Next Article in Journal
Multiparametric Whole-Body MRI: A Game Changer in Metastatic Prostate Cancer
Previous Article in Journal / Special Issue
miRNA Expression Profiling in G1 and G2 Pancreatic Neuroendocrine Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunotherapy in Neuroendocrine Neoplasms: A Diamond to Cut

by
Esmeralda García-Torralba
1,2,3,
Esther Garcia-Lorenzo
4,
Bernard Doger
4,
Francesca Spada
5 and
Angela Lamarca
6,7,8,*
1
Department of Medical Oncology, Hospital Universitario Morales Meseguer, 30008 Murcia, Spain
2
Department of Medicine, Medical School, University of Murcia, 30001 Murcia, Spain
3
IMIB-Arrixaca, 30120 Murcia, Spain
4
START Madrid-FJD, Early Phase Clinical Trials Unit, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain
5
European Institute of Oncology, European Institute of Oncology (IEO) IRCCS, 20141 Milan, Italy
6
Department of Oncology, OncoHealth Institute, Fundación Jiménez Díaz University Hospital, 28040 Madrid, Spain
7
Department of Medical Oncology, The Christie NHS Foundation, Manchester M20 4BX, UK
8
Division of Cancer Sciences, University of Manchester, Manchester M13 9PL, UK
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(14), 2530; https://doi.org/10.3390/cancers16142530
Submission received: 5 May 2024 / Revised: 8 July 2024 / Accepted: 10 July 2024 / Published: 13 July 2024
(This article belongs to the Special Issue Neuroendocrine Tumors: From Diagnosis to Therapy)

Abstract

:

Simple Summary

The development of new treatments for patients with neuroendocrine neoplasms (NENs) is imperative. Immunotherapy has shown efficacy in various neoplasms, such as small cell lung cancer and Merkel cell carcinoma. Although immunotherapy’s effectiveness is more limited in NENs, combining immune checkpoint inhibitors with other therapeutic strategies like chemotherapy or targeted therapies could improve outcomes. Additionally, identifying predictive immune biomarkers could enhance patient selection. Our objective was to review the current evidence of immunotherapy in NENs, covering efficacy results and potential predictive biomarkers.

Abstract

A raise in the incidence of NENs is expected. Therefore, the identification of new therapeutic strategies, such as immunotherapy, remains crucial. To date, immune checkpoint inhibitors as monotherapy have shown modest activity in unselected NENs. Although immunotherapy combos (plus another immune agents or chemotherapy, among others) are potentially more active than single agents, this has not been uniformly confirmed, even in high-grade NENs. Other immunotherapeutic strategies under development include bispecific antibodies, targeting specific tumor antigens like DLL3, and cell therapy. Currently, no predictive immune biomarkers are available to guide clinical decisions. A comprehensive tumor molecular profiling approach needs to be developed for the selection of patients with NEN who could potentially benefit from immunotherapy. Ideally, clinical trials should incorporate this tumor molecular profiling to identify predictive biomarkers and improve efficacy. Achieving this goal requires an international collaborative effort.

Graphical Abstract

1. Introduction

Neuroendocrine neoplasms (NENs) are a group of rare epithelial tumors that originate from the neuroendocrine system, and they are widely distributed throughout the body [1]. The global occurrence of NENs is on the rise, affecting all anatomical sites, histological grades, and stages [2]. Although a biological and clinical heterogeneity is the rule for these malignancies, they usually debut as low-grade, non-functioning, advanced-stage neoplasms (with 40–50% of patients having distant metastases at diagnosis) [3].
Currently, the standard treatment for well-differentiated NENs includes surgery, local ablative treatments, agents targeting somatostatin receptors, peptide receptor radionuclide therapy (PRRT), and therapies directed at specific targets [4]. However, emerging treatment approaches, such as exploring immunotherapy, are under investigation for managing NENs.
Immune evasion by tumors plays a crucial role in tumorigenesis and progression. Tumor cells employ various mechanisms to suppress the immune response, with a primary focus on reversing the inhibition of adaptive immunity through a blockade of T cell checkpoint pathways [1]. In this context, immunotherapy has emerged as a way to enhance a patient’s own immune system in combating cancer.
The identification of immune regulatory molecules, such as the PD1/PD-L1 axis, has led to the revolution of immunotherapy in cancer. Several phase III trials using monoclonal antibodies targeting the interaction between the PD-1 checkpoint and its ligands (PD-L1 and PD-L2), or the CTLA-4 checkpoint molecule, have demonstrated durable responses across different histologies [5].
While immunotherapy has transformed the standard of care in many cancers, its efficacy varies widely among histologies and even among patients with the same type of tumor. Moreover, immunotherapy can induce autoimmune side effects, including potentially life-threatening toxicities such as pneumonitis and myocarditis. Some of these side effects can also become chronic (e.g., endocrine disorders), significantly impacting patients’ quality of life. This underscores the need for new predictive and prognostic factors to identify patients who are most likely to benefit from these new therapies [6].
Like many carcinomas of various origins, clinical trials investigating immunotherapy have been conducted in NENs. This review focuses on the clinical efficacy of immunotherapies in NENs and discusses the role of current biomarkers in predicting responses to immune checkpoint inhibitors.

2. Immunotherapy in Neuroendocrine Neoplasms: Current Evidence of Efficacy

Several monotherapies, such as pembrolizumab, avelumab, spartalizumab, and toripalimab, have been evaluated in patients with gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs), yielding inconsistent results (Table 1).

2.1. Pembrolizumab

Pembrolizumab monotherapy was evaluated in two different KEYNOTE trials. The non-randomized phase 1b KEYNOTE-028 “basket” trial assessed pembrolizumab in a large cohort of patients with advanced solid tumors and positive PD-L1 (≥1% membranous expression in tumor or stromal cells). Among them, 25 patients with locally advanced or metastatic well/moderately differentiated gastrointestinal NENs (GI-NENs) and 16 pancreatic NETs (PanNETs) were enrolled. The objective response rate (ORR) was 12.0% and 6.3%, respectively. Interestingly, the median duration of response in the carcinoid cohort was 9.2 months, while it was not reached in the PanNET cohort. Overall survival at the first-year evaluation mark was 87% [7].
The phase 2 KEYNOTE-158 trial enrolled 107 previously treated patients with advanced, unresectable, or metastatic well-differentiated NENs (n = 40 PanNETs, n = 43 GI-NETs, and n = 14 lung NETs). Among them, 15.9% had PD-L1-positive tumors (combined positive score ≥ 1). The efficacy was limited, with an ORR of 3.7%. However, partial response (PR) was observed in 3 patients with PanNETs and in 1 patient with rectal NET [8].

2.2. Avelumab

Although evidence on the efficacy of avelumab in NENs is limited, the phase II AVENEC trial (ClinicalTrials.gov (accessed on 4 May 2024) NCT 03352934) reported an ORR of 6.9% in high-grade NENs from different origins (72.4% GEP). Promising results have been observed in patients with Merkel carcinoma (MCC) [9,19,20]. Avelumab’s activity and efficacy have been extensively studied in the large JAVELIN program, which included both pretreated metastatic MCC patients and those naïve to systemic therapy.
For the cohort of patients treated with avelumab as first-line therapy (n = 116), the ORR was 39.7% and the median OS was 20.3 months [21]. Among patients whose disease had progressed after more than one prior line of therapy (n = 88), the 60-month OS rate was 26%, with a median OS of 12.6 months [22]. The treatment was generally well tolerated. Based on the data from the JAVELIN Merkel 200 trial, avelumab is currently considered the standard of care for metastatic MCC and the only immunotherapeutic agent universally approved for systemic treatment across all NENs.

2.3. Spartalizumab

Spartalizumab was evaluated in a phase II trial that enrolled patients with well-/poorly differentiated NENs, including GI-NETs (n = 32), PanNETs (n = 33), thoracic NETs (n = 30), and gastroenteropancreatic neuroendocrine carcinomas (GEP-NECs, n = 21). The primary endpoint of the study (prespecified activity threshold of ≥10% in the NEN group) was not reached, with a global ORR of 4.8% in the GEP-NEC cohort and 7.4% in the NET group, resulting in the study being considered negative. Notably, a higher ORR was observed in the thoracic NET group (16.7%) compared to the GI (3.1%) and pancreatic (3.0%) cohorts, suggesting a potentially more active role of spartalizumb in thoracic NETs [10].
Patients with ≥1% PD-L1 expression showed a higher ORR, and PD-L1 overexpression was more common in GEP-NECs than in GEP-NENs. Another identified predictive factor was the presence of ≥1% CD8+ cells at baseline [10].

2.4. Toripalimab

The efficacy of the humanized IgG4 anti-PD-1 toripalimab was evaluated in a phase 1b trial involving 40 patients with advanced NENs (Ki-67 ≥ 10%) who progressed after first-line treatment. The ORR was 20% (8 partial responses and 6 stable diseases) and the median duration of response was 15.2 months [11]. As predictors of greater benefit, the authors suggested PD-L1 ≥ 10% (ORR 50.0% vs. 10.7%), a tumor mutational burden (TMB) ≥ 9.9 mut/Mb (ORR 75.0% vs. 16.1%), and/or high microsatellite instability (MSI-H) [11].

3. Dual Therapy

Based on the limited activity of monotherapy strategies mentioned above, combination strategies (immunotherapy combos and combinations of immunotherapy with targeted therapies or chemotherapy) were explored (Table 1).

3.1. Nivolumab Plus Ipilimumab

The combination of ipilimumab and nivolumab has been extensively evaluated in NENs. The prospective, multicenter, open-label, phase II “basket” trial DART SWOG 1609 evaluated the activity and safety of ipilimumab plus nivolumab across multiple rare tumor cohorts. A total of 32 patients with any grade of non-pancreatic primary localization NENs (45% GI-NENs and 18% lung NENs) were included. A 44% ORR was observed in patients with NECs (n = 18) compared to 0% in patients with low- and intermediate-grade NENs (n = 14) [12].
Similarly, in the subgroup analysis of the CA209-538 clinical trial for rare cancers, 29 patients with advanced NENs (excluding patients with small cell lung cancers) were included. This multicenter, open-label, phase II study was conducted in five centers in Australia. Overall, 90% of patients had intermediate- (45%) or high-grade (45%) NENs: 39% lung NENs and 23% GEP-NENs. The ORR was 43% in patients with PanNENs and 33% in those with atypical lung carcinoids [13].
Preliminary results from non-comparative, phase II, NIPINEC trial (ClinicalTrials.gov (accessed on 4 May 2024) NCT 03591731) were recently reported (ESMO annual meeting, 2021). This study evaluated 85 patients with refractory (24% of whom had asymptomatic brain metastases) GEP-NECs (n = 42 were treated with nivolumab, and n = 43 with nivolumab plus ipilimumab). The ORR at 8 weeks was 7.2% with the monotherapy and 14.9% with the combination. Median PFS was 1.8 months and 1.9 months, respectively [23].

3.2. Durvalumab Plus Tremelimumab

The DUNE phase II trial has a design similar to the previously mentioned phase II trial with spartalizumab as a single agent and included 123 patients with NENs divided into four different cohorts: (1) lung carcinoids (n = 27), (2) grade 1-2 GI-NETs (n = 31), (3) grade 1-2 PanNETs (n = 32), and (4) grade 3 GEP-NENs (n = 33, 91% NECs). The 9-month clinical benefit rate (CBR: complete/partial response and stable disease) was the primary endpoint for the cohorts 1–3, and it was 25.9%, 35.5%, and 25%, respectively. A CBR of 36% was observed at a median follow-up of 10.8 months in the PanNEN cohort. The 9-month overall survival (OS) rate was the primary endpoint for the cohort 4 [14,24].

3.3. Nivolumab Plus Platinum-Doublet Chemotherapy

It has been proposed that chemotherapy could increase the tumor mutational burden and immune tumor infiltration, thereby making immunotherapy more effective. Final survival results from the NICE-NEC trial have been recently reported [15]. They enrolled 38 patients with GEP or unknown origin grade 3 NENs who had not been previously treated. Overall, 68.4% had NECs and 65.8% had high Ki-67 (>55%). After a median follow-up of 18.6 months, the ORR was 54.1% and the median PFS was 5.7 months. The 12-month OS rate was 46.7% for high Ki-67 and 59.3% for low Ki-67, respectively [15].

3.4. Nivolumab Plus Temozolamide

Nivolumab combined with temozolomide was explored in a phase II, multi-cohort, single-center, non-randomized trial [16]. This study included 28 patients with NENs (71% NET, 29 NEC) from different primary locations (n = 11 GI-NENs, n = 11 lung NENs, n = 3 PanNENs, n = 1 head and neck NEN and n = 2 unknown NENs). The ORR was 32.1% (n = 9), with a significant difference in responses by primary tumor location (lung vs. others, p = 0.020). The median PFS was 8.8 months. Interestingly, patients who achieved a partial response had lower levels of LAG-3-expressing T cells [16].

3.5. Atezolizumab Plus Bevacizumab

The combination of atezolizumab and bevacizumab was evaluated in an open-label, phase 2 basket trial with 8 parallel rare tumor cohorts [17]. A total of 40 patients with advanced grade 1 to 2 NENs (20 with PanNENs and 20 with extra-pancreatic NENs (ePanNENs)) were included. An ORR was observed in 4 (20%) patients with PanNENs and 3 (15%) patients with ePanNENs. With a safe profile, the median PFS was 14.9 months and 14.2 months, respectively [17].

3.6. Spartalizumab Plus LAG525

Spartalizumab and LAG525 are monoclonal antibodies targeting PD-1 and LAG-3, respectively, which have shown synergistic antitumor activity in preclinical models. In a phase II study of advanced solid tumors, the primary endpoint was reached in the GEP-NEN cohort, with an 86% CBR at 24 weeks [25].

4. Other Immunotherapy Approaches

Additional immunotherapy approaches under investigation include bispecific antibodies or bispecific T-cell engagers, which target both CD3 on lymphocytes and specific tumor antigens like DLL3 simultaneously [26]. Oncolytic viruses are utilized to enhance the responsiveness of immunologically ‘cold’ tumors to immune checkpoint blockades [26]. Further details are provided in Section 8.

5. Combination of Surgical Procedures and Immunotherapy in Neuroendocrine Neoplasms

The integration of surgical procedures and immunotherapy in the treatment of NENs is an evolving field, aimed at improving patient outcomes through a multimodal approach. Combining surgery with immunotherapy can potentially induce an upgraded immune response. Surgical debulking can reduce the immunosuppressive tumor microenvironment, making residual tumors more susceptible to immunotherapy. Additionally, surgery may release tumor antigens into circulation, potentially increasing the immunogenicity of the tumor and improving the effectiveness of immune checkpoint blockades [27].
Early clinical trials and retrospective studies suggest that patients who undergo surgical resection followed by immunotherapy may experience prolonged survival and better disease control [28]. Moreover, neoadjuvant immunotherapy is being investigated for its potential to downstage tumors, making them more resectable and reducing the risk of recurrence [29].
While the combination of surgical procedures and immunotherapy holds promise, challenges remain, including determining the optimal timing and sequencing of these treatments, managing potential adverse effects, and identifying biomarkers to predict response.

6. Predictive Biomarkers

As previously mentioned, the benefit of immunotherapy in NENs remained limited, and not all patients with the same primary origin or extension experience the same response. This highlights the need to identify predictive biomarkers for patient subgroups that would benefit from this treatment strategy.

6.1. PD-L1

The programmed death receptor-1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway has been recognized as crucial for tumors to evade the immune response. PD-L1 expression has been proposed as a predictive biomarker of response to immunotherapy [1]. Consequently, PD-L1 quantification and its implications have been investigated in NENs of different grades and origins.
PD-L1 expression has been associated with high-grade (G3) neoplasms [30]. In a cohort reported by Kim et al., including 15 G2 NENs and 17 G3 NENs, no patients in the first group and 41% of G3 NENs were considered PD-L1-positive (≥1% membranous expression). PD-L1 positivity was significantly associated with undifferentiated NENs [31]. In an independent series, PD-L1 expression was investigated in 68 NEN samples from different origins (37% GEP) with a high proliferation rate (Ki-67 above 20%). The authors found PD-L1 expression in 31.6% of G3 NENs. The proportion of PD-L1-positive tumors was similar across different primary sites but varied based on tumor differentiation (none of the three well-differentiated NENs showed positive expression) and disease extent (more common in patients with locally advanced disease at diagnosis) [32].
In a series of 70 small bowel (sb) NEN specimens (67.1% from primary tumors, 32.9% from metastases), the percentage of PD-L1 positivity (≥5% membranous expression) was found in 12.8% of tumor cells and 24.3% of tumor-infiltrating lymphocytes. Most of the NENs were grade 1 (67.1%) or grade 2 (32.86%), with a median Ki-67 of 2% [6]. Similarly, Da Silva et al. assessed PD-L1 and PD-L2 expressions in a series of well-differentiated GEP-NENs (64 sb-NENs and 31 PanNENs).
PD-L1 positivity (≥5% membranous expression) was found in 12.8% of tumor cells and 24.3% of tumor-infiltrating lymphocytes. Most of the NENs were grade 1 (67.1%) or grade 2 (32.86%), with a median Ki-67 of 2% [6]. Similarly, Da Silva et al. assessed PD-L1 and PD-L2 expressions in a series of well-differentiated NENs.
PD-L1-positive expression (≥5% membranous expression) in tumor cells was low (0% of sb-NENs and 7.4% of PanNENs) [33]. Another series that quantified PD-L1 expression in surgical specimens from sb-NENs, which can provide a more accurate evaluation, found 39% positivity (≥1% membranous expression). Most of the samples were from ileal primary (87%) and grades 1–2 (92%) [34].
Although PD-L1 expression has been proposed as a plausible biomarker for prognosis and predicting improved survival in GEP-NENs [31,35,36], some limitations such as heterogeneous determination, the inclusion of primary and metastatic samples, and the unknown relationship between PD-L1 expression and other immune biomarkers can make data interpretation difficult.

6.2. Tumor Mutational Burden

As previously reported in various studies, the tumor mutational burden (TMB) tends to be low in well-differentiated neoplasms and higher in poorly differentiated carcinomas [37]. TMB has been proposed as a predictive marker for immunotherapy effectiveness [38]. Notably, the KEYNOTE-158 clinical trial (which included five patients with NENs) led to the approval of pembrolizumab for patients with a TMB of ≥10 mutations per megabase (mut/Mb), irrespective of the primary tumor site [39].
However, the universal TMB cut-off did not uniformly predict the same benefit across all tumor types. Possible explanations include the “quality” rather than the quantity of somatic mutations [40] and the dynamic biology with differences between primary tumors and metastatic sites [41].
For instance, a lower TMB of 1.09 mut/Mb in NENs compared to 5.45 mut/Mb in NECs has been reported in a cohort study of 69 NENs and 16 advanced NECs. Investigating the genomic alterations, the NEC cohort exhibited a greater frequency of InDels, structural variants, and polyploid genomes, suggesting a potentially more favorable landscape for immunotherapy when considering TMB as a single biomarker [42]. Similar findings have been replicated in independent cohorts, showing a higher TMB in G3 and NECs compared to well-differentiated NENs [43,44].
Like PD-L1 expression, TMB is not a perfect predictive biomarker of immunotherapy efficacy, but it is another component in the picture of the immune environment of NENs. In a multicenter phase Ib trial by Lu et al., patients with NENs (Ki-67 ≥ 10%) who had failed first line were treated with toripalimab. Among 40 patients, those with PD-L1 ≥ 10% or a high TMB (top 10%) showed a significantly higher ORR (50%), compared to those with PD-L1 expression < 10% (10.7%) and a low TMB (75% vs. 16.1%) [11].
Given the typically low TMB observed in NENs and the potential of the TMB as a biomarker for predicting immunotherapy responses, exploring sequential chemotherapy to induce an increase in the TMB before initiating immunotherapy warrants further investigation [45].

6.3. Immune Cell Infiltration in the Tumor

The presence of immune cells within the neuroendocrine neoplasm (NEN) tumor microenvironment (TME) is pivotal. The TME, which interacts dynamically with tumor cells, influences tumor regression, progression, and response to therapy [46]. The predominant immune cell subgroups extensively studied in the NEN TME include B and T lymphocytes, along with regulatory T cells (Tregs).
Overall, immune cell infiltration appears more pronounced in PanNENs compared to midgut carcinoids [33]. Da Silva et al. reported T cell infiltration in 68% of 87 PanNEN samples, which did not correlate with tumor grade. Takahashi et al. assessed immune cell infiltration across low-grade (n = 32, 61%), intermediate-grade (n = 15, 29%), and high-grade PanNENs (n = 3, 6%), as well as PanNECs (n = 2, 4%) [47]. Their findings indicated that PanNENs exhibited a ‘cold’ immune microenvironment characterized by limited tumor-infiltrating lymphocytes (TILs), whereas high-grade tumors showed abundant TILs [47]. In a cohort of 22 NENs from various primary sites with Ki-67 ≥ 20%, CD3+ T lymphocytes were present in 45.5% of samples, while CD8+ cytotoxic T cells were detected in 18.2% of samples [32].
In a study involving 87 primary tumors and 39 liver metastases from G1/G2 NENs originating from various sites, CD3+ lymphocytes were observed in 68% of primary lesions and 97% of liver metastases [48]. Potential reasons could include progressive immune system activation during tumor progression and accumulation of mutations. Similarly, in patients with grade 2 PanNENs, low infiltration of CD3+ T cells significantly predicted recurrence after tumor resection [48]. Cai et al. documented prolonged disease-free survival (DFS) associated with high CD8+ T cell infiltration in PanNENs, whereas elevated peritumoral CD4+ T cells were linked to worse DFS outcomes [49].
Strong infiltration of lymphocytes near or around tumor cells, coupled with a low density of regulatory T cells (Tregs), has been linked to an enhanced responsiveness to immunotherapy [37]. In a study involving 102 primary sb-NENs of grades 1 and 2, intratumoral immune infiltration was observed in 66% of samples. Approximately one-fifth of these tumors exhibited ectopic lymph nodes with activated germinal centers, which correlated with PD-L1 expression ≥ 50% in tumor cells [34]. In another study by Lamarca et al., including 62 sb-NEN, infiltration of CD8+ lymphocytes was focal in 93% of cases. However, this percentage decreased to 4.29% when considering moderate expression (infiltrates interspersed among tumor cells). Lymphoid aggregates were present in 27% of tumors [6]. The biological significance of these tertiary lymphoid structures in NENs remains ambiguous.
CD4+ FoxP3+ T regulatory (Treg) cells are thought to play a crucial role in immune evasion within patients with cancer. The presence of FoxP3+ cells has been noted to be more prevalent in high-grade compared to low-grade PanNENs [48]. Additionally, higher levels of FoxP3+ Tregs have been linked to poorer prognosis in patients with PanNEN, likely due to their immunosuppressive effects [50]. In another series of NENs, Tregs were identified in 55% of G2/G3 tumors, whereas only 16% of G1 metastatic NENs showed intratumoral Tregs [48]. Among a cohort of 39 metastatic midgut NENs, circulating Treg cells were detected in 6.5% of patients (compared to 3.7% in healthy donors), with levels reaching 9.0% in patients with a high tumor burden [51].
However, gaining a deeper understanding of the immune composition within these diverse tumors is essential to elucidate the prognostic implications of these NEN characteristics. Further insight into the genomic profiles and the NEN tumor microenvironment could aid in stratifying these patients for immunotherapy.

6.4. Notch Signaling Pathway

Notch pathway deregulation has been implicated in tumorigenesis, disease progression, therapeutic resistance, and the suppression of neuroendocrine differentiation. Specifically, DLL3 acts as an inhibitory ligand of the Notch receptor and plays a role in the tumorigenesis of NENs and NECs [52]. In normal tissues, DLL3 is typically expressed at minimal levels, whereas it is overexpressed in approximately 85% of small cell lung cancer (SCLC) cases [52,53]. This observation has led to the proposition of DLL3 as a new biomarker and potential therapeutic target in SCLC.
DLL3 expression has been detected in GEP, bladder, and cervical NENs [54,55,56]. It is notably upregulated in high-grade NENs and NECs [57]. In a cohort of 47 patients with GEP NENs, DLL3 was present in 76.9% of NECs, while it was absent in 5 patients with G3 NENs [54]. DLL3 expression showed a statistically significant association with negative Ga-PET/CT scans and poorer prognosis. Among 155 surgically resected lung NENs, high DLL3 expression was observed in 12.2% of typical carcinoids and 24.4% of atypical carcinoids [57]. While high DLL3 expression is commonly linked to NECs and advanced disease, and is negatively correlated with survival in most studies, its role as a prognostic factor has not been consistently reported [54,58].
In Ali et al.’s series, which included patients with resected lung NENs, DLL3 overexpression was associated with a moderate-to-high inflammatory infiltrate (65.6% vs. 27.7%) [57]. In contrast, some studies in SCLC patients found that DLL3 levels were lowest in the SCLC-inflamed subtype, characterized by the expression of numerous immune checkpoints and human leukocyte antigens [59]. Overall, a deeper understanding of the correlation between DLL3 expression and the immune system, inflammatory biomarkers, and whether DLL3 expression predicts the ICI response is essential.

7. Discussion

This narrative review outlines the landscape of clinical trials exploring the role of immunotherapy in NENs and highlights the main potential biomarkers.
While the incidence of GEP NENs remains relatively low, it is expected to rise in the coming years. At the time of initial diagnosis, many patients presenting with advanced disease require palliative local therapies. The majority of NENs demonstrate poor responsiveness to chemotherapy, making the identification of new therapeutic strategies, such as immunotherapy, crucial [36].
To date, clinical trials evaluating the efficacy of pembrolizumab, avelumab, spartalizumab, and toripalimab as monotherapies for NENs have been reported. While immune checkpoint inhibitors have shown a safety profile consistent with their use in other neoplasms, their efficacy as monotherapy in unselected NENs has been modest, with no complete responses achieved [7,8,10,11,21].
Given these underwhelming results, combination strategies aimed at overcoming the immunosuppressive tumor microenvironment and enhancing the immune response have been investigated. Although immunotherapy combinations show potential for greater efficacy than single-agent treatments, this was not consistently confirmed, even in high-grade NENs. This inconsistency underscores the need for a more precise definition of high-grade categories (differentiating between poorly differentiated and G3 NENs), proliferation index classification, histological subtypes (NENs versus NECs), and overall study design [24].
Despite the investigation of various classical biomarkers (such as PD-L1, TMB, and immune infiltration) and novel biomarkers (such as DLL3) in patients with NENs treated with immunotherapy, no clear correlations have been established. There are significant doubts regarding their predictive and prognostic implications, and they are not recommended for use in clinical practice. It is important to note that most studies evaluating immune biomarkers in NENs are small, retrospective, and not centrally reviewed, thus requiring a cautious interpretation of their results [60].
NENs are inherently challenging due to significant molecular differences among subtypes. While immunotherapy may benefit some patients, comprehensive tumor molecular profiling is essential for selecting potential candidates. Furthermore, specific NEN trials should consider the following factors: primary origin, histological subtype, proliferation index, and molecular background. An attractive strategy currently under evaluation involves combining immunotherapy with bispecific tumor-targeting antibodies, cell therapy, and biological agents like oncolytic viruses to achieve a more accurate immune response [61].
Ideally, clinical trials should incorporate tumor molecular profiling to identify predictive biomarkers that can enhance efficacy. Achieving this goal will require an international collaborative effort.

8. Future Directions

Apart from checkpoint inhibitors, there is increasing interest in bispecific antibodies targeting tumors, adoptive transfer of genetically modified autologous T cells, and oncolytic viruses as promising advancements in cancer immunotherapy. Table 2 summarizes some of the most notable ongoing clinical trials involving immunotherapy for NENs.

9. Conclusions

Immune checkpoint inhibitor monotherapy has shown modest activity for unselected NENs. Although combination immunotherapies are potentially more effective than single agents, this has not been homogeneously confirmed, even in G3-NENs. Other immunotherapeutic strategies in development include bispecific antibodies and oncolytic viruses.
The predictive significance of some biomarkers (including PD-L1 expression, TMB, and DLL3) has been explored, with inconclusive results. The heterogeneity of NENs presents challenges for translational research, but a better understanding of the interplay between local/systemic immunity and the host is essential to identify patients with NENs who may benefit from immunotherapy.

Author Contributions

Conceptualization, E.G.-T., F.S. and A.L.; methodology, E.G.-T., F.S. and A.L.; investigation, E.G.-T., E.G.-L., F.S. and A.L.; writing—original draft preparation, E.G.-T. and A.L.; writing—review and editing, E.G.-T., E.G.-L., B.D., F.S. and A.L.; visualization, E.G.-T., E.G.-L., B.D., F.S. and A.L.; supervision, F.S. and A.L.; project administration, F.S. and A.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of this study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Rizen, E.N.; Phan, A.T. Neuroendocrine Tumors: A Relevant Clinical Update. Curr. Oncol. Rep. 2022, 24, 703–714. [Google Scholar] [CrossRef] [PubMed]
  2. Dasari, A.; Shen, C.; Halperin, D.; Zhao, B.; Zhou, S.; Xu, Y.; Shih, T.; Yao, J.C. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients with Neuroendocrine Tumors in the United States. JAMA Oncol. 2017, 3, 1335–1342. [Google Scholar] [CrossRef] [PubMed]
  3. Borbath, I.; Garcia-Carbonero, R.; Bikmukhametov, D.; Jimenez-Fonseca, P.; Castaño, A.; Barkmanova, J.; Sedlackova, E.; Kollár, A.; Christ, E.; Kaltsas, G.; et al. The European Neuroendocrine Tumour Society Registry, a Tool to Assess the Prognosis of Neuroendocrine Neoplasms. Eur. J. Cancer Oxf. Engl. 1990 2022, 168, 80–90. [Google Scholar] [CrossRef] [PubMed]
  4. Pavel, M.; O’Toole, D.; Costa, F.; Capdevila, J.; Gross, D.; Kianmanesh, R.; Krenning, E.; Knigge, U.; Salazar, R.; Pape, U.-F.; et al. 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, 172–185. [Google Scholar] [CrossRef] [PubMed]
  5. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A Guide to Cancer Immunotherapy: From T Cell Basic Science to Clinical Practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  6. Lamarca, A.; Nonaka, D.; Breitwieser, W.; Ashton, G.; Barriuso, J.; McNamara, M.G.; Moghadam, S.; Rogan, J.; Mansoor, W.; Hubner, R.A.; et al. PD-L1 Expression and Presence of TILs in Small Intestinal Neuroendocrine Tumours. Oncotarget 2018, 9, 14922–14938. [Google Scholar] [CrossRef] [PubMed]
  7. Mehnert, J.M.; Bergsland, E.; O’Neil, B.H.; Santoro, A.; Schellens, J.H.M.; Cohen, R.B.; Doi, T.; Ott, P.A.; Pishvaian, M.J.; Puzanov, I.; et al. Pembrolizumab for the Treatment of Programmed Death-Ligand 1-Positive Advanced Carcinoid or Pancreatic Neuroendocrine Tumors: Results from the KEYNOTE-028 Study. Cancer 2020, 126, 3021–3030. [Google Scholar] [CrossRef] [PubMed]
  8. Strosberg, J.; Mizuno, N.; Doi, T.; Grande, E.; Delord, J.-P.; Shapira-Frommer, R.; Bergsland, E.; Shah, M.; Fakih, M.; Takahashi, S.; et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Neuroendocrine Tumors: Results From the Phase II KEYNOTE-158 Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2124–2130. [Google Scholar] [CrossRef] [PubMed]
  9. Fottner, C.; Apostolidis, L.; Ferrata, M.; Krug, S.; Michl, P.; Schad, A.; Roth, W.; Jaeger, D.; Galle, P.R.; Weber, M.M. A Phase II, Open Label, Multicenter Trial of Avelumab in Patients with Advanced, Metastatic High-Grade Neuroendocrine Carcinomas NEC G3 (WHO 2010) Progressive after First-Line Chemotherapy (AVENEC). J. Clin. Oncol. 2019, 37, 4103. [Google Scholar] [CrossRef]
  10. Yao, J.C.; Strosberg, J.; Fazio, N.; Pavel, M.E.; Bergsland, E.; Ruszniewski, P.; Halperin, D.M.; Li, D.; Tafuto, S.; Raj, N.; et al. Spartalizumab in Metastatic, Well/Poorly-Differentiated Neuroendocrine Neoplasms. Endocr. Relat. Cancer 2021, 28, 161–172. [Google Scholar] [CrossRef]
  11. Lu, M.; Zhang, P.; Zhang, Y.; Li, Z.; Gong, J.; Li, J.; Li, J.; Li, Y.; Zhang, X.; Lu, Z.; et al. Efficacy, Safety, and Biomarkers of Toripalimab in Patients with Recurrent or Metastatic Neuroendocrine Neoplasms: A Multiple-Center Phase Ib Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2337–2345. [Google Scholar] [CrossRef] [PubMed]
  12. Patel, S.P.; Othus, M.; Chae, Y.K.; Giles, F.J.; Hansel, D.E.; Singh, P.P.; Fontaine, A.; Shah, M.H.; Kasi, A.; Baghdadi, T.A.; et al. A Phase II Basket Trial of Dual Anti-CTLA-4 and Anti-PD-1 Blockade in Rare Tumors (DART SWOG 1609) in Patients with Nonpancreatic Neuroendocrine Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2290–2296. [Google Scholar] [CrossRef] [PubMed]
  13. Klein, O.; Kee, D.; Markman, B.; Michael, M.; Underhill, C.; Carlino, M.S.; Jackett, L.; Lum, C.; Scott, C.; Nagrial, A.; et al. Immunotherapy of Ipilimumab and Nivolumab in Patients with Advanced Neuroendocrine Tumors: A Subgroup Analysis of the CA209-538 Clinical Trial for Rare Cancers. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 4454–4459. [Google Scholar] [CrossRef] [PubMed]
  14. Capdevila, J.; Hernando, J.; Teule, A.; Lopez, C.; Garcia-Carbonero, R.; Benavent, M.; Custodio, A.; Garcia-Alvarez, A.; Cubillo, A.; Alonso, V.; et al. Durvalumab plus Tremelimumab for the Treatment of Advanced Neuroendocrine Neoplasms of Gastroenteropancreatic and Lung Origin. Nat. Commun. 2023, 14, 2973. [Google Scholar] [CrossRef] [PubMed]
  15. Riesco Martinez, M.C.; Capdevila Castillon, J.; Alonso, V.; Jimenez-Fonseca, P.; Teule, A.; Grande, E.; Sevilla, I.; Benavent, M.; Alonso-Gordoa, T.; Custodio, A.; et al. Final overall survival results from the NICE-NEC trial (GETNE-T1913): A phase II study of nivolumab and platinum-doublet chemotherapy (CT) in untreated advanced G3 neuroendocrine neoplasms (NENs) of gastroenteropancreatic (GEP) or unknown (UK) origin. Ann. Oncol. 2022, 33 (Suppl. S7), S225–S226. [Google Scholar] [CrossRef]
  16. Owen, D.H.; Benner, B.; Wei, L.; Sukrithan, V.; Goyal, A.; Zhou, Y.; Pilcher, C.; Suffren, S.-A.; Christenson, G.; Curtis, N.; et al. A Phase II Clinical Trial of Nivolumab and Temozolomide for Neuroendocrine Neoplasms. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2023, 29, 731–741. [Google Scholar] [CrossRef] [PubMed]
  17. Halperin, D.M.; Liu, S.; Dasari, A.; Fogelman, D.; Bhosale, P.; Mahvash, A.; Estrella, J.S.; Rubin, L.; Morani, A.C.; Knafl, M.; et al. Assessment of Clinical Response Following Atezolizumab and Bevacizumab Treatment in Patients with Neuroendocrine Tumors: A Nonrandomized Clinical Trial. JAMA Oncol. 2022, 8, 904–909. [Google Scholar] [CrossRef]
  18. Capdevila Castillon, J.; Molina-Cerrillo, J.; Benavent Viñuale, M.; Garcia-Carbonero, R.; Teule, A.; Custodio, A.; Jimenez-Fonseca, P.; López, C.; Hierro, C.; Carmona-Bayonas, A.; et al. 723O—Cabozantinib plus atezolizumab in advanced and progressive neoplasms of the endocrine system: A multi-cohort basket phase II trial (CABATEN/GETNE-T1914). Ann. Oncol. 2023, 34 (Suppl. S2), S498–S502. [Google Scholar] [CrossRef]
  19. Kaufman, H.L.; Russell, J.; Hamid, O.; Bhatia, S.; Terheyden, P.; D’Angelo, S.P.; Shih, K.C.; Lebbé, C.; Linette, G.P.; Milella, M.; et al. Avelumab in Patients with Chemotherapy-Refractory Metastatic Merkel Cell Carcinoma: A Multicentre, Single-Group, Open-Label, Phase 2 Trial. Lancet Oncol. 2016, 17, 1374–1385. [Google Scholar] [CrossRef]
  20. D’Angelo, S.P.; Russell, J.; Lebbé, C.; Chmielowski, B.; Gambichler, T.; Grob, J.-J.; Kiecker, F.; Rabinowits, G.; Terheyden, P.; Zwiener, I.; et al. Efficacy and Safety of First-Line Avelumab Treatment in Patients with Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysis of a Clinical Trial. JAMA Oncol. 2018, 4, e180077. [Google Scholar] [CrossRef]
  21. D’Angelo, S.P.; Lebbé, C.; Mortier, L.; Brohl, A.S.; Fazio, N.; Grob, J.-J.; Prinzi, N.; Hanna, G.J.; Hassel, J.C.; Kiecker, F.; et al. First-Line Avelumab in a Cohort of 116 Patients with Metastatic Merkel Cell Carcinoma (JAVELIN Merkel 200): Primary and Biomarker Analyses of a Phase II Study. J. Immunother. Cancer 2021, 9, e002646. [Google Scholar] [CrossRef] [PubMed]
  22. D’Angelo, S.P.; Bhatia, S.; Brohl, A.S.; Hamid, O.; Mehnert, J.M.; Terheyden, P.; Shih, K.C.; Brownell, I.; Lebbé, C.; Lewis, K.D.; et al. Avelumab in Patients with Previously Treated Metastatic Merkel Cell Carcinoma (JAVELIN Merkel 200): Updated Overall Survival Data after >5 Years of Follow-Up. ESMO Open 2021, 6, 100290. [Google Scholar] [CrossRef] [PubMed]
  23. Girard, N.; Mazieres, J.; Otto, J.; Lena, H.; Lepage, C.; Egenod, T.; Smith, D.; Madelaine, J.; Gérinière, L.; El Hajbi, F.; et al. LBA41—Nivolumab (nivo) ± ipilimumab (ipi) in pre-treated patients with advanced, refractory pulmonary or gastroenteropancreatic poorly differentiated neuroendocrine tumors (NECs) (GCO-001 NIPINEC). Ann. Oncol. 2021, 32 (Suppl. S5), S1283–S1346. [Google Scholar] [CrossRef]
  24. Capdevila, J.; Teule, A.; López, C.; Garcia-Carbonero, R.; Benavent, M.; Custodio, A.; Cubillo, A.; Alonso, V.; Gordoa, T.; Carmona-Bayonas, A.; et al. 1157O A Multi-Cohort Phase II Study of Durvalumab plus Tremelimumab for the Treatment of Patients (Pts) with Advanced Neuroendocrine Neoplasms (NENs) of Gastroenteropancreatic or Lung Origin: The DUNE Trial (GETNE 1601). Ann. Oncol. 2020, 31, S770–S771. [Google Scholar] [CrossRef]
  25. Uboha, N.V.; Milhem, M.M.; Kovacs, C.; Amin, A.; Magley, A.; Purkayastha, D.D.; Piha-Paul, S.A. Phase II Study of Spartalizumab (PDR001) and LAG525 in Advanced Solid Tumors and Hematologic Malignancies. J. Clin. Oncol. 2019, 37, 2553. [Google Scholar] [CrossRef]
  26. Garcia-Carbonero, R.; Anton-Pascual, B.; Modrego, A.; Riesco-Martinez, M.D.C.; Lens-Pardo, A.; Carretero-Puche, C.; Rubio-Cuesta, B.; Soldevilla, B. Advances in the Treatment of Gastroenteropancreatic Neuroendocrine Carcinomas: Are We Moving Forward? Endocr. Rev. 2023, 44, 724–736. [Google Scholar] [CrossRef] [PubMed]
  27. Bakos, O.; Lawson, C.; Rouleau, S.; Tai, L.-H. Combining Surgery and Immunotherapy: Turning an Immunosuppressive Effect into a Therapeutic Opportunity. J. Immunother. Cancer 2018, 6, 86. [Google Scholar] [CrossRef] [PubMed]
  28. Riechelmann, R.P.; Taboada, R.G.; de Jesus, V.H.F.; Iglesia, M.; Trikalinos, N.A. Therapy Sequencing in Patients with Advanced Neuroendocrine Neoplasms. Am. Soc. Clin. Oncol. Educ. Book Am. Soc. Clin. Oncol. Annu. Meet. 2023, 43, e389278. [Google Scholar] [CrossRef] [PubMed]
  29. Topalian, S.L.; Bhatia, S.; Amin, A.; Kudchadkar, R.R.; Sharfman, W.H.; Lebbé, C.; Delord, J.-P.; Dunn, L.A.; Shinohara, M.M.; Kulikauskas, R.; et al. Neoadjuvant Nivolumab for Patients with Resectable Merkel Cell Carcinoma in the CheckMate 358 Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 2476–2487. [Google Scholar] [CrossRef]
  30. Sampedro-Núñez, M.; Serrano-Somavilla, A.; Adrados, M.; Cameselle-Teijeiro, J.M.; Blanco-Carrera, C.; Cabezas-Agricola, J.M.; Martínez-Hernández, R.; Martín-Pérez, E.; Muñoz de Nova, J.L.; Díaz, J.Á.; et al. Analysis of Expression of the PD-1/PD-L1 Immune Checkpoint System and Its Prognostic Impact in Gastroenteropancreatic Neuroendocrine Tumors. Sci. Rep. 2018, 8, 17812. [Google Scholar] [CrossRef]
  31. Kim, S.T.; Ha, S.Y.; Lee, S.; Ahn, S.; Lee, J.; Park, S.H.; Park, J.O.; Lim, H.Y.; Kang, W.K.; Kim, K.-M.; et al. The Impact of PD-L1 Expression in Patients with Metastatic GEP-NETs. J. Cancer 2016, 7, 484–489. [Google Scholar] [CrossRef] [PubMed]
  32. Ferrata, M.; Schad, A.; Zimmer, S.; Musholt, T.J.; Bahr, K.; Kuenzel, J.; Becker, S.; Springer, E.; Roth, W.; Weber, M.M.; et al. PD-L1 Expression and Immune Cell Infiltration in Gastroenteropancreatic (GEP) and Non-GEP Neuroendocrine Neoplasms with High Proliferative Activity. Front. Oncol. 2019, 9, 343. [Google Scholar] [CrossRef] [PubMed]
  33. da Silva, A.; Bowden, M.; Zhang, S.; Masugi, Y.; Thorner, A.R.; Herbert, Z.T.; Zhou, C.W.; Brais, L.; Chan, J.A.; Hodi, F.S.; et al. Characterization of the Neuroendocrine Tumor Immune Microenvironment. Pancreas 2018, 47, 1123–1129. [Google Scholar] [CrossRef] [PubMed]
  34. Cives, M.; Strosberg, J.; Al Diffalha, S.; Coppola, D. Analysis of the Immune Landscape of Small Bowel Neuroendocrine Tumors. Endocr. Relat. Cancer 2019, 26, 119–130. [Google Scholar] [CrossRef] [PubMed]
  35. Bösch, F.; Brüwer, K.; Altendorf-Hofmann, A.; Auernhammer, C.J.; Spitzweg, C.; Westphalen, C.B.; Boeck, S.; Schubert-Fritschle, G.; Werner, J.; Heinemann, V.; et al. Immune Checkpoint Markers in Gastroenteropancreatic Neuroendocrine Neoplasia. Endocr. Relat. Cancer 2019, 26, 293–301. [Google Scholar] [CrossRef] [PubMed]
  36. Kole, C.; Charalampakis, N.; Vailas, M.; Tolia, M.; Sotiropoulou, M.; Tsakatikas, S.; Kouris, N.-I.; Tsoli, M.; Koumarianou, A.; Karamouzis, M.V.; et al. Immunotherapy for Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs): A 2021 Update. Cancer Immunol. Immunother. CII 2022, 71, 761–768. [Google Scholar] [CrossRef] [PubMed]
  37. Cives, M.; Pelle’, E.; Quaresmini, D.; Rizzo, F.M.; Tucci, M.; Silvestris, F. The Tumor Microenvironment in Neuroendocrine Tumors: Biology and Therapeutic Implications. Neuroendocrinology 2019, 109, 83–99. [Google Scholar] [CrossRef] [PubMed]
  38. Strickler, J.H.; Hanks, B.A.; Khasraw, M. Tumor Mutational Burden as a Predictor of Immunotherapy Response: Is More Always Better? Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2021, 27, 1236–1241. [Google Scholar] [CrossRef] [PubMed]
  39. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H.J.; et al. Association of Tumour Mutational Burden with Outcomes in Patients with Advanced Solid Tumours Treated with Pembrolizumab: Prospective Biomarker Analysis of the Multicohort, Open-Label, Phase 2 KEYNOTE-158 Study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef]
  40. Turajlic, S.; Litchfield, K.; Xu, H.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Wong, Y.N.S.; Rowan, A.; Kanu, N.; Al Bakir, M.; et al. Insertion-and-Deletion-Derived Tumour-Specific Neoantigens and the Immunogenic Phenotype: A Pan-Cancer Analysis. Lancet Oncol. 2017, 18, 1009–1021. [Google Scholar] [CrossRef]
  41. Salem, M.E.; Puccini, A.; Grothey, A.; Raghavan, D.; Goldberg, R.M.; Xiu, J.; Korn, W.M.; Weinberg, B.A.; Hwang, J.J.; Shields, A.F.; et al. Landscape of Tumor Mutation Load, Mismatch Repair Deficiency, and PD-L1 Expression in a Large Patient Cohort of Gastrointestinal Cancers. Mol. Cancer Res. MCR 2018, 16, 805–812. [Google Scholar] [CrossRef] [PubMed]
  42. van Riet, J.; van de Werken, H.J.G.; Cuppen, E.; Eskens, F.A.L.M.; Tesselaar, M.; van Veenendaal, L.M.; Klümpen, H.-J.; Dercksen, M.W.; Valk, G.D.; Lolkema, M.P.; et al. The Genomic Landscape of 85 Advanced Neuroendocrine Neoplasms Reveals Subtype-Heterogeneity and Potential Therapeutic Targets. Nat. Commun. 2021, 12, 4612. [Google Scholar] [CrossRef] [PubMed]
  43. Scarpa, A.; Chang, D.K.; Nones, K.; Corbo, V.; Patch, A.-M.; Bailey, P.; Lawlor, R.T.; Johns, A.L.; Miller, D.K.; Mafficini, A.; et al. Whole-Genome Landscape of Pancreatic Neuroendocrine Tumours. Nature 2017, 543, 65–71. [Google Scholar] [CrossRef]
  44. Venizelos, A.; Elvebakken, H.; Perren, A.; Nikolaienko, O.; Deng, W.; Lothe, I.M.B.; Couvelard, A.; Hjortland, G.O.; Sundlöv, A.; Svensson, J.; et al. The Molecular Characteristics of High-Grade Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr. Relat. Cancer 2021, 29, 1–14. [Google Scholar] [CrossRef] [PubMed]
  45. Albertelli, M.; Dotto, A.; Nista, F.; Veresani, A.; Patti, L.; Gay, S.; Sciallero, S.; Boschetti, M.; Ferone, D. Present and Future of Immunotherapy in Neuroendocrine Tumors. Rev. Endocr. Metab. Disord. 2021, 22, 615–636. [Google Scholar] [CrossRef] [PubMed]
  46. Milione, M.; Miceli, R.; Barretta, F.; Pellegrinelli, A.; Spaggiari, P.; Tagliabue, G.; Centonze, G.; Paolino, C.; Mangogna, A.; Kankava, K.; et al. Microenvironment and Tumor Inflammatory Features Improve Prognostic Prediction in Gastro-Entero-Pancreatic Neuroendocrine Neoplasms. J. Pathol. Clin. Res. 2019, 5, 217–226. [Google Scholar] [CrossRef] [PubMed]
  47. Takahashi, D.; Kojima, M.; Suzuki, T.; Sugimoto, M.; Kobayashi, S.; Takahashi, S.; Konishi, M.; Gotohda, N.; Ikeda, M.; Nakatsura, T.; et al. Profiling the Tumour Immune Microenvironment in Pancreatic Neuroendocrine Neoplasms with Multispectral Imaging Indicates Distinct Subpopulation Characteristics Concordant with WHO 2017 Classification. Sci. Rep. 2018, 8, 13166. [Google Scholar] [CrossRef] [PubMed]
  48. Katz, S.C.; Donkor, C.; Glasgow, K.; Pillarisetty, V.G.; Gönen, M.; Espat, N.J.; Klimstra, D.S.; D’Angelica, M.I.; Allen, P.J.; Jarnagin, W.; et al. T Cell Infiltrate and Outcome Following Resection of Intermediate-Grade Primary Neuroendocrine Tumours and Liver Metastases. HPB 2010, 12, 674–683. [Google Scholar] [CrossRef] [PubMed]
  49. Cai, L.; Michelakos, T.; Deshpande, V.; Arora, K.S.; Yamada, T.; Ting, D.T.; Taylor, M.S.; Castillo, C.F.-D.; Warshaw, A.L.; Lillemoe, K.D.; et al. Role of Tumor-Associated Macrophages in the Clinical Course of Pancreatic Neuroendocrine Tumors (PanNETs). Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 2644–2655. [Google Scholar] [CrossRef]
  50. de Reuver, P.R.; Mehta, S.; Gill, P.; Andrici, J.; D’Urso, L.; Clarkson, A.; Mittal, A.; Hugh, T.J.; Samra, J.S.; Gill, A.J. Immunoregulatory Forkhead Box Protein P3-Positive Lymphocytes Are Associated with Overall Survival in Patients with Pancreatic Neuroendocrine Tumors. J. Am. Coll. Surg. 2016, 222, 281–287. [Google Scholar] [CrossRef]
  51. Vikman, S.; Sommaggio, R.; De La Torre, M.; Oberg, K.; Essand, M.; Giandomenico, V.; Loskog, A.; Totterman, T.H. Midgut Carcinoid Patients Display Increased Numbers of Regulatory T Cells in Peripheral Blood with Infiltration into Tumor Tissue. Acta Oncol. Stockh. Swed. 2009, 48, 391–400. [Google Scholar] [CrossRef] [PubMed]
  52. Leonetti, A.; Facchinetti, F.; Minari, R.; Cortellini, A.; Rolfo, C.D.; Giovannetti, E.; Tiseo, M. Notch Pathway in Small-Cell Lung Cancer: From Preclinical Evidence to Therapeutic Challenges. Cell. Oncol. Dordr. 2019, 42, 261–273. [Google Scholar] [CrossRef] [PubMed]
  53. Saunders, L.R.; Bankovich, A.J.; Anderson, W.C.; Aujay, M.A.; Bheddah, S.; Black, K.; Desai, R.; Escarpe, P.A.; Hampl, J.; Laysang, A.; et al. A DLL3-Targeted Antibody-Drug Conjugate Eradicates High-Grade Pulmonary Neuroendocrine Tumor-Initiating Cells in Vivo. Sci. Transl. Med. 2015, 7, 302ra136. [Google Scholar] [CrossRef] [PubMed]
  54. Liverani, C.; Bongiovanni, A.; Mercatali, L.; Pieri, F.; Spadazzi, C.; Miserocchi, G.; Di Menna, G.; Foca, F.; Ravaioli, S.; De Vita, A.; et al. Diagnostic and Predictive Role of DLL3 Expression in Gastroenteropancreatic Neuroendocrine Neoplasms. Endocr. Pathol. 2021, 32, 309–317. [Google Scholar] [CrossRef] [PubMed]
  55. Batista da Costa, J.; Gibb, E.A.; Bivalacqua, T.J.; Liu, Y.; Oo, H.Z.; Miyamoto, D.T.; Alshalalfa, M.; Davicioni, E.; Wright, J.; Dall’Era, M.A.; et al. Molecular Characterization of Neuroendocrine-like Bladder Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 3908–3920. [Google Scholar] [CrossRef]
  56. Yao, J.; Bergsland, E.; Aggarwal, R.; Aparicio, A.; Beltran, H.; Crabtree, J.S.; Hann, C.L.; Ibrahim, T.; Byers, L.A.; Sasano, H.; et al. DLL3 as an Emerging Target for the Treatment of Neuroendocrine Neoplasms. Oncol. 2022, 27, 940–951. [Google Scholar] [CrossRef] [PubMed]
  57. Alì, G.; Di Stefano, I.; Poma, A.M.; Ricci, S.; Proietti, A.; Davini, F.; Lucchi, M.; Melfi, F.; Fontanini, G. Prevalence of Delta-Like Protein 3 in a Consecutive Series of Surgically Resected Lung Neuroendocrine Neoplasms. Front. Oncol. 2021, 11, 729765. [Google Scholar] [CrossRef]
  58. Tanaka, K.; Isse, K.; Fujihira, T.; Takenoyama, M.; Saunders, L.; Bheddah, S.; Nakanishi, Y.; Okamoto, I. Prevalence of Delta-like Protein 3 Expression in Patients with Small Cell Lung Cancer. Lung Cancer Amst. Neth. 2018, 115, 116–120. [Google Scholar] [CrossRef]
  59. Gay, C.M.; Stewart, C.A.; Park, E.M.; Diao, L.; Groves, S.M.; Heeke, S.; Nabet, B.Y.; Fujimoto, J.; Solis, L.M.; Lu, W.; et al. Patterns of Transcription Factor Programs and Immune Pathway Activation Define Four Major Subtypes of SCLC with Distinct Therapeutic Vulnerabilities. Cancer Cell 2021, 39, 346–360.e7. [Google Scholar] [CrossRef]
  60. Fazio, N.; Abdel-Rahman, O. Immunotherapy in Neuroendocrine Neoplasms: Where Are We Now? Curr. Treat. Options Oncol. 2021, 22, 19. [Google Scholar] [CrossRef]
  61. Cives, M.; Pelle’, E.; Strosberg, J. Emerging Treatment Options for Gastroenteropancreatic Neuroendocrine Tumors. J. Clin. Med. 2020, 9, 3655. [Google Scholar] [CrossRef] [PubMed]
Table 1. Clinical trials of immunotherapy in patients with NENs.
Table 1. Clinical trials of immunotherapy in patients with NENs.
Clinical TrialTreatmentTherapyPrimary SiteNMain Outcomes
KEYNOTE-028 [7]PembrolizumabMonotherapyAdvanced PD-L1+ carcinoids or PanNENs25 GI-NETs and 16 PanNENsORR: 12.0% (GI-NENs) and 6.3% (PanNENs)
KEYNOTE-158 [8]MonotherapyAdvanced, well-differentiated NENs40 PanNETs, 43 GI-NETs and 14 lung NETsORR: 3.7%
AVENEC [9]AvelumabMonotherapyAdvanced G3 NECs29 NENs from different origins (72.4% GEP)ORR: 6.9%
Yao et al. [10]SpartalizumabMonotherapyAdvanced thoracic/GEP-NENs and GEP-NECs31 GI-NETs, 33 PanNETs, 30 thoracic NETs and 21 GEP-NECsORR: 7.4% (NENs) and 4.8% (NECs)
Lu et al. [11]ToripalimabMonotherapyAdvanced NENs (Ki-67 ≥ 10%)8 WD-NENs and 32 NECs; 9 PanNENs and 31 ePanNENsORR: 25% (WD-NENs) and 18.7% (NECs)
DART SWOG 1609 [12]Nivolumab and ipilimumabDual therapy (anti-PD1–anti-CTLA4)Advanced, any grade NENs (excluding PanNENs)14 WD-NENs and 18 NECs; 45% GI-NENs and 18% lung NENsORR: 0% (WD-NENs) and 44% (NECs)
CA209-538 [13]Nivolumab and ipilimumabDual therapy (anti-PD1–anti-CTLA4)Advanced, any grade NENs29 NENs; 90% had G2 or G3; 39% lung NENs and 23% GEP-NENsORR: 24%; 43% in PanNENs, and 33% for atypical lung carcinoids
DUNE [14]Durvalumab and tremelimumabDual therapy (anti-PDL1–anti-CTLA4)Cohort 1: G1/2 lung NETs—Cohort 2: G1/G2 GI-NETs—Cohort 3: G1/2 PanNENs—Cohort 4: G3 GEP-NENs27 lung carcinoids, 31 grade 1-2 GI-NETs, 32 grade 1-2 PanNETs and 33 grade 3 GEP-NENs (91% NECs)ORR: 11.1% (cohort 1), 0% (cohort 2), 6.3% (cohort 3) and 9.1% (cohort 4)
NICE-NEC trial [15]Nivolumab and platinum-based chemotherapyDual therapy (anti-PD1–chemotherapy)GEP or unknown origin G3 NENs38 NENs; 68.4% NECsORR: 54.1%
Owen et al. [16]Nivolumab and temozolomideDual therapy (anti-PD1–chemotherapy)Advanced, any grade NENs28 NENs; 3 PanNENs, 11 lung NENs, 11 GI-NENs, 1 head and neck NENs and 2 unknown NENsORR: 32.1% (lung vs. others, p = 0.020)
Halperin D et al. [17]Atezolizumab and bevacizumabDual therapy (anti-PDL1–antiangiogenic)Advanced, G1/2 NENs40 NENs; 20 PanNENs and 20 ePanNENsORR: 20% PanNENs and 15% ePanNENs
CABATEN/GETNE-T1914 [18]Atezolizumab and cabozantinibDual therapy (anti-PDL1–kinase inhibitor)Cohort 1: WD lung NENs—Cohort 2: ATC—Cohort 3: ACC—Cohort 4: PPGL—Cohort 5: WD GEP NENs—Cohort 6: G3 extrapulmonary NENs9 WD lung NENs, 14 ATC, 24 ACC, 13 PPGL, 24 WD GEP NENs and 9 G3 extrapulmonary NENsORR: 0% (cohort 1), 21.4% (cohort 2), 8.3% (cohort 3), 7.7% (cohort 4), 16.7% (cohort 5) and 0% (cohort 6)
ACC: adrenocortical carcinoma. ATC: anaplastic thyroid cancer. ePanNENs: extra-pancreatic neuroendocrine neoplasms. G: grade. GEP: gastro-entero-pancreatic. GI: gastrointestinal. NEC: neuroendocrine carcinomas. NENs: neuroendocrine neoplasms. ORR: overall response rate. PanNENs: pancreatic neuroendocrine neoplasms. PPGL: pheochromocytoma/ paraganglioma. WD: well differentiated.
Table 2. Ongoing clinical trials involving immunotherapy combination in NENs.
Table 2. Ongoing clinical trials involving immunotherapy combination in NENs.
Clinical Trials.gov IdentifierStudy TitlePhaseNEN PopulationTreatment ArmsPrimary Endpoint
03457948Pembrolizumab With Liver-Directed or Peptide Receptor Radionuclide Therapy for Neuroendocrine Tumors and Liver MetastasesIIAdvanced grade 1–2 NENs
-
Arm 1: pembrolizumab + 177Lu-DOTATATE (4 sessions).
-
Arm 2: pembrolizumab + arterial embolization, 3–7 days following the first dose of pembrolizumab.
-
Arm 3: pembrolizumab + yttrium-90 microsphere radio embolization, 3–15 days following the first dose of pembrolizumab.
ORR
04525638A Clinical Study to Assess the Combination of Two Drugs (177Lu-DOTATATE and Nivolumab) in Neuroendocrine TumorsIIAdvanced, well-differentiated
grade 3 NENs or NECs of the pancreas, GI, lung, and unknown primary
All subjects will be treated with nivolumab + 177-Lu-DOTATATE (maximum 4 cycles).ORR
05058651Evaluating the Addition of the Immunotherapy Drug Atezolizumab to Standard Chemotherapy Treatment for Advanced or Metastatic Neuroendocrine Carcinomas That Originate Outside the LungII/IIIAdvanced extrapulmonary NECs
-
Arm 1: atezolizumab + cisplatin/carboplatin + etoposide (4 cycles). Maintenance: atezolizumab.
-
Arm 2: atezolizumab + cisplatin/carboplatin + etoposide (4 cycles). Observation for 1 year.
-
Arm 3: cisplatin/carboplatin + etoposide (4 cycles). Observation for 1 year.
OS
04969887Combination Immunotherapy in Rare Cancers Under InvesTigation (MOST-CIRCUIT)IIAtypical bronchial carcinoid, NECs, and grade 3 NENs independent of primary site (SCLC excluded)All subjects will be treated with nivolumab + ipilimumab at concurrently (4 cycles). Nivolumab until progression (up to 2 years).6 months PFS and ORR
01174121Immunotherapy Using Tumor Infiltrating Lymphocytes for Patients with Metastatic Cancer IIMetastatic NENs
-
Arm 1: CD8+-enriched TIL—cyclophosphamide and fludarabine (C+F) + young CD8+-enriched TIL + high-dose aldesleukin.
-
Arm 2: unselected TIL—C+F + young, unselected TIL + high-dose aldesleukin.
-
Arm 3: unselected TIL—C+F + young, unselected TIL + high-dose aldesleukin + pembrolizumab prior to cell administration and 3 additional doses following cell infusion.
-
Arm 4: unselected TIL—C+F + young, unselected TIL + high-dose aldesleukin + pembrolizumab within 4 weeks of progressive disease (for up to 8 doses).
ORR
03412877Administration of Autologous T-Cells Genetically Engineered to Express T-Cell Receptors Reactive Against Neoantigens in People with Metastatic CancerIINENs refractory to second-vb line therapy
-
Arm 1: cyclophosphamide and fludarabine + individual patient TCR-transduced PBL + high- or low-dose aldesleukin.
-
Arm 2: cyclophosphamide and fludarabine + individual patient TCR-transduced PBL + high- or low-dose aldesleukin + pembrolizumab prior to cell administration and 3 additional doses following cell infusion.
ORR
05882058A Study to Test Whether Different Doses of BI 764532 Help People with Small Cell Lung Cancer or Other Neuroendocrine CancersIISCLC, epNECs, LCNECDrug: BI 764532 (DLL3/CD3 T cell engaging bispecific antibody—two different doses).OR
04702737A Study of AMG 757 in Participants with Neuroendocrine Prostate CancerINeuroendocrine prostate cancerDrug: tarlatamab (bispecific T-cell engager molecule, binds both DLL3 and CD3—dose exploration/expansion).Security
5652686A Phase 1 Study of PT217 in Patients with Advanced Refrac-tory Cancers Expressing DLL3ILCNEC, neuroendocrine prostate cancer and GEP-NECDrug: PT217 (bispecific antibody (bsAb) against human DLL3 (huDLL3) and human CD47 (huCD47))Security
5879978A Study to Test How Well Different Doses of BI 764532 in Combination with Ezabenlimab Are Tolerated by People with SCLC and Other Neuroendocrine Tumos That Are Positive for DLL3ILCNEC and NEC of any originDrug: BI 764532 (a DLL3)/CD3 IgG-like T-cell engager)
Drug: ezabenlimab (anti-PD-1 mAb)
MTD
4471727Study in Patients with Advanced Cancers Associated with Expression of DLL3I/IINeuroendocrine prostate cancer, high grade neuroendocrine tumor types other than SCLCDrug: HPN328 (tri-specific DLL3-targeting T-cell engager—dose exploration/expansion)
Drug: atezolizumab (combination)
Security
4429087A Study to Test Different Doses of BI 764532 in Patients with SCLC and Other Neuroendocrine
Tumours That Are Positive for DLL3
ILCNEC and NEC Drug: BI 764532 (a DLL3)/CD3 IgG-like T-cell engager)MTD
epNECs: extra-pulmonary neuroendocrine carcinoma. GEP-NEC: gastroenteropancreatic neuroendocrine carcinoma. LCNEC: large cell neuroendocrine carcinoma. MTD: maximum tolerated dose. NECs: neuroendocrine carcinomas. NENs: neuroendocrine neoplasms. OR: objective response. ORR: overall response rate. OS: overall survival. PFS: progression-free survival. SCLC: small cell lung cancer.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

García-Torralba, E.; Garcia-Lorenzo, E.; Doger, B.; Spada, F.; Lamarca, A. Immunotherapy in Neuroendocrine Neoplasms: A Diamond to Cut. Cancers 2024, 16, 2530. https://doi.org/10.3390/cancers16142530

AMA Style

García-Torralba E, Garcia-Lorenzo E, Doger B, Spada F, Lamarca A. Immunotherapy in Neuroendocrine Neoplasms: A Diamond to Cut. Cancers. 2024; 16(14):2530. https://doi.org/10.3390/cancers16142530

Chicago/Turabian Style

García-Torralba, Esmeralda, Esther Garcia-Lorenzo, Bernard Doger, Francesca Spada, and Angela Lamarca. 2024. "Immunotherapy in Neuroendocrine Neoplasms: A Diamond to Cut" Cancers 16, no. 14: 2530. https://doi.org/10.3390/cancers16142530

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop