Systemic Therapies for Advanced Pancreatic Neuroendocrine Tumors




Pancreatic neuroendocrine tumors are an uncommon tumor type and compose 1% to 2% of all pancreatic neoplasms. They are rarely localized at presentation and are typically diagnosed in the presence of metastatic disease. The management poses a significant challenge because of the heterogeneous clinical presentations and varying degrees of aggressiveness. A variety of systemic therapies have been developed for the management of pancreatic neuroendocrine tumors, including somatostatin analogues, a select group of cytotoxic chemotherapy agents, and targeted or biological agents. This article reviews the available systemic therapy options for advanced pancreatic neuroendocrine tumors.


Key points








  • Pancreatic neuroendocrine tumors (NETs) are genetically and clinically different than extrapancreatic NETs (ie, carcinoid tumors) and often respond to cytotoxic and targeted treatments.



  • Asymptomatic patients with low-volume advanced pancreatic NETs often have indolent disease, and some can be followed expectantly. Careful evaluation of each individual patient with an initial interval of observation and assessment can help define who needs treatment sooner.



  • Somatostatin analogues (octreotide and lanreotide) can decrease hormone production in functional tumors and can control neuroendocrine tumor growth; given their favorable toxicity profile, they are generally used as first-line treatment in unresectable patients.



  • Sunitinib and everolimus are 2 targeted therapies approved for progressive pancreatic NETs and are generally reserved for use in tumors that have progressed on somatostatin analogue therapy.



  • Pancreatic NETs can respond to cytotoxic chemotherapy; the most commonly used agents include alkylating, fluorouracil, and platinum drugs.






Introduction


Well-differentiated neuroendocrine tumors (NETs) are an uncommon and heterogeneous group of neoplasms that arise throughout the body, most commonly in the lung and gastrointestinal tract. These tumors are subdivided into carcinoid tumors and pancreatic NETs (panNETs). Carcinoid tumors develop from the neuroendocrine tissues of the aerodigestive tract, and panNETs develop from the endocrine tissues of the pancreas (ie, islets of Langerhans). This group of well-differentiated NETs is both morphologically and clinically distinct from high-grade neuroendocrine carcinomas, tumors that are characterized by an extremely aggressive behavior and are treated along small cell lung cancer paradigms with platinum-based chemotherapy. Epidemiologic data from the last 30 years have demonstrated that the incidence of NETs continues to increase, although there have been no significant changes in survival from this disease.


panNETs are the second most common tumor of the pancreas and represent 1% to 2% of all pancreatic neoplasms. Although most panNETs are slow growing, after the development of metastatic disease (most commonly in the liver), median survival ranges from 2 to 5 years; most patients with liver metastases will die of the disease. About one-third of panNETs are functional tumors and produce clinical syndromes due to excessive hormone secretion; these functional panNETs are classified by the hormones they hypersecrete and include insulinoma (secrete insulin and cause hypoglycemia), gastrinoma (secrete gastrin and cause Zollinger-Ellison syndrome, which is characterized by severe peptic ulcer disease), glucagonoma (secrete glucagon and cause hyperglycemia), and vasoactive intestinal polypeptide (VIP) (VIPoma, secrete VIP and cause severe secretory diarrhea). Nonfunctional panNETs are tumors that do not secrete hormones or the products they secrete do not cause a clinical syndrome, such as pancreatic polypeptide, chromogranin A, ghrelin, neurotensin, subunits of chorionic gonadotropin, and neuron-specific enolase. Metastatic disease is a common presentation for most patients with panNETs, especially those with nonfunctioning tumors given the absence of clinical symptoms that would warrant earlier clinical evaluation.


Asymptomatic patients diagnosed with advanced, metastatic panNETs are often monitored initially; however, with time, often their disease will progress and require treatment. The typical indications for therapy are pain and symptoms due to tumor bulk, symptoms from hormone secretion for functional tumors, high tumor burden, or progression of disease under observation. Given the heterogeneous clinical presentations and complex spectrum of aggressiveness of panNETs, their treatment is challenging and requires multimodality management with surgeons, interventional radiologists, medical oncologists, endocrinologists, and gastroenterologists. This article focuses on the data and rationale supporting the use of systemic treatments for advanced, metastatic, well-differentiated panNETs.




Introduction


Well-differentiated neuroendocrine tumors (NETs) are an uncommon and heterogeneous group of neoplasms that arise throughout the body, most commonly in the lung and gastrointestinal tract. These tumors are subdivided into carcinoid tumors and pancreatic NETs (panNETs). Carcinoid tumors develop from the neuroendocrine tissues of the aerodigestive tract, and panNETs develop from the endocrine tissues of the pancreas (ie, islets of Langerhans). This group of well-differentiated NETs is both morphologically and clinically distinct from high-grade neuroendocrine carcinomas, tumors that are characterized by an extremely aggressive behavior and are treated along small cell lung cancer paradigms with platinum-based chemotherapy. Epidemiologic data from the last 30 years have demonstrated that the incidence of NETs continues to increase, although there have been no significant changes in survival from this disease.


panNETs are the second most common tumor of the pancreas and represent 1% to 2% of all pancreatic neoplasms. Although most panNETs are slow growing, after the development of metastatic disease (most commonly in the liver), median survival ranges from 2 to 5 years; most patients with liver metastases will die of the disease. About one-third of panNETs are functional tumors and produce clinical syndromes due to excessive hormone secretion; these functional panNETs are classified by the hormones they hypersecrete and include insulinoma (secrete insulin and cause hypoglycemia), gastrinoma (secrete gastrin and cause Zollinger-Ellison syndrome, which is characterized by severe peptic ulcer disease), glucagonoma (secrete glucagon and cause hyperglycemia), and vasoactive intestinal polypeptide (VIP) (VIPoma, secrete VIP and cause severe secretory diarrhea). Nonfunctional panNETs are tumors that do not secrete hormones or the products they secrete do not cause a clinical syndrome, such as pancreatic polypeptide, chromogranin A, ghrelin, neurotensin, subunits of chorionic gonadotropin, and neuron-specific enolase. Metastatic disease is a common presentation for most patients with panNETs, especially those with nonfunctioning tumors given the absence of clinical symptoms that would warrant earlier clinical evaluation.


Asymptomatic patients diagnosed with advanced, metastatic panNETs are often monitored initially; however, with time, often their disease will progress and require treatment. The typical indications for therapy are pain and symptoms due to tumor bulk, symptoms from hormone secretion for functional tumors, high tumor burden, or progression of disease under observation. Given the heterogeneous clinical presentations and complex spectrum of aggressiveness of panNETs, their treatment is challenging and requires multimodality management with surgeons, interventional radiologists, medical oncologists, endocrinologists, and gastroenterologists. This article focuses on the data and rationale supporting the use of systemic treatments for advanced, metastatic, well-differentiated panNETs.




Pathology


Since 2010, the classification of panNETs has been based on the revised criteria from the World Health Organization, which is defined by the cytologic grade and the proliferative index (as assessed by the Ki-67 and/or mitotic count). In these revised criteria, tumors are broken down by differentiation status (well and poorly differentiated) and grade (grade 1, low; grade 2, intermediate; and grade 3, high). Although the family of well-differentiated tumors are classically of the grade 1 or grade 2 type and generally have a more indolent, less aggressive course, grade 3 or high-grade neuroendocrine carcinomas are typically poorly differentiated and classified as large or small cell carcinomas; these grade 3 neuroendocrine carcinomas are highly aggressive, akin to small cell lung cancers, and are associated with a poor prognosis.


To support this belief, many studies have looked at the relationship between tumor grade and survival; not surprisingly, tumor grade seems to be correlated with survival; in one retrospective analysis of 425 patients with panNETs, the 5-year survival rates for grade 1, grade 2, and grade 3 tumors were 75%, 62%, and 7%, respectively. Unfortunately, this grading system is not universally incorporated into most of the clinical trials investigating panNETs and makes the interpretation of the published data somewhat challenging.


However, recent data also suggest that it may not be correct to consider all grade 3 gastroenteropancreatic (GEP)-NETs as a single entity. Specifically, it has been suggested that some well-differentiated grade 3 NETs may behave differently than poorly differentiated grade 3 NETs. Furthermore, data on treatment outcomes suggest that NETs with a Ki-67 proliferation index in the lower end of the G3 range respond less robustly to chemotherapy agents, such as platinum drugs. In one study, it was shown that grade 3 GEP-NETs with a Ki-67 proliferation index less than 55% were less responsive to first-line platinum-based chemotherapy, though this subgroup achieved longer survival in comparison with grade 3 GEP-NETs with a Ki-67 proliferation index of 55% or greater (14 months vs 10 months). Further corroborating these data, in an investigation of 45 patients specifically with grade 3 panNETs, studying survival and treatment response based on poorly or well-differentiated status, differences were appreciated both in survival and response by therapy type. Specifically, the well-differentiated subgroup demonstrated improved overall survival (OS) in comparison with the poorly differentiated subgroup (52.0 months vs 10.1 months). Looking at responses to therapy, although both poorly and well-differentiated grade 3 panNETs responded to alkylating chemotherapy agents, poorly differentiated tumors had a higher response to platinum agents. Based on the findings of these studies, current research efforts are directed toward a better understanding of grade 3 NETs, both pancreatic and extrapancreatic, as they seem to be a more heterogeneous group than originally thought.




Genetics


Inherited Pancreatic Neuroendocrine Tumors


Although panNETs often develop sporadically, inherited panNETs occur and are generally associated with 4 genetic disorders. These disorders include multiple endocrine neoplasia type 1 (MEN1), von Hippel Lindau (VHL) disease, neurofibromatosis 1 (NF1; von Recklinghausen disease), and tuberous sclerosis complex (TSC). All of these genetic disorders demonstrate autosomal dominant inheritance; additionally, the genes implicated in these disorders (MEN1, VHL, NF1, and TSC1/2) are all tumor suppressor genes and play a critical role in cellular development. The most frequent occurrence of panNETs is in MEN1, followed by VHL, then NF1, and finally TSC.


Nonfamilial (Sporadic) Pancreatic Neuroendocrine Tumors


More recently, effort has been made to better understand the genetic basis of nonfamilial panNETs through whole-exome sequencing; in this study, the exomic sequences of approximately 18,000 protein-coding genes in 10 nonfamilial panNETs were determined (small cell and large cell neuroendocrine carcinomas were excluded in order to ensure the set of tumors studied was clinically homogeneous). The most commonly mutated genes in these 10 tumor samples were then screened in an additional 58 panNETs. In addition to observing an increased number of mutations in genes implicated in chromatin remodeling, mutations in the mammalian target of rapamycin (mTOR) pathway were also identified. Specifically, 44% of the tumors had somatic inactivating mutations in MEN1, the gene that encodes menin, which is a component of the histone methyltransferase complex. Forty-three percent of the tumors had mutations in genes encoding death-domain-associated protein (DAXX) and alpha thalassemia/mental retardation syndrome X-linked (ATRX), these proteins are subunits of a chromatin remodeling complex; no tumor had mutations in both DAXX and ATRX, which was expected given that these proteins function within the same pathway. Approximately 18% of the tumors had mutations along the mammalian target of rapamycin (mTOR) pathway; 7.3% of these mutations were in phosphatase and tensin homolog (PTEN), 8.8% in TSC2, and 1.4% in phosphatidylinositol-4, 5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA).


In the aforementioned study, the patient population was composed of individuals pursuing surgical resection with curative intent, and those with metastatic disease. Looking specifically at survival in different subgroups, prolonged OS was appreciated in patients with mutations in MEN1, DAXX/ATRX, or the combination of both MEN1 and DAXX/ATRX. This survival benefit was most pronounced in those patients with the combination of mutations; in patients with mutations in both MEN1 and DAXX/ATRX, 100% survived for a minimum of 10 years; however, 60% of patients who lacked these mutations died within 5 years of diagnosis.


The aforementioned study has been critical to our understanding of the genomic basis of sporadic panNETs. The findings demonstrate that patients with mutations in chromatin remodeling genes may represent a more favorable panNET subgroup. In addition, this study also highlights the subgroup of patients with panNETs who may demonstrate a more favorable response to the mTOR inhibitor everolimus; further studies to test this hypothesis are ongoing.




Systemic treatment of advanced pancreatic neuroendocrine tumors


The treatment of patients with advanced, metastatic panNETs, whether inherited or sporadic, is approached in a multidisciplinary manner and may include surgical resection, liver-directed therapies, and/or systemic treatments. In unresectable patients, the goals of these therapies are to palliate tumor-related symptoms and prolong the life span.


There are multiple systemic therapy options available for the treatment of metastatic panNETs. These systemic options include somatostatin analogue therapy, targeted agents, and cytotoxic chemotherapy.


Somatostatin Analogues


Somatostatin and its synthetic analogues (ie, octreotide and lanreotide) bind to G-protein couple receptors on the cell surface to exert their effects. There are 5 known subtypes of somatostatin receptors (SST1–SST5), and binding of somatostatin to these receptors can inhibit the release of hormones and secretory proteins and also stall tumor growth, offering cytostatic control.


More than 75% of panNETs express somatostatin receptors (most commonly SST2) on their surface and are octreotide avid on somatostatin analogue scintigraphy (ie, indium-111 pentetreotide [Octreoscan]). In octreotide-positive disease, somatostatin analogues are often used as first line, as they are well tolerated, treat functional symptoms (in those tumors that are hormone secreting), and have been demonstrated to have an antiproliferative, cytostatic effect on the growth of tumors.


Somatostatin analogues and control of symptoms from hormone secretion


Therapy with octreotide and lanreotide has revolutionized the way we care for patients with hormone-producing, functional panNETs. As previously discussed, functional panNETs include insulinomas, gastrinomas, glucagonomas, and VIPomas. Somatostatin analogues seem to be highly useful in the treatment of functional symptoms from VIPomas and glucagonomas, with an improvement seem in secretory diarrhea in VIPomas and an improvement in necrolytic migratory erythema, a characteristic blistering skin rash, in glucagonomas.


Although insulinomas and gastrinomas are the most common types of functional panNETs, somatostatin analogues seem to have a more limited role in controlling their hormone-related symptoms. In particular, when initiating somatostatin analogue therapy on insulinomas, close monitoring of glucose levels is required, as there can be transient worsening of hypoglycemia; hypoglycemia can occur as nearly half of insulinomas do not express SST2 and somatostatin analogue therapy can blunt a compensatory glucagon response. In gastrinomas, rather than somatostatin analogues, proton pump inhibitors are the preferred treatment to blunt the effects of excessive gastric acid production.


Somatostatin analogues and control of tumor growth


In addition to treating hormone-related symptoms in functional tumors, octreotide and lanreotide have a role in controlling tumor growth. The earliest studies investigating a cytostatic role for somatostatin analogues included patients with many types of NETs, questioning the applicability to panNETs alone. The only randomized data to support an antiproliferative role for octreotide in the treatment of NETs came from the phase III Placebo-Controlled, Double-Blind, Prospective, Randomized Study on the Effect of Octreotide LAR in the Control of Tumor Growth in Patients with Metastatic Neuroendocrine Midgut Tumors (PROMID) study, but this study only included midgut tumors and not pancreatic NETs. In this study, 85 patients were randomized to either placebo or octreotide long-acting-release (LAR) 30 mg intramuscularly monthly until progression of disease or death. The primary end point was time to tumor progression (TTP), and the investigators observed a significant difference in TTP in the octreotide LAR and placebo groups (14.3 months vs 6 months, P = .000072). In clinical practice and by the National Comprehensive Cancer Network’s guidelines, physicians were extrapolating the use of octreotide in midgut tumors to use in pancreatic NET; but no prospective randomized data exist.


The Controlled Study of Lanreotide Antiproliferative Response in Neuroendocrine Tumors (CLARINET) study, however, confirmed the antiproliferative effect of somatostatin analogues in GEP-NETs. In this double-blind, placebo-controlled, multinational study in patients with low- or intermediate-grade, moderately or well-differentiated NETs (45% panNETs), 204 patients were randomized to receive an extended-release aqueous-gel formulation of lanreotide (Autogel) at a dose of 120 mg or placebo once every 28 days for 96 weeks; the primary end point was progression-free survival (PFS), defined as time to disease progression or death. The investigators observed that lanreotide was associated with significantly prolonged PFS in comparison with placebo (median not reached vs median of 18 months, P <.001). There were no significant differences between the two groups in quality of life (QOL) or OS. The most common adverse event was diarrhea, which was more prevalent in patients receiving lanreotide (26% in the lanreotide group and 9% in the placebo group). This study confirmed that in comparison with placebo, lanreotide is safely tolerated and significantly prolongs PFS in patients with metastatic NETs; based on these findings, lanreotide is approved by the Food and Drug Administration (FDA) to treat unresectable well-differentiated GEP-NETs. Of note, lanreotide is the only somatostatin analogue FDA approved for cytostatic tumor control in the treatment of NETs; however, octreotide is thought to have similar efficacy in controlling tumor growth.




Targeted therapies


In recent years, targeted agents have been evaluated as therapy options for panNETs. All NETs are known to be highly vascular; for this reason, targeted angiogenic inhibitors have been heavily investigated as a treatment option in the spectrum of NETs. Specific biological agents that have been studied include those directed against vascular endothelial growth factor (VEGF) as well as those targeting mTOR. Sunitinib and everolimus are the only targeted therapies approved for progressive panNETs. Both single and combination therapies have been tested in clinical trials.


Sunitinib


Sunitinib is a small-molecule tyrosine kinase inhibitor that blocks many receptor tyrosine kinases, including VEGF receptor 1 to 3, platelet-derived growth factor receptor (PDGFR), mast/stem cell growth factor receptor (c-KIT), RET, and Fms-Related Tyrosine Kinase 3 (FLT-3). A phase II multicenter study investigated the efficacy of sunitinib in both carcinoid and panNETs (107 treated patients; 41 carcinoid and 66 panNET). Patients were treated with sunitinib 50 mg for 4 weeks followed by a 2-week break. The overall objective response rate in panNETs was 16.7% (11 of 66 patients), and 68% (45 of 66 patients) had stable disease; in panNETs, the median TTP was 7.7 months and 1-year OS was 81.1%. In comparison, only 2.4% of patients with carcinoid tumors achieved a confirmed partial response. The findings suggested that sunitinib has antitumor activity in panNETs; however, a lack of efficacy was seen in carcinoid tumors.


Based on the findings from this phase II study, a multinational, randomized double-blind placebo-controlled phase III trial was conducted of sunitinib in patients with advanced, well-differentiated panNETs. In this study, 171 patients were randomly assigned to either sunitinib 37.5 mg/d or placebo; the study was discontinued early after observation of more serious adverse events and deaths in the placebo group as well as improved PFS in the sunitinib arm. Specifically, the median PFS was 11.4 months in the sunitinib group, compared with 5.5 months in the placebo group. The objective response rate was also improved with sunitinib in comparison with placebo (9.3% vs 0%). Sunitinib was most commonly associated with gastrointestinal side effects (diarrhea, nausea, vomiting) as well as asthenia and fatigue. These positive findings led to the FDA approval of sunitinib in progressive panNETs. In a more recent updated analysis of the survival data, at 2 years of additional follow-up, median OS was 33 months with sunitinib and 26.7 months with placebo ( P = .11).


Everolimus


Everolimus is an oral inhibitor of mTOR, a serine-threonine kinase that impacts cell proliferation, cell survival, and also controls angiogenic pathways via hypoxia-inducible factor-1a, VEGF, and by endothelial and smooth muscle cell proliferation. In the phase II setting, everolimus demonstrated promising antitumor activity in both carcinoid tumors and panNETs. Based on these findings, RAD001 in Advanced Neuroendocrine Tumors, Third Trial (RADIANT 3) Study Group, a randomized double-blind placebo-controlled study of 410 patients with low- or intermediate-grade, progressive, and advanced panNETs, was conducted. In this study, patients were randomized to receive everolimus or placebo, and the investigators observed a significantly improved median PFS of 11.0 months with everolimus compared with 4.6 months with placebo. In the everolimus arm, the response rate was 5%, in comparison with 2% in the placebo arm. The most common adverse events were grade 1 or 2 and included stomatitis, rash, diarrhea, fatigue, and infections. More serious grade 3 or 4 events seen with everolimus included anemia (6%) and hyperglycemia (5%). Based on the RADIANT 3 study, everolimus received FDA approval in the treatment of panNETs. The findings of RADIANT 3 were concordant with those from whole-exome sequencing of panNETs, in which mutations along the mTOR pathway were observed with increased frequency, suggestive that this tumor type may respond to mTOR inhibition.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Sep 16, 2017 | Posted by in HEMATOLOGY | Comments Off on Systemic Therapies for Advanced Pancreatic Neuroendocrine Tumors

Full access? Get Clinical Tree

Get Clinical Tree app for offline access