Guidelines

Endokrynologia Polska

DOI: 10.5603/EP.a2022.0053

ISSN 0423–104X, e-ISSN 2299–8306

Volume/Tom 73; Number/Numer 3/2022

Colorectal neuroendocrine neoplasms — update of the diagnostic and therapeutic guidelines (recommended by the Polish Network of Neuroendocrine Tumours)

Teresa Starzyńska1Magdalena Londzin-Olesik2Tomasz Bednarczuk*3Marek Bolanowski*4Małgorzata Borowska*5Ewa Chmielik*6Jarosław B. Ćwikła*7Wanda Foltyn*2Iwona Gisterek*8Daria Handkiewicz-Junak*9Alicja Hubalewska-Dydejczyk*10Michał Jarząb*11Roman Junik*12Dariusz Kajdaniuk*13Grzegorz Kamiński*14Agnieszka Kolasińska-Ćwikła*15Aldona Kowalska*16Leszek Królicki*17Jolanta Kunikowska*17Katarzyna Kuśnierz*18Andrzej Lewiński*19Łukasz Liszka*20Bogdan Marek*13Anna Malczewska*2Anna Nasierowska-Guttmejer*21Ewa Nowakowska-Duława*22Marianne E. Pavel*23Joanna Pilch-Kowalczyk*24Jarosław Reguła*25Violetta Rosiek*2Marek Ruchała*26Grażyna Rydzewska*27Lucyna Siemińska*13Anna Sowa-Staszczak*10Zoran Stojčev*28Janusz Strzelczyk*2Michał Studniarek*29Anhelli Syrenicz*30Marek Szczepkowski*31Ewa Wachuła*32Wojciech Zajęcki*5Anna Zemczak*2Wojciech Zgliczyński*33Krzysztof Zieniewicz*34Beata Kos-Kudła**2
1Department of Gastroenterology, Medical Pomeranian University in Szczecin, Szczecin, Poland
2Department of Endocrinology and Neuroendocrine Tumours, Department of Pathophysiology and Endocrinology, Medical University of Silesia, Katowice, Poland
3Department of Internal Medicine and Endocrinology, Medical University of Warsaw, Warsaw, Poland
4Chair and Department of Endocrinology, Diabetes and Isotope Therapy, Medical University of Wroclaw, Wroclaw, Poland
5Department of Endocrinology and Neuroendocrine Tumours, Medical University of Silesia, Katowice, Poland
6Tumor Pathology Department, Maria Sklodowska-Curie Memorial National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
7Department of Cardiology and Internal Medicine, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
8Chair of Oncology and Radiotherapy, Medical University of Silesia, Katowice, Poland
9Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie Memorial National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
10Chair and Department of Endocrinology, Jagiellonian University Medical College, Cracow, Poland
11Breast Unit, Maria Sklodowska-Curie Memorial National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland
12Department of Endocrinology and Diabetology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland
13Division of Pathophysiology, Department of Pathophysiology and Endocrinology, Medical University of Silesia, Katowice, Poland
14Department of Endocrinology and Radioisotope Therapy, Military Institute of Medicine, Warsaw, Poland
15Department of Oncology and Radiotherapy, Maria Sklodowska-Curie Memorial National Research Institute of Oncology, Warsaw, Poland
16Department of Endocrinology, Holycross Cancer Centre, Collegium Medicum, Jan Kochanowski University, Kielce, Poland
17Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
18Department of Gastrointestinal Surgery, Medical University of Silesia, Katowice, Poland
19Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, Lodz, Poland
20Department of Pathomorphology and Molecular Diagnostics, Medical University of Silesia, Katowice, Poland
21Faculty of Medicine, Lazarski University in Warsaw, Warsaw, Poland
22Department of Gastroenterology and Hepatology, Medical University of Silesia, Katowice, Poland
23Department of Medicine 1, Endocrinology and Diabetology, Friedrich Aleksander University of Erlangen-Nurnberg, Erlangen, Germany
24Department of Radiology and Nuclear Medicine, Medical University of Silesia, Katowice, Poland
25Department of Oncological Gastroenterology, Maria Sklodowska-Curie Memorial National Research Institute of Oncology, Warsaw, Poland
26Department of Endocrinology, Metabolism, and Internal Diseases, Medical University in Poznan, Poznan, Poland
27Department of Internal Medicine and Gastroenterology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
28Department of Oncology and Breast Diseases, Centre of Postgraduate Medical Education, Warsaw, Poland
29Department of Radiology, Medical University of Gdansk, Gdansk, Poland
30Department of Endocrinology, Metabolism, and Internal Diseases, Pomeranian Medical University, Szczecin, Poland
31Clinical Department of Colorectal, General, and Oncological Surgery, Centre of Postgraduate Medical Education, Warsaw, Poland
32Department of Clinical Oncology, Gdynia Oncology Centre of the Polish Red Cross Maritime Hospital, Gdynia, Poland
33Department of Endocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
34Chair and Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
*Authors arranged in alphabetical order, **Senior author

Teresa Starzyńska, MD PhD, Professor of Medicine, Department of Gastroenterology, Medical Pomeranian University in Szczecin, ul. Uni Lubelskiej St. 1, 71–252 Szczecin, Poland, tel/fax: (+48) 91 425 32 11; e-mail: testa@pum.edu.pl

Submitted: 24.04.2022

Accepted: 25.04.2022

Early publication date: 30.06.2022

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially

Abstract
Colorectal neuroendocrine neoplasms (CRNENs), especially rectal tumours, are diagnosed with increased frequency due to the widespread use of colonoscopy, including screening examinations. It is important to constantly update and promote the principles of optimal diagnostics and treatment of these neoplasms. Based on the latest literature and arrangements made at the working meeting of the Polish Network of Neuroendocrine Tumours (June 2021), this paper includes updated and supplemented data and guidelines for the management of CRNENs originally published in Endokrynologia Polska 2017; 68: 250260. (Endokrynol Pol 2022; 73 (3): 584–597)
Key words: colorectal neuroendocrine neoplasms; diagnosis; treatment; monitoring

Colorectal neuroendocrine neoplasms (CRNENs) are diagnosed with increasing frequency, mainly due to the widespread use of colonoscopy, including screening examinations [1–9]. Most of these lesions are located in the rectum. An increasing body of evidence suggests that rectal neuroendocrine neoplasms (RNENs) and colon neuroendocrine neoplasms (CNENs) are separate diseases. RNENs are characterised with a low to moderate degree of malignancy, good prognosis, and most of the lesions can be treated endoscopically. On the other hand, CNENs are often aggressive, poorly differentiated neoplasms with a higher degree of malignancy, and unfavourable prognosis, and surgery is the treatment of choice [5, 10].

1. Epidemiology

CNENs and RNENs constitute 17.6% and 26.3%, respectively, of all neuroendocrine neoplasms of the gastrointestinal tract [11, 12].

According to the 2015 Surveillance, Epidemiology and End Result (SEER) register, the incidence of CNEN is 0.35/100,000/year [13]. The mean age of CNEN onset is the seventh decade of life. Women suffer twice as often [14]. The most common location of colon tumours is the caecum [15].

There has been a ten-fold increase in the incidence of RNEN over the past 30 years, and it now amounts to 1.5 cases per 100,000 per year [13]. They are detected once in every 10002000 endoscopic examinations [1, 5, 6, 8, 16]. The average age of a patient diagnosed with RNEN is 56 years, and they are predominantly men [17].

2. Clinical characteristics

The clinical picture of a CRNEN case is related to its location and stage. These tumours are characterised by the lack of specific hormone secretion [8].

If the neuroendocrine neoplasm (NEN) is located in the colon, the main symptoms include a change in the nature of bowel movements (most often diarrhoea), and in advanced disease, abdominal pain and weight loss [7]. Abdominal pain is caused by the effect of the tumour mass or overgrowth of the connective tissue (desmoplastic effect). Weakness and decreased exercise tolerance related to blood loss through the gastrointestinal tract may also occur. Gastrointestinal obstruction is possible. At diagnosis, the mean CNEN size is approximately 5 cm. Distant metastases occur in 1640% of patients [18]. In the case of CNEN, the five-year survival is the lowest among all gastrointestinal neuroendocrine neoplasms and refers to 3342% of patients after surgical treatment [18].

Most RNENs are asymptomatic and are diagnosed incidentally during colonoscopy [5, 19–21].

Symptoms may include rectal bleeding, itching and discomfort in the area, change in bowel movements, and painful faecal urgency. The vast majority (7585%) of the lesions are local at diagnosis. Five-year survival is 75100% depending on the grade of histological maturity, the proliferation index, and the stage of clinical advancement [22]. Most RNENs do not exceed 10 mm at diagnosis. The tumours of this size have a low metastatic potential, but it should be kept in mind that metastases to regional lymph nodes and distant metastases occur in 3% and 1.6% of patients, respectively [23]. The risk of metastasis increases with the lesion size. In tumours of 1119 mm in size, regional metastases occur in 66% and distant metastases in 50% of patients. However, in tumours of20 mm in size, the risk of distant metastases is almost 100% [23].

Carcinoid syndrome in RNEN is almost unheard of due to the very rare occurrence of serotonin-secreting cells in this location (0.1%) [2, 24].

A second neoplasm develops in 13% of patients with CNEN [2, 4, 5, 25]. The gastrointestinal system, including the colon, is the most common location for synchronous tumours, while metachronous neoplasms mainly affect the lungs, prostate, and urinary system.

3. Diagnostics

3.1. Biochemical diagnostics

We do not have a biochemical marker specific for CRNEN. Determination of serum chromogranin A (CgA) is still the most valuable method of monitoring the disease treatment and prognosis. The CgA concentration may increase and correlate with the severity of the neoplastic disease [26].

Because tumours located in this part of the gastrointestinal tract rarely secrete serotonin, the concentration of 5-hydroxyindoleacetic acid (5-HIAA) in the 24-hour urine collection usually remains within the normal range.

The possibility of using other tests in the diagnosis and monitoring the course of the disease and treatment effects is described in the article Update of the diagnostic and therapeutic guidelines for gastro-entero-pancreatic neuroendocrine neoplasms (recommended by the Polish Network of Neuroendocrine Tumours) [27].

The minimal consensus statement on biochemical tests:
1. If CRNEN is diagnosed, CgA remains a biochemical marker before the initiation of treatment and during further monitoring [II, 2A]#.
3.2. Pathomorphological diagnosis

Similarly to other gastrointestinal NENs, CRNENs include neoplasms, the common element of which is the ability to produce neuroendocrine markers. These properties are most often confirmed by immunohistochemical tests, demonstrating mainly the presence of synaptophysin and CgA in neoplastic cells. The ability to produce neuroendocrine markers is not a factor determining the biology of these tumours, but only their common property. Based on their clinical course and the response to treatment, they can be divided into two main groups. These are neuroendocrine tumours (NETs) and neuroendocrine carcinomas (NECs). This division of pancreatic neuroendocrine neoplasms (PanNENs), initially proposed in 2017, is based on the assessment of two factors: proliferative activity determined by the Ki-67 proliferation index and the mitotic index, as well as by the histoformative features of neoplastic cells [28, 29]. This division effectively reflects the biological differences stemming from various genetic disorders in these groups. For example, in the NET group there are no mutations in the TP53 and RB genes, which are observed in the NEC group. Moreover, NECs, unlike NETs, respond well to platinum-based chemotherapy [30].

The group of well-differentiated NETs, due to proliferative activity defined by the Ki-67 proliferation index and the mitotic index, is divided into three subcategories: NETs G1 with the Ki-67 proliferation index < 3% and the mitotic index < 2/2 mm2, NETs G2 with the Ki-67 proliferation index in the range 320% and the mitotic index in the range 220/2 mm2, and NETs G3 with higher parameters of proliferative activity. It should be noted that we are not grading the group of poorly differentiated NENs, or NECs, previously graded as NENs G3 [30]. This has led to confusion in the interpretation of NETs G3. It is also worth noting that currently the group of NETs G3 and NECs has the same lower limit of the Ki-67 proliferation index (above 20%) and no upper limit of the Ki-67 proliferation index. Very rarely, neoplasms with the Ki-67 index of, e.g., 80% can be classified into the NET G3 group, and neoplasms with the Ki-67 index of, e.g., 30% can be calssified into the NEC group. The differences between these two groups of neoplasms, which are important from the point of view of treatment and prognosis, are based on cytological and histoformative features.

There are two groups of NECs classified according to their cytological and histological structure: small cell carcinoma (SCC) and large cell neuroendocrine carcinoma (LCNC). The nomenclature suggests significant morphological differences, while in practice this distinction is sometimes difficult, sometimes even impossible, especially when we are dealing with a small biopsy specimen. Due to the currently used treatment regimen, which is the same for both types of carcinomas, the appropriate procedure in difficult cases and the availability of only a small specimen is the diagnosis of poorly differentiated neuroendocrine carcinoma (without taking into account the division into two groups).

The Ki-67 proliferation index in neuroendocrine tumours is often uneven; therefore, the assessment is performed in the areas of the highest intensity, the so-called hot spots. For this reason, when we are dealing with a small section of neoplastic tissue, and not the entire tumour, the degree of differentiation may change after assessing the entire tumour in the postoperative material. The method of assessment of the mitotic index and Ki-67 proliferation index is presented in the pathomorphological part of the Update of the diagnostic and therapeutic guidelines for gastro-entero-pancreatic neuroendocrine neoplasms (recommended by the Polish Network of Neuroendocrine Tumours) [27].

As in other parts of the gastrointestinal tract, there also occur tumours in the large intestine with a mixed histological structure, including neuroendocrine and non-neuroendocrine tissues. Such tumours are classified as mixed neuroendocrine non-neuroendocrine neoplasms (MiNENs). In the large intestine, these neoplasms occur mainly as tumours composed of components of adenocarcinoma and neuroendocrine carcinoma. For this reason, they were formerly referred to as mixed adenoneuroendocrine carcinomas (MANECs). To classify the neoplasm to the MiNEN group, it is assumed that each component should constitute at least 30% of the tumour tissue. In cases that do not meet the quantitative criteria, the presence of a minority component should be considered in the diagnosis, especially when it is a neuroendocrine carcinoma component. In MiNEN, both components must be invasive. Thus, the presence of NEN tissue within the adenoma does not classify it as mixed tumour. There are rare cases when both neoplastic tissues do not constitute a single tumour with two components, but two independent tumours. Such tumours are not classified as MiNEN; they are referred to as “collision tumours”, and each component is described and graded separately. Neuroendocrine differentiation sometimes occurs in relapses of non-neuroendocrine cancers after treatment. These tumours also do not belong to the MiNEN group.

The presence of cells with neuroendocrine properties can be confirmed in immunohistochemical tests in 50% of cancers typical for the gastrointestinal system, but they do not constitute a basis for the diagnosis of NET. The histopathological structure, confirmed in immunohistochemical tests, is of decisive importance. Colon NETs are most often composed of serotonin-producing cells (EC-cells), which, histologically, form nest structures with peripheral nuclear palisades, similarly to tumours composed of histamine-producing cells (ECL-cell). These cells with eosinophilic cytoplasm demonstrate a strong positive reaction with the presence of CgA and synaptophysin. More rarely, NETs are composed of L-cells forming trabecular structures that are characterised with a positive immunohistochemical reaction to the presence of synaptophysin and, only focally, a weakly positive reaction to the presence of CgA, similarly to NEC. Tumours composed of these cells are more common in the rectum; therefore, synaptophysin is a more sensitive marker of immunohistochemical neuroendocrine differentiation for tumours located in this area.

CRNETs often demonstrate the presence of CDX2 in immunohistochemical tests, which is sometimes useful in the search for a primary tumour in the presence of metastases of a neuroendocrine tumour from an unknown primary site. Unfortunately, some of these tumours also demonstrate the presence of TTF1, an antigen associated mainly with lung and thyroid cancers.

The 2019 World Health Organization (WHO) histopathological classification of colorectal neuroendocrine neoplasms is presented in Table 1 [32].

Table 1. 2019 World Health Organization (WHO) classification of colorectal neuroendocrine neoplasms [32]

Neuroendocrine tumour NOS (8240/3)

Neuroendocrine tumours, grade 1 (G1) (8240/3)

Neuroendocrine tumours, grade 2 (G2) (8249/3)

Neuroendocrine tumours, grade 3 (G3) (8249/3)

L-cell tumour (8152/3)

Glucagon-like peptide-producing tumour (98152/3)

PP/PYY-producing tumour (8241/3)

Enterochromaffin-cell carcinoid (8241/3)

Serotonin-producing tumour (8241/3)

Neuroendocrine carcinoma (8246/3)

Large cell neuroendocrine carcinoma (8013/3)

Small cell neuroendocrine carcinoma (8041/3)

Mixed neuroendocrine non-neuroendocrine neoplasm (MiNEN) (8154/3)

The final postoperative histopathological report must include an assessment of the tumournodemetastasis (TNM) staging. In the case of neuroendocrine tumours, due to biological differences in relation to carcinomas, separate staging classification is used. Currently, the 2019 WHO TNM classification of gastrointestinal tumours is accepted, which in the case of colon tumours coincides with the 8th edition of the American Joint Committee on Cancer (AJCC) and Union for International Cancer Control (UICC) classifications [32–34] (Tab. 2). NEC and MiNEN are graded as non-neuroendocrine carcinomas.

Table 2. The stage of colorectal neuroendocrine tumour according to the 8th edition of the American Joint Committee on Cancer/Union for International Cancer Control (AJCC/UICC) pathological tumour–node–metastasis (pTNM) classification of 2017 [32–34]

T primary tumour

pTX

The main tumour has not been assessed

pT1

The tumour is confined to the mucosa and submucosa and does not exceed 2 cm in diameter

pT1a

The tumour is confined to the mucosa and submucosa and does not exceed 1 cm in diameter

pT1b

The tumour is confined to the mucosa and submucosa and its diameter is 1–2 cm

pT2

The tumour infiltrates the muscularis propria or is larger than 2 cm in diameter

pT3

The tumour infiltrates the subserous tissue or the pericolonic and perirectal tissues not covered by the peritoneum

pT4

The tumour infiltrates the peritoneum and other organs

N regional lymph nodes

pNX

Regional lymph nodes have not been assessed

pN0

No regional lymph node involvement

pN1

Regional lymph node involvement

M distant metastases

pM0

No distant metastases

pM1

The presence of distant metastases

  • M1a metastases limited to the liver
  • M1b metastases to at least one non-hepatic region (e.g. lungs, ovaries, distant lymph nodes, peritoneum, bones)
  • M1c metastases both to the liver and to non-hepatic regions

An important parameter in the CRNEN histopathological report is the assessment of the proximal, distal, and peripheral margins. The peripheral margin is assessed in the parts of the gastrointestinal tract not covered with serosa. It must be emphasised that during the macroscopic assessment of the surgical specimen, it should be marked with ink. It is recommended that the distance from the most deeply infiltrating tumour foci to the peripheral margin line is specified. The presence of residual neoplastic tissue following treatment, denoted by the letter R, is not a necessary factor for the assessment of pTNM and clinical advancement. However, this assessment is very important for further therapeutic decisions, especially in patients with rectal tumours. Due to doubts regarding the minimum width of the surgical margin relative to the tumour, in order to determine the lack of residual tissue at the tumour resection border, this assessment can be characterised according to the scheme proposed by Wittekind [35] (Tab. 3).

Table 3. Assessment of residual neoplastic tissue in the postoperative material according to Wittekind [35]

Assessment of residual tumour tissue (R) after treatment

Presence of tumour tissue

Rx

Presence of residual neoplastic tissue cannot be assessed

R0 > 1 mm

No neoplastic tissue in the resection margin located at a distance larger than 1 mm from the tumour

R0 ≤ 1 mm

No neoplastic tissue in the resection margin located at a distance smaller than or equal to 1 mm from the tumour

R1-dir

Neoplastic tissue present in the resection margin the resection line runs through the neoplastic infiltration

R2a

Neoplastic tissue visible in macroscopic picture at the site of tumour resection (locally)

R2b

Neoplastic tissue visible in macroscopic picture at a site distant from the tumour (e.g. in a metastatic lymph node cut within the margin of resection)

R2c

Neoplastic tissue visible in macroscopic picture at both sites

Statement on pathomorphological examinations

The macroscopic assessment method and the principles of collecting specimens for histopathological examination are the same as in the case of other neoplasms. It is included in the standards for the postoperative material processing published by the Polish Society of Pathologists. It is recommended that the final pathology report be a synaptic report, which includes the following:

name of the procedure during which the specimen was collected;
macroscopic description of the specimen;
histopathological diagnosis with International Classification of Disease (ICD) code.

This way of result reporting is transparent, first of all, to clinicians and oncologists. At the same time, because laboratories use various numbering systems for blocks and organs from which the specimens were collected, it is necessary for the report to include information on the labelling of preparations collected from a specific area. This avoids any ambiguity in the case of pathomorphological consultations.

Minimal consensus statement on pathomorphological examinations
1. The minimum histopathological report regarding CRNENs should include:
histological type of the neoplasm divided into NET, NEC, SCC, LCNC, MiNEN;
grade of histological maturity (G) relating to well-differentiated neoplasms (G1, G2, G3) and NECs with the division into large and small cell neuroendocrine carcinoma;
pTNM staging according to the TNM, AJCC/UICC classification, 8th edition of 2017 (in each case, it is important to provide information on the specific classification used);
the assessment of the tumour purity of the surgical margins and their location relative to the tumour;
the histopathological diagnosis of NEN must be confirmed by immunohistochemical examinations, assessment of the expression of neuroendocrine markers: synaptophysin and CgA, and Ki-67 proliferation activity using the mindbomb homolog 1 (MIB1) antibody [III, 1]#.
3.3. Location diagnostics
3.3.1. Endoscopic diagnostics

“Classic” white light colonoscopy combined with biopsy for morphological assessment is an essential tool in the diagnosis of CRNENs.

Diagnostic efficiency is increased by the use of next-generation image-enhanced endoscopy (IEE), including narrow-band imaging (NBI) and magnifying endoscopy (ME). Image enhancement endoscopy increases the sensitivity of early lesion detection. Additionally, it helps in the differential diagnosis (distinguishing NENs from epithelial lesions), which in turn determines the selection of the optimal treatment method [36]. The most accessible and most frequently used is endoscopic image enhancement with the use of a narrow light band.

In recent years, artificial intelligence has been a significant achievement. Equipping the devices with this type of software significantly increases the sensitivity and specificity of endoscopy [37, 38].

Imaging of the large intestine is also possible with the use of capsule endoscopy; however, in the diagnostics of CRNENs, this method has no practical application at present [39].

In most patients (83%) with CRNEN (compared to other subepithelial lesions), the biopsy is positive because NENs originate from the muscularis mucosae [40]. In patients qualified for endoscopic treatment, biopsy is not recommended, because fibrosis occurring after collecting the specimens may make the procedure more difficult and increase the risk of complications. The collection of material for pathomorphological examinations is necessary before surgical treatment.

The characteristic endoscopic morphological features of CRNEN include polyps growing from a broad base, with a smooth surface, covered with unchanged or injected mucosa with dilated vessels, often yellow/white (in the case of rectal lesions). CNENs are most often advanced lesions macroscopically resembling cancer infiltration, diagnosed early in the form of a polyp/subepithelial tumour. Most RNENs (80%) have the above-described characteristic morphological features [5–8]. The atypical features, occurring in about 20% of cases, include the following: a semi-pedunculated shape, a form resembling a mushroom cap with a central depression, a flat lesion, reddening of the mucosa, and the presence of erosions or ulcerations on the surface. Atypical features occur mainly in lesions larger than 1 cm. Minor NEN lesions (up to 57 mm) may resemble a hyperplastic polyp. The use of endoscopy with NBI and/or magnification is helpful in differentiating between both lesions due to the different pit pattern and microvessel image [36]. RNENs are usually single, most often occurring in the middle part. The majority of RNENs (7080%) are up to 10 mm in size at diagnosis [24]. In 2020, Chen et al. described a new RNEN endoscopic assessment system [41]. The size, shape and surface of the mucosa are considered in the scale. The division of individual features is as follows: size < 1 cm,1 cm,2 cm; shape sessile, semi-pedunculated, flat/mushroom-shaped; mucosa smooth, with a depression, erosion/ulceration (Tab. 4). The individual features are assigned points, with a cut-off value of 110. The accuracy of the scale in identifying poor NEN prognosis is high and reaches 95% (change110 points).

Table 4. Endoscopic scale of rectal neuroendocrine neoplasms prognosis assessment according to Chen et al. [41]

Endoscopic picture

Number of points

Lesion size

< 1 cm

0

≥ 1 cm

76

≥ 2 cm

100

Lesion shape

Non-pedunculated

0

Semi-pedunculated

34

Flat/mushroom-shaped

75

Surface of the mucosa

Smooth/unchanged

0

With a depression

21

With erosion/ulceration

86

In the case of NENs located in the rectum, endoscopic ultrasound (EUS) is an important complement to colonoscopy. The latest ENETS (European Neuroendocrine Tumour Society) guidelines recommend EUS in RNENs starting from 5 mm [9]. EUS is recommended in three main clinical situations: at diagnosis before planned treatment, in the case of incomplete lesion removal before selecting “rescue” therapy, and as a follow-up examination. The examination is primarily used to determine the degree of local advancement (depth of infiltration of the intestinal wall, assessment of regional lymph nodes for metastases) and is useful in the differential diagnosis [8, 9, 42]. The sensitivity and specificity of this examination in determining the depth of infiltration are 87% and 93%, respectively [6]. A typical picture of NEN in EUS is a well-limited, hypoechoic, homogeneous lesion originating from the muscularis mucosa [43, 44]. After NEN removal, EUS is of little use in assessing the completeness of the procedure or looking for recurrence in the intestinal wall; it is mainly used to assess regional lymph nodes [45].

Minimal consensus statement on endoscopic examinations
1. “Classic” colonoscopy with the use of white light with biopsy for morphological evaluation (* I, 1) (we do not collect specimens when planning endoscopic treatment).
2. EUS in RNEN5 mm [IV/V, 2B]#.
3. colonoscopy using image enhancement techniques narrow-band imaging and/or magnification (differential diagnosis of NENs and epithelial polyps) [II, 2A]#.
3.3.2. Imaging diagnostics

Imaging diagnostics in patients with CRNEN is primarily used to determine the stage of the disease, and to assess and monitor the effects of treatment. Assessment of the primary lesion in the large intestine is usually based on endoscopic examinations. The following methods are used in imaging diagnostics.

Computed tomography (CT) before and after intravenous administration of a contrast agent

Multiphase CT examination (native, usually involving the abdominal cavity, in particular the liver) before and after the intravenous administration of a contrast agent, involving the abdominal cavity and pelvis in the arterial phase (approx. 2530 s) and portal venous phase (approx. 5575 s), because some highly vascularised NENs are only visible in one phase or the other after intravenous (i.v.) administration of the contrast agent. This examination is performed to detect metastatic lesions to regional lymph nodes and distant metastases, as well as in the assessment of NEN infiltration of the neighbouring organs [46, 47].

If there is a suspicion of dissemination of the neoplastic process (in particular, when the primary feature of the primary tumour pT is assessed as T34, and/or N1, Mx), CT of the chest and base of the neck is performed to assess potential metastases.

In the case of lung parenchyma assessment, the examination is performed without administering a contrast agent, in the case of mediastinal assessment, examination before and after the i.v. administration of a contrast agent is preferable. Generally, the CT scan of the CNEN is performed in the same way as a standard CT scan for colorectal cancer. In the case of therapy, the standard examination description must include a summary of the effectiveness of therapy based on the Respons Evaluation Criteria In Solid Tumors (RECIST) 1.0 or RECIST 1.1 criteria [48].

Magnetic resonance imaging (MRI) before and after intravenous administration of a contrast agent

In patients with CRNETs, MRI mainly concerns the assessment of the pelvis and the abdominal cavity. In selected cases, MRI of the spine, central nervous system (CNS), or whole body should be performed. MRI is very helpful in the case of suspected metastases in the course of CRNENs due to the excellent contrast resolution, which is superior to that of CT.

MRI is the preferred examination in the assessment of local advancement in the case of RNENs that do not meet the size criterion of the primary lesion as T1a, size < 1 cm with R0 > 1 mm or R01 mm. Rectal MRI includes all other T1 resections with R1 and R2, and at least T1 moderately differentiated NETs G2. MRI should be performed before and after intravenous administration of a contrast agent (DCE dynamic contrast enhancement) along with diffusion-weighted imaging (DWI) and apparent diffusion coefficient (ADC) maps.

MRI is the optimal examination to assess the degree of local advancement, it is preferable to CT, it is more easily accessible, and it has a broader scope than EUS in the assessment of pelvic structures. Pelvic MRI should be performed according to a dedicated protocol to assess the rectum, mesorectum, and mesorectal fascia and the presence of regional lymph node metastases.

MRI is more sensitive and specific than CT in detecting metastases to the liver, pelvic lymph nodes, bones, and the CNS.

It is also recommended that MRI of the abdominal cavity be performed alongside chest CT in order to obtain a comprehensive assessment of possible distant metastases, especially small lesions in the liver. If liver metastases can be detected only by MRI, MRI should be used to assess their presence and continued as the main follow-up examination [46].

In patients with liver metastases, additional MRI of the spine or MRI of the whole body may be performed. Whole body MRI is less sensitive in assessing potential metastasis, so segmented MRI is preferred. After the identification of liver metastases, extended MRI is used to identify other potential distant metastases, especially to bone or peritoneal implants. In the case of the latter, the preferred examination is CT with i.v. administration of a contrast agent, without filling the loops of the intestine with the contrast agent. In every case, a neutral contrast medium is administered orally (water). The sensitivity of MRI in the detection of focal (metastatic) lesions seems to be similar to that of positron emission tomography/computed tomography (PET/CT) with the use of [68Ga]Ga-DOTATATE/TOC (91% vs. 92%), except for the liver, for which MRI sensitivity is higher, 99% vs. 92% and bone 96% vs. 82%, respectively [49].

CNS MRI is recommended in patients with symptoms suggesting metastases to the CNS, which, however, are rare even in gastroenteropancreatic neuroendocrine carcinomas (GEP-NECs) (in 4% of patients) [50]. The exception is advanced RNEN, in which metastases to the CNS should be ruled out each time, in almost every patient.

There is currently no consensus on the need for systematic performance of MRI of the CNS at first diagnosis in patients with advanced CNEC and distant metastases [51].

Minimal consensus statement on CT and MRI examinations
1. Multiphase CT examination performed to detect metastatic lesions to regional lymph nodes and distant metastases as well as in the assessment of NEN infiltration of the neighbouring organs [III, 2B]#.
2. MRI is more sensitive and specific than CT in detecting metastases to the liver, pelvic lymph nodes, bones, and the CNS [III, 2B]#.
3.3.3. Radioisotope diagnostics

Somatostatin receptor imaging (SRI) is performed by means of scintigraphy (planar, single photon emission computed tomography (SPECT) or single photon emission computed tomography/computed tomography (SPECT/CT), or PET/CT.

PET/CT imaging with [68Ga]Ga-DOTA-SSA (somatostatin analogue) demonstrates higher sensitivity than scintigraphic examination and should be performed in cases of doubtful or negative results of scintigraphy as a conclusive examination, especially in the case of lesions < 1 cm in size [52, 53]

To assess the stage of advancement, SRI may be considered, especially in patients with T2 or higher clinical progression. In the case of radical surgery, there are no indications for patient follow-up using SRI.

SRI should be performed in all patients qualified for treatment with somatostatin analogues (SSAs) and peptide receptor radionuclide therapy (PRRT) [54].

Additionally, in the case of negative [68Ga]Ga-DOTA-SSA PET/CT and rapidly growing NETs and NECs, 2-deoxy-2-[fluorine-18]fluoro-D-glucose ([18F]FDG) PET/CT is indicated.

High accumulation of [18F] FDG is considered a hallmark of malignant neoplasms with high metabolism and proliferation. It correlates with higher stage of advancement and is an independent negative prognostic factor [53, 55, 56].

Minimal consensus statement of radioisotope examinations
1. SRI is required to qualify patients for antiproliferative treatment with SSA and PRRT [III, 2B]#.
2. [18F]FDG PET/CT is indicated in patients with NEC [III, 2B]#.

4. Treatment

4.1. Surgical treatment of CNENs

Recommendations for the surgical treatment of CNENs are analogous to the recommendations for the treatment of colon adenocarcinoma [8]. It is recommended that a resection procedure (open or laparoscopic approach) with lymphadenectomy be performed in patients with tumours without distant metastases. In the case of CNENs G1 and G2 with distant metastases (usually to the liver), palliative resection with regional lymphadenectomy or, if possible, cytoreduction of the tumour are recommended, even if complete resection cannot be achieved.

In the case of infiltration of adjacent organs, if technically possible, multi-organ excision combined with left or right hemicolectomy or extension of the procedure according to the extent of the lymphatic drainage is also proposed [8].

4.1.1. Surgical treatment of RNENs

Most RNENs are detected early and can be treated endoscopically.

In the case of lesions that do not qualify for endoscopic treatment, the method of choice is local full-thickness resection using transanal endoscopic microsurgery (TEM) or resection of the rectum with the mesorectum using a classic (open) approach or with the use of minimally invasive techniques. The recommended treatment algorithm for rectal neuroendocrine neoplasms is presented in Figure 1 [8].

191495.png
Figure 1. Modified algorithm for the treatment of rectal neuroendocrine neoplasms (RNENs) in individual cases [8]; NEN neuroendocrine neoplasm; TEM transanal endoscopic microsurgery; *look at Figure 2

TEM is mainly used in so-called salvage therapy [57–60]. This treatment is used in the event of failure to achieve radical resection confirmed in pathomorphological examination or uncertainty regarding radical resection following an endoscopic procedure or in the case of local recurrence. Recent data show that endoscopic full-thickness resection (EFTR) is as effective as TEM in these patients [61].

In tumours larger than 2 cm in diameter (and according to some authors even > 1.5 cm), the muscularis mucosa is frequently infiltrated. In these cases, resection is recommended, preserving the sphincters if possible. The procedure of choice is anterior rectal resection with total mesorectal excision (TME) and the possible creation of a protective stoma [8, 62].

Radical resection is also recommended in the presence of other risk factors, including metastases to regional lymph nodes, such as the following: high mitotic index of the tumour (G2, G3, Ki-67 > 2%), infiltration of lymphatic and blood vessels, and positive HES77 expression [23, 63].

In the case of T3 and T4 tumours with local lymph node involvement, it is possible to achieve oncological radicality if there are no distant metastases.

If the tumour is located in the lower part or infiltrates the sphincters, it is advisable to perform abdomino-sacral or abdomino-perineal amputation.

In special situations, especially when the patient does not consent to radical surgery, the optimal solution is to use the TEM technique. However, the patient should be informed that radical rectal resection may be necessary in the event of an unfavourable histopathological examination result of the tumour resected using TEM.

One work has questioned the advantage of radical resection over local excision in the case of RNENs of 1020 mm in diameter with and without lymph node involvement, because radical excision may have a negative impact on the quality of life [64].

In locally and systemically advanced tumours with distant metastases, radical resection surgeries are not recommended because in this group the survival is 69 months from diagnosis [9].

The indication for palliative surgery is tumour bleeding with ineffective local haemostasis or bowel obstruction. In RNETs G1 and G2 with metastases confined to the liver, local tumour excision followed by liver parenchyma resection (metastasectomy) may be considered or, in selected cases, liver transplantation [65].

The minimal consensus statement on surgical treatment
1. CNENs, in most cases, require surgery [IV, 2A]#.
2. RNENs with unfavourable prognostic factors (regardless of size) and tumours 2 cm in diameter or larger require surgical treatment [IV, 2A]#.
4.2. Endoscopic treatment of CRNENs

Patients with negligible risk of metastasis are eligible for endoscopic treatment of CRNEN. Surgical treatment is recommended in CNEN, even in the case of small/early diagnosed lesions, due to the high risk of metastases [66, 67]. In contrast, most RNENs can be treated endoscopically. At diagnosis, most of these lesions are up to 1 cm in size, which is associated with a very good prognosis [68, 69].

According to the latest reports, the highest R0 resection rates (over 90%) are provided by three main groups of endoscopic methods: modified endoscopic mucosal resection (mEMR), endoscopic submucosal dissection (ESD), and EFTR [61, 70–80]. RNEN removal cannot be performed using classic polypectomy or biopsy forceps due to the very low rate of R0 resections (approximately 30%). Classic endoscopic mucosal resection (EMR), consisting only of injecting fluid under the lesion followed by excision with a standard diathermy loop, should not be performed either, because comparative studies have shown that the modified EMR (mEMR) allows for significantly better results [81].

In rescue therapy (incomplete/uncertain radicality), ESD or EFTR is recommended with the optimal procedure being full-thickness resection.

Indications for endoscopic treatment of RNENs should primarily take into account the size, stage (T and N), and proliferation index of the tumour (G). The optimal situation assumed by most guidelines is that the physician performing the colonoscopy correctly recognises the neuroendocrine lesion and refers the patient for EUS examination, and then for optimal treatment. However, in clinical practice, the correct diagnosis of RNENs at the time of the first endoscopy concerns only 1830% of patients. Most lesions are removed incorrectly and non-radically, using a diathermy loop or biopsy forceps the R0 resection rate is 2430% [24, 82]. It may also occur that the correct endoscopic method of NEN treatment is used but R0 resection is not achieved.

In the case of RNENs of 5 mm in size and above, EUS should be performed before the planned endoscopic treatment, with assessment of the depth of infiltration and the condition of regional lymph nodes.

Patients with RNEN lesions up to 10 mm in size without risk factors are eligible for endoscopic treatment [74, 75]. The risk factors are as follows: infiltration of the muscular membrane proper, involvement of regional lymph nodes, infiltration of lymphatic and/or blood vessels, and a proliferation index above 2%. In the case of lesions of 1020 mm in diameter, without muscularis propria infiltration and without metastases to regional lymph nodes, the tumour can be removed endoscopically, and after histopathological evaluation it is possible to decide on further treatment, taking into account (as well as the pathomorphological examination result) the patient’s age, general condition, and comorbidities. RNENs greater than 20 mm in size, all NENs G3, and lesions with markers of poor prognosis should undergo radical surgical treatment.

The scale developed by Chen et al. [41] may be useful in the initial endoscopic assessment of RNEN prognosis (Tab. 4).

One of the recommended methods of endoscopic treatment of RNENs is the above-mentioned ESD [75–78]. Considering the fact that ESD is a technically difficult and time-consuming procedure, recent reports on the very good effectiveness of the modified EMR (mEMR) method, achieving R0 resection in over 93% of cases, facilitate the use of this easier procedure, especially in the case of lesions with a diameter of up to 10 mm. There are three main types of mEMR: cap-assisted endoscopic mucosal resection (EMR-C), endoscopic mucosal resection with ligation (EMR-L), and endoscopic mucosal resection with precut of the mucosa around the lesion (EMR-precut) [70–74].

The highest R0 resection rate was achieved using EMR-L.

According to the latest publications, EFTR may become the method of choice in the treatment of RNEN due to its high efficiency (100% of R0 resection) and its safety [61, 79, 80].

In a situation where the rectal neuroendocrine lesion has been removed endoscopically and the histopathologist identifies the lack of radicality or cannot confirm the radicality following resection, the patient should be referred for a “rescue” procedure. A rescue endoscopic procedure is possible if there is a visible scar after NEN removal.

The rescue endoscopic procedures include ESD and EFTR. The use of EFTR provides a higher rate of R0 resection (100%) than ESD (90%) [61]. The effectiveness of EFTR is comparable to that of TEM, with a shorter procedure time (18 minutes on average), without the need for general anaesthesia or the use of an operating room [79]. It is worth emphasising that even in the case of small (up to 5 mm) RNENs that had been incorrectly removed with a diathermy loop, neoplastic infiltration was found in the material obtained during rescue treatment in more than 20% of patients [83]. The RNEN endoscopic treatment algorithm is presented in Figure 2 [84].

191507.png
Figure 2. Algorithm for the endoscopic treatment of rectal neuroendocrine neoplasm (RNEN) [84]. EUS endoscopic ultrasound; mEMR modified endoscopic mucosal resection; EMR-L endoscopic mucosal resection with ligation; EMR-C cap-assisted endoscopic mucosal resection; EMR-precut endoscopic mucosal resection with precut of the mucosa around the lesion; ESD endoscopic submucosal dissection; EFTR endoscopic full-thickness resection; TEM transanal endoscopic microsurgery. *Invasion of the muscularis propria, blood, or lymph vessels; **individual decision based on the patient’s condition (age, comorbidities) and preferences

Minimal consensus statement on endoscopic treatment
1. Endoscopic treatment of CRNEN concerns only rectal lesions [IV, 2A]#.
2. RNENs up to 1 cm in diameter, without invasion of the muscularis propria, and without metastases to regional lymph nodes identified in the EUS constitute the optimal indication for endoscopic treatment [II, 1]#.
3. RNENs of 12 cm in diameter can be removed endoscopically, and after pathomorphological assessment (risk factors) it is possible to decide on further treatment [IV, 2A]#.
4. mEMR, ESD, and EFTR are the preferred methods [IV, 2A]#.
5. EFTR is the most effective endoscopic rescue procedure in the case of the lack of radicality or unconfirmed radicality of endoscopic treatment [IV, 2A]#.
4.3. Pharmacological treatment
4.3.1. Biotherapy
Somatostatin analogues

SSAs are used in NENs of the ascending colon. The CLARINET study demonstrated the antiproliferative activity of SSAs in all non-functional, highly or moderately differentiated GEP-NENs (Ki-67 < 10%). Hence, SSAs are the first-line treatment in NETs G1 and G2 [85, 86].

Both octreotide and lanreotide are effective in controlling the hormonal symptoms associated with NETs and inhibiting tumour growth. In clinical trials of the use of SSAs in the treatment of tumours located in the hindgut, there has been little evidence of their efficacy, among others, due to the size of the study group being too small. However, in the case of lesions with exposed somatostatin receptors, the use of SSA should be considered.

Targeted therapy — m-TOR inhibitors (everolimus)

Analysis of the subgroup of patients with advanced functional CRNETs (G1/G2) in the RADIANT-2 study demonstrated that the combination of everolimus and octreotide resulted in a significant increase in progression-free survival [87]. Additionally, the data from the RADIANT-4 study, including patients with non-functional CRNETs G1 and G2, confirmed the effectiveness of m-TOR inhibitors in this group of patients [88].

The m-TOR inhibitors are effective in the treatment of unresectable, locally advanced, and/or metastatic neoplasms that demonstrate progression during active follow-up or SSA therapy, and are recommended by scientific societies. The drug is approved in G1/G2 neoplasms, but currently it is not reimbursed in this indication [89].

4.3.2. Chemotherapy and immunotherapy

The importance of chemotherapy (ChT) in neoplasms with well- and intermediate grade of differentiation (G1/G2) is limited, and its application should apply to neoplasms resistant to other forms of systemic treatment, radioisotope therapy, or radiotherapy (NET G2 with Ki-67 >10 %, aggressive progression according to RECIST within 36 months, or without SSTR expression).

ChT is primarily indicated in the treatment of patients diagnosed with NEC, as well as those who show features of NET G3 progression. In the case of NECs, the use of platinum derivatives should be standard practice [89].

As well as the combination of cisplatin and etoposide (important especially in small cell neoplasms with high Ki-67), the use of oxaliplatin (FOLFOX, XELOX) or irinotecan (FOLFIRI, IP) regimens can be considered [90–94]. Temozolomide- and/or capecitabine-based regimens are justified especially in patients with NETs G2/G3 [95, 96].

In some patients with neuroendocrine colorectal neoplasms with a higher degree of biological aggressiveness (including NEC, NET G3), DNA repair disorders, including microsatellite instability (MSI), are identified [97, 98]. Despite many reports on the possibility of using PD-1 inhibitors in this population and the site-agnostic approval of pembrolizumab in this indication, it is argued that the effectiveness of therapy is not equal in every organ location. Considering that the large intestine location is associated with potentially higher efficacy in the treatment of adenocarcinomas, the MSI assessment is indicated in situations where it is necessary to seek effective systemic treatment of advanced disease. Due to the approval of pembrolizumab as monotherapy for metastatic colorectal cancer with high microsatellite instability (MSI-H) or deficient DNA mismatch repair (dMMR), with very limited data in the population of patients with neuroendocrine neoplasms, consideration of this therapy must include a comprehensive patient assessment and evaluation of the tumour’s histoclinical and molecular features. The presence of the MSI-H phenotype in patients after radical treatment may indicate a potentially better prognosis and speak against the considered NEC/NET G3 adjuvant treatment.

Minimal consensus statement on pharmacological treatment:
1. In patients with CRNET with symptoms of carcinoid syndrome, SSAs are the drugs of choice [II, 2A]#.
2. In patients with inoperable, locally advanced, and/or metastatic CRNETs G1/G2 after assessing the disease dynamics, everolimus (currently without direct reimbursement in Poland), PRRT, or independent use of SSA (off-label treatment) should be considered. In patients with Ki-67 > 10% or in the case of further dynamic progression, an attempt at chemotherapy with temozolomide and/or capecitabine is justified [IV, 2A]#.
3. In patients with NEC or patients with dynamic disease progression, chemotherapy with platinum derivatives is the treatment of choice [II, 2A]#.
4.4. Radioisotope therapy — PRRT

There are no prospective randomised clinical trials in patients with CRNENs. Few reports come from retrospective studies [99–102]. The observed survival parameters in a Polish multicentre study in this group of patients amounted to PFS 40.6 months and OS 131.2 months and were longer than those previously observed in the literature [100–104].

Qualification for treatment takes place according to the principles described in the Update of the diagnostic and therapeutic guidelines for gastro-entero-pancreatic neuroendocrine neoplasms (recommended by the Polish Network of Neuroendocrine Tumours) [27].

Minimal consensus statement on radioisotope therapy
1. PRRT should be considered in patients with disseminated or unresectable NETs G1/G2 with increased expression of somatostatin receptors confirmed in SRI [III, 2B]#.
4.5. Treatment monitoring

An endoscopic and imaging follow-up after R0 resection in patients with RNENs is presented in Table 5 [105]. Serum CgA provides additional information: if the elevated concentration observed before tumour resection normalises after surgery and then increases, it may suggest NET recurrence. According to the SEER Norwegian Register Cancer database, RNENs are associated with the best prognosis of all gastrointestinal tumours, with a five-year survival rate of 7488% [106]. GEP-NENs are typically slow-growing tumours, so tumour marker determination should be performed infrequently, i.e. at intervals ranging from 4 to 12 months; and after radical surgery, every 6 months up to 2 years [20]. According to 2020 NANETS GUIDELINES, monitoring of CgA levels and 5-HIAA excretion should be considered if these results were abnormal at the onset of the disease [107].

Table 5. Rectal neuroendocrine neoplasm (RNEN) follow-up after R0 resection [105]

Tumour size

Grading

Examination

Incidence

Duration

< 1 cm

1 or 2

No need for follow-up

< 1 cm

3

Colonoscopy

Annual

For 5 years

1–2 cm

Regardless of grade

Colonoscopy

EUS

MRI

After 12 months then every 5 months

After 12 months

After 12 months

For 5 years

> 2 cm

1 or 2

Colonoscopy

EUS

MRI

Every year

Every year

Every year

For 5 years

For 5 years

For 5 years

> 2 cm

3

Colonoscopy

EUS

MRI

Every 4–6 months for a year, then every year

For 5 years

For 5 years

For 5 years

Evidence quality and the strength of recommendations

#Evidence quality and the strength of recommendations has been established on the basis of the following tables according to ESMO and NCCN [108, 109] (Supplementary File — Tab. S1 and S2).

Conflict of interest

For the Conflict of Interest Statement, please see the Supplementary File.

Author contributions

All authors contributed to the idea, gathered the information, interpreted the data, and wrote and accepted the final version of the manuscript.

References

  1. Regula J, Rupinski M, Kraszewska E, et al. Colonoscopy in colorectal-cancer screening for detection of advanced neoplasia. N Engl J Med. 2006; 355(18): 18631872, doi: 10.1056/NEJMoa054967, indexed in Pubmed: 17079760.
  2. Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003; 97(4): 934959, doi: 10.1002/cncr.11105, indexed in Pubmed: 12569593.
  3. Modlin IM, Kidd M, Latich I, et al. Current status of gastrointestinal carcinoids. Gastroenterology. 2005; 128(6): 17171751, doi: 10.1053/j.gastro.2005.03.038, indexed in Pubmed: 15887161.
  4. Tichansky DS, Cagir B, Borrazzo E, et al. Risk of second cancers in patients with colorectal carcinoids. Dis Colon Rectum. 2002; 45(1): 9197, doi: 10.1007/s10350-004-6119-y, indexed in Pubmed: 11786770.
  5. Bogacka B, Marlicz W, Białek A, et al. Trends in colorectal neuroendocrine tumors: A 10 years review. Gut. 2009; 58: A296.
  6. Kamiński MF, Polkowski M, Reguła J, et al. Prevalence and endoscopic features of rectal neuro-endocrine tumours among 50 148 participants of the Polish Colorectal-Cancer Screening Pro-gramme. Gut. 2007; 56: A310.
  7. Caplin M, Sundin A, Nillson O, et al. Barcelona Consensus Conference participants. ENETS Consensus Guidelines for the management of patients with digestive neuroendocrine neoplasms: colorectal neuroendocrine neoplasms. Neuroendocrinology. 2012; 95(2): 8897, doi: 10.1159/000335594, indexed in Pubmed: 22261972.
  8. Starzyńska T, Londzin-Olesik M, Bałdys-Waligórska A, et al. Colorectal neuroendocrine neoplasms management guidelines (recommended by the Polish Network of Neuroendocrine Tumours). Endokrynol Pol. 2017; 68(2): 250260, doi: 10.5603/EP.2017.0019, indexed in Pubmed: 28540975.
  9. Ramage JK, De Herder WW, Delle Fave G, et al. Vienna Consensus Conference participants. ENETS Consensus Guidelines Update for Colorectal Neuroendocrine Neoplasms. Neuroendocrinology. 2016; 103(2): 139143, doi: 10.1159/000443166, indexed in Pubmed: 26730835.
  10. Zhang Yu, Chen HY, Zhou XL, et al. Diagnostic efficacy of the Japan Narrow-band-imaging Expert Team and Pit pattern classifications for colorectal lesions: A meta-analysis. World J Gastroenterol. 2020; 26(40): 62796294, doi: 10.3748/wjg.v26.i40.6279, indexed in Pubmed: 33177800.
  11. Bellizzi AM. Pathologic Considerations in Gastroenteropancreatic Neuroendocrine Tumors. Surg Oncol Clin N Am. 2020; 29(2): 185208, doi: 10.1016/j.soc.2019.11.003, indexed in Pubmed: 32151355.
  12. Federspiel B, Burke A, Sobin L, et al. Rectal and colonic carcinoids. A clinicopathologic study of 84 cases. Cancer. 1990; 65(1): 135140, doi: 10.1002/1097-0142(19900101)65:1<135::aid-cncr2820650127>3.0.co;2-a, indexed in Pubmed: 2293859.
  13. Xu Z, Wang Li, Dai S, et al. Epidemiologic Trends of and Factors Associated With Overall Survival for Patients With Gastroenteropancreatic Neuroendocrine Tumors in the United States. JAMA Netw Open. 2021; 4(9): e2124750, doi: 10.1001/jamanetworkopen.2021.24750, indexed in Pubmed: 34554237.
  14. Rosenberg J, Welch J. Carcinoid tumors of the colon. Am J Surg. 1985; 149(6): 775779, doi: 10.1016/s0002-9610(85)80184-7, indexed in Pubmed: 2409828.
  15. Anthony LB, Strosberg JR, Klimstra DS, et al. North American Neuroendocrine Tumor Society (NANETS). The NANETS consensus guidelines for the diagnosis and management of gastrointestinal neuroendocrine tumors (nets): well-differentiated nets of the distal colon and rectum. Pancreas. 2010; 39(6): 767774, doi: 10.1097/MPA.0b013e3181ec1261, indexed in Pubmed: 20664474.
  16. Soga J. Early-stage carcinoids of the gastrointestinal tract: an analysis of 1914 reported cases. Cancer. 2005; 103(8): 15871595, doi: 10.1002/cncr.20939, indexed in Pubmed: 15742328.
  17. Yoon SN, Yu CS, Shin UiS, et al. Clinicopathological characteristics of rectal carcinoids. Int J Colorectal Dis. 2010; 25(9): 10871092, doi: 10.1007/s00384-010-0949-y, indexed in Pubmed: 20397020.
  18. 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(18): 30633072, doi: 10.1200/JCO.2007.15.4377, indexed in Pubmed: 18565894.
  19. Ahmed M. Gastrointestinal neuroendocrine tumors in 2020. World J Gastrointest Oncol. 2020; 12(8): 791807, doi: 10.4251/wjgo.v12.i8.791, indexed in Pubmed: 32879660.
  20. Cives M, Strosberg JR. Gastroenteropancreatic Neuroendocrine Tumors. CA Cancer J Clin. 2018; 68(6): 471487, doi: 10.3322/caac.21493, indexed in Pubmed: 30295930.
  21. Modlin I, Sandor A. An analysis of 8305 cases of carcinoid tumors. Cancer. 2000; 79(4): 813829, doi: 10.1002/(sici)1097-0142(19970215)79:4<813::aid-cncr19>3.0.co;2-2, indexed in Pubmed: 9024720.
  22. Jetmore AB, Ray JE, Gathright JB, et al. Rectal carcinoids: the most frequent carcinoid tumor. Dis Colon Rectum. 1992; 35(8): 717725, doi: 10.1007/BF02050318, indexed in Pubmed: 1643994.
  23. Gleeson FC, Levy MJ, Dozois EJ, et al. Endoscopically identified well-differentiated rectal carcinoid tumors: impact of tumor size on the natural history and outcomes. Gastrointest Endosc. 2014; 80(1): 144151, doi: 10.1016/j.gie.2013.11.031, indexed in Pubmed: 24462168.
  24. Fine C, Roquin G, Terrebonne E, et al. Endoscopic management of 345 small rectal neuroendocrine tumours: A national study from the French group of endocrine tumours (GTE). United European Gastroenterol J. 2019; 7(8): 11021112, doi: 10.1177/2050640619861883, indexed in Pubmed: 31662867.
  25. Mełeń-Mucha G, Mucha S, Komorowski J. Early detection of gastric GIST tumor in a patient with rectal neuroendocrine cancer a case report. 8th Annual ENETS Conference for the Diag-nosis and Treatment of Neuroendocrine Tumor Disease, 911 March 2011, Lisbon, Portugal. Neuroendocrinology. 2011; 94(Suppl 1): 3637.
  26. Kölby L, Bernhardt P, Swärd C, et al. Chromogranin A as a determinant of midgut carcinoid tumour volume. Regul Pept. 2004; 120(1-3): 269273, doi: 10.1016/j.regpep.2004.03.017, indexed in Pubmed: 15177946.
  27. Kos-Kudła B, Foltyn W, Malczewska A, et al. Update of the diagnostic and therapeutic guidelines for gastro-entero-pancreatic neuroendocrine neoplasms (recommended by the Polish Network of Neuroendocrine Tumours). Endokrynol Pol. 2022; 73(3): 387423; doi: 10.5603/EP.a2022.0049.
  28. Klöppel G, Couvelard A, Hruban RH. et al. Tumours of the endocrine pancreas. In: Lloyd RV, Hruban RH, Osamura RY, Klöppel G, Rosai J, eds WHO. ed. Classification of the Tumours of Endocrine Organs. 4th ed. International Agency for Research on Cancer, Lyon 2017: 175207.
  29. Heetfeld M, Chougnet CN, Olsen IH, et al. other Knowledge Network members. Characteristics and treatment of patients with G3 gastroenteropancreatic neuroendocrine neoplasms. Endocr Relat Cancer. 2015; 22(4): 657664, doi: 10.1530/ERC-15-0119, indexed in Pubmed: 26113608.
  30. Fazio N, Milione M. Heterogeneity of grade 3 gastroenteropancreatic neuroendocrine carcinomas: New insights and treatment implications. Cancer Treat Rev. 2016; 50: 6167, doi: 10.1016/j.ctrv.2016.08.006, indexed in Pubmed: 27636009.
  31. Rindi G, Klimstra DS, Abedi-Ardekani B, et al. A common classification framework for neuroendocrine neoplasms: an International Agency for Research on Cancer (IARC) and World Health Organization (WHO) expert consensus proposal. Mod Pathol. 2018; 31(12): 17701786, doi: 10.1038/s41379-018-0110-y, indexed in Pubmed: 30140036.
  32. WHO Classification of Tumours Editorial Boards. Digestive system tumours. In: WHO classification of tumours series. 5th ed. International Agency for Research on Cancer, Lyon 2019.
  33. Brierley J, Gospodarowicz MK, Wittekind Ch. TNM classification of malignat tumours. 8th ed. Wiley Blackwell, Oxford 2017: Wiley.
  34. Woltering EA, Bergsland EK, Beyer DT. et al. Neuroendocrine tumors of the jejunum and ileum. American Joint Committee on Cancer 2017. In: Amin MB. ed. ed. AJCC Cancer Staging Manual. 8th ed. Springer, New York 2017: 375387.
  35. Wittekind C, Compton C, Quirke P, et al. A uniform residual tumor (R) classification: integration of the R classification and the circumferential margin status. Cancer. 2009; 115(15): 34833488, doi: 10.1002/cncr.24320, indexed in Pubmed: 19536900.
  36. Lin CK, Chung CS, Huang WC. Rectal carcinoid tumour observed by magnifying colonoscopy with narrow band imaging. Dig Liver Dis. 2014; 46(7): e7, doi: 10.1016/j.dld.2013.12.003, indexed in Pubmed: 24411486.
  37. Mori Y, Kudo SE, Misawa M, et al. Simultaneous detection and characterization of diminutive polyps with the use of artificial intelligence during colonoscopy. VideoGIE. 2019; 4(1): 710, doi: 10.1016/j.vgie.2018.10.006, indexed in Pubmed: 30623149.
  38. Mori Y, Kudo SE, East JE, et al. Cost savings in colonoscopy with artificial intelligence-aided polyp diagnosis: an add-on analysis of a clinical trial (with video). Gastrointest Endosc. 2020; 92(4): 905911.e1, doi: 10.1016/j.gie.2020.03.3759, indexed in Pubmed: 32240683.
  39. Hosoe N, Limpias Kamiya KJL, Hayashi Y, et al. Current status of colon capsule endoscopy. Dig Endosc. 2021; 33(4): 529537, doi: 10.1111/den.13769, indexed in Pubmed: 32542702.
  40. Hawes RH, Fockens P, Varadarajulu S. Endosonography. Saunders Elsevier 2015.
  41. Chen L, Guo Yu, Zhang Y, et al. Development of a novel scoring system based on endoscopic appearance for management of rectal neuroendocrine tumors. Endoscopy. 2021; 53(7): 702709, doi: 10.1055/a-1274-0161, indexed in Pubmed: 32992347.
  42. Fu KI, Mashimo Y, Matsuda T, et al. Is endoscopic ultrasonography necessary for depth evaluation of rectal carcinoid tumors 10.1007/s10350-006-0589-z, indexed in Pubmed: 16752204.
  43. Gu Q, Lin YM, Cen Li, et al. Endoscopic ultrasonography is useful in the diagnosis and treatment of rectal neuroendocrine neoplasms: a case series. J Zhejiang Univ Sci B. 2019; 20(10): 861864, doi: 10.1631/jzus.B1900168, indexed in Pubmed: 31489805.
  44. Hamada Y, Tanaka K, Hattori A, et al. Clinical utility of endoscopic submucosal dissection using the pocket-creation method with a HookKnife and preoperative evaluation by endoscopic ultrasonography for the treatment of rectal neuroendocrine tumors. Surg Endosc. 2022; 36(1): 375384, doi: 10.1007/s00464-021-08292-6, indexed in Pubmed: 33492506.
  45. Stier MW, Chapman CG, Shamah S, et al. Endoscopic resection is more effective than biopsy or EUS to detect residual rectal neuroendocrine tumor. Endosc Int Open. 2021; 9(1): E4E8, doi: 10.1055/a-1300-1017, indexed in Pubmed: 33403229.
  46. Sundin A, Arnold R, Baudin E, et al. Antibes Consensus Conference participants. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Tumors: Radiological, Nuclear Medicine & Hybrid Imaging. Neuroendocrinology. 2017; 105(3): 212244, doi: 10.1159/000471879, indexed in Pubmed: 28355596.
  47. Ronot M, Cuccioli F, Dioguardi Burgio M, et al. Neuroendocrine liver metastases: Vascular patterns on triple-phase MDCT are indicative of primary tumour location. Eur J Radiol. 2017; 89: 156162, doi: 10.1016/j.ejrad.2017.02.007, indexed in Pubmed: 28267533.
  48. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009; 45(2): 228247, doi: 10.1016/j.ejca.2008.10.026, indexed in Pubmed: 19097774.
  49. Schraml C, Schwenzer NF, Sperling O, et al. Staging of neuroendocrine tumours: comparison of [⁶⁸Ga]DOTATOC multiphase PET/CT and whole-body MRI. Cancer Imaging. 2013; 13: 6372, doi: 10.1102/1470-7330.2013.0007, indexed in Pubmed: 23466785.
  50. Sorbye H, Welin S, Langer SW, et al. Predictive and prognostic factors for treatment and survival in 305 patients with advanced gastrointestinal neuroendocrine carcinoma (WHO G3): the NORDIC NEC study. Ann Oncol. 2013; 24(1): 152160, doi: 10.1093/annonc/mds276, indexed in Pubmed: 22967994.
  51. Garcia-Carbonero R, Sorbye H, Baudin E, et al. Vienna Consensus Conference participants. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology. 2016; 103(2): 186194, doi: 10.1159/000443172, indexed in Pubmed: 26731334.
  52. Kunikowska J, Lewington V, Krolicki L. Optimizing Somatostatin Receptor Imaging in Patients With Neuroendocrine Tumors: The Impact of 99mTc-HYNICTOC SPECT/SPECT/CT Versus 68Ga-DOTATATE PET/CT Upon Clinical Management. Clin Nucl Med. 2017; 42(12): 905911, doi: 10.1097/RLU.0000000000001877, indexed in Pubmed: 29076910.
  53. Ambrosini V, Kunikowska J, Baudin E, et al. Consensus on molecular imaging and theranostics in neuroendocrine neoplasms. Eur J Cancer. 2021; 146: 5673, doi: 10.1016/j.ejca.2021.01.008, indexed in Pubmed: 33588146.
  54. Hicks RJ, Kwekkeboom DJ, Krenning E, et al. Antibes Consensus Conference participants. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasia: Peptide Receptor Radionuclide Therapy with Radiolabeled Somatostatin Analogues. Neuroendocrinology. 2017; 105(3): 295309, doi: 10.1159/000475526, indexed in Pubmed: 28402980.
  55. Binderup T, Knigge U, Loft A, et al. 18F-fluorodeoxyglucose positron emission tomography predicts survival of patients with neuroendocrine tumors. Clin Cancer Res. 2010; 16(3): 978985, doi: 10.1158/1078-0432.CCR-09-1759, indexed in Pubmed: 20103666.
  56. Bahri H, Laurence L, Edeline J, et al. High prognostic value of 18F-FDG PET for metastatic gastroenteropancreatic neuroendocrine tumors: a long-term evaluation. J Nucl Med. 2014; 55(11): 17861790, doi: 10.2967/jnumed.114.144386, indexed in Pubmed: 25286923.
  57. Dąbkowski K, Szczepkowski M, Kos-Kudła B, et al. Endoscopic management of rectal neuroendocrine tumours. How to avoid a mistake and what to do when one is made? Endokrynol Pol. 2020; 71(4): 343349, doi: 10.5603/EP.a2020.0045, indexed in Pubmed: 32852049.
  58. Chen WJ, Wu N, Zhou JL, et al. Full-thickness excision using transanal endoscopic microsurgery for treatment of rectal neuroendocrine tumors. World J Gastroenterol. 2015; 21(30): 91429149, doi: 10.3748/wjg.v21.i30.9142, indexed in Pubmed: 26290641.
  59. Shao Q, Lin G, Qiu H. [Transanal endoscopic microsurgery for treatment of rectal neuroendocrine tumors]. Zhonghua Wei Chang Wai Ke Za Zhi. 2017; 20(9): 10091014, indexed in Pubmed: 28900991.
  60. Kumar AS, Sidani SM, Kolli K, et al. Transanal endoscopic microsurgery for rectal carcinoids: the largest reported United States experience. Colorectal Dis. 2012; 14(5): 562566, doi: 10.1111/j.1463-1318.2011.02726.x, indexed in Pubmed: 21831099.
  61. Meier B, Albrecht H, Wiedbrauck T, et al. Full-thickness resection of neuroendocrine tumors in the rectum. Endoscopy. 2020; 52(1): 6872, doi: 10.1055/a-1008-9077, indexed in Pubmed: 31614372.
  62. Szczepkowski M, Banasiewicz T, Krokowicz P, et al. et al.. Polski konsensus w sprawie stomii protekcyjnej. Przeg Chir. 2014; 86: 717741.
  63. Jernman J, Hagström J, Mäenpää H, et al. Expression of Stem Cell-associated Marker HES77 in Rectal Neuroendocrine Tumors. Anticancer Res. 2015; 35(7): 37673772, indexed in Pubmed: 26124320.
  64. Al-Jiffry BO, Al-Malki O. Neuroendocrine small cell rectal cancer metastasizing to the liver: a unique treatment strategy, case report, and review of the literature. World J Surg Oncol. 2013; 11: 153, doi: 10.1186/1477-7819-11-153, indexed in Pubmed: 23844568.
  65. Ringe B, Lorf T, Döpkens K, et al. Treatment of hepatic metastases from gastroenteropancreatic neuroendocrine tumors: role of liver transplantation. World J Surg. 2001; 25(6): 697699, doi: 10.1007/s00268-001-0016-5, indexed in Pubmed: 11376400.
  66. Pavel M, O’Toole D, Costa F, et al. Vienna Consensus Conference participants. ENETS Consensus Guidelines Update for the Management of Distant Metastatic Disease of Intestinal, Pancreatic, Bronchial Neuroendocrine Neoplasms (NEN) and NEN of Unknown Primary Site. Neuroendocrinology. 2016; 103(2): 172185, doi: 10.1159/000443167, indexed in Pubmed: 26731013.
  67. Al Natour RH, Saund MS, Sanchez VM, et al. Tumor size and depth predict rate of lymph node metastasis in colon carcinoids and can be used to select patients for endoscopic resection. J Gastrointest Surg. 2012; 16(3): 595602, doi: 10.1007/s11605-011-1786-1, indexed in Pubmed: 22143420.
  68. Zhao B, Hollandsworth HM, Lopez NE, et al. Outcomes for a Large Cohort of Patients with Rectal Neuroendocrine Tumors: an Analysis of the National Cancer Database. J Gastrointest Surg. 2021; 25(2): 484491, doi: 10.1007/s11605-020-04525-6, indexed in Pubmed: 32016672.
  69. Ngamruengphong S, Kamal A, Akshintala V, et al. Prevalence of metastasis and survival of 788 patients with T1 rectal carcinoid tumors. Gastrointest Endosc. 2019; 89(3): 602606, doi: 10.1016/j.gie.2018.11.010, indexed in Pubmed: 30447216.
  70. Gu MG, Lee SiH. [Endoscopic Treatment Outcome of Rectal Neuroendocrine Tumors Removed by Ligation-assisted Endoscopic Submucosal Resection]. Korean J Gastroenterol. 2018; 72(3): 128134, doi: 10.4166/kjg.2018.72.3.128, indexed in Pubmed: 30270594.
  71. Lim HK, Lee SJ, Baek DH, et al. Resectability of Rectal Neuroendocrine Tumors Using Endoscopic Mucosal Resection with a Ligation Band Device and Endoscopic Submucosal Dissection. Gastroenterol Res Pract. 2019; 2019: 8425157, doi: 10.1155/2019/8425157, indexed in Pubmed: 31687016.
  72. Wang XY, Chai NL, Linghu EQ, et al. The outcomes of modified endoscopic mucosal resection and endoscopic submucosal dissection for the treatment of rectal neuroendocrine tumors and the value of endoscopic morphology classification in endoscopic resection. BMC Gastroenterol. 2020; 20(1): 200, doi: 10.1186/s12876-020-01340-w, indexed in Pubmed: 32586282.
  73. Lee J, Park YE, Choi JH, et al. Comparison between cap-assisted and ligation-assisted endoscopic mucosal resection for rectal neuroendocrine tumors. Ann Gastroenterol. 2020; 33(4): 385390, doi: 10.20524/aog.2020.0485, indexed in Pubmed: 32624659.
  74. So H, Yoo SuH, Han S, et al. Efficacy of Precut Endoscopic Mucosal Resection for Treatment of Rectal Neuroendocrine Tumors. Clin Endosc. 2017; 50(6): 585591, doi: 10.5946/ce.2017.039, indexed in Pubmed: 29020763.
  75. Wang XY, Chai NL, Linghu EQ, et al. Efficacy and safety of hybrid endoscopic submucosal dissection compared with endoscopic submucosal dissection for rectal neuroendocrine tumors and risk factors associated with incomplete endoscopic resection. Ann Transl Med. 2020; 8(6): 368, doi: 10.21037/atm.2020.02.25, indexed in Pubmed: 32355812.
  76. Suzuki S, Ishii N, Uemura M, et al. Endoscopic submucosal dissection (ESD) for gastrointestinal carcinoid tumors. Surg Endosc. 2012; 26(3): 759763, doi: 10.1007/s00464-011-1948-y, indexed in Pubmed: 21993939.
  77. Lee EJ, Lee JB, Lee SH, et al. Endoscopic submucosal dissection for colorectal tumors 1,000 colorectal ESD cases: one specialized institute’s experiences. Surg Endosc. 2013; 27(1): 3139, doi: 10.1007/s00464-012-2403-4, indexed in Pubmed: 22729707.
  78. Kim JiH, Baek IlH, Kim KOh, et al. Usefulness and feasibility of endoscopic submucosal dissection for colorectal tumor: a nationwide multicenter retrospective study in Korea. J Gastrointest Oncol. 2016; 7(6): 924930, doi: 10.21037/jgo.2016.06.08, indexed in Pubmed: 28078115.
  79. Brand M, Reimer S, Reibetanz J, et al. Endoscopic full thickness resection vs. transanal endoscopic microsurgery for local treatment of rectal neuroendocrine tumors - a retrospective analysis. Int J Colorectal Dis. 2021; 36(5): 971976, doi: 10.1007/s00384-020-03800-x, indexed in Pubmed: 33215239.
  80. Aepli P, Criblez D, Baumeler S, et al. Endoscopic full thickness resection (EFTR) of colorectal neoplasms with the Full Thickness Resection Device (FTRD): Clinical experience from two tertiary referral centers in Switzerland. United European Gastroenterol J. 2018; 6(3): 463470, doi: 10.1177/2050640617728001, indexed in Pubmed: 29774161.
  81. Kim J, Kim JH, Lee JY, et al. Clinical outcomes of endoscopic mucosal resection for rectal neuroendocrine tumor. BMC Gastroenterol. 2018; 18(1): 77, doi: 10.1186/s12876-018-0806-y, indexed in Pubmed: 29866049.
  82. Dąbkowski K, Rusiniak-Rossińska N, Michalska K, et al. Endoscopic treatment of rectal neuroendocrine tumors in a 13-year retrospective single-center study: are we following the guidelines? Pol Arch Intern Med. 2021; 131(3): 241248, doi: 10.20452/pamw.15823, indexed in Pubmed: 33620189.
  83. Pagano N, Ricci C, Brighi N, et al. Incidental diagnosis of very small rectal neuroendocrine neoplasms: when should endoscopic submucosal dissection be performed? A single ENETS centre experience. Endocrine. 2019; 65(1): 207212, doi: 10.1007/s12020-019-01907-y, indexed in Pubmed: 30919286.
  84. Frei R. ENOSWISS 2021.
  85. Caplin ME, Pavel M, Ćwikła JB, et al. CLARINET Investigators. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014; 371(3): 224233, doi: 10.1056/NEJMoa1316158, indexed in Pubmed: 25014687.
  86. Berardi R, Rinaldi S, Torniai M, et al. Gastrointestinal neuroendocrine tumors: Searching the optimal treatment strategy--A literature review. Crit Rev Oncol Hematol. 2016; 98: 264274, doi: 10.1016/j.critrevonc.2015.11.003, indexed in Pubmed: 26643525.
  87. Pavel M, Hainsworth J, 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(9808): 20052012, doi: 10.1016/s0140-6736(11)61742-x, indexed in Pubmed: 22119496.
  88. Yao J, 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(10022): 968977, doi: 10.1016/s0140-6736(15)00817-x, indexed in Pubmed: 26703889.
  89. Pavel M, Öberg K, Falconi M, et al. ESMO Guidelines Committee. Electronic address: clinicalguidelines@esmo.org. Gastroenteropancreatic neuroendocrine neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2020; 31(7): 844860, doi: 10.1016/j.annonc.2020.03.304, indexed in Pubmed: 32272208.
  90. Bajetta E, Catena L, Procopio G, et al. Are capecitabine and oxaliplatin (XELOX) suitable treatments for progressing low-grade and high-grade neuroendocrine tumours? Cancer Chemother Pharmacol. 2007; 59(5): 637642, doi: 10.1007/s00280-006-0306-6, indexed in Pubmed: 16937105.
  91. Hentic O, Hammel P, Couvelard A, et al. FOLFIRI regimen: an effective second-line chemotherapy after failure of etoposide-platinum combination in patients with neuroendocrine carcinomas grade 3. Endocr Relat Cancer. 2012; 19(6): 751757, doi: 10.1530/ERC-12-0002, indexed in Pubmed: 22940375.
  92. Welin S, Sorbye H, Sebjornsen S, et al. Clinical effect of temozolomide-based chemotherapy in poorly differentiated endocrine carcinoma after progression on first-line chemotherapy. Cancer. 2011; 117(20): 46174622, doi: 10.1002/cncr.26124, indexed in Pubmed: 21456005.
  93. Olsen IH, Sørensen JB, Federspiel B, et al. Temozolomide as second or third line treatment of patients with neuroendocrine carcinomas. ScientificWorldJournal. 2012; 2012: 170496, doi: 10.1100/2012/170496, indexed in Pubmed: 22973169.
  94. Koumarianou A, Kaltsas G, Kulke MH, et al. Temozolomide in Advanced Neuroendocrine Neoplasms: Pharmacological and Clinical Aspects. Neuroendocrinology. 2015; 101(4): 274288, doi: 10.1159/000430816, indexed in Pubmed: 25924937.
  95. Mitry E, Walter T, Baudin E, et al. Bevacizumab plus capecitabine in patients with progressive advanced well-differentiated neuroendocrine tumors of the gastro-intestinal (GI-NETs) tract (BETTER trial)--a phase II non-randomised trial. Eur J Cancer. 2014; 50(18): 31073115, doi: 10.1016/j.ejca.2014.10.001, indexed in Pubmed: 25454413.
  96. Sahu A, Jefford M, Lai-Kwon J, et al. CAPTEM in Metastatic Well-Differentiated Intermediate to High Grade Neuroendocrine Tumors: A Single Centre Experience. J Oncol. 2019; 2019: 9032753, doi: 10.1155/2019/9032753, indexed in Pubmed: 30915122.
  97. Luong TuV, Nisa Z, Watkins J, et al. Should immunohistochemical expression of mismatch repair (MMR) proteins and microsatellite instability (MSI) analysis be routinely performed for poorly differentiated colorectal neuroendocrine carcinomas? Endocrinol Diabetes Metab Case Rep. 2020 [Epub ahead of print]; 2020, doi: 10.1530/EDM-20-0058, indexed in Pubmed: 32729847.
  98. Fraune C, Simon R, Hube-Magg C, et al. Homogeneous MMR Deficiency Throughout the Entire Tumor Mass Occurs in a Subset of Colorectal Neuroendocrine Carcinomas. Endocr Pathol. 2020; 31(2): 182189, doi: 10.1007/s12022-020-09612-7, indexed in Pubmed: 32144630.
  99. Kong G, Grozinsky-Glasberg S, Hofman MS, et al. Highly favourable outcomes with peptide receptor radionuclide therapy (PRRT) for metastatic rectal neuroendocrine neoplasia (NEN). Eur J Nucl Med Mol Imaging. 2019; 46(3): 718727, doi: 10.1007/s00259-018-4196-8, indexed in Pubmed: 30343432.
  100. Kunikowska J, Pawlak D, Bąk MI, et al. Long-term results and tolerability of tandem peptide receptor radionuclide therapy with Y/Lu-DOTATATE in neuroendocrine tumors with respect to the primary location: a 10-year study. Ann Nucl Med. 2017; 31(5): 347356, doi: 10.1007/s12149-017-1163-6, indexed in Pubmed: 28316066.
  101. Kunikowska J, Zemczak A, Kołodziej M, et al. Tandem peptide receptor radionuclide therapy using Y/Lu-DOTATATE for neuroendocrine tumors efficacy and side-effects - polish multicenter experience. Eur J Nucl Med Mol Imaging. 2020; 47(4): 922933, doi: 10.1007/s00259-020-04690-5, indexed in Pubmed: 31980909.
  102. Zemczak A, Kołodziej M, Gut P, et al. Effect of peptide receptor radionuclide therapy (PRRT) with tandem isotopes - [90Y]Y/[177Lu]Lu-DOTATATE in patients with disseminated neuroendocrine tumours depending on [18F]FDG PET/CT qualification in Polish multicentre experience - do we need [18F]FDG PET/CT for qualification to PRRT? Endokrynol Pol. 2020; 71(3): 240248, doi: 10.5603/EP.a2020.0014, indexed in Pubmed: 32293704.
  103. Hörsch D, Ezziddin S, Haug A, et al. Effectiveness and side-effects of peptide receptor radionuclide therapy for neuroendocrine neoplasms in Germany: A multi-institutional registry study with prospective follow-up. Eur J Cancer. 2016; 58: 4151, doi: 10.1016/j.ejca.2016.01.009, indexed in Pubmed: 26943056.
  104. Baum RP, Kulkarni HR, Singh A, et al. Results and adverse events of personalized peptide receptor radionuclide therapy with Yttrium and Lutetium in 1048 patients with neuroendocrine neoplasms. Oncotarget. 2018; 9(24): 1693216950, doi: 10.18632/oncotarget.24524, indexed in Pubmed: 29682195.
  105. Garcia-Carbonero R, Sorbye H, Baudin E, et al. ENETS Consensus Guidelines for High-Grade Gastroenteropancreatic Neuroendocrine Tumors and Neuroendocrine Carcinomas. Neuroendocrinology. 2016; 103(2): 186194, doi: 10.1159/000443172, indexed in Pubmed: 26731334.
  106. Hauso O, Gustafsson BI, Kidd M, et al. Neuroendocrine tumor epidemiology: contrasting Norway and North America. Cancer. 2008; 113(10): 26552664, doi: 10.1002/cncr.23883, indexed in Pubmed: 18853416.
  107. Halfdanarson TR, Strosberg JR, Tang L, et al. The North American Neuroendocrine Tumor Society Consensus Guidelines for Surveillance and Medical Management of Pancreatic Neuroendocrine Tumors. Pancreas. 2020; 49(7): 863881, doi: 10.1097/MPA.0000000000001597, indexed in Pubmed: 32675783.
  108. European Society for Medical Oncology. Standard Operating Procedures (SOPs) for Authors and templates for ESMO Clinical Practice Guidelines (CPGs) and ESMO-MCBS Scores 2021. https://www.esmo.org/content/download/77789/1426712/file/ESMO-Clinical-Practice-Guidelines-Standard-Operating-Procedures.pdf.
  109. The National Comprehensive Cancer Network. About the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) 2020. https://www.nccn.org/professionals/default.aspx.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

Via MedicaWydawcą jest  VM Media Group sp. z o.o., Grupa Via Medica, ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl