n=300)

    ◆ G1 or G2 metastatic or locally advanced well

    diff, functioning or non-functioning GEP-NETs

    ◆ SSTR +ve

    ◆ PD as per RECIST 1.1

    ◆ Randomisation 2:1

    R

    177Lu-DOTA-TOC 7.5 Gbq

    (4 cycles; 1 dose/12 wks)

    Everolimus 10 mg PO OD

    Primary endpoint: Progression free survival at 2 years

    Primary completion date: Dec 2020

  • CHALLENGES FOR TREATMENT SEQUENCING

    Targeted or Chemotherapy: SEQTOR study (n=180)

    ◆ G1 or G2 metastatic or locally advanced well

    diff, functioning or non-functioning Pan-NETs

    ◆ Randomisation 1:1

    R

    Everolimus 10 mg PO OD

    Streptozocin + 5-FU

    3/6-weekly

    Primary endpoint: Progression free survival

    Accrual completed: October 2018

    Expected results: 2020

  • IMMUNOTHERAPY

    Current data in PanNETS

    Pembrolizumab (KEYNOTE-028 study; anti-PD-1)

    ◆ 16 PanNETs (PD-L1 positive )

    ◆ 6% objective responses

    Spartalizumab (PRD001; anti PD-1) in NETs

    ◆ Cohort of patients with PanNETS (n=30)

    ◆ Partial response rate: 3%

    ◆ Disease-control rate: 58%

    Further studies are required in PanNETS to assess the role of immunotherapy

    Mehnert ESMO 2017; Yao J, et al. Ann Oncol 2018;29 (suppl_8): viii467-viii478. Presented at ESMO 2018. By permission of Dr J. Yao.

  • TAKE HOME MESSAGES

  • TAKE HOME MESSAGES

    ◆ PanNETs are rare types of neoplasms whose incidence and prevalence are increasing

    ◆ Proliferation index and morphology are the cornerstone of tumour classification, which impacts both treatment

    and prognosis

    ◆ Surgery is the only curative treatment for localised disease

    ◆ Surgery for metastatic disease (if resectable) does have a role

    ◆ Liver directed therapies are of use for patients with unresectable liver disease if liver predominant or functional

    symptoms are present

    ◆ Systemic treatment includes somatostatin analogues, targeted therapies (everolimus, sunintinib), chemotherapy

    (TemCap) and PRRT

    ◆ Most adequate treatment sequencing is unknown and is a current challenge

    ◆ Discussion of patients in expert MDTs is recommended for adequate treatment planning at time of presentation

    ◆ Selection of systemic treatment relies on tumour proliferation rate, Ki-67 and disease burden

  • THANK YOU!