Next Article in Journal
Intravenous Idursulfase for the Treatment of Mucopolysaccharidosis Type II: A Systematic Literature Review
Previous Article in Journal
Mechanism of the Oxidative Ring-Closure Reaction during Gliotoxin Biosynthesis by Cytochrome P450 GliF
Previous Article in Special Issue
Analysis of Carcinogenic Involvement of MicroRNA Pattern in Peripheral Non-Cancerous Tissues and Chronic Viral Liver Injury
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of Altered Pathways Associated with Sensitivity to Platinum-Based Chemotherapy in Neuroendocrine Tumors: Strengths and Prospects

by
Erika Stefàno
,
Federica De Castro
,
Antonella Ciccarese
,
Antonella Muscella
,
Santo Marsigliante
,
Michele Benedetti
* and
Francesco Paolo Fanizzi
Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Monteroni, I-73100 Lecce, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(16), 8568; https://doi.org/10.3390/ijms25168568
Submission received: 30 May 2024 / Revised: 26 July 2024 / Accepted: 2 August 2024 / Published: 6 August 2024
(This article belongs to the Collection Feature Papers in Molecular Oncology)

Abstract

:
Neuroendocrine neoplasms (NENs) are a diverse group of malignancies with a shared phenotype but varying prognosis and response to current treatments. Based on their morphological features and rate of proliferation, NENs can be classified into two main groups with a distinct clinical behavior and response to treatment: (i) well-differentiated neuroendocrine tumors (NETs) or carcinoids (with a low proliferation rate), and (ii) poorly differentiated small- or large-cell neuroendocrine carcinomas (NECs) (with a high proliferation rate). For certain NENs (such as pancreatic tumors, higher-grade tumors, and those with DNA damage repair defects), chemotherapy is the main therapeutic approach. Among the different chemotherapic agents, cisplatin and carboplatin, in combination with etoposide, have shown the greatest efficacy in treating NECs compared to NETs. The cytotoxic effects of cisplatin and carboplatin are primarily due to their binding to DNA, which interferes with normal DNA transcription and/or replication. Consistent with this, NECs, which often have mutations in pathways involved in DNA repair (such as Rb, MDM2, BRCA, and PTEN), have a high response to platinum-based chemotherapy. Identifying mutations that affect molecular pathways involved in the initiation and progression of NENs can be crucial in predicting the response to platinum chemotherapy. This review aims to highlight targetable mutations that could serve as predictors of therapeutic response to platinum-based chemotherapy in NENs.

1. Introduction

Neuroendocrine neoplasms (NENs) are a diverse group of malignancies that originate from neuroendocrine (NE) cells, which are characterized by both “neuro” and “endocrine” properties. These cells release hormones into the bloodstream in response to nervous system stimulation [1]. The classification of neuroendocrine neoplasms is based on the primary site of origin, proliferation index (Ki-67), and symptoms caused by the production of biologically active amines (functioning and non-functioning NENs) [2].
Since NE cells are found throughout the body, neuroendocrine tumors can arise in various tissues, including the skin, nervous system, respiratory tract, gastrointestinal tract, larynx, thyroid, breast, and urogenital system. The neuroendocrine system includes endocrine glands (such as the parathyroid, pituitary, and adrenal glands), as well as endocrine islet tissue embedded within glandular tissue (such as the thyroid or pancreas) and scattered cells in the exocrine parenchyma (such as the endocrine cells of the digestive and respiratory tracts in the diffuse endocrine system) [1,3]. The most common primary tumor sites for neuroendocrine neoplasms are the gastroenteropancreatic (GEP) and bronchopulmonary (BP) tracts, although they can develop in any organ or system in the human body, with similar features due to their neuroendocrine nature (Figure 1).
Neuroendocrine neoplasms (NENs) can also be classified based on the specific hormones they secrete [1,4,5,6,7,8]. In fact, they can oversecrete bioactive substances that regulate certain body functions, which results in a clinical syndrome known as carcinoid syndrome. Tumors associated with this syndrome are currently defined as “carcinoids”, which are well established and distinct clinical entities [9]. The degree of biological aggressiveness and response to therapies of NENs [7,10] is influenced by their secretory properties and syndromes of uncontrolled hormone hypersecretion (such as Cushing, Verner–Morrison, Zollinger–Ellison, and other eponymic syndromes). Therefore, the challenging management of NENs is due to their heterogeneous clinical presentations and varying degrees of aggressiveness [11].
In order to standardize the nomenclature of NENs, the 2015 World Health Organization (WHO) proposed a universal definition system based on mitotic count and/or Ki-67 index and/or the presence of necrosis, classifying NENs into three tiers (grades 1–3). Based on this concept, NENs are divided into well-differentiated neuroendocrine tumors (NETs) (G1, G2 and G3 grade) and poorly differentiated neuroendocrine carcinomas (NECs), which are high-grade neoplasms (G3) (Figure 1) [7,10]. NETs and NECs have different risk factors, hereditary predispositions, relationships to non-NEN, and genetic factors (for example, NECs are most frequently characterized by p53 and Rb gene alterations compared to NETs) [7,10]. Furthermore, NECs should be distinguished from carcinoids: both are composed of chromogranin-positive neuroendocrine cells but, while carcinoids are low-grade malignancies, NECs are highly aggressive malignancies [12]. These types of neoplasms can also be composed of different combinations of NENs (NET or NEC) and non-NENs, which are referred to as mixed NENs/non-NENs (MiNENs) [13].
Neuroendocrine neoplasms can also be classified by identifying conventional biomarkers of NE lineage and differentiation, which are useful in distinguishing G3 NETs from NECs, epithelial from non-epithelial NENs, and so on. Some examples of biochemical markers for NENs include chromogranin A (CgA), pancreatic polypeptide (PP), human chorionic gonadotropin (HCG), alpha-fetoprotein (AFP), neuron specific enolase (NSE) [10,14], insulinoma-associated protein-1 (INSM-1), synaptophysin (SYN), and somatostatin receptors (SSTRs). Transcription factors (e.g., thyroid transcription factor-1, TTF-1; Islet 1, Isl-1; paired box 8, PAX 8), enzymes, keratins, and hormones can also be useful in functional and structural correlation. For example, insulinomas, glucagonomas, gastrinomas, somatostatinomas, and vipomas are named after the hormones they produce (insulin, glucagon, gastrin, somatostatin, and vasoactive intestinal peptide (VIP), respectively) [7,14,15,16].
Generally, NENs are sporadic, but they may also arise due to hereditary syndromes that predispose individuals to the onset of neuroendocrine neoplasms, such as multiple endocrine neoplasia type 1 and 2 (MEN-1 and MEN-2 hereditary cancer syndromes), von Hippel–Lindau (VHL) syndrome, neurofibromatosis, and tuberous sclerosis [1,17]. NENs can also be associated with mutations in different pathway genes, as rearranged during transfection (RET) proto-oncogene, mTOR (mammalian target of rapamycin), and VEGF (vascular endothelial growth factor)/VEGF-receptor pathway genes [2,18]. The PI3K-Akt-mTOR (phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin) pathway plays an important role in NENs. In fact, mTOR inhibitors (rapamycin analogs) have been approved by the FDA (Food and Drug Administration) for the treatment of advanced pancreatic NETs [19]. The treatment of NENs generally involves a variety of therapeutic approaches (surgery, radiotherapy, immunotherapy, molecular-targeted agents, and chemotherapy), depending on the tumor grade and differentiation [7,20] (Figure 1).
Currently, platinum-based chemotherapy is considered a first-line palliative treatment for various types of tumors, including NENs [21,22,23]. For poorly differentiated NECs, such as gastroenteropancreatic neuroendocrine cancers (GEP-NECs) and bronchopulmonary neuroendocrine cancers (BP-NECs), the most effective therapy involves the use of cisplatin or carboplatin in combination with etoposide (Figure 2) [21]. Despite the widespread use of platinum agents in various tumor types, their mechanism of action in NE tumor cells remains unclear.
This review stems from the need to better understand the predictive and prognostic features of NENs. These features remain uncertain, making it crucial to identify the major molecular genetic alterations in each type of tumor. Establishing correlations between specific genetic abnormalities involved in tumorigenesis and metastasis could reveal potential targets for cancer therapy. Due to the heterogeneity of NENs, there is a significant need to re-evaluate chemotherapeutic approaches, focusing on combined and personalized therapies that offer greater selectivity and effectiveness. The review incorporates recent and relevant studies in the field, providing a comprehensive analysis of current knowledge and developments in NENs and responses to platinum-based chemotherapy. This analysis considers data and research findings up to the year 2024 and discusses the role of altered molecular pathways in NENs that contribute to sensitivity to platinum drugs. These genomic alterations are typically involved in tumor initiation and progression and serve as biomarkers for predicting therapeutic outcomes. Therefore, the purpose of this work is to identify the main affected signaling pathways in NENs and their relationship to response to platinum chemotherapy.

1.1. Most Common Neuroendocrine Neoplasms

1.1.1. Gastroenteropancreatic Neuroendocrine Neoplasms (GEP-NENs)

Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) affect organs of the gastrointestinal tract. Approximately 30% of GEP-NENs are hormonally active and can produce and secrete peptides and neuroamines causing specific clinical syndromes. Poorly differentiated neuroendocrine carcinomas (NECs) account for 10–20% of malignant GEP-NENs and are primarily found in the esophagus, pancreas, ampulla of Vater, large bowel, and rarely in the ileum (Figure 1) [24].
Their clinical features mainly depend on the primary site of the tumor and its functionality [25]. Based on the primary tumor site, GEP-NENs are divided into two sub-categories: carcinoid tumors of the luminal gastrointestinal (GI) tract and pancreatic (P) neoplasms [1,6]. Among GEP neoplasms, pancreatic NETs (P-NETs) account for approximately 1% of pancreatic cancers. An estimated 40–91% of P-NETs are non-functioning, while the others manifest evident hormonal symptoms (e.g., insulinoma, gastrinoma, glucagonoma, vipoma, somatostatinoma) [26,27]. GI-NETs and P-NETs may have similar histological features but variable clinical behavior and biology. P-NETs have a relatively worse prognosis than GI-NETs and respond differently to therapies. Several agents have shown higher response rates in P-NETs compared to GI-NETs [28,29,30].

1.1.2. Bronchopulmonary Neuroendocrine Neoplasms (BP-NENs)

Similar to GEP-NENs, bronchopulmonary neuroendocrine tumors (BP-NENs) are classified based on morphology and/or mitotic count. The World Health Organization (WHO) classification (2015) groups both, lung and thymic neuroendocrine tumors (NETs) (referred to as neoplasms in the digestive WHO classification) into one category, subdivided into two main groups: (a) BP-NETs, which include low-grade typical carcinoid (TC) (0–1 mitoses per 2 mm2) and intermediate-grade atypical carcinoid (AC) (2–10 mitoses per 2 mm2), and (b) BP-NECs, which consist of large-cell neuroendocrine carcinoma (LCNEC) and small-cell lung carcinoma (SCLC) (≥11 mitoses per 2 mm2) (Figure 1) [6,31].
The accurate identification and differentiation of AC from TC or NECs (LCNEC and SCLC) is crucial for determining treatment options and prognosis [32]. Currently, the standard treatment for patients with SCLC involves platinum-based chemotherapy combined with an immune checkpoint inhibitor (ICI) such as atezolizumab or durvalumab [29] (Figure 1). The histological features of LCNEC can overlap with non-small-cell lung cancer (NSCLC) and, in some cases, SCLC, making a histological diagnosis challenging and requiring the establishment of an optimal systemic treatment. On the other hand, chemotherapy for SCLC is considered the most appropriate treatment (Figure 1) [33,34].

2. Approved Therapeutic Options for Neuroendocrine Neoplasms

As previously discussed, neuroendocrine neoplasms (NENs) encompass a wide range of tumors with varying biological and clinical features. As a result, there has been a noticeable expansion in therapeutic options for NENs in recent years, particularly for well-differentiated NETs [6]. NENs can be differentiated based on laboratory tests (secretory vs. non-secretory), clinical symptoms (functioning vs. non-functioning), morphological features (growth patterns of cancer cells, mitotic counts, Ki-67 index, necrosis, expression of somatostatin receptors, SSTRs), and the degree of cellular differentiation. Due to the significant differences in clinical behavior between G1 and G2 NETs compared to G3 NETs, treatment approaches vary between these two malignancies [35]. Additionally, G3 NETs have distinct biological features compared to poorly differentiated G3 neuroendocrine carcinomas (NECs) [28,36,37,38].
The identification of altered pathways involved in the pathogenesis of NENs has led to the development of specific therapies [39]. Surgery is the preferred option for resectable tumors, while for inoperable or metastatic disease, therapeutic options include radiation therapy, somatostatin analogs (SSAs), mTOR inhibitors, receptor tyrosine kinase receptors inhibitors (TKIs), chemotherapy, peptide receptor radionuclide therapy (PRRT), and targeted therapy (Figure 1) [40,41].
In recent years, molecular-targeted therapies have emerged as a treatment approach for advanced neuroendocrine tumors (NETs) [42]. Among these therapies, somatostatin analogs (SSAs) have been shown to delay tumor progression and decrease hormone overproduction by interacting with somatostatin receptors (SSTRs), which are often overexpressed in NETs [43]. Somatostatin (SST) plays a role in regulating cell growth and hormone secretion, making SSTRs a potential target for treating neuroendocrine neoplasms (NENs) [43]. The first synthetic SSA to be approved by the FDA was octreotide, an octapeptide available in both conventional and long-acting release (LAR) injections (approved in 1988 and 1998, respectively). Lanreotide was initially approved for treating acromegaly but has since been approved for treating unresectable, well- or moderately differentiated, locally advanced, or metastatic gastroenteropancreatic (GEP) NETs (in 2014) and carcinoid syndrome (in 2017).
Another type of targeted therapy is represented by everolimus and sunitinib, which were approved by the FDA in 2011. Everolimus is an mTOR inhibitor that plays a role in the tumorigenesis and progression of NENs, while sunitinib is a multi-targeted tyrosine kinase inhibitor (TKI) that blocks the activation of VEGFRs 1–3 (vascular endothelial growth factor receptor 1–3), PDGFR-α and -β (platelet-derived growth factors-α and -β), KIT (stem-cell growth factor receptor), FLT3 (fms-related tyrosine kinase 3), RET and CSF1R (colony-stimulating factor receptor 1), resulting in antiangiogenic and antitumor activity against a broad range of neoplasms. The FDA has approved these targeted agents for managing advanced GEP-NETs: everolimus for both primary gastrointestinal NETs (GI-NETs) and pancreatic NETs (P-NETs) and sunitinib for primary P-NETs [44,45,46,47]. Vandetanib and cabozantinib are TKIs currently used to treat patients with unresectable, progressive, and symptomatic medullary thyroid carcinoma (MTC) [48,49,50] (Figure 1).
Peptide receptor radionuclide therapy (PRRT) is a treatment option for patients with progressive somatostatin receptor (SSTR)-positive neuroendocrine tumors (NETs) [43,51]. PRRT involves attaching a radioisotope to a chelating molecule, which is then linked to a peptide that targets SSTRs on the surface of cancer cells. This allows for the precise delivery of radiation to the tumor. In the case of advanced NETs, the peptide used can be a somatostatin analog (SSA) or antagonist that binds to SSTRs. In 2018, the FDA approved the radiopharmaceutical lutetium (177Lu) for the treatment of patients with SSTR-positive gastroenteropancreatic NETs (GEP-NETs) (Figure 1) [52].
Immunotherapy is another treatment option for neuroendocrine neoplasms (NENs), that has achieved significant success in treating small-cell lung cancer (SCLC) and Merkel cell carcinoma (MCC) [53]. CTLA-4 (cytotoxic T-lymphocyte antigen 4), PD-1 (programmed death-1) and PD-L1 (programmed death-1 ligand) are involved in cancer cells evading immune surveillance. Immune checkpoint inhibitors (ICIs) are antibodies that target these molecules and have proven effective in various cancers, included NENs. Avelumab and pembrolizumab are the only two FDA-approved ICIs for treating metastatic MCC, a rare and aggressive neuroendocrine tumor of the skin (Figure 1) [53,54,55].
Cytotoxic chemotherapy is the standard treatment for patients with poorly differentiated neuroendocrine carcinomas (NECs), but its role in patients with well-differentiated neuroendocrine tumors (NETs) is not well defined [56]. Streptozotocin (STZ) is an alkylating agent that was the first drug to show efficacy in NETs.
It was approved in 1982 for treating pancreatic NETs. Subsequent studies have shown that STZ in combination with 5-fluorouracil (5-FU) and/or doxorubicin is effective for treating well-differentiated pancreatic NETs (P-NETs) [57,58,59,60]. In 2021, the FDA approved belzutifan, a hypoxia-inducible factor inhibitor (HIF-2α), for certain types of cancers, including P-NETs associated with von Hippel-Lindau disease [61]. Recently, the combination therapy of capecitabine and temozolomide (CAPTEM) showed improved anticancer activity in metastatic P-NETs [62] (Figure 1).
Among the chemotherapeutic agents mentioned, cisplatin/carboplatin-based therapy is the first-line regimen (Figure 1 and Figure 2). However, not all platinum compounds have the same activity in neuroendocrine neoplasms [63]. Cisplatin with etoposide-based therapy appears to be more effective in G3 NET (with Ki-67 > 50%) and metastatic NECs (Figure 3) [21]. On the other hand, NEC patients with Ki-67 < 55% are less responsive to platinum-based chemotherapy but have a longer survival than those with a higher Ki-67.
Of the FDA-approved platinum compounds, oxaliplatin has shown significant clinical results in patients with well differentiated NETs, rather than cisplatin or carboplatin. The most commonly tested oxaliplatin combination regimens in patients with NETs are fluorouracil plus oxaliplatin (FOLFOX) or capecitabine plus oxaliplatin (CAPOX) [56]. Oxaliplatin has also demonstrated significant anticancer activity in G2 NETs and carcinoids, but there is currently no universally accepted standard chemotherapy for these tumors [40,63].
Significant effects have been observed when platinum-based drugs are combined with molecular-targeted therapy, immunotherapy, or other cytotoxic agents [29,53,64,65]. In 2019, the FDA approved atezolizumab in combination with etoposide and carboplatin [65] and, in 2020, durvalumab (an IgG1 kappa anti-PD-L1 monoclonal human antibody) in combination with etoposide and carboplatin/cisplatin as first-line treatments for patients with extensive-stage (ES) SCLC (ES-SCLC) (Figure 1) [53].

3. Platinum-Based Chemotherapy

Platinum-based antitumor drugs are a successful class of chemotherapy agents [66,67,68]. Cisplatin was the first metal-based anticancer drug introduced into clinical use in 1978 for several types of solid tumors [69]. However, its use as an anticancer drug is limited due to side effects such as neurotoxicity, nephrotoxicity, hepatotoxicity, and myelosuppression [22]. To overcome the high toxicity and chemoresistance associated with cisplatin-based therapy, a very high number of new platinum complexes have been synthesized and tested for antitumor activity [67,68,70,71,72,73,74,75,76,77,78]. However, only its analogs carboplatin and oxaliplatin have been approved by the FDA as chemotherapeutic drugs. Other platinum agents, such as nedaplatin, lobaplatin, and heptaplatin, have been approved only in Japan, China, and South Korea, respectively [66,67,68].
Currently, cisplatin and its derivatives are used to treat lymphomas, lung, colon, ovary, testicular, bladder, cervical, and other types of cancer, including neuroendocrine cancers [22,40]. These drugs induce cytotoxic effects by binding to DNA, interfering with its normal transcription and/or replication (Figure 3) [68,70,72,79]. In order to enhance and optimize the antitumor activity of cisplatin analogs, new coordination compounds are under evaluation as alternative platinum drugs. In recent years, platinum-based anticancer complexes have made significant progress in cancer therapy. New anticancer molecules have been synthesized or developed by modifying existing platinum drugs and attempting to mimic the mechanism of action of cisplatin [67,68,70,71,72,73,75,76,77,80,81,82,83,84,85,86,87,88]. However, no new platinum complexes have been approved for cancer therapy, neuroendocrine neoplasms included.

General Mechanism of Action of Cisplatin and Carboplatin

Cisplatin (cis-diamminedichloroplatinum(II)) and carboplatin (cis-diammine(1,1-cyclobutanedicarboxylato)platinum(II)), in combination with etoposide (Figure 2), represent the current standard first-line chemotherapy for various types of tumors, including neuroendocrine cancers. By 1979, pre-clinical data had shown synergistic effects of the combination of cisplatin and etoposide [89,90].
Platinum-based anticancer drugs can generally enter cells through passive diffusion and also using various cell membrane transport proteins, such as copper transporter 1 and 2 (CTR1 and CTR2), P-type copper-transporting ATPases (ATP7A and ATP7B), the organic cation transporter 2 (OCT2), the multidrug extrusion transporter 1 (MATE1), and LRRC8 volume-regulated anion channels (VRACs) (Figure 3) [91,92,93]. In the extracellular matrix, the concentration of chloride ions is higher (~100 mM) than in intracellular environment (~4 mM). As a result, after entering the cell, cisplatin undergoes an activation step where chloro-ligands are replaced by water molecules or other molecules containing sulfhydryl groups. This “aquation” of cisplatin promotes the formation of mono- and di-aquo species, such as cis-[Pt(NH3)2Cl(OH2)]+, cis-[Pt(NH3)2(OH)(OH2)]+ and cis-[Pt(NH3)2(OH2)2]2+ [83]. Due to the chelation of the leaving ligand, carboplatin and oxaliplatin are more stable and their activation is allowed by nucleophiles containing sulfhydryl groups, such as glutathione (GSH), aspartic acid, and other molecules [83,94,95]. The cytotoxicity induced by cisplatin and carboplatin is primarily due to their binding to DNA, through the formation of mono- and bis-adducts, producing intra- and inter-strand cross-links with DNA. However, it was estimated that only about 1% of intracellular cisplatin interacts with nuclear DNA [96]. It has been demonstrated that in the cytoplasm, platinum drugs interact with other biomolecules, such as cellular proteins, membrane phospholipids, and RNA [96], and induce cytotoxicity through the acidification of the cytoplasm, ER stress, the inhibition of RNA transcription and translation, the inhibition of important oncogenic proteins, and a decrease in metabolic plasticity of cancer cells [88,97,98]. The general accepted mechanism of action of cisplatin/carboplatin etoposide combined therapy essentially showing the platinum drugs contribution is reported in Figure 3.
In the nucleus, the diamineplatinum(II) units are coordinated by the N7 of purine bases, leading to the inhibition of replication and/or transcription, DNA damage, interference with DNA repair mechanisms, and ultimately, cell cycle arrest at S, G1 or G2-M and the induction of apoptosis/necrosis in cancer cells [67,83,99,100,101]. The interaction of cisplatin and carboplatin with DNA activates several signal transduction pathways, including those involving ATR, p53, p73, MAPKs (such as ERK, p38, and JNK), and PI3K/Akt, which ultimately result in the induction of apoptosis through both intrinsic and extrinsic pathways [68,102,103,104,105,106,107]. The formation of cisplatin/carboplatin adducts to DNA also activates the tumor suppressor p53, which can activate genes involved in cell cycle progression, DNA repair, and apoptosis [86,105,107]. The p53 protein can also activate genes of the Bcl family, which include pro-apoptotic (e.g., Bax and Bak) and anti-apoptotic (e.g., Bcl-2) factors (Figure 3) [108].
The combination of cisplatin or carboplatin drugs with etoposide improves DNA damage and induces cancer cell death [83,109]. Etoposide, a derivative of podophyllotoxin, was first synthesized in 1966 and approved for cancer therapy by the FDA in 1983. It targets DNA topoisomerase II (topo II) activity, inhibiting the faithful rejoining of DNA breaks and affecting various aspects of cell metabolism. Specifically, it causes topo II-linked DNA double- or single-strand breaks by inhibiting the rejoining of cleaved DNA [109]. Studies have shown that etoposide-induced DNA damage activates p53, leading to cell death [110,111]. In the apoptotic cascade, the activation of DNA-PK (DNA-dependent protein kinase) is crucial as it links the recognition of DNA damage to downstream signaling events. The activation of p53 by etoposide results in the upregulation of the pro-apoptotic protein Bax and the release of cytochrome c (Cyt C) (Figure 3) [112,113]. Robertson and colleagues hypothesized that etoposide-induced DNA damage leads to the activation of caspase-2, which acts as an intermediary in the induction of the mitochondrial apoptotic pathway [113,114]. On the other hand, while it is well known that etoposide triggers apoptotic pathways, recent findings also suggest its involvement in autophagic pathways [115,116]. Specifically, etoposide-induced autophagy seems linked to the activation of AMPK (AMP-activated protein kinase) [115]. Indeed, etoposide could induce an autophagy-associated surge in ATP, which contributes to cell survival and drug resistance [117].

4. Altered Pathways in NENs and Platinum-Based Chemotherapy Sensitivity

The anticancer activity of cisplatin and carboplatin was extensively studied in vitro and in vivo by various research groups for several types of tumors (Figure 3) [98,118]. Despite their widespread use in treating different types of neoplasms, platinum-based chemotherapy is often ineffective in treating NENs, and the mechanism of action in NE tumor cells remains unclear. Several pathways have been found to be altered in cisplatin/carboplatin sensitivity and are discussed below (Figure 4).

4.1. PTEN/PI3K/Akt/mTOR Pathway

The PI3K/Akt/mTOR pathway plays a crucial role in the development, progression, and angiogenesis of NENs, making it a promising target for treatment [119,120,121,122,123]. Akt, a major downstream regulator of PI3K, promotes cell proliferation by deactivating pro-apoptotic genes, such as caspase and Bcl-2 family members [107]. Moreover, mTOR is essential for the activation of the autophagic process, a key homoeostatic machinery of cellular self-degradation. Interestingly, across different tumor types, autophagy exhibits promoting or inhibitory effects to tumorigenesis by favoring resistance to anticancer treatments or inducing tumor cell cycle arrest, respectively [27,124,125,126]. The autophagic activity or the expression of autophagy-associated genes to influence survival in NENs has not been investigated yet, although it seems that a lower expression level of autophagic genes is associated with a metastatic stage [126].
Since the mTOR pathway is consistently activated in NETs, the development of mTOR inhibitors has provided a new therapeutic option for these tumors (Figure 1 and Table S1) [19,33,37,38,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171]. In fact, the PI3K/Akt/mTOR pathway plays an important role in tumorigenesis and the tumor progression of NENs [122]. It was observed that the PI3K/Akt/mTOR axis can cause resistance to cisplatin [172] and carboplatin [173] treatment. Conversely, the inhibition of Akt/mTOR can promoted cisplatin-induced apoptosis in resistant cells [174].
Despite these advancements, there is still much to learn about how platinum drugs interact with NENs, particularly in patients with an abnormal expression of PI3K/Akt/mTOR components (Figure 4). In this regard, we summarize below some studies that have investigated the relationships between the PTEN/PI3K/Akt/mTOR signaling system and the differential sensitivity to platinum-based chemotherapy.
PTEN (phosphatase and tensin homolog) is a tumor suppressor protein that negatively regulates the PI3K/Akt/mTOR signaling axis [122] and stimulates various DNA repair pathways, including homologous recombination (HR), non-homologous end joining (NHEJ), and nucleotide excision repair (NER). Its absence can sensitize cancer cells to DNA-damaging agents, including platinum drugs (Figure 3 and Figure 4) [128,129,175,176,177]. In NECs, the loss or reduced expression of PTEN has been associated with rapid tumor growth, metastasis, and poor survival (Table S1) [37,129,130,178,179]. Omura and colleagues observed that PTEN loss or downregulation was linked to a better response to platinum drugs. They reported the first case of a patient with advanced castration-resistant prostate NEC who showed a significant response to platinum-based chemotherapy and had mutations in both BRCA2 and PTEN (Table 1) [177]. Nuclear PTEN promotes genomic stability and DNA repair through the upregulation of RAD51, a key protein involved in double-strand break repair. Cytoplasmic PTEN also inhibits the Akt-mediated cytoplasmic sequestration of the checkpoint kinase CHEK1, preventing genomic instability and the accumulation of double-strand breaks [180]. By contrast, the loss of PTEN promotes genomic instability and the accumulation of double-strand break repair in tumor cells, enhancing platinum drugs’ anticancer activity [180,181]. However, the frequency of somatic PTEN alterations in patients with neuroendocrine prostate cancer is unknown because of its rarity and the lack of available genomic analysis in the literature [177].

4.2. Mitogen-Activated Protein Kinase (MAPK) Pathway

The MAPK pathway is an important regulator of the survival and proliferation of NENs, activated by various growth factors [105,197,198,199,200]. Platinum drugs can also induce apoptosis through the activation of MAPKs, contributing to tumor regression [105,197,201,202,203]. It has been demonstrated that the MAPK signal transduction pathway is associated with NE differentiation, cell growth, autophagy and metastasis (Table S1) [133,202,204]. Downstream of the growth factor receptors, the ERK cascade is initiated by RAS, which activates RAF and recruits it from the cytosol to the cell membrane. RAF then activates MEK, which in turn phosphorylates and activates ERK (Figure 4) [201]. Mutations in RAS and RAF family members have been linked to different clinical behaviors and responses to chemotherapy in NENs (Table S1).
The KRAS oncogene, a member of the RAS family, has the highest mutation rate among all cancers and is associated with high mortality [183,205]. In NENs, it is rarely mutated [201], but its mutational status appears to be related to different patterns of sensitivity or resistance to platinum drugs [182,183,206]. Tanaka and colleagues showed that patients with P-NEC who had KRAS mutations demonstrated an improved response to platinum-containing therapy [183,205]. Hijioka et al. demonstrated that high-grade P-NETs and P-NECs have distinct clinicopathologic features and that pancreatic NENs (G3) with mutated KRAS had a significantly higher response rate (77%) to platinum-based chemotherapy than those without (23%) [182]. Elvebakke et al. reported no influence of KRAS mutations in treatment efficacy or survival for patients with colon NEC receiving first-line platinum/etoposide chemotherapy. Conversely, BRAF mutations were associated with a limited effect of first-line chemotherapy, although they did not affect progression-free survival or overall survival (Figure 4; Table 1) [184].

4.3. Notch/ASCL1 Pathway

Notch (neurogenic locus notch homolog)/ASCL1 (Achaete-scute homolog 1) signaling is known to regulate cellular differentiation, proliferation and survival. However, contradictory findings have shown that Notch can act as both an oncogene and a tumor suppressor, indicating that its role is highly dependent on the specific cellular context. The Notch signaling pathway plays a crucial role in the growth and differentiation of GI-NETs [134], and its low expression has been linked to a better prognosis in SCLC patients (Table S1) [131,207]. Conversely, the overexpression of Notch has been shown to inhibit cell proliferation and to induce apoptosis in NET rather than promoting tumor growth [129,208,209]. This can also lead to the modulation of ASCL1 expression, as ASCL1 levels decrease when Notch signaling is active (Figure 4). ASCL1 is a transcription factor that is essential for the development and neuroendocrine differentiation of pulmonary NE cells, SCLC, thyroid C cells, and adrenal chromaffin cells [123]. Additionally, ASCL1 has been found to promote more aggressive growth of pulmonary adenocarcinoma in vivo and can interact with the Rb-p53 axis in the carcinogenesis of NE lung cancers (Table S1) [38,123,210,211,212,213,214].
Notch-negative and ASCL1-positive NE cells appear to be particularly susceptible to cytotoxic chemotherapy during initial treatment. However, epigenetic mechanisms that induce Notch expression in residual cancer cells may lead to recurrence in patients after repeated chemotherapy [38,207,215,216].
DLL3 (Delta-like protein 3) is a member of the Notch receptor ligand family that inhibits Notch signaling and is considered a predictive marker of sensitivity to platinum-based chemotherapy for LCNEC. Among patients with DLL3-negative LCNEC, platinum-based adjuvant chemotherapy has been shown to significantly improve overall survival and recurrence-free survival (Figure 4). However, patients with DLL3-positive LCNEC do not demonstrate improved response to chemotherapy (Table 1) [185]. In contrast, Tendler et al. found that an abnormal expression of ASCL1 and DLL3 in SCLC did not result in differences in clinical outcome. However, patients with a low expression of Notch-1 had a better prognosis and higher sensitivity to platinum-based chemotherapeutic drugs (Figure 4; Table 1) [131].

4.4. Pathways Involved in DNA Repair

Several studies have demonstrated that aberrations in DNA repair genes, including MDM2, RB, BRCA2, and MEN-1 mutations, serve as biomarkers for a heightened response of platinum-based chemotherapy (Figure 4). The combination of platinum drugs and etoposide in NENs results in increased DNA damage, leading to the inhibition of DNA repair and replication [217]. In P-NETs, mutations in PTEN, MEN1, and DAXX/ATRX genes were found to be common. The loss of these tumor suppressors in NENs may render them more susceptible to the cytotoxic effects of platinum-based drugs (Figure 4) [20,135,136,137,218].
The tumor suppressor p53, which is involved in cell cycle progression, DNA repair, and apoptosis, is activated after the formation of platinum drug adducts to DNA [86,105,107]. Some researchers have suggested that the inactivation of p53 and downstream DNA repair-related genes are responsible for platinum resistance mechanisms in NENs [219]. The role of p53 regulation in NENs and its consequences on chemotherapy efficacy are controversial. Mutant p53 proteins commonly lose wild-type function but can also acquire novel functions in promoting metastasis and resistance to platinum drugs and etoposide [220]. In NSCLC with NE differentiation, no significant correlation was found between altered p53 expression and response to platinum-based chemotherapy, although the increased expression of p53 was related to progressive disease following chemotherapy [132]. Similarly, Elvebakken and colleagues observed a limited response to treatment with platinum/etoposide in patients with high-grade GEP-NEN. Moreover, also a significantly better survival was observed in small-cell NEC [189]. In high-grade NENs, which often have alterations in the p53 gene (Table S1), p53 expression was not related to improved chemotherapy response [20,191]. On the other hand, some studies associated TP53 mutations or abnormal p53 protein expression with higher sensitivity to platinum therapy [187,188]. Some discrepancies in these results could be explained by the existence of different TP53 mutations which can differently modulate p53 accumulation in the cell nuclei. Moreover, the prognostic value of TP53 mutations might depend on co-mutations and tumor type. Finally, mutational analyses are recommended for the evaluation of clinical outcomes in order to bypass discordance between protein p53 measurements and TP53 mutation frequency [189].
Although TP53 gene mutations are rare in NENs [138,219,221], epigenetic and regulatory aberrations interfere with p53 network activity and influence response to platinum therapy (e.g., p53 negative regulators MDM2, MDM4 and WIP1) [139,186,219]. The E3 ubiquitin ligase MDM2 directly binds to p53 and promotes its nuclear export and proteasomal degradation, thus suppressing p53’s transcriptional activity [222]. In NEN, Akt activation and DAXX mutation may influence the stability of MDM2, regulating p53’s location, stability, and transcriptional activity, and sustaining proliferation or tumorigenesis [219,223,224]. In neuroendocrine prostate cancer, MDM2 amplification was related to the major effectiveness of platinum-based chemotherapy [186]. MDM2 can be inactivated after cisplatin-induced DNA damage, thus stabilizing the p53 protein and enabling it to induce cell cycle arrest and apoptosis [219,221].
Alterations of Rb (retinoblastoma protein), which are closely related to p53 mutations in NECs (Table S1) [33,38,140,183], were associated with better prognosis and response to platinum drugs-based therapy, especially in high-grade NENs (Figure 4; Table 1) [20,182,183,186,190,191]. The absence of Rb seemed to be more frequent in G3 NECs compared to G3 NETs and in SCNEC compared to LCNEC (Table S1) [182,191]. Derks and colleagues observed that patients with LCNEC tumors that carry a wild-type RB1 gene or express the Rb protein have a more favorable outcome when treated with platinum plus gemcitabine or taxanes compared to standard platinum plus etoposide chemotherapy, whereas no differences were observed when RB1 was mutated or the Rb protein not expressed [33]. However, in general, chemotherapeutic outcomes were better in tumors with abnormal Rb expression, sometimes associated with other genomic aberrations, such as KRAS [182,183], p16 [191], and MDM2 [186] (Table 1).
The overexpression of p16, a tumor suppressor that inhibits the CDK4/6 cell cycle regulators, can indicate a disruption of the Rb pathway. Several studies have confirmed an inverse relationship between the expression of Rb and p16 proteins in high-grade lung NETs. SCLC usually shows moderate or strong p16 staining in about 90% of the neoplastic cells [225]. Lacombe at al. observed a significantly higher response rate in NEC presenting high p16 levels, together with Rb loss (Table 1) [191].
A significant response to platinum therapy was observed in patients with advanced neuroendocrine prostate cancer who exhibited BRCA (breast cancer susceptibility gene) mutations [177,186,192,193,194,226]. BRCA1 and BRCA2 are important DNA repair genes that act as tumor suppressors. Their loss prevents DNA repair, leading to cell death after the formation of cisplatin DNA cross-links [226]. The positive association between BRCA2 mutations and response to platinum-based chemotherapy was clearly demonstrated in patients with prostate NEC. Both germline [192] and somatic [177,192,193] mutations of BRCA2 were found in these patients and were correlated with a high response to cisplatin/carboplatin and etoposide. Wood et al. also found somatic mutations in BRCA1 and BAP1 (BRCA1-associated protein) in colon LCNEC. Treatment with platinum-based therapy resulted in a complete response of the metastases, with no evidence of recurrence after 6.5 years. This led to the hypothesis that the loss or mutation of BRCA1 and/or BAP1 can predict response to platinum-based therapy, confirming the important role of genes involved in DNA repair in response to platinum drugs (Table 1) [195].

4.5. Other Genomic Alterations

Other mutations have recently been linked to the efficacy of chemotherapy in NENs. For instance, the speckle-type POZ protein, SPOP, is a zing finger protein with an oncogenic role that is frequently mutated in prostate and endometrial cancers [227,228]. It promotes the ubiquitination and degradation of proteins involved in tumor progression, such as PTEN and DAXX, thereby facilitating proliferation and inhibiting apoptosis in cancer cells [229]. Watanabe and colleagues showed that SPOP mutations in enzalutamide-resistant prostate cancer with NE differentiation improved treatment with platinum drugs (Table 1) [227].
The involvement of Wnt/β-catenin signaling in cancer was thoroughly described, and the altered expression of its components has also been observed in NETs [230]. Furukawa et al. demonstrated that β-catenin may serve as a reliable predictive biomarker for response to platinum-based chemotherapy in pancreatobiliary NEC [196], as evidenced by the increased expression of β-catenin in NET tissues and its correlation with tumor severity [231] (Table 1).

5. Influence of Specific Cellular Pathways in the Response to Platinum-Based Therapy in NENs

Bcl-2 is also closely related to NEN differentiation, as Bcl-2 expression is closely linked to chromogranin A (CgA) positivity, and tumor progression [232]. The Bcl system includes oncoproteins that affect apoptosis (such as Bax, Bad, Bid, and Bak) and proliferation (such as Bcl-2, Bcl-xL, and Raf), making it a key factor in regulating these processes [141]. Chemotherapeutic agents, including platinum drugs, can exert their cytotoxic effects by inducing intrinsic apoptosis through the mitochondrial pathway by modulating Bcl-2/Bax levels (Figure 3). The overexpression of Bcl-2 and reduction in Bax levels have been linked to resistance to platinum drugs in several cancers, including NENs [140,141,142,232,233]. In SCLC patients, Bcl-2 has been found to be overexpressed [234] and related to increased resistance to platinum chemotherapy in vitro [235], but its role in patients with NENs undergoing platinum-based chemotherapy has not yet been established.
Genomic analysis has revealed that NENs show alterations in chromatin remodeling genes, such as MEN1, DAXX (death domain-associated protein), and ATRX (α thalassemia/mental retardation syndrome X-linked) [236]. DAXX and ATRX cooperate with other genes in chromatin remodeling complexes. DAXX can also modulate the distribution of PTEN between the nucleus and the cytoplasm (Figure 4) [229,237]. Menin, encoded by MEN1, is involved in the regulation of DAXX and SMADs, ref. [238] and its loss of function has been associated with impaired DNA repair capability in NETs (Figure 4) [218]. Although Menin acts as a tumor suppressor in endocrine tissues, recent studies have shown that it can also promote tumorigenesis in various tumors. In some neoplasms, MEN1 has been found to act as a hub gene, interacting with and modulating several pathways. For example, Menin can inhibit the PI3K/Akt/mTOR pathway (by binding to Akt and preventing its translocation to the plasma membrane) [239,240] and the RAS-RAF-MEK1/2-ERK1/2 signaling pathway. Cherif et al. studied the in vitro response of prostate cancer PC-3 cells (which are positive for NE markers) to cisplatin treatment after Menin inhibition. They found that Menin activates the PI3K/Akt signaling pathway, which is associated with platinum drug resistance, and that Menin inhibition enhanced cisplatin sensitivity (by 69%) in PC-3 cells [240]. The role of MEN1 and DAXX/ATRX in the effectiveness of platinum-based treatments has not yet been defined.
Finally, other pathways have been linked to the differentiation and progression of NENs, but their involvement in cisplatin/carboplatin therapy has not yet been considered. Among these, the SMAD signaling pathway operates downstream of TGF-β and BMP ligands, regulating a diverse set of biological processes including proliferation, differentiation, and apoptosis [241,242]. Murai et al. were the first to show that in small-cell lung cancer (SCLC), TGF-β inhibited proliferation in vivo and tumor formation in vitro through the TGF-β-SMAD-ASCL1 pathway [243]. The loss of the tumor suppressor SMAD4 occurs in numerous solid organ neoplasms, included NENs, and it is associated with poor prognosis (Table S1) [143,144,145].

6. Conclusions

This review stems from the need to better understand the predictive and prognostic features of NENs. These features are still uncertain, and it is crucial to identify the major molecular genetic alterations in each type of tumor. Establishing correlations between specific abnormalities involved in tumorigenesis and the metastatic process could provide potential targets for cancer therapy. Due to the heterogeneity of NENs, there is a significant need to re-evaluate chemotherapeutic approaches, with a focus on studying combined and personalized therapies that can offer greater selectivity and effectiveness. The review incorporates recent and relevant studies in the field, providing a comprehensive analysis of current knowledge and developments in NENs and platinum-based chemotherapy responses, taking into account data and research findings up to the year 2024.
Among the available therapeutic options, platinum-based chemotherapy is considered the first-line treatment for well-differentiated G3 NETs and NECs, particularly when the Ki-67 index is higher than 55% or in cases of rapid clinical progression. The cellular response to platinum-based chemotherapy is a complex process that typically begins with the induction of DNA damage, followed by a series of events involving signal transduction and the activation of transcription factors. These factors induce the expression of numerous genes involved in various cellular functions, such as DNA repair, cell cycle arrest, cell death, and the inhibition of epithelial–mesenchymal transition (EMT) (Figure 3). Additionally, there may be crosstalk between different signaling pathways, resulting in diverse downstream effects. Also, with this review, we underscore the pivotal role of molecular genetic alterations in NENs and their impact on treatment outcomes. It is very important to identify the major molecular genetic alterations specific to each tumor type, essential for advancing personalized medicine in NENs. Current guidelines recommend using MEN1, DAXX/ATRX, and RB1/TP53 to distinguish between P-NET G3 and P-NEC. The distinct genetic abnormalities between P-NETs (G3) and P-NEC may explain why P-NETs have a lower sensitivity to platinum-based chemotherapy compared to P-NECs. Consistently, NECs are often characterized by the aberrant expression of p53 and/or Rb proteins, as well as KRAS mutations (Table S1), which have been linked to a higher response to platinum-based chemotherapy (Figure 4; Table 1). On the other hand, diagnosing LCNEC can be challenging due to its histological similarities with non-small-cell lung cancer (NSCLC) and, in some cases, small-cell lung cancer (SCLC) (as evidenced by the PI3K/Akt/mTOR pathway and other gene alterations) (Table S1). As platinum-based chemotherapy is the standard treatment for SCLC, it is often used for both LCNEC and NSCLC. Interestingly, although platinum-based chemotherapy is not typically used for neuroendocrine prostate cancer, it has been reported to be effective in some cases. This is likely due to the presence of BRCA2, PTEN, and MDM2 mutations, which make this type of cancer responsive to platinum-based chemotherapy (Table 1).
The relationship between specific genetic abnormalities and the processes of tumorigenesis and metastasis presents a promising area for future research. Understanding these correlations could lead to the identification of novel targets for cancer therapy, thereby enhancing treatment efficacy. The heterogeneity of NENs suggests that personalized treatments, based on the genetic characteristics of each tumor, could offer better selectivity and effectiveness. The prospects for improved treatment outcomes through personalized and combined therapeutic approaches represent a significant advancement in the field of neuroendocrine neoplasms.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms25168568/s1.

Author Contributions

Conceptualization, M.B., F.P.F., E.S. and F.D.C.; validation, M.B., F.P.F., A.M. and S.M.; investigation, E.S. and F.D.C.; writing—original draft preparation, E.S.; writing—review and editing, F.D.C., M.B., F.P.F., A.M. and S.M.; visualization, A.C.; supervision, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors acknowledge the University of Salento (Italy) for its support.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

5-FU: 5-Fluorouracil; ABC: ATP-binding cassette; AC: atypical carcinoid; AFP: alpha-fetoprotein; Akt/PKB: protein kinase B; AMPK: AMP-activated protein kinase; ASCL1: achaete-scute homolog 1; ASCO: American Society of Clinical Oncology; ATP: adenosine triphosphate; ATPase: adenosine triphosphatases; ATR: ataxia-telangiectasia-mutated and Rad 3-related; ATRX: X-linked mental retardation and α-thalassaemia syndrome protein; BAP1: BRCA1-associated protein; BCL-2: B-cell lymphoma 2; BMP: bone morphogenetic protein; BP: bronchopulmonary; BRCA1/2: breast cancer gene 1/2; CAPOX: capecitabine plus oxaliplatin; CAPTEM: capecitabine plus temozolomide; CDK: cyclin-dependent kinase; CgA: chromogranin A; CGP: comprehensive genomic profiling; CSF1R: colony-stimulating factor receptor 1; CTLA-4: cytotoxic T-lymphocyte antigen 4; CTR: copper transporter; Cyt-C: cytochrome C; DAXX: death domain-associated protein; DLL3: delta-like protein 3; DNA: deoxyribonucleic acid; DNA-PK: DNA-dependent protein kinase; DOXO: doxorubicin; EGF: epidermal growth factor; ERK: extracellular signal-regulated kinase; ES: extensive-stage; ESMO: European Society for Medical Oncology; FGF: fibroblast growth factor; FLT3: Fms-related tyrosine kinase 3; FOLFOX: fluorouracil plus oxaliplatin; GEP: gastroenteropancreatic; GI: gastrointestinal; GSH: glutathione; HCG: human chorionic gonadotropin; HIF: hypoxia-inducible factor; HR: homologous recombination; ICI: immune checkpoint inhibitor; IGF: insulin-like growth factor; IHC: immunohistochemistry; INSM-1: insulinoma-associated protein-1; Isl-1: Islet 1; JNK: c-Jun N-terminal kinase; KIT: stem-cell growth factor receptor; LAR: long-acting release; LCNEC: large-cell neuroendocrine carcinoma; LRRC: leucine-rich repeat containing; MAPK: mitogen-activated protein kinase; MATE: multidrug extrusion transporter; MCC: Merkel cell carcinoma; MDM: murine double minute; MEN: multiple endocrine neoplasia; MTC: medullary thyroid carcinoma; mTOR: mammalian target of rapamycin; NE: neuroendocrine; NEC: neuroendocrine cancer; NEN: neuroendocrine neoplasm; NER: nucleotide excision repair; NET: neuroendocrine tumor; NGS: next-generation sequencing; NHEJ: non-homologous end joining; Notch: neurogenic locus notch homolog; NSCLC: non-small-cell lung cancer; NSE: neuron-specific enolase; OCT: organic cation transporter; P: pancreatic; p-Akt: phosphorylate Akt; PARP: poly(ADP-ribose) polymerase; PAX 8: paired box 8; PCR: polymerase chain reaction; PD-1: programmed death-1; PDGFR: platelet-derived growth factor; PD-L1: programmed death-1 ligand; PI3K: phosphatidylinositol 3-kinase; p-mTOR: phosforilate mTOR; PP: pancreatic polypeptide; PRRT: peptide receptor radionuclide therapy; PTEN: phosphatase and tensin homolog; Rb: retinoblastoma protein; RET: rearranged during transfection; ROS: reactive oxygen species; SCLC: small-cell lung carcinoma; SCNEC: small-cell neuroendocrine carcinoma; SMAD: small mother against decapentaplegic; SPOP: speckle-type POZ protein; SSA: somatostatin analog; SSTR: somatostatin receptor; STZ: streptozotocin; SYN: synaptophysin; TC: typical carcinoid; TGF-α/β: transforming growth factor-α/β; TGF-βR1,2: transforming growth factor-β receptor; TKI: tyrosine kinase receptor inhibitors; TTF-1: thyroid transcription factor-1; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; VHL: von Hippel–Lindau syndrome; VIP: vasoactive intestinal peptide; VRAC: volume-regulated anion channel; WHO: World Health Organization; WIP1: wild-type p53-induced phosphatase 1; Wnt: wingless-related integration site.

References

  1. Oronsky, B.; Ma, P.C.; Morgensztern, D.; Carter, C.A. Nothing But NET: A Review of Neuroendocrine Tumors and Carcinomas. Neoplasia 2017, 19, 991–1002. [Google Scholar] [CrossRef]
  2. Lamberti, G.; Brighi, N.; Maggio, I.; Manuzzi, L.; Peterle, C.; Ambrosini, V.; Ricci, C.; Casadei, R.; Campana, D. The Role of mTOR in Neuroendocrine Tumors: Future Cornerstone of a Winning Strategy? Int. J. Mol. Sci. 2018, 19, 747. [Google Scholar] [CrossRef]
  3. Kaltsas, G.A.; Besser, G.M.; Grossman, A.B. The Diagnosis and Medical Management of Advanced Neuroendocrine Tumors. Endocr. Rev. 2004, 25, 458–511. [Google Scholar] [CrossRef] [PubMed]
  4. Schimmack, S.; Svejda, B.; Lawrence, B.; Kidd, M.; Modlin, I.M. The Diversity and Commonalities of Gastroenteropancreatic Neuroendocrine Tumors. Langenbeck’s Arch. Surg. 2011, 396, 273–298. [Google Scholar] [CrossRef]
  5. Rossi, R.E.; Massironi, S. The Increasing Incidence of Neuroendocrine Neoplasms Worldwide: Current Knowledge and Open Issues. J. Clin. Med. 2022, 11, 3794. [Google Scholar] [CrossRef] [PubMed]
  6. Espinosa-Olarte, P.; La Salvia, A.; Riesco-Martinez, M.C.; Anton-Pascual, B.; Garcia-Carbonero, R. Chemotherapy in NEN: Still Has a Role? Rev. Endocr. Metab. Disord. 2021, 22, 595–614. [Google Scholar] [CrossRef]
  7. Rindi, G.; Mete, O.; Uccella, S.; Basturk, O.; La Rosa, S.; Brosens, L.A.A.; Ezzat, S.; de Herder, W.W.; Klimstra, D.S.; Papotti, M.; et al. Overview of the 2022 WHO Classification of Neuroendocrine Neoplasms. Endocr. Pathol. 2022, 33, 115–154. [Google Scholar] [CrossRef]
  8. La Rosa, S.; Uccella, S. Classification of Neuroendocrine Neoplasms: Lights and Shadows. Rev. Endocr. Metab. Disord. 2021, 22, 527–538. [Google Scholar] [CrossRef]
  9. Rorstad, O. Prognostic Indicators for Carcinoid Neuroendocrine Tumors of the Gastrointestinal Tract. J. Surg. Oncol. 2005, 89, 151–160. [Google Scholar] [CrossRef]
  10. Rindi, G.; Klimstra, D.S.; Abedi-Ardekani, B.; Asa, S.L.; Bosman, F.T.; Brambilla, E.; Busam, K.J.; de Krijger, R.R.; Dietel, M.; El-Naggar, A.K.; et al. A Common Classification Framework for Neuroendocrine Neoplasms: An International Agency for Research on Cancer (IARC) and World Health Organization (WHO) Expert Consensus Proposal. Mod. Pathol. 2018, 31, 1770–1786. [Google Scholar] [CrossRef]
  11. Kunz, P.L.; Reidy-Lagunes, D.; Anthony, L.B.; Bertino, E.M.; Brendtro, K.; Chan, J.A.; Chen, H.; Jensen, R.T.; Kim, M.K.; Klimstra, D.S.; et al. Consensus Guidelines for the Management and Treatment of Neuroendocrine Tumors. Pancreas 2013, 42, 557–577. [Google Scholar] [CrossRef] [PubMed]
  12. Kimura, N.; Nagura, H. A Comparative Study of Neuroendocrine Carcinoma and Carcinoid Tumor with Special Reference to Expression of HLA-DR Antigen and PCNA. Zentralbl. Pathol. 1993, 139, 171–175. [Google Scholar] [PubMed]
  13. La Rosa, S.; Sessa, F.; Uccella, S. Mixed Neuroendocrine-Nonneuroendocrine Neoplasms (MiNENs): Unifying the Concept of a Heterogeneous Group of Neoplasms. Endocr. Pathol. 2016, 27, 284–311. [Google Scholar] [CrossRef] [PubMed]
  14. Bocchini, M.; Nicolini, F.; Severi, S.; Bongiovanni, A.; Ibrahim, T.; Simonetti, G.; Grassi, I.; Mazza, M. Biomarkers for Pancreatic Neuroendocrine Neoplasms (PanNENs) Management—An Updated Review. Front. Oncol. 2020, 10, 831. [Google Scholar] [CrossRef]
  15. La Salvia, A.; Siciliani, A.; Rinzivillo, M.; Verrico, M.; Baldelli, R.; Puliani, G.; Modica, R.; Zanata, I.; Persano, I.; Fanciulli, G.; et al. Thyroid Transcription Factor-1 Expression in Lung Neuroendocrine Tumours: A Gender-Related Biomarker? Endocrine 2024, 83, 519–526. [Google Scholar] [CrossRef] [PubMed]
  16. Asa, S.L.; Mete, O. Cytokeratin Profiles in Pituitary Neuroendocrine Tumors. Hum. Pathol. 2021, 107, 87–95. [Google Scholar] [CrossRef]
  17. Gut, P.; Komarowska, H.; Czarnywojtek, A.; Waligórska-Stachura, J.; Bączyk, M.; Ziemnicka, K.; Fischbach, J.; Wrotkowska, E.; Ruchała, M. Familial Syndromes Associated with Neuroendocrine Tumours. Contemp. Oncol./Współczesna Onkol. 2015, 3, 176–183. [Google Scholar] [CrossRef]
  18. Murakumo, Y.; Jijiwa, M.; Asai, N.; Ichihara, M.; Takahashi, M. RET and Neuroendocrine Tumors. Pituitary 2006, 9, 179–192. [Google Scholar] [CrossRef]
  19. Estrella, J.S.; Broaddus, R.R.; Mathews, A.; Milton, D.R.; Yao, J.C.; Wang, H.; Rashid, A. Progesterone Receptor and PTEN Expression Predict Survival in Patients with Low- and Intermediate-Grade Pancreatic Neuroendocrine Tumors. Arch. Pathol. Lab. Med. 2014, 138, 1027–1036. [Google Scholar] [CrossRef]
  20. Pulvirenti, A.; Raj, N.; Cingarlini, S.; Pea, A.; Tang, L.H.; Luchini, C.; Chou, J.F.; Grego, E.; Marinova, I.; Capanu, M.; et al. Platinum-Based Treatment for Well- and Poorly Differentiated Pancreatic Neuroendocrine Neoplasms. Pancreas 2021, 50, 138–146. [Google Scholar] [CrossRef]
  21. Rinke, A.; Gress, T.M. Neuroendocrine Cancer, Therapeutic Strategies in G3 Cancers. Digestion 2017, 95, 109–114. [Google Scholar] [CrossRef]
  22. Brown, A.; Kumar, S.; Tchounwou, P.B. Cisplatin-Based Chemotherapy of Human Cancers. J. Cancer Sci. Ther. 2019, 11, 97. [Google Scholar]
  23. Sorbye, H.; Welin, S.; Langer, S.W.; Vestermark, L.W.; Holt, N.; Osterlund, P.; Dueland, S.; Hofsli, E.; Guren, M.G.; Ohrling, K.; et al. Predictive and Prognostic Factors for Treatment and Survival in 305 Patients with Advanced Gastrointestinal Neuroendocrine Carcinoma (WHO G3): The NORDIC NEC Study. Ann. Oncol. 2013, 24, 152–160. [Google Scholar] [CrossRef] [PubMed]
  24. Sorbye, H.; Strosberg, J.; Baudin, E.; Klimstra, D.S.; Yao, J.C. Gastroenteropancreatic High-Grade Neuroendocrine Carcinoma. Cancer 2014, 120, 2814–2823. [Google Scholar] [CrossRef]
  25. Fottner, C.; Ferrata, M.; Weber, M.M. Hormone Secreting Gastro-Entero-Pancreatic Neuroendocrine Neoplasias (GEP-NEN): When to Consider, How to Diagnose? Rev. Endocr. Metab. Disord. 2017, 18, 393–410. [Google Scholar] [CrossRef]
  26. Kulke, M.H.; Shah, M.H.; Benson, A.B.; Bergsland, E.; Berlin, J.D.; Blaszkowsky, L.S.; Emerson, L.; Engstrom, P.F.; Fanta, P.; Giordano, T.; et al. Neuroendocrine Tumors, Version 1.2015. J. Natl. Compr. Cancer Netw. 2015, 13, 78–108. [Google Scholar] [CrossRef]
  27. Daskalakis, K. Functioning and Nonfunctioning pNENs. Curr. Opin. Endocr. Metab. Res. 2021, 18, 284–290. [Google Scholar] [CrossRef]
  28. Zhang, J.Y.; Kunz, P.L. Making Sense of a Complex Disease: A Practical Approach to Managing Neuroendocrine Tumors. JCO Oncol. Pract. 2022, 18, 258–264. [Google Scholar] [CrossRef] [PubMed]
  29. Ooki, A.; Osumi, H.; Fukuda, K.; Yamaguchi, K. Potent Molecular-Targeted Therapies for Gastro-Entero-Pancreatic Neuroendocrine Carcinoma. Cancer Metastasis Rev. 2023, 42, 1021–1054. [Google Scholar] [CrossRef]
  30. Riihimäki, M.; Hemminki, A.; Sundquist, K.; Sundquist, J.; Hemminki, K. The Epidemiology of Metastases in Neuroendocrine Tumors: Epidemiology of Metastases. Int. J. Cancer 2016, 139, 2679–2686. [Google Scholar] [CrossRef]
  31. Rekhtman, N. Lung Neuroendocrine Neoplasms: Recent Progress and Persistent Challenges. Mod. Pathol. 2022, 35, 36–50. [Google Scholar] [CrossRef] [PubMed]
  32. Fisseler-Eckhoff, A.; Demes, M. Neuroendocrine Tumors of the Lung. Cancers 2012, 4, 777–798. [Google Scholar] [CrossRef] [PubMed]
  33. Derks, J.L.; Van Suylen, R.J.; Thunnissen, E.; den Bakker, M.A.; Groen, H.J.; Smit, E.F.; Damhuis, R.A.; Van Den Broek, E.C.; Speel, E.-J.M.; Dingemans, A.-M.C.; et al. Chemotherapy for Pulmonary Large Cell Neuroendocrine Carcinomas: Does the Regimen Matter? Eur. Respir. J. 2017, 49, 1601838. [Google Scholar] [CrossRef] [PubMed]
  34. Naidoo, J.; Santos-Zabala, M.L.; Iyriboz, T.; Woo, K.M.; Sima, C.S.; Fiore, J.J.; Kris, M.G.; Riely, G.J.; Lito, P.; Iqbal, A.; et al. Large Cell Neuroendocrine Carcinoma of the Lung: Clinico-Pathologic Features, Treatment, and Outcomes. Clin. Lung Cancer 2016, 17, e121–e129. [Google Scholar] [CrossRef] [PubMed]
  35. Zappi, A.; Persano, I.; Galvani, L.; Parlagreco, E.; Andrini, E.; Campana, D.; Brizzi, M.P.; Lamberti, G.; La Salvia, A. Chemotherapy in Well Differentiated Neuroendocrine Tumors (NET) G1, G2, and G3: A Narrative Review. J. Clin. Med. 2023, 12, 717. [Google Scholar] [CrossRef] [PubMed]
  36. Tang, L.H.; Basturk, O.; Sue, J.J.; Klimstra, D.S. A Practical Approach to the Classification of WHO Grade 3 (G3) Well-Differentiated Neuroendocrine Tumor (WD-NET) and Poorly Differentiated Neuroendocrine Carcinoma (PD-NEC) of the Pancreas. Am. J. Surg. Pathol. 2016, 40, 1192–1202. [Google Scholar] [CrossRef] [PubMed]
  37. Vijayvergia, N.; Boland, P.M.; Handorf, E.; Gustafson, K.S.; Gong, Y.; Cooper, H.S.; Sheriff, F.; Astsaturov, I.; Cohen, S.J.; Engstrom, P.F. Molecular Profiling of Neuroendocrine Malignancies to Identify Prognostic and Therapeutic Markers: A Fox Chase Cancer Center Pilot Study. Br. J. Cancer 2016, 115, 564–570. [Google Scholar] [CrossRef] [PubMed]
  38. George, J.; Lim, J.S.; Jang, S.J.; Cun, Y.; Ozretić, L.; Kong, G.; Leenders, F.; Lu, X.; Fernández-Cuesta, L.; Bosco, G.; et al. Comprehensive Genomic Profiles of Small Cell Lung Cancer. Nature 2015, 524, 47–53. [Google Scholar] [CrossRef] [PubMed]
  39. Torniai, M.; Rinaldi, S.; Morgese, F.; Ricci, G.; Onofri, A.; Ghroé, C.; Berardi, R. Medical Therapy for Advanced Gastro-Entero-Pancreatic and Bronchopulmonary Neuroendocrine Tumors. J. Cancer Metastasis Treat. 2016, 2, 329. [Google Scholar] [CrossRef]
  40. Spada, F.; Valente, M. Review of Recents Advances in Medical Treatment for Neuroendocrine Neoplasms: Somatostatin Analogs and Chemotherapy. J. Cancer Metastasis Treat. 2016, 2, 313. [Google Scholar] [CrossRef]
  41. Zandee, W.T.; de Herder, W.W. The Evolution of Neuroendocrine Tumor Treatment Reflected by ENETS Guidelines. Neuroendocrinology 2018, 106, 357–365. [Google Scholar] [CrossRef] [PubMed]
  42. Gomes-Porras, M.; Cárdenas-Salas, J.; Álvarez-Escolá, C. Somatostatin Analogs in Clinical Practice: A Review. Int. J. Mol. Sci. 2020, 21, 1682. [Google Scholar] [CrossRef]
  43. Rogoza, O.; Megnis, K.; Kudrjavceva, M.; Gerina-Berzina, A.; Rovite, V. Role of Somatostatin Signalling in Neuroendocrine Tumours. Int. J. Mol. Sci. 2022, 23, 1447. [Google Scholar] [CrossRef]
  44. Nuñez, J.E.; Donadio, M.; Filho, D.R.; Rego, J.F.; Barros, M.; Formiga, M.N.; Lopez, R.; Riechelmann, R. The Efficacy of Everolimus and Sunitinib in Patients with Sporadic or Germline Mutated Metastatic Pancreatic Neuroendocrine Tumors. J. Gastrointest. Oncol. 2019, 10, 645–651. [Google Scholar] [CrossRef]
  45. Yoo, C.; Cho, H.; Song, M.J.; Hong, S.-M.; Kim, K.; Chang, H.-M.; Chae, H.; Kim, T.W.; Hong, Y.S.; Ryu, M.-H.; et al. Efficacy and Safety of Everolimus and Sunitinib in Patients with Gastroenteropancreatic Neuroendocrine Tumor. Cancer Chemother. Pharmacol. 2017, 79, 139–146. [Google Scholar] [CrossRef]
  46. Raymond, E.; Dahan, L.; Raoul, J.-L.; Bang, Y.-J.; Borbath, I.; Lombard-Bohas, C.; Valle, J.; Metrakos, P.; Smith, D.; Vinik, A.; et al. Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors. N. Engl. J. Med. 2011, 364, 501–513. [Google Scholar] [CrossRef] [PubMed]
  47. Yao, J.C.; Fazio, N.; Singh, S.; Buzzoni, R.; Carnaghi, C.; Wolin, E.; Tomasek, J.; Raderer, M.; Lahner, H.; Voi, M.; et al. Everolimus for the Treatment of Advanced, Non-Functional Neuroendocrine Tumours of the Lung or Gastrointestinal Tract (RADIANT-4): A Randomised, Placebo-Controlled, Phase 3 Study. Lancet 2016, 387, 968–977. [Google Scholar] [CrossRef]
  48. Okafor, C.; Hogan, J.; Raygada, M.; Thomas, B.J.; Akshintala, S.; Glod, J.W.; Del Rivero, J. Update on Targeted Therapy in Medullary Thyroid Cancer. Front. Endocrinol. 2021, 12, 708949. [Google Scholar] [CrossRef] [PubMed]
  49. Wells, S.A.; Gosnell, J.E.; Gagel, R.F.; Moley, J.; Pfister, D.; Sosa, J.A.; Skinner, M.; Krebs, A.; Vasselli, J.; Schlumberger, M. Vandetanib for the Treatment of Patients with Locally Advanced or Metastatic Hereditary Medullary Thyroid Cancer. J. Clin. Oncol. 2010, 28, 767–772. [Google Scholar] [CrossRef]
  50. Carra, S.; Gaudenzi, G.; Dicitore, A.; Saronni, D.; Cantone, M.C.; Plebani, A.; Ghilardi, A.; Borghi, M.O.; Hofland, L.J.; Persani, L.; et al. Vandetanib versus Cabozantinib in Medullary Thyroid Carcinoma: A Focus on Anti-Angiogenic Effects in Zebrafish Model. Int. J. Mol. Sci. 2021, 22, 3031. [Google Scholar] [CrossRef]
  51. Pavel, M.; Valle, J.W.; Eriksson, B.; Rinke, A.; Caplin, M.; Chen, J.; Costa, F.; Falkerby, J.; Fazio, N.; Gorbounova, V.; et al. ENETS Consensus Guidelines for the Standards of Care in Neuroendocrine Neoplasms: Systemic Therapy—Biotherapy and Novel Targeted Agents. Neuroendocrinology 2017, 105, 266–280. [Google Scholar] [CrossRef] [PubMed]
  52. Das, S.; Al-Toubah, T.; El-Haddad, G.; Strosberg, J. 177Lu-DOTATATE for the Treatment of Gastroenteropancreatic Neuroendocrine Tumors. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 1023–1031. [Google Scholar] [CrossRef] [PubMed]
  53. 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]
  54. 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]
  55. Fang, L.; Arvind, D.; Dowlati, A.; Mohamed, A. Role of Immunotherapy in Gastro-enteropancreatic Neuroendocrine Neoplasms (Gep-nens): Current Advances and Future Directions. J. Neuroendocrinol. 2021, 33, e12943. [Google Scholar] [CrossRef] [PubMed]
  56. Das, S.; Al-Toubah, T.; Strosberg, J. Chemotherapy in Neuroendocrine Tumors. Cancers 2021, 13, 4872. [Google Scholar] [CrossRef] [PubMed]
  57. Moertel, C.G.; Hanley, J.A.; Johnson, L.A. Streptozocin Alone Compared with Streptozocin plus Fluorouracil in the Treatment of Advanced Islet-Cell Carcinoma. N. Engl. J. Med. 1980, 303, 1189–1194. [Google Scholar] [CrossRef] [PubMed]
  58. Murraylyon, I. Treatment of Multiple-Hormone-Producing Malignant Islet-Cell Tumour with Streptozotocin. Lancet 1968, 292, 895–898. [Google Scholar] [CrossRef]
  59. Capdevila, J.; Ducreux, M.; García Carbonero, R.; Grande, E.; Halfdanarson, T.; Pavel, M.; Tafuto, S.; Welin, S.; Valentí, V.; Salazar, R. Streptozotocin, 1982–2022: Forty Years from the FDA’s Approval to Treat Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2022, 112, 1155–1167. [Google Scholar] [CrossRef]
  60. Moertel, C.G.; Lefkopoulo, M.; Lipsitz, S.; Hahn, R.G.; Klaassen, D. Streptozocin–Doxorubicin, Streptozocin–Fluorouracil, or Chlorozotocin in the Treatment of Advanced Islet-Cell Carcinoma. N. Engl. J. Med. 1992, 326, 519–523. [Google Scholar] [CrossRef]
  61. Pelle, E.; Al-Toubah, T.; Morse, B.; Strosberg, J. Belzutifan in a Patient with VHL-Associated Metastatic Pancreatic Neuroendocrine Tumor. J. Natl. Compr. Cancer Netw. 2022, 20, 1285–1287. [Google Scholar] [CrossRef]
  62. Cives, M.; Ghayouri, M.; Morse, B.; Brelsford, M.; Black, M.; Rizzo, A.; Meeker, A.; Strosberg, J. Analysis of Potential Response Predictors to Capecitabine/Temozolomide in Metastatic Pancreatic Neuroendocrine Tumors. Endocr.-Relat. Cancer 2016, 23, 759–767. [Google Scholar] [CrossRef]
  63. Bajetta, E.; Catena, L.; Procopio, G.; De Dosso, S.; Bichisao, E.; Ferrari, L.; Martinetti, A.; Platania, M.; Verzoni, E.; Formisano, B.; et al. Are Capecitabine and Oxaliplatin (XELOX) Suitable Treatments for Progressing Low-Grade and High-Grade Neuroendocrine Tumours? Cancer Chemother. Pharmacol. 2007, 59, 637–642. [Google Scholar] [CrossRef]
  64. Lassen, U.; Kristjansen, P.E.G.; Østerlind, K.; Bergman, B.; Sigsgaard, T.C.; Hirsch, F.R.; Hansen, M.; Dombernowsky, P.; Hansen, H.H. Superiority of Cisplatin or Carboplatin in Combination with Teniposide and Vincristine in the Induction Chemotherapy of Small-Cell Lung Cancer. A Randomized Trial with 5 Years Follow Up. Ann. Oncol. 1996, 7, 365–371. [Google Scholar] [CrossRef] [PubMed]
  65. Horn, L.; Mansfield, A.S.; Szczęsna, A.; Havel, L.; Krzakowski, M.; Hochmair, M.J.; Huemer, F.; Losonczy, G.; Johnson, M.L.; Nishio, M.; et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2220–2229. [Google Scholar] [CrossRef] [PubMed]
  66. Wilson, J.J.; Lippard, S.J. Synthetic Methods for the Preparation of Platinum Anticancer Complexes. Chem. Rev. 2014, 114, 4470–4495. [Google Scholar] [CrossRef]
  67. Baik, M.-H.; Friesner, R.A.; Lippard, S.J. Theoretical Study of Cisplatin Binding to Purine Bases: Why Does Cisplatin Prefer Guanine over Adenine? J. Am. Chem. Soc. 2003, 125, 14082–14092. [Google Scholar] [CrossRef] [PubMed]
  68. Johnstone, T.C.; Suntharalingam, K.; Lippard, S.J. The Next Generation of Platinum Drugs: Targeted Pt(II) Agents, Nanoparticle Delivery, and Pt(IV) Prodrugs. Chem. Rev. 2016, 116, 3436–3486. [Google Scholar] [CrossRef]
  69. Tomida, A.; Tsuruo, T. Drug Resistance Mediated by Cellular Stress Response to the Microenvironment of Solid Tumors. Anti-Cancer Drug Des. 1999, 14, 169–177. [Google Scholar]
  70. De Castro, F.; Stefàno, E.; De Luca, E.; Benedetti, M.; Fanizzi, F.P. Platinum-Nucleos(t)Ide Compounds as Possible Antimetabolites for Antitumor/Antiviral Therapy: Properties and Perspectives. Pharmaceutics 2023, 15, 941. [Google Scholar] [CrossRef]
  71. Stefàno, E.; De Castro, F.; De Luca, E.; Muscella, A.; Marsigliante, S.; Benedetti, M.; Fanizzi, F.P. Synthesis and Comparative Evaluation of the Cytotoxic Activity of Cationic Organometallic Complexes of the Type [Pt(η1-CH2-CH2-OR)(DMSO)(Phen)]+ (R = Me, Et, Pr, Bu). Inorganica Chim. Acta 2023, 546, 121321. [Google Scholar] [CrossRef]
  72. De Castro, F.; De Luca, E.; Benedetti, M.; Fanizzi, F.P. Platinum Compounds as Potential Antiviral Agents. Coord. Chem. Rev. 2022, 451, 214276. [Google Scholar] [CrossRef]
  73. De Castro, F.; Stefàno, E.; Migoni, D.; Iaconisi, G.N.; Muscella, A.; Marsigliante, S.; Benedetti, M.; Fanizzi, F.P. Synthesis and Evaluation of the Cytotoxic Activity of Water-Soluble Cationic Organometallic Complexes of the Type [Pt(η1-C2H4OMe)(L)(Phen)]+ (L = NH3, DMSO; Phen = 1,10-Phenanthroline). Pharmaceutics 2021, 13, 642. [Google Scholar] [CrossRef]
  74. Benedetti, M.; De Castro, F.; Romano, A.; Migoni, D.; Piccinni, B.; Verri, T.; Lelli, M.; Roveri, N.; Fanizzi, F.P. Adsorption of the Cis-[Pt(NH3)2(P2O7)]2− (Phosphaplatin) on Hydroxyapatite Nanocrystals as a Smart Way to Selectively Release Activated Cis-[Pt(NH3)2Cl2] (Cisplatin) in Tumor Tissues. J. Inorg. Biochem. 2016, 157, 73–79. [Google Scholar] [CrossRef]
  75. Benedetti, M.; Antonucci, D.; De Castro, F.; Girelli, C.R.; Lelli, M.; Roveri, N.; Fanizzi, F.P. Metalated Nucleotide Chemisorption on Hydroxyapatite. J. Inorg. Biochem. 2015, 153, 279–283. [Google Scholar] [CrossRef] [PubMed]
  76. Benedetti, M.; Girelli, C.R.; Antonucci, D.; De Pascali, S.A.; Fanizzi, F.P. New Method for the Synthesis of [PtCl{η1-CH2C(O)R}(N-N)] Ketonyl Derivatives Starting from the Zeise’s Salt. Inorganica Chim. Acta 2014, 413, 109–114. [Google Scholar] [CrossRef]
  77. Carrisi, C.; Antonucci, D.; Lunetti, P.; Migoni, D.; Girelli, C.R.; Dolce, V.; Fanizzi, F.P.; Benedetti, M.; Capobianco, L. Transport of Platinum Bonded Nucleotides into Proteoliposomes, Mediated by Drosophila Melanogaster Thiamine Pyrophosphate Carrier Protein (DmTpc1). J. Inorg. Biochem. 2014, 130, 28–31. [Google Scholar] [CrossRef]
  78. Benedetti, M.; Lamacchia, V.; Antonucci, D.; Papadia, P.; Pacifico, C.; Natile, G.; Fanizzi, F.P. Insertion of Alkynes into Pt–X Bonds of Square Planar [PtX2(N^N)] (X = Cl, Br, I) Complexes. Dalton Trans. 2014, 43, 8826–8834. [Google Scholar] [CrossRef]
  79. Paprocka, R.; Wiese-Szadkowska, M.; Janciauskiene, S.; Kosmalski, T.; Kulik, M.; Helmin-Basa, A. Latest Developments in Metal Complexes as Anticancer Agents. Coord. Chem. Rev. 2022, 452, 214307. [Google Scholar] [CrossRef]
  80. De Castro, F.; Ciardullo, G.; Fanizzi, F.P.; Prejanò, M.; Benedetti, M.; Marino, T. Incorporation of N7-Platinated Guanines into Thermus Aquaticus (Taq) DNA Polymerase: Atomistic Insights from Molecular Dynamics Simulations. Int. J. Mol. Sci. 2023, 24, 9849. [Google Scholar] [CrossRef]
  81. De Castro, F.; Benedetti, M.; Antonaci, G.; Del Coco, L.; De Pascali, S.; Muscella, A.; Marsigliante, S.; Fanizzi, F. Response of Cisplatin Resistant Skov-3 Cells to [Pt(O,O′-Acac)(γ-Acac)(DMS)] Treatment Revealed by a Metabolomic 1H-NMR Study. Molecules 2018, 23, 2301. [Google Scholar] [CrossRef]
  82. De Castro, F.; Vergaro, V.; Benedetti, M.; Baldassarre, F.; Del Coco, L.; Dell’Anna, M.M.; Mastrorilli, P.; Fanizzi, F.P.; Ciccarella, G. Visible Light-Activated Water-Soluble Platicur Nanocolloids: Photocytotoxicity and Metabolomics Studies in Cancer Cells. ACS Appl. Bio Mater. 2020, 3, 6836–6851. [Google Scholar] [CrossRef]
  83. Zhang, C.; Xu, C.; Gao, X.; Yao, Q. Platinum-Based Drugs for Cancer Therapy and Anti-Tumor Strategies. Theranostics 2022, 12, 2115–2132. [Google Scholar] [CrossRef]
  84. Lucaciu, R.L.; Hangan, A.C.; Sevastre, B.; Oprean, L.S. Metallo-Drugs in Cancer Therapy: Past, Present and Future. Molecules 2022, 27, 6485. [Google Scholar] [CrossRef]
  85. González-Ballesteros, M.M.; Mejía, C.; Ruiz-Azuara, L. Metallodrugs: An Approach against Invasion and Metastasis in Cancer Treatment. FEBS Open Bio 2022, 12, 880–899. [Google Scholar] [CrossRef] [PubMed]
  86. Dasari, S.; Bernard Tchounwou, P. Cisplatin in Cancer Therapy: Molecular Mechanisms of Action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef]
  87. Barry, N.P.E.; Sadler, P.J. Exploration of the Medical Periodic Table: Towards New Targets. Chem. Commun. 2013, 49, 5106. [Google Scholar] [CrossRef]
  88. Saad, J.S.; Benedetti, M.; Natile, G.; Marzilli, L.G. NMR Studies of Models Having the Pt(d(GpG)) 17-Membered Macrocyclic Ring Formed in DNA by Platinum Anticancer Drugs: Pt Complexes with Bulky Chiral Diamine Ligands. Inorg. Chem. 2011, 50, 4559–4571. [Google Scholar] [CrossRef]
  89. Skowron, M.A.; Oing, C.; Bremmer, F.; Ströbel, P.; Murray, M.J.; Coleman, N.; Amatruda, J.F.; Honecker, F.; Bokemeyer, C.; Albers, P.; et al. The Developmental Origin of Cancers Defines Basic Principles of Cisplatin Resistance. Cancer Lett. 2021, 519, 199–210. [Google Scholar] [CrossRef]
  90. Schabel, F.M.; Trader, M.W.; Laster, W.R.; Corbett, T.H.; Griswold, D.P. Cis-Dichlorodiammineplatinum(II): Combination Chemotherapy and Cross-Resistance Studies with Tumors of Mice. Cancer Treat. Rep. 1979, 63, 1459–1473. [Google Scholar]
  91. Ciarimboli, G. Membrane Transporters as Mediators of Cisplatin Effects and Side Effects. Scientifica 2012, 2012, 473829. [Google Scholar] [CrossRef] [PubMed]
  92. Fujita, S.; Hirota, T.; Sakiyama, R.; Baba, M.; Ieiri, I. Identification of Drug Transporters Contributing to Oxaliplatin-induced Peripheral Neuropathy. J. Neurochem. 2019, 148, 373–385. [Google Scholar] [CrossRef] [PubMed]
  93. Planells-Cases, R.; Lutter, D.; Guyader, C.; Gerhards, N.M.; Ullrich, F.; Elger, D.A.; Kucukosmanoglu, A.; Xu, G.; Voss, F.K.; Reincke, S.M.; et al. Subunit Composition of VRAC Channels Determines Substrate Specificity and Cellular Resistance to P T-based Anti-cancer Drugs. EMBO J. 2015, 34, 2993–3008. [Google Scholar] [CrossRef]
  94. Frezza, M.; Hindo, S.; Chen, D.; Davenport, A.; Schmitt, S.; Tomco, D.; Ping Dou, Q. Novel Metals and Metal Complexes as Platforms for Cancer Therapy. Curr. Pharm. Des. 2010, 16, 1813–1825. [Google Scholar] [CrossRef]
  95. Sousa, G.F.D.; Wlodarczyk, S.R.; Monteiro, G. Carboplatin: Molecular Mechanisms of Action Associated with Chemoresistance. Braz. J. Pharm. Sci. 2014, 50, 693–701. [Google Scholar] [CrossRef]
  96. Yu, F.; Megyesi, J.; Price, P.M. Cytoplasmic Initiation of Cisplatin Cytotoxicity. Am. J. Physiol. Ren. Physiol. 2008, 295, F44–F52. [Google Scholar] [CrossRef]
  97. Raudenska, M.; Balvan, J.; Fojtu, M.; Gumulec, J.; Masarik, M. Unexpected Therapeutic Effects of Cisplatin. Metallomics 2019, 11, 1182–1199. [Google Scholar] [CrossRef]
  98. Becker, J.P.; Weiss, J.; Theile, D. Cisplatin, Oxaliplatin, and Carboplatin Unequally Inhibit in Vitro mRNA Translation. Toxicol. Lett. 2014, 225, 43–47. [Google Scholar] [CrossRef]
  99. Galluzzi, L.; Vitale, I.; Michels, J.; Brenner, C.; Szabadkai, G.; Harel-Bellan, A.; Castedo, M.; Kroemer, G. Systems Biology of Cisplatin Resistance: Past, Present and Future. Cell Death Dis. 2014, 5, e1257. [Google Scholar] [CrossRef]
  100. Lazarević, T.; Rilak, A.; Bugarčić, Ž.D. Platinum, Palladium, Gold and Ruthenium Complexes as Anticancer Agents: Current Clinical Uses, Cytotoxicity Studies and Future Perspectives. Eur. J. Med. Chem. 2017, 142, 8–31. [Google Scholar] [CrossRef]
  101. Hah, S.S.; Stivers, K.M.; De Vere White, R.W.; Henderson, P.T. Kinetics of Carboplatin−DNA Binding in Genomic DNA and Bladder Cancer Cells As Determined by Accelerator Mass Spectrometry. Chem. Res. Toxicol. 2006, 19, 622–626. [Google Scholar] [CrossRef]
  102. Aldossary, S.A. Review on Pharmacology of Cisplatin: Clinical Use, Toxicity and Mechanism of Resistance of Cisplatin. Biomed. Pharmacol. J. 2019, 12, 07–15. [Google Scholar] [CrossRef]
  103. Anthony, E.J.; Bolitho, E.M.; Bridgewater, H.E.; Carter, O.W.L.; Donnelly, J.M.; Imberti, C.; Lant, E.C.; Lermyte, F.; Needham, R.J.; Palau, M.; et al. Metallodrugs Are Unique: Opportunities and Challenges of Discovery and Development. Chem. Sci. 2020, 11, 12888–12917. [Google Scholar] [CrossRef] [PubMed]
  104. Siddik, Z.H. Cisplatin: Mode of Cytotoxic Action and Molecular Basis of Resistance. Oncogene 2003, 22, 7265–7279. [Google Scholar] [CrossRef]
  105. Singh, S.; Upadhyay, A.K.; Ajay, A.K.; Bhat, M.K. P53 Regulates ERK Activation in Carboplatin Induced Apoptosis in Cervical Carcinoma: A Novel Target of P53 in Apoptosis. FEBS Lett. 2007, 581, 289–295. [Google Scholar] [CrossRef]
  106. Guégan, J.-P.; Ezan, F.; Théret, N.; Langouët, S.; Baffet, G. MAPK Signaling in Cisplatin-Induced Death: Predominant Role of ERK1 over ERK2 in Human Hepatocellular Carcinoma Cells. Carcinogenesis 2013, 34, 38–47. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, K.-B.; Byun, H.-J.; Park, S.H.; Park, C.-Y.; Lee, S.-H.; Rho, S.B. CYR61 Controls P53 and NF-κB Expression through PI3K/Akt/mTOR Pathways in Carboplatin-Induced Ovarian Cancer Cells. Cancer Lett. 2012, 315, 86–95. [Google Scholar] [CrossRef] [PubMed]
  108. Kleih, M.; Böpple, K.; Dong, M.; Gaißler, A.; Heine, S.; Olayioye, M.A.; Aulitzky, W.E.; Essmann, F. Direct Impact of Cisplatin on Mitochondria Induces ROS Production That Dictates Cell Fate of Ovarian Cancer Cells. Cell Death Dis. 2019, 10, 851. [Google Scholar] [CrossRef]
  109. Muslimović, A.; Nyström, S.; Gao, Y.; Hammarsten, O. Numerical Analysis of Etoposide Induced DNA Breaks. PLoS ONE 2009, 4, e5859. [Google Scholar] [CrossRef]
  110. Zhang, H.-H.; Li, S.-Z.; Zhang, Z.-Y.; Hu, X.-M.; Hou, P.-N.; Gao, L.; Du, R.-L.; Zhang, X.-D. Nemo-like Kinase Is Critical for P53 Stabilization and Function in Response to DNA Damage. Cell Death Differ. 2014, 21, 1656–1663. [Google Scholar] [CrossRef]
  111. Jamil, S.; Lam, I.; Majd, M.; Tsai, S.-H.; Duronio, V. Etoposide Induces Cell Death via Mitochondrial-Dependent Actions of P53. Cancer Cell Int. 2015, 15, 79. [Google Scholar] [CrossRef]
  112. Karpinich, N.O.; Tafani, M.; Rothman, R.J.; Russo, M.A.; Farber, J.L. The Course of Etoposide-Induced Apoptosis from Damage to DNA and P53 Activation to Mitochondrial Release of Cytochromec. J. Biol. Chem. 2002, 277, 16547–16552. [Google Scholar] [CrossRef]
  113. Day, T.W.; Wu, C.-H.; Safa, A.R. Etoposide Induces Protein Kinase Cδ- and Caspase-3-Dependent Apoptosis in Neuroblastoma Cancer Cells. Mol. Pharmacol. 2009, 76, 632–640. [Google Scholar] [CrossRef] [PubMed]
  114. Robertson, J.D.; Enoksson, M.; Suomela, M.; Zhivotovsky, B.; Orrenius, S. Caspase-2 Acts Upstream of Mitochondria to Promote Cytochromec Release during Etoposide-Induced Apoptosis. J. Biol. Chem. 2002, 277, 29803–29809. [Google Scholar] [CrossRef]
  115. Xie, B.-S.; Zhao, H.-C.; Yao, S.-K.; Zhuo, D.-X.; Jin, B.; Lv, D.-C.; Wu, C.-L.; Ma, D.-L.; Gao, C.; Shu, X.-M.; et al. Autophagy Inhibition Enhances Etoposide-Induced Cell Death in Human Hepatoma G2 Cells. Int. J. Mol. Med. 2011, 27, 599–606. [Google Scholar] [CrossRef]
  116. Montecucco, A.; Zanetta, F.; Biamonti, G. Molecular Mechanisms of Etoposide. EXCLI J. 2015, 14, 95. [Google Scholar] [CrossRef]
  117. Katayama, M.; Kawaguchi, T.; Berger, M.S.; Pieper, R.O. DNA Damaging Agent-Induced Autophagy Produces a Cytoprotective Adenosine Triphosphate Surge in Malignant Glioma Cells. Cell Death Differ. 2007, 14, 548–558. [Google Scholar] [CrossRef] [PubMed]
  118. Abdel-Rahman, O.; Koski, S.L. Cisplatin-Based versus Carboplatin-Based Chemotherapy for Extrapulmonary Neuroendocrine Carcinomas: A Real-World Study. Neuroendocrinology 2022, 112, 777–783. [Google Scholar] [CrossRef]
  119. Robbins, H.L.; Hague, A. The PI3K/Akt Pathway in Tumors of Endocrine Tissues. Front. Endocrinol. 2016, 6, 188. [Google Scholar] [CrossRef]
  120. Wolin, E.M. PI3K/Akt/mTOR Pathway Inhibitors in the Therapy of Pancreatic Neuroendocrine Tumors. Cancer Lett. 2013, 335, 1–8. [Google Scholar] [CrossRef]
  121. Antonuzzo, L.; Del Re, M.; Barucca, V.; Spada, F.; Meoni, G.; Restante, G.; Danesi, R.; Di Costanzo, F.; Fazio, N. Critical Focus on Mechanisms of Resistance and Toxicity of M-TOR Inhibitors in Pancreatic Neuroendocrine Tumors. Cancer Treat. Rev. 2017, 57, 28–35. [Google Scholar] [CrossRef] [PubMed]
  122. Gajate, P.; Alonso-Gordoa, T.; Martínez-Sáez, O.; Molina-Cerrillo, J.; Grande, E. Prognostic and Predictive Role of the PI3K–AKT–mTOR Pathway in Neuroendocrine Neoplasms. Clin. Transl. Oncol. 2018, 20, 561–569. [Google Scholar] [CrossRef]
  123. Zarebczan, B.; Chen, H. Signaling Mechanisms in Neuroendocrine Tumors as Targets for Therapy. Endocrinol. Metab. Clin. N. Am. 2010, 39, 801–810. [Google Scholar] [CrossRef]
  124. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef]
  125. Wiedmer, T.; Blank, A.; Pantasis, S.; Normand, L.; Bill, R.; Krebs, P.; Tschan, M.P.; Marinoni, I.; Perren, A. Autophagy Inhibition Improves Sunitinib Efficacy in Pancreatic Neuroendocrine Tumors via a Lysosome-Dependent Mechanism. Mol. Cancer Ther. 2017, 16, 2502–2515. [Google Scholar] [CrossRef] [PubMed]
  126. Matrood, S.; De Prisco, N.; Wissniowski, T.T.; Wiese, D.; Jabari, S.; Griesmann, H.; Wanzel, M.; Stiewe, T.; Neureiter, D.; Klieser, E.; et al. Modulation of Pancreatic Neuroendocrine Neoplastic Cell Fate by Autophagy-Mediated Death. Neuroendocrinology 2021, 111, 965–985. [Google Scholar] [CrossRef]
  127. Gagliano, T.; Bellio, M.; Gentilin, E.; Molè, D.; Tagliati, F.; Schiavon, M.; Cavallesco, N.G.; Andriolo, L.G.; Ambrosio, M.R.; Rea, F.; et al. mTOR, p70S6K, AKT, and ERK1/2 Levels Predict Sensitivity to mTOR and PI3K/mTOR Inhibitors in Human Bronchial Carcinoids. Endocr.-Relat. Cancer 2013, 20, 463–475. [Google Scholar] [CrossRef]
  128. Perren, A.; Komminoth, P.; Saremaslani, P.; Matter, C.; Feurer, S.; Lees, J.A.; Heitz, P.U.; Eng, C. Mutation and Expression Analyses Reveal Differential Subcellular Compartmentalization of PTEN in Endocrine Pancreatic Tumors Compared to Normal Islet Cells. Am. J. Pathol. 2000, 157, 1097–1103. [Google Scholar] [CrossRef] [PubMed]
  129. Missiaglia, E.; Dalai, I.; Barbi, S.; Beghelli, S.; Falconi, M.; Della Peruta, M.; Piemonti, L.; Capurso, G.; Di Florio, A.; Delle Fave, G.; et al. Pancreatic Endocrine Tumors: Expression Profiling Evidences a Role for AKT-mTOR Pathway. J. Clin. Oncol. 2010, 28, 245–255. [Google Scholar] [CrossRef]
  130. Jayakumar, R.; Lanjewar, S.; Axiotis, C.A. Loss of PTEN and Increased pAKT Expression Distinguishes Aggressive Low-Grade Neuroendocrine Tumors. Ann. Clin. Lab. Sci. 2018, 48, 565–572. [Google Scholar]
  131. Tendler, S.; Kanter, L.; Lewensohn, R.; Ortiz-Villalón, C.; Viktorsson, K.; De Petris, L. The Prognostic Implications of Notch1, Hes1, Ascl1, and DLL3 Protein Expression in SCLC Patients Receiving Platinum-Based Chemotherapy. PLoS ONE 2020, 15, e0240973. [Google Scholar] [CrossRef]
  132. Gajra, A.; Tatum, A.H.; Newman, N.; Gamble, G.P.; Lichtenstein, S.; Rooney, M.T.; Graziano, S.L. The Predictive Value of Neuroendocrine Markers and P53 for Response to Chemotherapy and Survival in Patients with Advanced Non-Small Cell Lung Cancer. Lung Cancer 2002, 36, 159–165. [Google Scholar] [CrossRef] [PubMed]
  133. Chen, Y.; Nowak, I.; Huang, J.; Keng, P.C.; Sun, H.; Xu, H.; Wei, G.; Lee, S.O. Erk/MAP Kinase Signaling Pathway and Neuroendocrine Differentiation of Non–Small-Cell Lung Cancer. J. Thorac. Oncol. 2014, 9, 50–58. [Google Scholar] [CrossRef] [PubMed]
  134. Nakakura, E.K.; Sriuranpong, V.R.; Kunnimalaiyaan, M.; Hsiao, E.C.; Schuebel, K.E.; Borges, M.W.; Jin, N.; Collins, B.J.; Nelkin, B.D.; Chen, H.; et al. Regulation of Neuroendocrine Differentiation in Gastrointestinal Carcinoid Tumor Cells by Notch Signaling. J. Clin. Endocrinol. Metab. 2005, 90, 4350–4356. [Google Scholar] [CrossRef] [PubMed]
  135. Jiao, Y.; Shi, C.; Edil, B.H.; De Wilde, R.F.; Klimstra, D.S.; Maitra, A.; Schulick, R.D.; Tang, L.H.; Wolfgang, C.L.; Choti, M.A.; et al. DAXX/ATRX, MEN1, and mTOR Pathway Genes Are Frequently Altered in Pancreatic Neuroendocrine Tumors. Science 2011, 331, 1199–1203. [Google Scholar] [CrossRef] [PubMed]
  136. Wong, H.; Yang, K.C.; Shen, Y.; Zhao, E.Y.; Loree, J.M.; Kennecke, H.F.; Kalloger, S.E.; Karasinska, J.M.; Lim, H.J.; Mungall, A.J.; et al. Molecular Characterization of Metastatic Pancreatic Neuroendocrine Tumors (PNETs) Using Whole-Genome and Transcriptome Sequencing. Cold Spring Harb. Mol. Case Stud. 2018, 4, a002329. [Google Scholar] [CrossRef] [PubMed]
  137. Umetsu, S.E.; Kakar, S.; Basturk, O.; Kim, G.E.; Chatterjee, D.; Wen, K.W.; Hale, G.; Shafizadeh, N.; Cho, S.-J.; Whitman, J.; et al. Integrated Genomic and Clinicopathologic Approach Distinguishes Pancreatic Grade 3 Neuroendocrine Tumor from Neuroendocrine Carcinoma and Identifies a Subset with Molecular Overlap. Mod. Pathol. 2023, 36, 100065. [Google Scholar] [CrossRef] [PubMed]
  138. Couce, M.E.; Bautista, D.; Costa, J.; Carter, D. Analysis of K-Ras, N-Ras, H-Ras, and P53 in Lung Neuroendocrine Neoplasms. Diagn. Mol. Pathol. 1999, 8, 71–79. [Google Scholar] [CrossRef]
  139. Hu, W.; Feng, Z.; Modica, I.; Klimstra, D.S.; Song, L.; Allen, P.J.; Brennan, M.F.; Levine, A.J.; Tang, L.H. Gene Amplifications in Well-Differentiated Pancreatic Neuroendocrine Tumors Inactivate the P53 Pathway. Genes Cancer 2010, 1, 360–368. [Google Scholar] [CrossRef] [PubMed]
  140. Yachida, S.; Vakiani, E.; White, C.M.; Zhong, Y.; Saunders, T.; Morgan, R.; de Wilde, R.F.; Maitra, A.; Hicks, J.; Demarzo, A.M.; et al. Small Cell and Large Cell Neuroendocrine Carcinomas of the Pancreas Are Genetically Similar and Distinct from Well-Differentiated Pancreatic Neuroendocrine Tumors. Am. J. Surg. Pathol. 2012, 36, 173–184. [Google Scholar] [CrossRef]
  141. Pal’tsev, M.A.; Demura, S.A.; Kogan, E.A.; Jaques, G.; Zende, B. Role ofBcl-2, Bax, andBak in Spontaneous Apoptosis and Proliferation in Neuroendocrine Lung Tumors: Immunohistochemical Study. Bull. Exp. Biol. Med. 2000, 130, 697–700. [Google Scholar] [CrossRef]
  142. Brambilla, E.; Negoescu, A.; Gazzeri, S.; Lantuejoul, S.; Moro, D.; Brambilla, C.; Coll, J.L. Apoptosis-Related Factors P53, Bcl2, and Bax in Neuroendocrine Lung Tumors. Am. J. Pathol. 1996, 149, 1941–1952. [Google Scholar] [PubMed]
  143. Roland, C.L.; Starker, L.F.; Kang, Y.; Chatterjee, D.; Estrella, J.; Rashid, A.; Katz, M.H.; Aloia, T.A.; Lee, J.E.; Dasari, A.; et al. Loss of DPC4/SMAD4 Expression in Primary Gastrointestinal Neuroendocrine Tumors Is Associated with Cancer-Related Death after Resection. Surgery 2017, 161, 753–759. [Google Scholar] [CrossRef] [PubMed]
  144. Hofving, T.; Elias, E.; Rehammar, A.; Inge, L.; Altiparmak, G.; Persson, M.; Kristiansson, E.; Johansson, M.E.; Nilsson, O.; Arvidsson, Y. SMAD4 Haploinsufficiency in Small Intestinal Neuroendocrine Tumors. BMC Cancer 2021, 21, 101. [Google Scholar] [CrossRef]
  145. Banck, M.S.; Kanwar, R.; Kulkarni, A.A.; Boora, G.K.; Metge, F.; Kipp, B.R.; Zhang, L.; Thorland, E.C.; Minn, K.T.; Tentu, R.; et al. The Genomic Landscape of Small Intestine Neuroendocrine Tumors. J. Clin. Investig. 2013, 123, 2502–2508. [Google Scholar] [CrossRef] [PubMed]
  146. Cejas, P.; Drier, Y.; Dreijerink, K.M.A.; Brosens, L.A.A.; Deshpande, V.; Epstein, C.B.; Conemans, E.B.; Morsink, F.H.M.; Graham, M.K.; Valk, G.D.; et al. Enhancer Signatures Stratify and Predict Outcomes of Non-Functional Pancreatic Neuroendocrine Tumors. Nat. Med. 2019, 25, 1260–1265. [Google Scholar] [CrossRef]
  147. Marinoni, I.; Kurrer, A.S.; Vassella, E.; Dettmer, M.; Rudolph, T.; Banz, V.; Hunger, F.; Pasquinelli, S.; Speel, E.; Perren, A. Loss of DAXX and ATRX Are Associated with Chromosome Instability and Reduced Survival of Patients with Pancreatic Neuroendocrine Tumors. Gastroenterology 2014, 146, 453–460.e5. [Google Scholar] [CrossRef]
  148. Cives, M.; Partelli, S.; Palmirotta, R.; Lovero, D.; Mandriani, B.; Quaresmini, D.; Pelle’, E.; Andreasi, V.; Castelli, P.; Strosberg, J.; et al. DAXX Mutations as Potential Genomic Markers of Malignant Evolution in Small Nonfunctioning Pancreatic Neuroendocrine Tumors. Sci. Rep. 2019, 9, 18614. [Google Scholar] [CrossRef]
  149. Krausch, M.; Kroepil, F.; Lehwald, N.; Lachenmayer, A.; Schott, M.; Anlauf, M.; Cupisti, K.; Knoefel, W.T.; Raffel, A. Notch 1 Tumor Expression Is Lacking in Highly Proliferative Pancreatic Neuroendocrine Tumors. Endocrine 2013, 44, 182–186. [Google Scholar] [CrossRef]
  150. Panelos, J.; Batistatou, A.; Paglierani, M.; Zioga, A.; Maio, V.; Santi, R.; Pimpinelli, N.; De Giorgi, V.; Santucci, M.; Massi, D. Expression of Notch-1 and Alteration of the E-Cadherin/β-Catenin Cell Adhesion Complex Are Observed in Primary Cutaneous Neuroendocrine Carcinoma (Merkel Cell Carcinoma). Mod. Pathol. 2009, 22, 959–968. [Google Scholar] [CrossRef]
  151. Kikuchi, H.; Sakakibara-Konishi, J.; Furuta, M.; Yokouchi, H.; Nishihara, H.; Yamazaki, S.; Uramoto, H.; Tanaka, F.; Harada, M.; Akie, K.; et al. Expression of Notch1 and Numb in Small Cell Lung Cancer. Oncotarget 2017, 8, 10348–10358. [Google Scholar] [CrossRef]
  152. George, J.; Walter, V.; Peifer, M.; Alexandrov, L.B.; Seidel, D.; Leenders, F.; Maas, L.; Müller, C.; Dahmen, I.; Delhomme, T.M.; et al. Integrative Genomic Profiling of Large-Cell Neuroendocrine Carcinomas Reveals Distinct Subtypes of High-Grade Neuroendocrine Lung Tumors. Nat. Commun. 2018, 9, 1048. [Google Scholar] [CrossRef]
  153. Lohmann, D.R.; Funk, A.; Niedermeyer, H.P.; Häupel, S.; Höfler, H. Identification of P53 Gene Mutations in Gastrointestinal and Pancreatic Carcinoids by Nonradioisotopic SSCA. Virchows Arch. B Cell Pathol. 1993, 64, 293–296. [Google Scholar] [CrossRef]
  154. Weckström, P.; Hedrum, A.; Makridis, C.; Åkerström, G.; Rastad, J.; Scheibenpflug, L.; Uhlén, M.; Juhlin, C.; Wilander, E. Midgut Carcinoids and Solid Carcinomas of the Intestine: Differences in Endocrine Markers and P53 Mutations. Endocr. Pathol. 1996, 7, 273–279. [Google Scholar] [CrossRef]
  155. Przygodzki, R.M.; Finkelstein, S.D.; Langer, J.C.; Swalsky, P.A.; Fishback, N.; Bakker, A.; Guinee, D.G.; Koss, M.; Travis, W.D. Analysis of P53, K-Ras-2, and C-Raf-1 in Pulmonary Neuroendocrine Tumors. Correlation with Histological Subtype and Clinical Outcome. Am. J. Pathol. 1996, 148, 1531–1541. [Google Scholar]
  156. Voortman, J.; Lee, J.-H.; Killian, J.K.; Suuriniemi, M.; Wang, Y.; Lucchi, M.; Smith, W.I.; Meltzer, P.; Wang, Y.; Giaccone, G. Array Comparative Genomic Hybridization-Based Characterization of Genetic Alterations in Pulmonary Neuroendocrine Tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 13040–13045. [Google Scholar] [CrossRef] [PubMed]
  157. Tan, H.-L.; Sood, A.; Rahimi, H.A.; Wang, W.; Gupta, N.; Hicks, J.; Mosier, S.; Gocke, C.D.; Epstein, J.I.; Netto, G.J.; et al. Rb Loss Is Characteristic of Prostatic Small Cell Neuroendocrine Carcinoma. Clin. Cancer Res. 2014, 20, 890–903. [Google Scholar] [CrossRef]
  158. Takizawa, N.; Ohishi, Y.; Hirahashi, M.; Takahashi, S.; Nakamura, K.; Tanaka, M.; Oki, E.; Takayanagi, R.; Oda, Y. Molecular Characteristics of Colorectal Neuroendocrine Carcinoma; Similarities with Adenocarcinoma Rather than Neuroendocrine Tumor. Hum. Pathol. 2015, 46, 1890–1900. [Google Scholar] [CrossRef]
  159. Rekhtman, N.; Pietanza, M.C.; Hellmann, M.D.; Naidoo, J.; Arora, A.; Won, H.; Halpenny, D.F.; Wang, H.; Tian, S.K.; Litvak, A.M.; et al. Next-Generation Sequencing of Pulmonary Large Cell Neuroendocrine Carcinoma Reveals Small Cell Carcinoma–like and Non–Small Cell Carcinoma–like Subsets. Clin. Cancer Res. 2016, 22, 3618–3629. [Google Scholar] [CrossRef]
  160. Miyoshi, T.; Umemura, S.; Matsumura, Y.; Mimaki, S.; Tada, S.; Makinoshima, H.; Ishii, G.; Udagawa, H.; Matsumoto, S.; Yoh, K.; et al. Genomic Profiling of Large-Cell Neuroendocrine Carcinoma of the Lung. Clin. Cancer Res. 2017, 23, 757–765. [Google Scholar] [CrossRef]
  161. Shamir, E.R.; Devine, W.P.; Pekmezci, M.; Umetsu, S.E.; Krings, G.; Federman, S.; Cho, S.-J.; Saunders, T.A.; Jen, K.-Y.; Bergsland, E.; et al. Identification of High-Risk Human Papillomavirus and Rb/E2F Pathway Genomic Alterations in Mutually Exclusive Subsets of Colorectal Neuroendocrine Carcinoma. Mod. Pathol. 2019, 32, 290–305. [Google Scholar] [CrossRef] [PubMed]
  162. Hillman, R.T.; Cardnell, R.; Fujimoto, J.; Lee, W.-C.; Zhang, J.; Byers, L.A.; Ramalingam, P.; Leitao, M.; Swisher, E.; Futreal, P.A.; et al. Comparative Genomics of High Grade Neuroendocrine Carcinoma of the Cervix. PLoS ONE 2020, 15, e0234505. [Google Scholar] [CrossRef]
  163. Xing, J.; Chen, J.; You, T.; Lu, T.; Wu, H.; Bai, C.; Sun, Z.; Cheng, Y. P53 and Rb Immunohistochemistry Staining Reveal Subtypes of Gastric Neuroendocrine Carcinoma with Distinct Prognosis. J. Clin. Oncol. 2022, 40, e16210. [Google Scholar] [CrossRef]
  164. Tannapfel, A.; Vomschloss, S.; Karhoff, D.; Markwarth, A.; Hengge, U.R.; Wittekind, C.; Arnold, R.; Hörsch, D. BRAF Gene Mutations Are Rare Events in Gastroenteropancreatic Neuroendocrine Tumors. Am. J. Clin. Pathol. 2005, 123, 256–260. [Google Scholar] [CrossRef] [PubMed]
  165. Kleist, B.; Kempa, M.; Novy, M.; Oberkanins, C.; Xu, L.; Li, G.; Loland, C.; Poetsch, M. Comparison of Neuroendocrine Differentiation and KRAS/NRAS/BRAF/PIK3CA/TP53 Mutation Status in Primary and Metastatic Colorectal Cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 5927–5939. [Google Scholar]
  166. Ganesh, K.; Raj, N.P.; Yaeger, R.D.; Berger, M.F.; Shia, J.; Reidy, D.L. BRAF Mutations in Patients with Large Cell Neuroendocrine Carcinoma of the Colon (LCNECC). J. Clin. Oncol. 2016, 34, 567. [Google Scholar] [CrossRef]
  167. Kim, M.K.; Ye, F.; Wang, D.; Cui, M.; Ward, S.C.; Warner, R.R.P.; Roayaie, S.; Shafir, M.; Schwartz, M.; Zhang, D.; et al. Differential Protein Expression in Small Intestinal Neuroendocrine Tumors and Liver Metastases. Pancreas 2016, 45, 528–532. [Google Scholar] [CrossRef]
  168. Shida, T.; Kishimoto, T.; Furuya, M.; Nikaido, T.; Koda, K.; Takano, S.; Kimura, F.; Shimizu, H.; Yoshidome, H.; Ohtsuka, M.; et al. Expression of an Activated Mammalian Target of Rapamycin (mTOR) in Gastroenteropancreatic Neuroendocrine Tumors. Cancer Chemother. Pharmacol. 2010, 65, 889–893. [Google Scholar] [CrossRef]
  169. Catena, L.; Bajetta, E.; Milione, M.; Ducceschi, M.; Valente, M.; Dominoni, F.; Colonna, V. Mammalian Target of Rapamycin Expression in Poorly Differentiated Endocrine Carcinoma: Clinical and Therapeutic Future Challenges. Target. Oncol. 2011, 6, 65–68. [Google Scholar] [CrossRef]
  170. Kasajima, A.; Pavel, M.; Darb-Esfahani, S.; Noske, A.; Stenzinger, A.; Sasano, H.; Dietel, M.; Denkert, C.; Rocken, C.; Wiedenmann, B.; et al. mTOR Expression and Activity Patterns in Gastroenteropancreatic Neuroendocrine Tumours. Endocr.-Relat. Cancer 2011, 18, 181–192. [Google Scholar] [CrossRef]
  171. Zhou, C.-F.; Ji, J.; Yuan, F.; Shi, M.; Zhang, J.; Liu, B.-Y.; Zhu, Z.-G. mTOR Activation in Well Differentiated Pancreatic Neuroendocrine Tumors: A Retrospective Study on 34 Cases. Hepato-Gastroenterology 2011, 58, 2140–2143. [Google Scholar] [CrossRef]
  172. Peng, D.-J.; Wang, J.; Zhou, J.-Y.; Wu, G.S. Role of the Akt/mTOR Survival Pathway in Cisplatin Resistance in Ovarian Cancer Cells. Biochem. Biophys. Res. Commun. 2010, 394, 600–605. [Google Scholar] [CrossRef]
  173. Yu, C.; Yu, Q.; Lu, J.; Zhou, J. KRT17 Enhances Carboplatin Resistance in Ovarian Cancer through the AKT/mTOR Pathway. Eur. J. Gynaecol. Oncol. 2022, 43, 49–57. [Google Scholar] [CrossRef]
  174. Nasrpour Navaei, Z.; Khalili-Tanha, G.; Sadra Zangouei, A.; Reza Abbaszadegan, M.; Moghbeli, M. PI3K/AKT Signaling Pathway as a Critical Regulator of Cisplatin Response in Tumor Cells. Oncol. Res. 2021, 29, 235–250. [Google Scholar] [CrossRef]
  175. Li, D.; Zhang, Y.; Xie, Y.; Xiang, J.; Zhu, Y.; Yang, J. Enhanced Tumor Suppression by Adenoviral PTEN Gene Therapy Combined with Cisplatin Chemotherapy in Small-Cell Lung Cancer. Cancer Gene Ther. 2013, 20, 251–259. [Google Scholar] [CrossRef]
  176. Minami, D.; Takigawa, N.; Takeda, H.; Takata, M.; Ochi, N.; Ichihara, E.; Hisamoto, A.; Hotta, K.; Tanimoto, M.; Kiura, K. Synergistic Effect of Olaparib with Combination of Cisplatin on PTEN -Deficient Lung Cancer Cells. Mol. Cancer Res. 2013, 11, 140–148. [Google Scholar] [CrossRef]
  177. Omura, M.; Kosaka, T.; Aimono, E.; Nakamura, K.; Hongo, H.; Mikami, S.; Nishihara, H.; Oya, M. First Successful Case of Platinum-based Chemotherapy for Neuroendocrine Prostate Cancer with BRCA2 and PTEN Alterations. IJU Case Rep. 2022, 5, 41–44. [Google Scholar] [CrossRef]
  178. Zhang, L.; Zhu, X.; Liu, C.; Zhang, B.; Zheng, J.; Singh, P.K.; Bshara, W.; Wang, J.; Gomez, E.C.; Zhang, X.; et al. PTEN Loss Expands the Histopathologic Diversity and Lineage Plasticity of Lung Cancers Initiated by Rb1/Trp53 Deletion. J. Thorac. Oncol. 2023, 18, 324–338. [Google Scholar] [CrossRef]
  179. Chang, T.-M.; Chu, P.-Y.; Lin, H.-Y.; Huang, K.-W.; Hung, W.-C.; Shan, Y.-S.; Chen, L.-T.; Tsai, H.-J. PTEN Regulates Invasiveness in Pancreatic Neuroendocrine Tumors through DUSP19-Mediated VEGFR3 Dephosphorylation. J. Biomed. Sci. 2022, 29, 92. [Google Scholar] [CrossRef]
  180. Song, M.S.; Salmena, L.; Pandolfi, P.P. The Functions and Regulation of the PTEN Tumour Suppressor. Nat. Rev. Mol. Cell Biol. 2012, 13, 283–296. [Google Scholar] [CrossRef]
  181. Puc, J.; Keniry, M.; Li, H.S.; Pandita, T.K.; Choudhury, A.D.; Memeo, L.; Mansukhani, M.; Murty, V.V.V.S.; Gaciong, Z.; Meek, S.E.M.; et al. Lack of PTEN Sequesters CHK1 and Initiates Genetic Instability. Cancer Cell 2005, 7, 193–204. [Google Scholar] [CrossRef]
  182. Hijioka, S.; Hosoda, W.; Matsuo, K.; Ueno, M.; Furukawa, M.; Yoshitomi, H.; Kobayashi, N.; Ikeda, M.; Ito, T.; Nakamori, S.; et al. Rb Loss and KRAS Mutation Are Predictors of the Response to Platinum-Based Chemotherapy in Pancreatic Neuroendocrine Neoplasm with Grade 3: A Japanese Multicenter Pancreatic NEN-G3 Study. Clin. Cancer Res. 2017, 23, 4625–4632. [Google Scholar] [CrossRef]
  183. Tanaka, H.; Hijioka, S.; Hosoda, W.; Ueno, M.; Kobayashi, N.; Ikeda, M.; Ito, T.; Kodama, Y.; Morizane, C.; Notohara, K.; et al. Pancreatic Neuroendocrine Carcinoma G3 May Be Heterogeneous and Could Be Classified into Two Distinct Groups. Pancreatology 2020, 20, 1421–1427. [Google Scholar] [CrossRef]
  184. Elvebakken, H.; Perren, A.; Scoazec, J.-Y.; Tang, L.H.; Federspiel, B.; Klimstra, D.S.; Vestermark, L.W.; Ali, A.S.; Zlobec, I.; Myklebust, T.Å.; et al. A Consensus-Developed Morphological Re-Evaluation of 196 High-Grade Gastroenteropancreatic Neuroendocrine Neoplasms and Its Clinical Correlations. Neuroendocrinology 2021, 111, 883–894. [Google Scholar] [CrossRef]
  185. Ogawa, H.; Sakai, Y.; Nishio, W.; Fujibayashi, Y.; Nishikubo, M.; Nishioka, Y.; Tane, S.; Kitamura, Y.; Sudo, T.; Sakuma, T.; et al. DLL3 Expression Is a Predictive Marker of Sensitivity to Adjuvant Chemotherapy for Pulmonary LCNEC. Thorac. Cancer 2020, 11, 2561–2569. [Google Scholar] [CrossRef]
  186. Daimon, T.; Kosaka, T.; Hongo, H.; Aimono, E.; Nakamura, K.; Mikami, S.; Nishihara, H.; Oya, M. Prominent Response to Platinum-based Chemotherapy in a Patient with BRCA2 Mutant-neuroendocrine Prostate Cancer and MDM2 Amplification. IJU Case Rep. 2021, 4, 216–219. [Google Scholar] [CrossRef]
  187. Chen, D.; Xu, J.; Qiao, R.; Zhao, Y.; Chu, T.; Han, B.; Zhong, R. Detection of Genetic Mutations by Next-Generation Sequencing for Predicting Prognosis of Extensive-Stage Small-Cell Lung Cancer. J. Oncol. 2020, 2020, 8811487. [Google Scholar] [CrossRef]
  188. Hongo, H.; Kosaka, T.; Nakatsuka, S.; Oya, M. A Long-Term Survivor of Metastatic Neuroendocrine Prostate Cancer Treated with Multimodal Therapy: Genetic Consideration from next-Generation Sequencing. Int. Cancer Conf. J. 2021, 10, 174–180. [Google Scholar] [CrossRef]
  189. Elvebakken, H.; Venizelos, A.; Perren, A.; Couvelard, A.; Lothe, I.M.B.; Hjortland, G.O.; Myklebust, T.Å.; Svensson, J.; Garresori, H.; Kersten, C.; et al. Treatment Outcome According to Genetic Tumour Alterations and Clinical Characteristics in Digestive High-Grade Neuroendocrine Neoplasms. Br. J. Cancer 2024. [Google Scholar] [CrossRef]
  190. Dowlati, A.; Lipka, M.B.; McColl, K.; Dabir, S.; Behtaj, M.; Kresak, A.; Miron, A.; Yang, M.; Sharma, N.; Fu, P.; et al. Clinical Correlation of Extensive-Stage Small-Cell Lung Cancer Genomics. Ann. Oncol. 2016, 27, 642–647. [Google Scholar] [CrossRef] [PubMed]
  191. Lacombe, C.; De Rycke, O.; Couvelard, A.; Turpin, A.; Cazes, A.; Hentic, O.; Gounant, V.; Zalcman, G.; Ruszniewski, P.; Cros, J.; et al. Biomarkers of Response to Etoposide-Platinum Chemotherapy in Patients with Grade 3 Neuroendocrine Neoplasms. Cancers 2021, 13, 643. [Google Scholar] [CrossRef]
  192. Kosaka, T.; Hongo, H.; Aimono, E.; Matsumoto, K.; Hayashida, T.; Mikami, S.; Nishihara, H.; Oya, M. A First Japanese Case of Neuroendocrine Prostate Cancer Accompanied by Lung and Brain Metastasis with Somatic and Germline BRCA2 Mutation. Pathol. Int. 2019, 69, 715–720. [Google Scholar] [CrossRef] [PubMed]
  193. Turina, C.B.; Coleman, D.J.; Thomas, G.V.; Fung, A.W.; Alumkal, J.J. Molecular Testing Identifies Determinants of Exceptional Response and Guides Precision Therapy in a Patient with Lethal, Treatment-Emergent Neuroendocrine Prostate Cancer. Cureus 2019, 11, e5197. [Google Scholar] [CrossRef] [PubMed]
  194. Pandya, D.; Shah, M.; Kaplan, F.; Martino, C.; Levy, G.; Kazanjian, M.; Batter, S.; Martignetti, J.; Frank, R.C. Treatment-Emergent Neuroendocrine Prostate Cancer with a Germline BRCA2 Mutation: Identification of a Candidate Reversion Mutation Associated with Platinum/PARP-Inhibitor Resistance. Cold Spring Harb. Mol. Case Stud. 2021, 7, a005801. [Google Scholar] [CrossRef]
  195. Wood, R.; Arnason, T.; DeCoste, R.; Gaston, D.; Carter, M.D.; Rayson, D. Complete Response of a Colonic High-Grade Neuroendocrine Carcinoma to Platinum-Based Therapy: Insights from Comprehensive Genomic Profiling. Am. J. Clin. Pathol. 2021, 156, S143. [Google Scholar] [CrossRef]
  196. Furukawa, T.; Ozaka, M.; Takamatsu, M.; Okamoto, T.; Ishitsuka, T.; Yamada, M.; Nakagawa, H.; Mie, T.; Takeda, T.; Kasuga, A.; et al. MO69-3 Pathological Predictors of the Response to Platinum-Based Chemotherapy in Pancreatobiliary Neuroendocrine Carcinoma. Ann. Oncol. 2023, 34, S1438. [Google Scholar] [CrossRef]
  197. Wang, X.; Martindale, J.L.; Holbrook, N.J. Requirement for ERK Activation in Cisplatin-Induced Apoptosis. J. Biol. Chem. 2000, 275, 39435–39443. [Google Scholar] [CrossRef] [PubMed]
  198. Sanchez-Prieto, R.; Rojas, J.M.; Taya, Y.; Gutkind, J.S. A Role for the P38 Mitogen-Acitvated Protein Kinase Pathway in the Transcriptional Activation of P53 on Genotoxic Stress by Chemotherapeutic Agents. Cancer Res. 2000, 60, 2464–2472. [Google Scholar]
  199. Jones, E.V.; Dickman, M.J.; Whitmarsh, A.J. Regulation of P73-Mediated Apoptosis by c-Jun N-Terminal Kinase. Biochem. J. 2007, 405, 617–623. [Google Scholar] [CrossRef]
  200. Kim, K.W.; Krajewski, K.M.; Nishino, M.; Jagannathan, J.P.; Shinagare, A.B.; Tirumani, S.H.; Ramaiya, N.H. Update on the Management of Gastroenteropancreatic Neuroendocrine Tumors with Emphasis on the Role of Imaging. Am. J. Roentgenol. 2013, 201, 811–824. [Google Scholar] [CrossRef]
  201. Fazio, N.; Abdel-Rahman, O.; Spada, F.; Galdy, S.; De Dosso, S.; Capdevila, J.; Scarpa, A. RAF Signaling in Neuroendocrine Neoplasms: From Bench to Bedside. Cancer Treat. Rev. 2014, 40, 974–979. [Google Scholar] [CrossRef]
  202. Ning, L.; Chen, H.; Kunnimalaiyaan, M. Focal Adhesion Kinase, a Downstream Mediator of Raf-1 Signaling, Suppresses Cellular Adhesion, Migration, and Neuroendocrine Markers in BON Carcinoid Cells. Mol. Cancer Res. 2010, 8, 775–782. [Google Scholar] [CrossRef]
  203. Stefàno, E.; Muscella, A.; Benedetti, M.; De Castro, F.; Fanizzi, F.P.; Marsigliante, S. Antitumor and Antimigration Effects of a New Pt Compound on Neuroblastoma Cells. Biochem. Pharmacol. 2022, 202, 115124. [Google Scholar] [CrossRef]
  204. Inoue, Y.; Lockwood, W. MA22.02 Activation of MAPK Suppresses Neuroendocrine Transcription Factors and Causes Transdifferentiation of Small Cell Lung Cancer. J. Thorac. Oncol. 2018, 13, S433–S434. [Google Scholar] [CrossRef]
  205. Huang, L.; Feng, Y.; Xie, T.; Zhu, H.; Tang, L.; Shi, Y. Incidence, Survival Comparison, and Novel Prognostic Evaluation Approaches for Stage iii-iv Pulmonary Large Cell Neuroendocrine Carcinoma and Small Cell Lung Cancer. BMC Cancer 2023, 23, 312. [Google Scholar] [CrossRef]
  206. Caiola, E.; Salles, D.; Frapolli, R.; Lupi, M.; Rotella, G.; Ronchi, A.; Garassino, M.C.; Mattschas, N.; Colavecchio, S.; Broggini, M.; et al. Base Excision Repair-Mediated Resistance to Cisplatin in KRAS(G12C) Mutant NSCLC Cells. Oncotarget 2015, 6, 30072–30087. [Google Scholar] [CrossRef]
  207. 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. 2019, 42, 261–273. [Google Scholar] [CrossRef]
  208. Carter, Y.; Jaskula-Sztul, R.; Chen, H.; Mazeh, H. Signaling Pathways as Specific Pharmacologic Targets for Neuroendocrine Tumor Therapy: RET, PI3K, MEK, Growth Factors, and Notch. Neuroendocrinology 2013, 97, 57–66. [Google Scholar] [CrossRef]
  209. Kunnimalaiyaan, M.; Chen, H. Tumor Suppressor Role of Notch-1 Signaling in Neuroendocrine Tumors. Oncologist 2007, 12, 535–542. [Google Scholar] [CrossRef]
  210. Meder, L.; König, K.; Ozretić, L.; Schultheis, A.M.; Ueckeroth, F.; Ade, C.P.; Albus, K.; Boehm, D.; Rommerscheidt-Fuss, U.; Florin, A.; et al. NOTCH, ASCL1, P53 and RB Alterations Define an Alternative Pathway Driving Neuroendocrine and Small Cell Lung Carcinomas. Int. J. Cancer 2016, 138, 927–938. [Google Scholar] [CrossRef]
  211. Borges, M.; Linnoila, R.I.; Van De Velde, H.J.K.; Chen, H.; Nelkin, B.D.; Mabry, M.; Baylin, S.B.; Ball, D.W. An Achaete-Scute Homologue Essential for Neuroendocrine Differentiation in the Lung. Nature 1997, 386, 852–855. [Google Scholar] [CrossRef] [PubMed]
  212. Linnoila, R.I.; Zhao, B.; DeMayo, J.L.; Nelkin, B.D.; Baylin, S.B.; DeMayo, F.J.; Ball, D.W. Constitutive Achaete-Scute Homologue-1 Promotes Airway Dysplasia and Lung Neuroendocrine Tumors in Transgenic Mice. Cancer Res. 2000, 60, 4005–4009. [Google Scholar] [PubMed]
  213. Demelash, A.; Rudrabhatla, P.; Pant, H.C.; Wang, X.; Amin, N.D.; McWhite, C.D.; Naizhen, X.; Linnoila, R.I. Achaete-Scute Homologue-1 (ASH1) Stimulates Migration of Lung Cancer Cells through Cdk5/P35 Pathway. Mol. Biol. Cell 2012, 23, 2856–2866. [Google Scholar] [CrossRef] [PubMed]
  214. Crabtree, J.S.; Singleton, C.S.; Miele, L. Notch Signaling in Neuroendocrine Tumors. Front. Oncol. 2016, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  215. Ito, T. Intratumoral Heterogeneity of Notch1 Expression in Small Cell Lung Cancer. J. Thorac. Dis. 2018, 10, 1272–1275. [Google Scholar] [CrossRef] [PubMed]
  216. Lim, J.S.; Ibaseta, A.; Fischer, M.M.; Cancilla, B.; O’Young, G.; Cristea, S.; Luca, V.C.; Yang, D.; Jahchan, N.S.; Hamard, C.; et al. Intratumoural Heterogeneity Generated by Notch Signalling Promotes Small-Cell Lung Cancer. Nature 2017, 545, 360–364. [Google Scholar] [CrossRef] [PubMed]
  217. Lawson, M.H.; Cummings, N.M.; Rassl, D.M.; Russell, R.; Brenton, J.D.; Rintoul, R.C.; Murphy, G. Two Novel Determinants of Etoposide Resistance in Small Cell Lung Cancer. Cancer Res. 2011, 71, 4877–4887. [Google Scholar] [CrossRef]
  218. Lakiza, O.; Lutze, J.; Vogle, A.; Williams, J.; Abukdheir, A.; Miller, P.; Liao, C.-Y.; Pitroda, S.P.; Martinez, C.; Olivas, A.; et al. Loss of MEN1 Function Impairs DNA Repair Capability of Pancreatic Neuroendocrine Tumors. Endocr.-Relat. Cancer 2022, 29, 225–239. [Google Scholar] [CrossRef]
  219. Briest, F.; Grass, I.; Sedding, D.; Möbs, M.; Christen, F.; Benecke, J.; Fuchs, K.; Mende, S.; Kaemmerer, D.; Sänger, J.; et al. Mechanisms of Targeting the MDM2-P53-FOXM1 Axis in Well-Differentiated Intestinal Neuroendocrine Tumors. Neuroendocrinology 2018, 107, 1–23. [Google Scholar] [CrossRef]
  220. Phatak, V.; Von Grabowiecki, Y.; Janus, J.; Officer, L.; Behan, C.; Aschauer, L.; Pinon, L.; Mackay, H.; Zanivan, S.; Norman, J.C.; et al. Mutant P53 Promotes RCP-Dependent Chemoresistance Coinciding with Increased Delivery of P-Glycoprotein to the Plasma Membrane. Cell Death Dis. 2021, 12, 207. [Google Scholar] [CrossRef]
  221. Briest, F.; Grabowski, P. The P53 Network as Therapeutic Target in Gastroenteropancreatic Neuroendocrine Neoplasms. Cancer Treat. Rev. 2015, 41, 423–430. [Google Scholar] [CrossRef] [PubMed]
  222. Pellot Ortiz, K.I.; Rechberger, J.S.; Nonnenbroich, L.F.; Daniels, D.J.; Sarkaria, J.N. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023, 11, 1879. [Google Scholar] [CrossRef] [PubMed]
  223. Ogawara, Y.; Kishishita, S.; Obata, T.; Isazawa, Y.; Suzuki, T.; Tanaka, K.; Masuyama, N.; Gotoh, Y. Akt Enhances Mdm2-Mediated Ubiquitination and Degradation of P53. J. Biol. Chem. 2002, 277, 21843–21850. [Google Scholar] [CrossRef] [PubMed]
  224. Brazina, J.; Svadlenka, J.; Macurek, L.; Andera, L.; Hodny, Z.; Bartek, J.; Hanzlikova, H. DNA Damage-Induced Regulatory Interplay between DAXX, P53, ATM Kinase and Wip1 Phosphatase. Cell Cycle 2015, 14, 375–387. [Google Scholar] [CrossRef] [PubMed]
  225. Švajdler, M.; Mezencev, R.; Ondič, O.; Šašková, B.; Mukenšnábl, P.; Michal, M. P16 Is a Useful Supplemental Diagnostic Marker of Pulmonary Small Cell Carcinoma in Small Biopsies and Cytology Specimens. Ann. Diagn. Pathol. 2018, 33, 23–29. [Google Scholar] [CrossRef] [PubMed]
  226. Chedgy, E.C.P.; Annala, M.; Beja, K.; Warner, E.W.; Gleave, M.E.; Chi, K.N.; Wyatt, A.W. Moving Toward Personalized Care: Liquid Biopsy Predicts Response to Cisplatin in an Unusual Case of BRCA2-Null Neuroendocrine Prostate Cancer. Clin. Genitourin. Cancer 2016, 14, e233–e236. [Google Scholar] [CrossRef] [PubMed]
  227. Watanabe, K.; Kosaka, T.; Aimono, E.; Hongo, H.; Mikami, S.; Nishihara, H.; Oya, M. Japanese Case of Enzalutamide-Resistant Prostate Cancer Harboring a SPOP Mutation with Scattered Allelic Imbalance: Response to Platinum-Based Therapy. Clin. Genitourin. Cancer 2019, 17, e897–e902. [Google Scholar] [CrossRef]
  228. Mani, R.-S. The Emerging Role of Speckle-Type POZ Protein (SPOP) in Cancer Development. Drug Discov. Today 2014, 19, 1498–1502. [Google Scholar] [CrossRef] [PubMed]
  229. Song, M.S.; Salmena, L.; Carracedo, A.; Egia, A.; Lo-Coco, F.; Teruya-Feldstein, J.; Pandolfi, P.P. The Deubiquitinylation and Localization of PTEN Are Regulated by a HAUSP–PML Network. Nature 2008, 455, 813–817. [Google Scholar] [CrossRef]
  230. Kim, J.T.; Li, J.; Jang, E.R.; Gulhati, P.; Rychahou, P.G.; Napier, D.L.; Wang, C.; Weiss, H.L.; Lee, E.Y.; Anthony, L.; et al. Deregulation of Wnt/β-Catenin Signaling through Genetic or Epigenetic Alterations in Human Neuroendocrine Tumors. Carcinogenesis 2013, 34, 953–961. [Google Scholar] [CrossRef]
  231. Taghavi, S.F.; Ghorbani, M.; Panahi, M.; Nazem, S.; Karimi, M.; Salimi, V.; Tavakoli-Yaraki, M. Differential Expression Levels of β-Catenin Are Associated with Invasive Behavior of Both Functional and Non-Functional Pituitary Neuroendocrine Tumor (PitNET). Mol. Biol. Rep. 2023, 50, 6425–6434. [Google Scholar] [CrossRef] [PubMed]
  232. Atasoy, P.; Bozdoğan, Ö.; Öztürk, S.; Ensari, A. BCL2 Expression and Its Correlation with Neuroendocrine Differentiation in Colon Carcinomas. Tumori 2004, 90, 233–238. [Google Scholar] [CrossRef]
  233. Gumulec, J.; Balvan, J.; Sztalmachova, M.; Raudenska, M.; Dvorakova, V.; Knopfova, L.; Polanska, H.; Hudcova, K.; Ruttkay-Nedecky, B.; Babula, P.; et al. Cisplatin-Resistant Prostate Cancer Model: Differences in Antioxidant System, Apoptosis and Cell Cycle. Int. J. Oncol. 2014, 44, 923–933. [Google Scholar] [CrossRef] [PubMed]
  234. De M Rêgo, J.F.; Salles Scortegagna De Medeiros, R.; Braghiroli, M.I.; Galvão, B.; Evangelista Bezerra Neto, J.; Ramella Munhoz, R.; Guerra, J.; Nonogaki, S.; Kimura, L.; Pfiffer, T.E.; et al. Expression of ERCC1, Bcl-2, Lin28a, and Ki-67 as Biomarkers of Response to First-Line Platinum-Based Chemotherapy in Patients with High-Grade Extrapulmonary Neuroendocrine Carcinomas or Small Cell Lung Cancer. Ecancermedicalscience 2017, 11, 767. [Google Scholar] [CrossRef] [PubMed]
  235. Sartorius, U.A.; Krammer, P.H. Upregulation of Bcl-2 Is Involved in the Mediation of Chemotherapy Resistance in Human Small Cell Lung Cancer Cell Lines. Int. J. Cancer 2002, 97, 584–592. [Google Scholar] [CrossRef] [PubMed]
  236. Ohmoto, A.; Rokutan, H.; Yachida, S. Pancreatic Neuroendocrine Neoplasms: Basic Biology, Current Treatment Strategies and Prospects for the Future. Int. J. Mol. Sci. 2017, 18, 143. [Google Scholar] [CrossRef] [PubMed]
  237. Benitez, J.A.; Ma, J.; D’Antonio, M.; Boyer, A.; Camargo, M.F.; Zanca, C.; Kelly, S.; Khodadadi-Jamayran, A.; Jameson, N.M.; Andersen, M.; et al. PTEN Regulates Glioblastoma Oncogenesis through Chromatin-Associated Complexes of DAXX and Histone H3.3. Nat. Commun. 2017, 8, 15223. [Google Scholar] [CrossRef] [PubMed]
  238. Maharjan, C.; Ear, P.; Tran, C.; Howe, J.; Chandrasekharan, C.; Quelle, D. Pancreatic Neuroendocrine Tumors: Molecular Mechanisms and Therapeutic Targets. Cancers 2021, 13, 5117. [Google Scholar] [CrossRef] [PubMed]
  239. Pipinikas, C.P.; Berner, A.M.; Sposito, T.; Thirlwell, C. The Evolving (Epi)Genetic Landscape of Pancreatic Neuroendocrine Tumours. Endocr.-Relat. Cancer 2019, 26, R519–R544. [Google Scholar] [CrossRef]
  240. Cherif, C.; Nguyen, D.T.; Paris, C.; Le, T.K.; Sefiane, T.; Carbuccia, N.; Finetti, P.; Chaffanet, M.; Kaoutari, A.E.; Vernerey, J.; et al. Menin Inhibition Suppresses Castration-Resistant Prostate Cancer and Enhances Chemosensitivity. Oncogene 2022, 41, 125–137. [Google Scholar] [CrossRef]
  241. Baas, R.; Sijm, A.; Van Teeffelen, H.A.A.M.; Van Es, R.; Vos, H.R.; Marc Timmers, H.T. Quantitative Proteomics of the SMAD (Suppressor of Mothers against Decapentaplegic) Transcription Factor Family Identifies Importin 5 as a Bone Morphogenic Protein Receptor SMAD-Specific Importin. J. Biol. Chem. 2016, 291, 24121–24132. [Google Scholar] [CrossRef] [PubMed]
  242. Blank, U.; Karlsson, S. The Role of Smad Signaling in Hematopoiesis and Translational Hematology. Leukemia 2011, 25, 1379–1388. [Google Scholar] [CrossRef] [PubMed]
  243. Murai, F.; Koinuma, D.; Shinozaki-Ushiku, A.; Fukayama, M.; Miyaozono, K.; Ehata, S. EZH2 Promotes Progression of Small Cell Lung Cancer by Suppressing the TGF-β-Smad-ASCL1 Pathway. Cell Discov. 2015, 1, 15026. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Classification and treatment options for neuroendocrine neoplasms (NENs). Well-differentiated neuroendocrine tumors (NETs) are categorized as G1, G2 and G3, while poorly differentiated neuroendocrine carcinomas (NECs) are classified as G3. Resectable tumors are best treated with surgery, while systemic therapeutic options such as somatostatin analogs (SSAs, e.g., lanreotide), mTOR inhibitors (e.g., everolimus), tyrosine kinase inhibitors (TKIs, e.g., sunitinib, vandetanib, cabozantinib), immunotherapy (e.g., avelumab, pembrolizumab), chemotherapy (e.g., streptozotocin, etoposide with cisplatin/carboplatin, belzutifan), and peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE (177Lutetium-[DOTA(0),Tyr(3)]octreotate) are available for inoperable or metastatic disease.
Figure 1. Classification and treatment options for neuroendocrine neoplasms (NENs). Well-differentiated neuroendocrine tumors (NETs) are categorized as G1, G2 and G3, while poorly differentiated neuroendocrine carcinomas (NECs) are classified as G3. Resectable tumors are best treated with surgery, while systemic therapeutic options such as somatostatin analogs (SSAs, e.g., lanreotide), mTOR inhibitors (e.g., everolimus), tyrosine kinase inhibitors (TKIs, e.g., sunitinib, vandetanib, cabozantinib), immunotherapy (e.g., avelumab, pembrolizumab), chemotherapy (e.g., streptozotocin, etoposide with cisplatin/carboplatin, belzutifan), and peptide receptor radionuclide therapy (PRRT) with 177Lu-DOTATATE (177Lutetium-[DOTA(0),Tyr(3)]octreotate) are available for inoperable or metastatic disease.
Ijms 25 08568 g001
Figure 2. Chemical structures of cisplatin, carboplatin and etoposide. Cisplatin or carboplatin (a), in combination with etoposide (b), are the cornerstone of chemotherapy treatment for various types of cancers, including poorly differentiated or undifferentiated, high-grade neuroendocrine tumors (NETs), small-cell lung cancer (SCLC), and large-cell neuroendocrine carcinoma (LCNEC).
Figure 2. Chemical structures of cisplatin, carboplatin and etoposide. Cisplatin or carboplatin (a), in combination with etoposide (b), are the cornerstone of chemotherapy treatment for various types of cancers, including poorly differentiated or undifferentiated, high-grade neuroendocrine tumors (NETs), small-cell lung cancer (SCLC), and large-cell neuroendocrine carcinoma (LCNEC).
Ijms 25 08568 g002
Figure 3. General accepted mechanism of action of cisplatin/carboplatin etoposide combined therapy essentially showing the platinum drugs contribution. Cisplatin and carboplatin enter cells through passive diffusion and various transport proteins, including copper transporters (CTR1–2), organic cation transporters (OCT1–3), and LRRC8 volume-regulated anion channels (VRACs). Some of these transporters are associated with the export of platinum drugs and drug resistance, such as P-type copper-transporting ATPases (ATP7A and ATP7B), multidrug extrusion transporters (MATE 1–3), and ATP-binding cassette (ABC) transporters (MRP1–2). Once inside the cell, cisplatin and carboplatin undergo hydrolysis of chloro-ligand(s) and 1,1-cyclobutanedicarboxylate, respectively, resulting in a positively charged form. This allows the platinum compounds to interact with nucleophilic molecules within the cell, including DNA, RNA, and proteins, leading to the formation of platinum adducts. The formation of DNA adducts inhibits the cell cycle and impairs DNA repair, ultimately causing DNA damage and p53 activation, which induces apoptosis. Additionally, cisplatin and carboplatin induce intrinsic apoptosis by increasing mitochondrial ROS generation and activating pro-apoptotic proteins, such as Bax, which promotes the release of cytochrome C (Cyt-C) and the subsequent activation of caspases. Etoposide is a topoisomerase II inhibitor, which is considered a major anticancer mechanism of this drug. The combined actions of cisplatin or carboplatin and etoposide enhance DNA damage and promote cancer cell death.
Figure 3. General accepted mechanism of action of cisplatin/carboplatin etoposide combined therapy essentially showing the platinum drugs contribution. Cisplatin and carboplatin enter cells through passive diffusion and various transport proteins, including copper transporters (CTR1–2), organic cation transporters (OCT1–3), and LRRC8 volume-regulated anion channels (VRACs). Some of these transporters are associated with the export of platinum drugs and drug resistance, such as P-type copper-transporting ATPases (ATP7A and ATP7B), multidrug extrusion transporters (MATE 1–3), and ATP-binding cassette (ABC) transporters (MRP1–2). Once inside the cell, cisplatin and carboplatin undergo hydrolysis of chloro-ligand(s) and 1,1-cyclobutanedicarboxylate, respectively, resulting in a positively charged form. This allows the platinum compounds to interact with nucleophilic molecules within the cell, including DNA, RNA, and proteins, leading to the formation of platinum adducts. The formation of DNA adducts inhibits the cell cycle and impairs DNA repair, ultimately causing DNA damage and p53 activation, which induces apoptosis. Additionally, cisplatin and carboplatin induce intrinsic apoptosis by increasing mitochondrial ROS generation and activating pro-apoptotic proteins, such as Bax, which promotes the release of cytochrome C (Cyt-C) and the subsequent activation of caspases. Etoposide is a topoisomerase II inhibitor, which is considered a major anticancer mechanism of this drug. The combined actions of cisplatin or carboplatin and etoposide enhance DNA damage and promote cancer cell death.
Ijms 25 08568 g003
Figure 4. Signal transduction pathways influencing the efficacy of platinum-based chemotherapy in neuroendocrine neoplasms (NENs). Genetic alterations or abnormal expression of pathway members, indicated in green and red respectively, have been linked to increased or decreased sensitivity to platinum-based therapy. Growth factors (EGF, FGF, IGF, TGF-α, etc.) can bind to tyrosine kinase receptors (RTKs) and activate the (a) PTEN/PI3K/Akt and (b) RAS/RAF/MAPK pathways, resulting in the transcription of genes associated with cell proliferation, invasion, and metastasis. Mutations in (a) PTEN and (b) KRAS have been associated with heightened cytotoxic effects following platinum drug administration, while (b) BRAF mutations have been linked to limited response to chemotherapy. TGF-β can interact with TGF-β receptors (TGF-βR1,2) and activate the (a) PTEN/PI3K/Akt, (b) MAPK, and (c) SMAD pathways. In NE cells, the disruption of TGF-β signaling leads to an increased expression of (c) ASCL1, which in turn protects cancer cells from apoptosis. Additionally, (c) Notch signaling can activate ASCL1 through SMAD-mediated activation. The low expression of both Notch1 and DLL3 has been associated with better prognosis and increased sensitivity to platinum chemotherapy. Mutation in DNA repair-related genes can also impact response to platinum drugs. (d) The role of p53 in chemotherapy response is still unclear, with alterations in this gene being linked to both negative and neutral effects. However, the overexpression of the p53-negative regulator MDM2 in NENs has been shown to improve response to platinum-based therapy. Mutations of Rb and BRCA2 have also been associated with improved response to cisplatin chemotherapy. (a–c) Menin is implicated in the regulation of several of the aforementioned pathways.
Figure 4. Signal transduction pathways influencing the efficacy of platinum-based chemotherapy in neuroendocrine neoplasms (NENs). Genetic alterations or abnormal expression of pathway members, indicated in green and red respectively, have been linked to increased or decreased sensitivity to platinum-based therapy. Growth factors (EGF, FGF, IGF, TGF-α, etc.) can bind to tyrosine kinase receptors (RTKs) and activate the (a) PTEN/PI3K/Akt and (b) RAS/RAF/MAPK pathways, resulting in the transcription of genes associated with cell proliferation, invasion, and metastasis. Mutations in (a) PTEN and (b) KRAS have been associated with heightened cytotoxic effects following platinum drug administration, while (b) BRAF mutations have been linked to limited response to chemotherapy. TGF-β can interact with TGF-β receptors (TGF-βR1,2) and activate the (a) PTEN/PI3K/Akt, (b) MAPK, and (c) SMAD pathways. In NE cells, the disruption of TGF-β signaling leads to an increased expression of (c) ASCL1, which in turn protects cancer cells from apoptosis. Additionally, (c) Notch signaling can activate ASCL1 through SMAD-mediated activation. The low expression of both Notch1 and DLL3 has been associated with better prognosis and increased sensitivity to platinum chemotherapy. Mutation in DNA repair-related genes can also impact response to platinum drugs. (d) The role of p53 in chemotherapy response is still unclear, with alterations in this gene being linked to both negative and neutral effects. However, the overexpression of the p53-negative regulator MDM2 in NENs has been shown to improve response to platinum-based therapy. Mutations of Rb and BRCA2 have also been associated with improved response to cisplatin chemotherapy. (a–c) Menin is implicated in the regulation of several of the aforementioned pathways.
Ijms 25 08568 g004
Table 1. Genomic alterations and aberrant regulations impacting response to platinum-based chemotherapy in neuroendocrine neoplasms (NENs). The utilization of IHC (immunohistochemistry) staining, NGS (next-generation sequencing), PCR (polymerase chain reaction), CGP (comprehensive genomic profiling) techniques for the identification of therapeutic targets and deregulated pathways with positive (green) or negative (red) influence on chemotherapy response. Yellow represents members with uncorrelated effects on chemotherapy.
Table 1. Genomic alterations and aberrant regulations impacting response to platinum-based chemotherapy in neuroendocrine neoplasms (NENs). The utilization of IHC (immunohistochemistry) staining, NGS (next-generation sequencing), PCR (polymerase chain reaction), CGP (comprehensive genomic profiling) techniques for the identification of therapeutic targets and deregulated pathways with positive (green) or negative (red) influence on chemotherapy response. Yellow represents members with uncorrelated effects on chemotherapy.
Altered Gene/
Biomarker
NE Tumor
Type
Type of AnalysesObservationsReferences
MAPK Pathway
KRASG3 P-NET
(n = 21)
P-NEC (n = 31 SCNEC; n = 18 LCNEC)
PCR
IHC
KRAS mutations are not detected in NET-G3 (0%), while NEC-G3 harbors KRAS mutations in 48.7% of cases. There are no significant differences between SCNEC and LCNEC in the prevalence of KRAS mutations. KRAS mutations are associated with a higher response to platinum-based chemotherapy compared to those without mutations (mutated KRAS, 77% vs. wild-type, 23%).[182]
G3 P-NET
(n = 21)
P-NEC
(n = 18 LCNEC; n = 31 SCNEC)
IHC
Real-Time PCR
KRAS is mutated in 48.7% of G3 P-NEC. Patients with a KRAS mutation exhibit a better response to first-line platinum-based therapy compared to those with wild-type KRAS but tend to have shorter overall survival rates.[183]
G3 NET (n = 6)
NEC (n = 77)
Real-Time PCR
NGS
KRAS mutations do not affect treatment effectiveness or survival rates following initial chemotherapy.[184]
BRAFG3 NET (n = 6)
NEC (n = 77)
Real-Time PCR
NGS
A higher frequency of BRAF mutations is found in colon NEC and predicts failure to first-line treatment with cisplatin/carboplatin and etoposide.[184]
PTEN/PI3K/Akt/mTOR Pathway
PTENprostate NEC
(n = 1)
NGS
IHC
Somatic mutations in PTEN (and BRCA2) were identified in the tumor tissue. The tumor cells exhibited decreased staining for PTEN, indicating a loss of protein expression, which is also associated with a significant response to platinum therapy.[177]
Notch/ASCL1 pathway
Notch1SCLC (n = 46)IHCHes1, ASCL1, and DLL3 protein expression levels are not associated with sensitivity to platinum chemotherapy or prognosis. However, SCLC with low Notch-1 expression has a better survival rate.[131]
DLL3LCNEC (n = 70)IHCDLL3 is a predictive marker for sensitivity to platinum-based adjuvant chemotherapy in LCNEC. Patients with DLL3-negative tumors who receive chemotherapy show significantly higher overall survival and recurrence-free survival rates.[185]
Members of pathways involved in DNA repair
MDM2prostate NEC
(n = 1)
NGSPlatinum-based chemotherapy was found to be effective in a patient with pancreatic neuroendocrine carcinoma (NEC) exhibiting an aggressive course and MDM2 amplification.[186]
p53NSCLC-NE
(n = 157)
IHCThere is no statistically significant correlation between the p53 marker and response to chemotherapy. However, patients with an increased expression of p53 are more likely to experience progressive disease after undergoing chemotherapy.[132]
G3 NET (n = 10)
LCNEC (n = 31)
SCNEC (n = 48)
IHCThere is no statistically significant correlation between the p53 marker and response to chemotherapy. However, patients with an increased expression of p53 are more likely to experience progressive disease after undergoing chemotherapy.[132]
ES-SCLC
(n = 75)
NGSPatients with mutant TP53 had a better PFS than those with wild-type TP53.[187]
P-NET (n = 50)
P-NEC (n = 29)
IHCAbnormal p53 expression is not associated with response to platinum-based therapy.[20]
Prostate NEC
(n = 1)
NGS
IHC
The TP53 p.P72R variant is correlated with higher platinum sensitivity and longer survival of patient with aggressive prostate cancer.[188]
G3 GEP-NET
(n = 41)
GEP-NEC
(n = 188)
NGSTP53 mutation predicts an inferior response rate to cisplatin/carboplatin for NEC but does not correlate with overall survival (except for small-cell NEC).[189]
RbSCLC (n = 50)Whole/Targeted Genome Sequencing
IHC
Western Blotting
The RB1 mutation status had the most significant impact of any gene. SCLC patients with wild-type RB1 demonstrated a significantly lower response to chemotherapy compared to patients with mutant RB1.[190]
G3 P-NET (n = 21)
P-NEC (n = 31 SCNEC; n = 18 LCNEC)
PCR
IHC
The loss of Rb expression was not observed in NET-G3 (0%), while NEC-G3 showed a loss of expression in 54.5% of cases. There were no significant differences in the prevalence of abnormal Rb expression between SCNEC and LCNEC. The loss of Rb in NECs was associated with a significantly higher response rate to platinum-based chemotherapy compared to those without (80% vs. 24% with normal Rb expression).[182]
G3 P-NET
(n = 21)
P-NEC
(n = 18 LCNEC; n = 31 SCNEC)
IHC
Real-Time PCR
The rate of Rb loss in G3 P-NEC is 54.5% and is associated with a higher response rate to first-line platinum-based regimens compared to those without Rb loss. However, patients with Rb loss tended to have shorter overall survival rates than those without Rb loss.[183]
prostate NEC
(n = 1)
NGSA patient with heterozygosity loss in the RB1 gene displayed an aggressive course and responded favorably to chemotherapy containing platinum.[186]
G3 NET (n = 10)
LCNEC (n = 31)
SCNEC
(n = 48)
IHCPatients with G3 neuroendocrine neoplasms (NENs) exhibit varying responses to treatment with etoposide and platinum. However, the objective response rate was notably higher in NENs lacking the retinoblastoma (Rb) gene (63% vs. 42%).[191]
BRCAprostate NEC with metastatic lung nodule and brain metastases (n = 1)NGSCombined platinum and etoposide chemotherapy yields partial and complete remissions of brain and lung metastases, respectively, in a patient with a somatic and germline BRCA2 mutation.[192]
prostate NEC
(n = 1)
NGSA patient with a complete copy number loss of BRCA2 and ATM in prostate NEC (but not in his original adenocarcinoma) exhibited a complete response to carboplatin plus etoposide chemotherapy.[193]
prostate NEC
(n = 1)
NGSA patient with a BRCA2 mutation (along with a PTEN mutation) displays an aggressive disease progression and showed a positive response to chemotherapy containing platinum.[186]
prostate SCNEC
(n = 1)
PCRThe patient with a germline BRCA2 mutation achieved a complete response to platinum-based chemotherapy but experienced a limited duration of remission when treated with olaparib (a PARP inhibitor) as maintenance therapy.[194]
colon LCNEC (n = 1)CGPTreatment with platinum-based therapy leads to a full radiographic remission of the metastases, with no indication of recurrence after 6.5 years. The response to the therapy is probably attributed to the loss of BRCA1 and/or BAP1 function.[195]
prostate NEC
(n = 1)
NGC
IHC
BRCA2 is mutated in tumors but not in normal tissue. BRCA2 somatic mutations are associated with a strong response to platinum therapy.[177]
Other Markers
β-cateninpancreatobiliary NEC (n = 30)IHCHigher levels of β-catenin are a predictive factor for response to platinum-based chemotherapy.[196]
p16G3 NET (n = 10)
LCNEC (n = 31)
SCNEC (n = 48)
IHCThe objective response rate is significantly higher in NEN with high p16 levels (66% vs. 35%).[191]
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

Stefàno, E.; De Castro, F.; Ciccarese, A.; Muscella, A.; Marsigliante, S.; Benedetti, M.; Fanizzi, F.P. An Overview of Altered Pathways Associated with Sensitivity to Platinum-Based Chemotherapy in Neuroendocrine Tumors: Strengths and Prospects. Int. J. Mol. Sci. 2024, 25, 8568. https://doi.org/10.3390/ijms25168568

AMA Style

Stefàno E, De Castro F, Ciccarese A, Muscella A, Marsigliante S, Benedetti M, Fanizzi FP. An Overview of Altered Pathways Associated with Sensitivity to Platinum-Based Chemotherapy in Neuroendocrine Tumors: Strengths and Prospects. International Journal of Molecular Sciences. 2024; 25(16):8568. https://doi.org/10.3390/ijms25168568

Chicago/Turabian Style

Stefàno, Erika, Federica De Castro, Antonella Ciccarese, Antonella Muscella, Santo Marsigliante, Michele Benedetti, and Francesco Paolo Fanizzi. 2024. "An Overview of Altered Pathways Associated with Sensitivity to Platinum-Based Chemotherapy in Neuroendocrine Tumors: Strengths and Prospects" International Journal of Molecular Sciences 25, no. 16: 8568. https://doi.org/10.3390/ijms25168568

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