Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 14.
Published in final edited form as: Curr Treat Options Oncol. 2018 Nov 5;19(12):69. doi: 10.1007/s11864-018-0592-3

Management of Advanced Small Bowel Cancer

Alberto Puccini 1,2,#, Francesca Battaglin 1,3,#, Heinz-Josef Lenz 1,*
PMCID: PMC7489287  NIHMSID: NIHMS1623857  PMID: 30397729

Opinion statement

Small bower cancer is a rare disease, despite its incidence is increasing in the last decade. Both benign and malignant tumors can arise from the small intestine. The main histological cancer types are adenocarcinomas, neuroendocrine tumors, sarcomas, gastrointestinal stromal tumors (GISTs), and lymphomas. Due to the rarity of these malignances, all the currently available data are based on small studies or retrospective series, although recent breakthroughs are redirecting our approach to these patients. Immunotherapy for small bowel adenocarcinomas, several multikinase inhibitors in resistant GIST patients, as well as everolimus and 177Lu-DOTATATE in neuroendocrine tumors are only few of the novel therapeutic options that have changed, or may change in the future, the therapeutic landscape of these rare cancers. Larger and more powerful studies on the molecular profile of these tumors may lead to a better design of clinical trials, which eventually would provide our patients with more efficacious treatments to improve both overall survival and quality of life.

Keywords: Small bowel cancer, Adenocarcinoma, GIST, Neuroendocrine

Introduction

Small bowel cancer is a rare disease that represents 0.6% of all new cancer cases in the USA [1]. Nonetheless, rates for new small intestine cancer cases have been rising by an average of 2.3% each year over the last 10 years [24]. In 2018, it is estimated that there will be 10,470 new cases of small bowel cancer and an estimated 1450 people will die of this disease.

Several different histological types of cancer can arise from the small intestine, such as adenocarcinomas, neuroendocrine tumors, sarcomas, gastrointestinal stromal tumors (GISTs), and lymphomas (Table 1). Of note, benign lesions that may arise in the small bowel include adenomas, leiomyomas, fibromas, and lipomas.

Table 1. Features of the main histological types of small bowel cancers.

Histotype Incidence among small bowel cancers Main location Management of advanced disease Future perspectives
Adenocarcinomas 30–50% Duodenum Chemotherapy
First line: FOLFOX/CAPOX
Second line: FOLFIRI
Immunotherapy and targeted therapy development based on novel molecular profiling data
GIST 10–20% Stomach (60%), jejunum and ileum (30%), duodenum (5%) First line: imatinib
Second line: sunitinib
Third line: regorafenib
Additional multikinase inhibitors (e.g., sorafenib, pazopanib, and ponatinib)
Neuroendocrine tumors 35–40% Ileum First line: SSAs, octreotide LAR, and lanreotide
Further lines: everolimus, 177Lu-dotatate, chemotherapy
Multitargeted tyrosine kinase inhibitors and PRRT

GIST gastrointestinal stromal tumors, LAR long-acting release, PRRT peptide receptor radiotherapy, SSAs somatostatin analogs

Due to their rarity and the heterogenous clinical presentation, small bowel cancers are often diagnosed in later stages, which are associated with a poor treatment outcome. Moreover, treatment and prognosis differ for each histological subtype and therapeutic strategies should be tailored to each separate class of tumor.

All data available in terms of clinicopathological features, molecular profile, treatment options, and survival outcomes of this neoplasia usually derive from small studies, often retrospective in nature. Larger and more powerful studies are eagerly awaited to expand our knowledge and implement the treatment scenario of these malignancies.

Herein, we aim to provide an overview on the actual body of knowledge on carcinogenesis, risk factors, and treatment of the main histological subtypes of small bower cancer: adenocarcinoma, GIST, and neuroendocrine tumors.

Epidemiology and etiopathogenetic factors

The mean age at diagnosis of a small bowel neoplasm is 65 years old, with sarcoma and lymphoma presenting at a slightly younger age (60 to 62) than adenocarcinoma and neuroendocrine tumors (67 to 68) [5, 6]. Adenocarcinoma has been the most common histologic type among small intestine malignancies, yet in the last few decades, neuroendocrine tumors incidence remarkably increased surpassing adenocarcinomas as the most frequent tumor type of the small bowel [2]. Of note, adenocarcinomas are most frequently diagnosed in the duodenum, while neuroendocrine tumors are most frequently found in the ileum.

A number of factors have been associated with an increased risk of developing a small bowel cancer including alcohol, tobacco, refined sugar and carbohydrates, red meat or smoked food, while a reduced risk has been linked with higher intakes of coffee, fish, fruit, and vegetables [711].

Small bowel cancers have been demonstrated to be associated with several predisposing conditions [12], such as inflammatory bowel disease (i.e., Crohn’s disease), coeliac disease, as well as hereditary syndromes: familial adenomatous polyposis (FAP), Lynch syndrome (otherwise called Hereditary non polyposis colorectal cancer or HNPCC), and Peutz–Jeghers syndrome.

Crohn’s disease induces a chronic transmural inflammation of the gastrointestinal tract which eventually may promote carcinogenesis. Cancers related to Crohn’s disease are usually adenocarcinomas of the ileum, with an increased relative risk (RR) of small bowel adenocarcinoma (SBA) in Crohn’s disease estimated in several population-based studies to range from 17 to 41 compared to the general population [12]. A meta-analysis showed a RR of 33.2 (95% CI 15.9–60.9) for small bowel cancer in patients with Crohn’s disease [13].

Moreover, an increased risk of small bowel lymphoma and SBA has been observed in patients affected by coeliac disease [14].

FAP, an autosomal dominant genetic disorder caused by a germline mutation of the APC gene, is a condition characterized by the early onset of numerous adenomatous polyps mainly in the epithelium of the large intestine. Although all patients affected by FAP will eventually develop colon cancer by age 35–40 years, if left untreated, other malignancies may occur. In fact, the prevalence of duodenal adenomas is 50–90% and these patients carry a RR of 330 for SBA or up to a 5% lifetime risk [15].

Lynch syndrome is a hereditary disorder due to the germline mutations of mismatch repair (MMR) genes (MLH1, MSH2, MSH6, and PMS2) that lead to microsatellite instability (MSI). Patients with this condition have an increased risk of colorectal and endometrial cancers and less frequently gastric, ovarian, biliary tract, urothelial cancers and SBA. The lifetime cumulative risk of SBA in Lynch syndrome patients remains low: 0.6–1% [16]. Therefore, international guidelines do not recommend to screen Lynch syndrome patients for SBA. However, analysis of the MMR phenotype is systematically recommended in SBA, because it could reveal the presence of Lynch syndrome and thus inform management and surveillance of patients and counseling for their relatives [17].

Finally, Peutz–Jeghers syndrome is an autosomal dominant condition due to the germline mutation of the STK11 gene associated with an increased risk of malignancies. The most common malignancy is colorectal cancer (CRC), followed by breast, small bowel, gastric, and pancreatic cancers. The reported lifetime risk for any cancer varied between 37 and 93%, with RRs ranging from 9.9 to 18 in comparison with the general population [18]. More specifically, the lifetime incidence for SBA is 1.7–13% and rises rapidly in elderly [19, 20].

Interestingly, data suggest that the carcinogenesis of SBA differs from that of CRC. In the former, the nuclear accumulation of β-catenin is most probably due to a gain-of-function mutation in the β-catenin gene rather than a loss of regulation due to the APC mutation, which is found in ~ 80% of all human colon tumors and it is considered one of the main events of CRC carcinogenesis [17, 21]. Due to the small sample size of the few studies that investigated the molecular profile of SBA, it is difficult to draw a definitive assessment of the frequencies of the gene mutations and protein expression involved in SBA carcinogenesis. However, it seems that SBAs are characterized by an overexpression of the p53 protein, vascular endothelial growth factor-A (VEGF-A), and the epidermal growth factor receptor (EGFR), as well a loss of SMAD4 expression [22]. While MSI or MMR-deficient (dMMR) tumors represent around 5% of CRC, the frequency of dMMR phenotype in SBA varies largely based on different studies; therefore, larger series are warranted to improve our knowledge on the biological mechanisms underlying these tumors and to implement the design of therapeutic trials.

Small bowel adenocarcinoma

As aforementioned, SBAs are rare and unique tumors, which are often diagnosed at an advanced stage due a delayed clinical presentation and non-specific symptoms (i.e., abdominal pain, weight loss, nausea and vomiting, occult GI tract bleeding), coupled with a limited sensitivity of conventional radiological imaging for the detection of this type of cancer [2, 17]. Few studies have specifically addressed the optimal management of SBA, and the general approach to the treatment of these tumors is to follow recommendations from expert agreements or by analogy with CRC [23, 24•]. However, more recently, new data are emerging on the molecular characterization of SBA and targeted treatment approaches in the advanced setting.

Overall, the 5-year survival for SBA patients is poor (14 to 33%), decreasing from 50 to 60% for stage I tumors (approximately 6–12% of patients) to 3–5% for stage IV (32–37% of patients at diagnosis) [2, 25, 26]. Among locally advanced SBA (stage III, 21–27%), the degree of lymph nodal involvement represents the main prognostic factor. In fact, the 5-year disease-free survival (DFS) is worse when three or more nodes are involved compared to two or less positive nodes (5-year DFS rates of 37% vs 58%, respectively, P < 0.01) [27]. A number of additional factors have been associated with a poor prognosis in different studies, including older age, impaired performance status, male gender, black ethnicity, a duodenal primary tumor location, poor tumor differentiation, positive margins, inadequate lymph nodal sampling (< 8 or < 10 according to different studies), low serum albumin, high lactate dehydrogenase, and high CEA or CA 19.9 [2, 26, 2830].

Surgical complete resection (R0) of primary tumor coupled with locoregional lymph node resection represents the only potentially curative treatment for localized disease. On the other hand, primary tumor resection is generally not recommended in the metastatic setting except in case of acute bowel obstruction, perforation, or uncontrolled bleeding. Of note, a retrospective study reported that the relapse pattern after R0 resection of localized SBA is predominantly systemic, with locoregional and distant relapse accounting for 18 and 86% of all recurrences, respectively [31].

The management of advanced SBA (i.e., unresectable or metastatic) is based on systemic treatment, although no randomized studies have been performed to demonstrate a benefit of systemic chemotherapy in patients with advanced disease. In fact, results from several retrospective comparative studies and small patients series in this setting demonstrated a significant benefit for systemic chemotherapy, both in terms of median progression-free survival (PFS) and overall survival (OS) (6 months vs 1 month and 9–19 vs 2–13 months, respectively) [3137]. Similarly, no prospective randomized data are available to determine which chemotherapeutic regimen is the most effective. However, retrospective series comparing different regimens consistently reported best results in terms of response, survival, and toxicity with the use of 5-fluorouracil/leucovorin plus oxaliplatin (FOLFOX) [3842]. More recently, several prospective phase II studies confirmed this retrospective evidence, showing response rates of 45–50%, disease control rates of 80–90%, and a median OS of 15–20 months following frontline treatment with either FOLFOX [43, 44] or capecitabine plus oxaliplatin (CAPOX) [45]. The addition of irinotecan to oxaliplatin and fluoropyrimidine appears to be feasible, but its benefit remains unclear. In a recent study, McWilliams and colleagues tested the efficacy of pharmacogenetic-based dosing of irinotecan (according to the UGT1A1 genotype), oxaliplatin, and capecitabine (CAPIRINOX) as first-line therapy in 33 patients with advanced SBA [46]. Results of this study showed overall an objective response rate of 38%, a disease control rate of 81%, a median PFS of 8.9 months, and a median OS of 13.4 months. Hematological toxicity as well as response rates were comparable among different genotype groups. In the second-line setting, the FOLFIRI regimen has shown modest activity in a small series of patients after failure on a platinum-based therapy (objective response rate 20%, disease control rate 52%, median PFS and OS 3.2 and 10.5 months, respectively) [47]. Of note, a recent small phase II study including ten SBA patients showed signs of activity of nab-paclitaxel in the treatment of refractory SBA, with two partial responses and in vivo testing of paclitaxel in a SBA patient-derived xenograft (PDX) supporting the activity of taxanes in this disease type [48]. Although encouraging, these results require further validation.

To date, contrary to CRC, few data are available exploring the benefit of specific biologic or targeted therapies in the treatment of advanced SBA. However, recent findings from a large-scale genomic profiling study of a series of 317 SBA highlighted that 92% of these tumors harbored a potentially actionable genomic alteration associated with available targeted agents or therapeutics under study in clinical trials. Among the most frequent alterations in SBA were HER2 mutation/amplification, EGFR mutation/amplification, MSI, high tumor mutational burden, and PIK3CA pathway activation [49•]. Similar findings were reported in a different exome sequencing study on a population-based set of 106 SBA [50]. Hence, SBA appears to have distinct molecular characteristics and may benefit from targeted therapeutic strategies. Notably, the rates of KRAS and BRAF mutations in SBA were similar to CRC, although BRAF V600E mutations were less common in SBA (10.3% of BRAF-mutated cases) and case reports have described responses to anti-EGFR therapy in patients with KRAS wild-type tumors [51, 52]. Nonetheless, recently published results from a small phase II single-arm trial investigating the efficacy of second-line panitumumab monotherapy in RAS wild-type SBA and ampullary adenocarcinoma failed to show any clinically meaningful activity of this treatment in this setting of patients [53]. A single published trial has tested, at present, the combination of the vascular endothelial growth factor (VEGF) inhibitor bevacizumab with chemotherapy in SBA. In this study, 30 patients with metastatic SBA and ampullary adenocarcinoma were treated with CAPOX plus bevacizumab, obtaining a response rate of 48% associated to a median PFS and OS of 8.7 and 12.9 months, respectively [54]. These results were not significantly different from results of the previous study with CAPOX alone from the same institution [45]. Finally, the high incidence of MSI, ranging from 5 to 45% in different studies, and the higher tumor mutational burden of SBA, alongside data showing high PD-1 and PDL-1 expression in this tumor type, support a potential benefit from immunotherapy [49•, 55]. Pembrolizumab, an anti-PD1 monoclonal antibody, has already been approved by the U.S. Food and Drug Administration (FDA) in May 2017 for the treatment of a variety of advanced MSI-high or dMMR solid tumors including SBA [56]. Results from dedicated ongoing trials investigating treatments with immune checkpoint inhibitors, such as pembrolizumab (NCT02949219) and the anti-PD-L1 monoclonal antibody avelumab (NCT03000179) in refractory SBA, are eagerly awaited.

Limited information and conflicting evidence are available regarding metastasectomy for advanced SBA. Cytoreductive surgery and intraperitoneal chemotherapy (IPC) have been proposed as an alternative treatment option for selected patients in case of peritoneal metastases. Peritoneal involvement characterizes about one third of stage IV patients and is more common with tumors arising from the jejunum or ileum rather than from the duodenum [57]. A review of literature including 15 observational and 3 phase II studies reporting outcome data from first-line systemic therapy or cytoreductive surgery with IPC for advanced SBA concluded that fluoropyrimidine plus oxaliplatin should be regarded as optimal first-line systemic treatment, although in selected patients, cytoreductive surgery plus IPC appeared safe and possibly more effective than systemic therapy as single treatment [58]. In the presented studies investigating cytoreduction plus IPC, median DFS and median OS ranged from 10 to 12 months and from 16 to 47 months, respectively, while grade III–V morbidity rates were 12 to 35%. More recently, outcome data have been published from a multiinstitutional international data registry including 152 patients with peritoneal metastases from SBA treated with cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy (HIPEC) between 1989 and 2016 in 21 institutions [59]. The combined treatment strategy of cytoreduction plus HIPEC achieved prolonged survival for selected patients (median DFS after completeness of cytoreduction [CCR] 0/1 of 14 months, range 1–100 months) with acceptable morbidity and mortality (treatment-related mortality rate 2%, grade 3 or 4 operative complications 19.1%). Absence of lymph nodal involvement, well-differentiated tumor, and peritoneal cancer index < 15 were independently associated with improved OS in the multivariate analysis. Generalization of these results however warrants some caution due to relatively small number of patients included in these studies and the possible significant bias in treatment results by patient selection and treatment heterogeneity across different studies. Furthermore, recently presented results of the randomized multicentric phase III PRODIGE 7 trial, investigating cytoreductive surgery plus HIPEC in association with systemic chemotherapy in metastatic CRC with isolated peritoneal carcinomatosis, showed no benefit from the addition of HIPEC to surgery alone plus systemic chemotherapy in this setting [60].

Sarcomas and GIST

Sarcomas represent around 10–20% of the small bowel cancers [61, 62]. Among all sarcomas arising in the small bowel tract, the vast majority is considered to be gastrointestinal stromal tumors (GISTs). GISTs are hypothesized to arise from interstitial cells of Cajal, the intestinal pacemaker cells, and are the most common mesenchymal neoplasms of the gastrointestinal tract. However, GISTs are rare tumors, with an incidence estimated to be between 6.8 and 20 per million population [63, 64]. They are usually found in stomach (60%), small intestine, jejunum and ileum (30%), duodenum (5%), rectum (2–3%), and colon (1–2%) [65•].

Activating mutations in the c-KIT (CD117) oncogene are seen in about 60–85% of all GISTs [66]. A subset of GISTs lacking c-KIT mutations (about 5–10% of all GISTs) has activating mutations in a related receptor tyrosine kinase, the platelet-derived growth factor receptor alpha (PDGFRA).

Imatinib mesylate (Gleevec®; Novartis Pharmaceuticals Corporation, East Hanover, NJ) was approved by the FDA for the treatment of malignant metastatic and/or unresectable GISTs on February 1, 2002, with the dose for GIST patients of 400 or 600 mg daily [67], based on a study which included 147 patients enrolled at three US centers and one center in Finland [68]. An overall response rate of 38% across the two dose levels and a response duration ranged from 7 to 38 weeks, with a median of 13 weeks, were observed. In 2012, the FDA granted regular approval of imatinib mesylate tablets for the adjuvant treatment of adult patients following complete resection of c-KIT+ (CD117+) GISTs, with the recommended dose of 400 mg/day administered daily for 3 years. The initial approval (2008) was based on a single phase III randomized, double-blinded, placebo-controlled study that enrolled a total of 713 adult patients with a c-KIT+ GIST, and a resected tumor size ≥ 3 cm. Imatinib mesylate, 400 mg orally once daily, was administered for 1 year [69]. In the intent-to-treat (ITT) population, imatinib treatment led to a significantly longer recurrence-free survival (RFS) interval than placebo (hazard ratio [HR] = 0.398; 95% confidence interval [CI], 0.259–0.610; P < .0001) [70]. Following these results, a phase III, multicenter, open-label, randomized adjuvant trial was conducted comparing imatinib treatment for 3 years with imatinib treatment for 1 year in a higher risk population [71]. Three hundred ninety-seven patients were enrolled and patients assigned in the 3-year arm had longer RFS (5-year RFS, 65.6% vs 47.9%, respectively HR = 0.46; 95% CI 0.32–0.65; P < 0.001) and longer overall survival (HR = 0.45; 95% CI 0.22–0.89; P = 0.02; 5-year survival, 92.0% vs 81.7%) compared with those assigned in the 1-year arm, leading to the aforementioned FDA approval in 2012.

Two main therapeutic options are available for patients with advanced GIST who progress on imatinib treatment: either imatinib dose escalation [72] or sunitinib, a multitargeted tyrosine kinase inhibitor (TKI) active against PDGFRα, PDGFRβ, and VEGFR receptors, which has demonstrated a 5-month PFS improvement compared to placebo in a randomized phase III trial [73]. Based on these results, the European Medicines Agency (EMA) and the FDA granted approval for sunitinib (Sutent, Pfizer) in the second-line treatment of GIST at the recommended dose of 50 mg orally once a day over 4 weeks followed by a 2-week rest period.

Finally, regorafenib (Stirvarga, Bayer HealthCare Pharmaceuticals Inc.), an orally active multikinase inhibitor with activity against several kinases including c-KIT, was recently approved by FDA and EMA for the treatment of patients with unresectable or metastatic GIST after failure or intolerance to imatinib and sunitinib [74].

Several other multikinase inhibitors have shown promising results in resistant GIST patients, although data derive mostly from small phase II studies. Therefore, sorafenib, pazopanib, and ponatinib represent valuable options in patients that progressed after all current standard treatments for advanced/metastatic disease (i.e., imatinib, sunitinib, and regorafenib). Moreover, the enrollment of patients in clinical trials is highly encouraged in this setting.

It is noteworthy that about 10 to 15% of GISTs in adults and 85% of GISTs in children are negative for c-KIT and PDGFRA mutations, which are designated as wild-type GIST [75]. This subset of GISTs is known to be resistant to kinase inhibitor therapies (i.e., imatinib), primarily affects young females, is multifocal but indolent, and the primary tumor has generally a gastric location. Wildtype GISTs may have several different driver mutations and may comprise heterogeneous entities, such as succinate dehydrogenase (SDH)-deficient GISTs, BRAF-mutated GISTs, and neurofibromatosis type I-associated GISTs (NF1-GISTs) [76]. In a recent study by Boikos et al. [77], wild-type GIST specimens from 95 patients were classified into 3 molecular subtypes: SDH-competent (n = 11) and 2 types of SDH-deficient GIST (by SDH-B mutation or SDH-C promoter methylation). The authors found that SDH-competent tumors retain SDHB expression and a normal methylation pattern. This subset of wild-type GIST displays tumor and patient demographic features similar to those seen in patients with c-KIT/PDGFRA-mutant tumors and has a more aggressive behavior compared with SDH-deficient tumors. Thus, investigating the molecular subtype of GIST has direct implications for treatment strategies and for the development of new approaches to systemic therapy.

Neuroendocrine tumors

Neuroendocrine tumors (NETs) of the gastrointestinal (GI) tract are rare diseases, but the incidence and prevalence of NETs are steadily rising, possibly owing to detection of early-stage disease and stage migration [78]. Survival for all NETs has improved over time, especially for advanced-stage GI-NETs, and in particular for pancreatic NETs in particular, reflecting major therapeutic improvements [79]. However, molecular data which may explain the clinical heterogeneity of this class of tumors, from indolent to highly aggressive, and divergent treatment responses, are lacking [80].

GI-NETs represent approximately 35–40% of primary small intestinal malignancies [2]. They comprise a genetically diverse spectrum of malignant solid tumors arising from the secretory cells of the neuroendocrine cell system that may produce peptides causing characteristic hormonal syndromes [81]. Therefore, GI-NETs are categorized as “functioning” when patients are clinically symptomatic (carcinoid syndrome) or “non-functioning.”

The majority of NETs occur sporadically. However, they may occur as part of complex familial endocrine cancer syndromes such as type 1 multiple endocrine neoplasia (MEN 1) [82], von Hippel-Lindau Disease [83], and neurofibromatosis type 1 (NF1) [84].

Many advances have been made in recent years for the treatment of patients with metastatic GI-NETs, and here we will briefly report the current therapeutic standard and the most recent trails which led to a change in clinical practice.

Somatostatin analogs (SSAs), octreotide LAR [85] and lanreotide [86], are appropriate initial therapy in most patients with unresectable metastatic midgut NETs for both control of carcinoid syndrome and tumor progression. Generally, SSAs are associated with major improvements in flushing and diarrhea in roughly 75% of patients with carcinoid syndrome. Because of their relatively benign side effect profile, SSAs are typically selected as first-line systemic therapy.

The RADIANT-2 [87] and RADIANT-4 [88] studies evaluated Everolimus (Afinitor®) in functioning and non-functioning NETs of the GI tract and lungs. RADIANT-4 results showed that median PFS improved from 3.9 to 11 months (P < 0.00001) with everolimus compared to placebo. These data led to the approval of everolimus (Afinitor®, Novartis) by the FDA for the treatment of patients with progressive, nonfunctional NETs of the GI tract and lung. An earlier study, the RADIANT-2 trial, randomized patients with progressive NETs and a history of carcinoid syndrome to receive everolimus plus octreotide LAR versus placebo plus octreotide LAR. It was noted that everolimus efficacy appears stronger in non-midgut NETs (which represented the majority of patients on the RADIANT-4 study) compared with midgut NETs (which represented the majority of patients on the RADIANT-2 study).

A large number of metastatic midgut NETs (> 50% in some studies) secrete serotonin and are associated with the carcinoid syndrome [89]. Therefore, the TELESTAR study [90] evaluated telotristat ethyl, a tryptophan hydroxylase inhibitor, in patients with refractory carcinoid syndrome, which has become the treatment of choice in patients with stable radiographic disease and refractory carcinoid syndrome characterized by suboptimal control of diarrhea.

Finally, the NETTER-1 trial [91] evaluated 177Lu-DOTATATE in NETs of the small intestine and proximal colon (midgut). 177Lu-DOTATATE is a radiolabeled SSA, a form of treatment also known as peptide receptor radiotherapy (PRRT). In this study, median PFS was 8 months on the high-dose octreotide arm and was not yet reached on the 177Lu-DOTATATE arm, translating to a 79% improvement in PFS (P < 0.00001).

Although the treatment landscape for GI-NETs patients has been revolutionized and evolved significantly in the last decade, several questions remain unanswered. Further clinical research will be needed to address key issues pertaining to the management of midgut NETs [92].

Conclusion

Small bowel cancers are a heterogenous group of malignancies, which require a dedicated and personalized approach. Many advances have been made for the treatment of adenocarcinomas, GIST, and NETs arising from the small intestine, although more clinical trials are strongly warranted to provide patients with more therapeutic options which may lead to a longer survival and a better quality of life.

Acknowledgments

Funding This manuscript was partly supported in part by the National Cancer Institute (grant number P30CA014089), the Gloria Borges WunderGlo Foundation-The Wunder Project, the Dhont Family Foundation, the San Pedro Peninsula Cancer Guild, the Daniel Butler Research Fund, and the Call to Cure Research Fund. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institutes of Health.

Footnotes

Conflict of Interest

Alberto Puccini declares that he has no conflict of interest.

Francesca Battaglin declares that she has no conflict of interest.

Heinz-Josef Lenz has received clinical trial financial support from Merck Serono and Roche and honoraria for advisory board membership and lectures from Bayer, Boehringer Ingelheim, Genentech, Merck Serono, and Roche.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Ethics Approval and Consent to Participate

Not applicable.

Consent for Publication

Not applicable.

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as:

• Of importance

  • 1.https://seer.cancer.gov/statfacts/html/smint.html. SCSFSICNCI.
  • 2.Bilimoria KY, Bentrem DJ, Wayne JD, Ko CY, Bennett CL, Talamonti MS. Small bowel cancer in the United States: changes in epidemiology, treatment, and survival over the last 20 years. Ann Surg. 2009;249:63–71. [DOI] [PubMed] [Google Scholar]
  • 3.Chow JS, Chen CC, Ahsan H, Neugut AI. A population-based study of the incidence of malignant small bowel tumours: SEER, 1973–1990. Int J Epidemiol. 1996;25:722–8. [DOI] [PubMed] [Google Scholar]
  • 4.Severson RK, Schenk M, Gurney JG, et al. Increasing incidence of adenocarcinomas and carcinoid tumors of the small intestine in adults. Cancer Epidemiol Biomark Prev. 1996;5:81–4. [PubMed] [Google Scholar]
  • 5.Hatzaras I, Palesty JA, Abir F, Sullivan P, Kozol RA, Dudrick SJ, et al. Small-bowel tumors: epidemiologic and clinical characteristics of 1260 cases from the Connecticut Tumor Registry. Arch Surg. 2007;142:229–35. [DOI] [PubMed] [Google Scholar]
  • 6.Lepage C, Bouvier AM, Manfredi S, Dancourt V, Faivre J. Incidence and management of primary malignant small bowel cancers: a well-defined French population study. Am J Gastroenterol. 2006;101:2826–32. [DOI] [PubMed] [Google Scholar]
  • 7.Chow WH, Linet MS, McLaughlin JK, et al. Risk factors for small intestine cancer. Cancer Causes Control. 1993;4:163–9. [DOI] [PubMed] [Google Scholar]
  • 8.Cross AJ, Leitzmann MF, Subar AF, Thompson FE, Hollenbeck AR, Schatzkin A. A prospective study of meat and fat intake in relation to small intestinal cancer. Cancer Res. 2008;68:9274–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bjorge T, Tretli S, Engeland A. Height and body mass index in relation to cancer of the small intestine in two million Norwegian men and women. Br J Cancer. 2005;93:807–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Boffetta P, Hazelton WD, Chen Y, Sinha R, Inoue M, Gao YT, et al. Body mass, tobacco smoking, alcohol drinking and risk of cancer of the small intestine—a pooled analysis of over 500,000 subjects in the Asia Cohort Consortium. Ann Oncol. 2012;23:1894–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bennett CM, Coleman HG, Veal PG, Cantwell MM, Lau CCL, Murray LJ. Lifestyle factors and small intestine adenocarcinoma risk: a systematic review and meta-analysis. Cancer Epidemiol. 2015;39:265–73. [DOI] [PubMed] [Google Scholar]
  • 12.Pan SY, Morrison H. Epidemiology of cancer of the small intestine. World J Gastrointest Oncol. 2011;3:33–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Canavan C, Abrams KR, Mayberry J. Meta-analysis: colorectal and small bowel cancer risk in patients with Crohn’s disease. Aliment Pharmacol Ther. 2006;23:1097–104. [DOI] [PubMed] [Google Scholar]
  • 14.Rampertab SD, Forde KA, Green PH. Small bowel neoplasia in coeliac disease. Gut. 2003;52:1211–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shenoy S Genetic risks and familial associations of small bowel carcinoma. World J Gastrointest Oncol. 2016;8:509–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bonadona V, Bonaiti B, Olschwang S, et al. Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome. JAMA. 2011;305:2304–10. [DOI] [PubMed] [Google Scholar]
  • 17.Aparicio T, Zaanan A, Svrcek M, Laurent-Puig P, Carrere N, Manfredi S, et al. Small bowel adenocarcinoma: epidemiology, risk factors, diagnosis and treatment. Dig Liver Dis. 2014;46:97–104. [DOI] [PubMed] [Google Scholar]
  • 18.van Lier MG, Wagner A, Mathus-Vliegen EM, et al. High cancer risk in Peutz-Jeghers syndrome: a systematic review and surveillance recommendations. Am J Gastroenterol. 2010;105:1258–64 author reply 1265. [DOI] [PubMed] [Google Scholar]
  • 19.Hearle N, Schumacher V, Menko FH, Olschwang S, Boardman LA, Gille JJ, et al. Frequency and spectrum of cancers in the Peutz-Jeghers syndrome. Clin Cancer Res. 2006;12:3209–15. [DOI] [PubMed] [Google Scholar]
  • 20.Giardiello FM, Brensinger JD, Tersmette AC, Goodman SN, Petersen GM, Booker SV, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology. 2000;119:1447–53. [DOI] [PubMed] [Google Scholar]
  • 21.Christie M, Jorissen RN, Mouradov D, Sakthianandeswaren A, Li S, Day F, et al. Different APC genotypes in proximal and distal sporadic colorectal cancers suggest distinct WNT/beta-catenin signalling thresholds for tumourigenesis. Oncogene. 2013;32:4675–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zaaimi Y, Aparicio T, Laurent-Puig P, Taieb J, Zaanan A. Advanced small bowel adenocarcinoma: molecular characteristics and therapeutic perspectives. Clin Res Hepatol Gastroenterol. 2016;40:154–60. [DOI] [PubMed] [Google Scholar]
  • 23.National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Colon Cancer. Version 3.2018. Avalable at: https://www.nccn.org/professionals/physician_gls/pdf/colon.pdf. Last Accessed 01 Sept 2018.
  • 24.•.Locher C, Batumona B, Afchain P, et al. Small bowel adenocarcinoma: French intergroup clinical practice guidelines for diagnosis, treatments and follow-up (SNFGE, FFCD, GERCOR, UNICANCER, SFCD, SFED, SFRO). Dig Liver Dis. 2018;50:15–.Updated clinical guidelines for the management of small bowel adenocarcinoma.
  • 25.Faivre J, Trama A, De Angelis R, et al. Incidence, prevalence and survival of patients with rare epithelial digestive cancers diagnosed in Europe in 1995–2002. Eur J Cancer. 2012;48:1417–24. [DOI] [PubMed] [Google Scholar]
  • 26.Nicholl MB, Ahuja V, Conway WC, Vu VD, Sim MS, Singh G. Small bowel adenocarcinoma: understaged and undertreated? Ann Surg Oncol. 2010;17:2728–32. [DOI] [PubMed] [Google Scholar]
  • 27.Overman MJ, Hu CY, Wolff RA, Chang GJ. Prognostic value of lymph node evaluation in small bowel adenocarcinoma: analysis of the surveillance, epidemiology, and end results database. Cancer. 2010;116:5374–82. [DOI] [PubMed] [Google Scholar]
  • 28.Sakae H, Kanzaki H, Nasu J, Akimoto Y, Matsueda K, Yoshioka M, et al. The characteristics and outcomes of small bowel adenocarcinoma: a multicentre retrospective observational study. Br J Cancer. 2017;117:1607–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ecker BL, McMillan MT, Datta J, et al. Efficacy of adjuvant chemotherapy for small bowel adenocarcinoma: a propensity score-matched analysis. Cancer. 2016;122:693–701. [DOI] [PubMed] [Google Scholar]
  • 30.Ecker BL, McMillan MT, Datta J, et al. Lymph node evaluation and survival after curative-intent resection of duodenal adenocarcinoma: a matched cohort study. Eur J Cancer. 2016;69:135–41. [DOI] [PubMed] [Google Scholar]
  • 31.Dabaja BS, Suki D, Pro B, Bonnen M, Ajani J. Adenocarcinoma of the small bowel: presentation, prognostic factors, and outcome of 217 patients. Cancer. 2004;101:518–26. [DOI] [PubMed] [Google Scholar]
  • 32.Halfdanarson TR, McWilliams RR, Donohue JH, Quevedo JF. A single-institution experience with 491 cases of small bowel adenocarcinoma. Am J Surg. 2010;199:797–803. [DOI] [PubMed] [Google Scholar]
  • 33.Czaykowski P, Hui D. Chemotherapy in small bowel adenocarcinoma: 10-year experience of the British Columbia Cancer Agency. Clin Oncol (R Coll Radiol). 2007;19:143–9. [DOI] [PubMed] [Google Scholar]
  • 34.Koo DH, Yun SC, Hong YS, Ryu MH, Lee JL, Chang HM, et al. Systemic chemotherapy for treatment of advanced small bowel adenocarcinoma with prognostic factor analysis: retrospective study. BMC Cancer. 2011;11:205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Khan K, Peckitt C, Sclafani F, Watkins D, Rao S, Starling N, et al. Prognostic factors and treatment outcomes in patients with small bowel adenocarcinoma (SBA): the Royal Marsden Hospital (RMH) experience. BMC Cancer. 2015;15:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Crawley C, Ross P, Norman A, Hill A, Cunningham D. The Royal Marsden experience of small bowel adenocarcinoma treated with protracted venous infusion 5-fluorouracil. Br J Cancer. 1998;78:508–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhang L, Wang LY, Deng YM, Wang FH, Feng F, Chen YC, et al. Efficacy of the FOLFOX/CAPOX regimen for advanced small bowel adenocarcinoma: a three-center study from China. J buon. 2011;16:689–96. [PubMed] [Google Scholar]
  • 38.Zaanan A, Costes L, Gauthier M, Malka D, Locher C, Mitry E, et al. Chemotherapy of advanced small-bowel adenocarcinoma: a multicenter AGEO study. Ann Oncol. 2010;21:1786–93. [DOI] [PubMed] [Google Scholar]
  • 39.Locher C, Malka D, Boige V, Lebray P, Elias D, Lasser P, et al. Combination chemotherapy in advanced small bowel adenocarcinoma. Oncology. 2005;69:290–4. [DOI] [PubMed] [Google Scholar]
  • 40.Overman MJ, Kopetz S, Wen S, Hoff PM, Fogelman D, Morris J, et al. Chemotherapy with 5-fluorouracil and a platinum compound improves outcomes in metastatic small bowel adenocarcinoma. Cancer. 2008;113:2038–45. [DOI] [PubMed] [Google Scholar]
  • 41.Gibson MK, Holcroft CA, Kvols LK, Haller D. Phase II study of 5-fluorouracil, doxorubicin, and mitomycin C for metastatic small bowel adenocarcinoma. Oncologist. 2005;10:132–7. [DOI] [PubMed] [Google Scholar]
  • 42.Tsushima T, Taguri M, Honma Y, Takahashi H, Ueda S, Nishina T, et al. Multicenter retrospective study of 132 patients with unresectable small bowel adenocarcinoma treated with chemotherapy. Oncologist. 2012;17:1163–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Xiang XJ, Liu YW, Zhang L, Qiu F, Yu F, Zhan ZY, et al. A phase II study of modified FOLFOX as first-line chemotherapy in advanced small bowel adenocarcinoma. Anti-Cancer Drugs. 2012;23:561–6. [DOI] [PubMed] [Google Scholar]
  • 44.Horimatsu T, Nakayama N, Moriwaki T, Hirashima Y, Fujita M, Asayama M, et al. A phase II study of 5-fluorouracil/L-leucovorin/oxaliplatin (mFOLFOX6) in Japanese patients with metastatic or unresectable small bowel adenocarcinoma. Int J Clin Oncol. 2017;22:905–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Overman MJ, Varadhachary GR, Kopetz S, Adinin R, Lin E, Morris JS, et al. Phase II study of capecitabine and oxaliplatin for advanced adenocarcinoma of the small bowel and ampulla of Vater. J Clin Oncol. 2009;27:2598–603. [DOI] [PubMed] [Google Scholar]
  • 46.McWilliams RR, Foster NR, Mahoney MR, et al. North Central Cancer Treatment Group N0543 (Alliance): a phase 2 trial of pharmacogenetic-based dosing of irinotecan, oxaliplatin, and capecitabine as first-line therapy for patients with advanced small bowel adenocarcinoma. Cancer. 2017;123:3494–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zaanan A, Gauthier M, Malka D, Locher C, Gornet JM, Thirot-Bidault A, et al. Second-line chemotherapy with fluorouracil, leucovorin, and irinotecan (FOLFIRI regimen) in patients with advanced small bowel adenocarcinoma after failure of first-line platinum-based chemotherapy: a multicenter AGEO study. Cancer. 2011;117:1422–8. [DOI] [PubMed] [Google Scholar]
  • 48.Overman MJ, Adam L, Raghav K, Wang J, Kee B, Fogelman D, et al. Phase II study of nab-paclitaxel in refractory small bowel adenocarcinoma and CpG island methylator phenotype (CIMP)-high colorectal cancer. Ann Oncol. 2018;29:139–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.•.Schrock AB, Devoe CE, McWilliams R, et al. Genomic profiling of small-bowel adenocarcinoma. JAMA Oncol. 2017;3:1546–53A large-scale genomic study of small bowel adenocarcinoma samples that provides insights into the molecular profile of this type of cancer.
  • 50.Hanninen UA, Katainen R, Tanskanen T, et al. Exome-wide somatic mutation characterization of small bowel adenocarcinoma. PLoS Genet. 2018;14:e1007200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Santini D, Fratto ME, Spoto C, Russo A, Galluzzo S, Zoccoli A, et al. Cetuximab in small bowel adenocarcinoma: a new friend? Br J Cancer. 2010;103:1305 author reply 1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Falcone R, Roberto M, Filetti M, Anselmi E, Marchetti P. Anti epidermal growth factor receptor therapy in small bowel adenocarcinoma: case report and literature review. Medicine (Baltimore). 2018;97:e9672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gulhati P, Raghav K, Shroff R, Varadhachary G, Javle M, Qiao W, et al. Phase II study of panitumumab in RAS wild-type metastatic adenocarcinoma of small bowel or ampulla of Vater. Oncologist. 2018;23:277–e226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gulhati P, Raghav K, Shroff RT, Varadhachary GR, Kopetz S, Javle M, et al. Bevacizumab combined with capecitabine and oxaliplatin in patients with advanced adenocarcinoma of the small bowel or ampulla of vater: a single-center, open-label, phase 2 study. Cancer. 2017;123:1011–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Thota R, Gonzalez RS, Berlin J, Cardin DB, Shi C. Could the PD-1 pathway be a potential target for treating small intestinal adenocarcinoma? Am J Clin Pathol. 2017;148:208–14. [DOI] [PubMed] [Google Scholar]
  • 56.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Legué LM, Bernards N, Gerritse SL, van Oudheusden TR, de Hingh IHJT, Creemers GJM, et al. Trends in incidence, treatment and survival of small bowel adenocarcinomas between 1999 and 2013: a population-based study in the Netherlands. Acta Oncol. 2016;55:1183–9. [DOI] [PubMed] [Google Scholar]
  • 58.Rovers KP, de Bree E, Yonemura Y, de Hingh IH. Treatment of peritoneal metastases from small bowel adenocarcinoma. Int J Hyperth. 2017;33:571–8. [DOI] [PubMed] [Google Scholar]
  • 59.Liu Y, Yonemura Y, Levine EA, et al. Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy for peritoneal metastases from a small bowel adenocarcinoma: multi-institutional experience. Ann Surg Oncol. 2018;25:1184–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Quenet F, Elias D, Roca L, et al. A UNICANCER phase III trial of hyperthermic intra-peritoneal chemotherapy (HIPEC) for colorectal peritoneal carcinomatosis (PC): PRODIGE 7. J Clin Oncol. 2018;36(suppl LBA3503). 10.1200/JCO.2018.36.18_suppl.LBA3503. [DOI] [Google Scholar]
  • 61.Howe JR, Karnell LH, Scott-Conner C. Small bowel sarcoma: analysis of survival from the National Cancer Data Base. Ann Surg Oncol. 2001;8(6):496–508. [DOI] [PubMed] [Google Scholar]
  • 62.Talamonti MS, Goetz LH, Rao S, Joehl RJ. Primary cancers of the small bowel: analysis of prognostic factors and results of surgical management. Arch Surg. 2002;137:564–70 discussion 570–561. [DOI] [PubMed] [Google Scholar]
  • 63.Corless CL, Fletcher JA, Heinrich MC. Biology of gastrointestinal stromal tumors. J Clin Oncol. 2004;22:3813–25. [DOI] [PubMed] [Google Scholar]
  • 64.Tran T, Davila JA, El-Serag HB. The epidemiology of malignant gastrointestinal stromal tumors: an analysis of 1,458 cases from 1992 to 2000. Am J Gastroenterol. 2005;100:162–8. [DOI] [PubMed] [Google Scholar]
  • 65.•.Poveda A, Garcia Del Muro X, Lopez-Guerrero JA, et al. GEIS guidelines for gastrointestinal sarcomas (GIST). Cancer Treat Rev. 2017;55:107–1.Updated guidelines for the management of GISTs.
  • 66.Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279:577–80. [DOI] [PubMed] [Google Scholar]
  • 67.Dagher R, Cohen M, Williams G, Rothmann M, Gobburu J, Robbie G, et al. Approval summary: imatinib mesylate in the treatment of metastatic and/or unresectable malignant gastrointestinal stromal tumors. Clin Cancer Res. 2002;8:3034–8. [PubMed] [Google Scholar]
  • 68.Lenke LG, Engsberg JR, Ross SA, Reitenbach A, Blanke K, Bridwell KH. Prospective dynamic functional evaluation of gait and spinal balance following spinal fusion in adolescent idiopathic scoliosis. Spine (Phila Pa 1976). 2001;26:E330–7. [DOI] [PubMed] [Google Scholar]
  • 69.Cohen MH, Johnson JR, Justice R, Pazdur R. Approval summary: imatinib mesylate for one or three years in the adjuvant treatment of gastrointestinal stromal tumors. Oncologist. 2012;17:992–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Dematteo RP, Ballman KV, Antonescu CR, Maki RG, Pisters PW, Demetri GD, et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;373:1097–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Joensuu H, Eriksson M, Sundby Hall K, Hartmann JT, Pink D, Schütte J, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: a randomized trial. JAMA. 2012;307:1265–72. [DOI] [PubMed] [Google Scholar]
  • 72.Zalcberg JR, Verweij J, Casali PG, le Cesne A, Reichardt P, Blay JY, et al. Outcome of patients with advanced gastro-intestinal stromal tumours crossing over to a daily imatinib dose of 800 mg after progression on 400 mg. Eur J Cancer. 2005;41:1751–7. [DOI] [PubMed] [Google Scholar]
  • 73.Demetri GD, van Oosterom AT, Garrett CR, Blackstein ME, Shah MH, Verweij J, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet. 2006;368:1329–38. [DOI] [PubMed] [Google Scholar]
  • 74.Demetri GD, Reichardt P, Kang YK, Blay JY, Rutkowski P, Gelderblom H, et al. Efficacy and safety of regorafenib for advanced gastrointestinal stromal tumours after failure of imatinib and sunitinib (GRID): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet. 2013;381:295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wada R, Arai H, Kure S, Peng WX, Naito Z. “Wild type” GIST: Clinicopathological features and clinical practice. Pathol Int. 2016;66:431–7. [DOI] [PubMed] [Google Scholar]
  • 76.Nishida T Therapeutic strategies for wild-type gastrointestinal stromal tumor: is it different from KIT or PDGFRA-mutated GISTs? Transl Gastroenterol Hepatol. 2017;2:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Boikos SA, Pappo AS, Killian JK, LaQuaglia MP, Weldon CB, George S, et al. Molecular subtypes of KIT/PDGFRA wild-type gastrointestinal stromal tumors: a report from the National Institutes of Health Gastrointestinal Stromal Tumor Clinic. JAMA Oncol. 2016;2:922–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lewis MA, Yao JC. Molecular pathology and genetics of gastrointestinal neuroendocrine tumours. Curr Opin Endocrinol Diabetes Obes. 2014;21:22–7. [DOI] [PubMed] [Google Scholar]
  • 79.Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3:1335–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Bijlsma MF, Sadanandam A, Tan P, Vermeulen L. Molecular subtypes in cancers of the gastrointestinal tract. Nat Rev Gastroenterol Hepatol. 2017;14:333–42. [DOI] [PubMed] [Google Scholar]
  • 81.Oberg KE. Gastrointestinal neuroendocrine tumors. Ann Oncol. 2010;21(Suppl 7):vii72–80. [DOI] [PubMed] [Google Scholar]
  • 82.Sadowski SM, Triponez F. Management of pancreatic neuroendocrine tumors in patients with MEN 1. Gland Surg. 2015;4:63–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tirosh A, Sadowski SM, Linehan WM, Libutti SK, Patel D, Nilubol N, et al. Association of VHL genotype with pancreatic neuroendocrine tumor phenotype in patients with von Hippel-Lindau disease. JAMA Oncol. 2018;4:124–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lodish MB, Stratakis CA. Endocrine tumours in neurofibromatosis type 1, tuberous sclerosis and related syndromes. Best Pract Res Clin Endocrinol Metab. 2010;24:439–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rinke A, Muller HH, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomized study on the effect of octreotide LAR in the control of tumor growth in patients with metastatic neuroendocrine midgut tumors: a report from the PROMID Study Group. J Clin Oncol. 2009;27:4656–63. [DOI] [PubMed] [Google Scholar]
  • 86.Caplin ME, Pavel M, Cwikla JB, et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N Engl J Med. 2014;371:224–33. [DOI] [PubMed] [Google Scholar]
  • 87.Pavel ME, Hainsworth JD, Baudin E, Peeters M, Hörsch D, Winkler RE, et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet. 2011;378:2005–12. [DOI] [PubMed] [Google Scholar]
  • 88.Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi C, Wolin E, et al. Everolimus for the treatment of advanced, nonfunctional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016;387:968–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Strosberg JR, Weber JM, Feldman M, Coppola D, Meredith K, Kvols LK. Prognostic validity of the American Joint Committee on Cancer staging classification for midgut neuroendocrine tumors. J Clin Oncol. 2013;31:420–5. [DOI] [PubMed] [Google Scholar]
  • 90.Kulke MH, Horsch D, Caplin ME, et al. Telotristat ethyl, a tryptophan hydroxylase inhibitor for the treatment of carcinoid syndrome. J Clin Oncol. 2017;35:14–23. [DOI] [PubMed] [Google Scholar]
  • 91.Strosberg J, El-Haddad G, Wolin E, et al. Phase 3 trial of (177)Lu-dotatate for midgut neuroendocrine tumors. N Engl J Med. 2017;376:125–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Strosberg JR, Halfdanarson TR, Bellizzi AM, Chan JA, Dillon JS, Heaney AP, et al. The North American Neuroendocrine Tumor Society consensus guidelines for surveillance and medical management of midgut neuroendocrine tumors. Pancreas. 2017;46:707–14. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES