Review
Vol. 117: Issue 3 - June 2025
Clinical needs and pathology’s answers in neuroendocrine neoplasms of the lung
Abstract
Lung neuroendocrine neoplasms (NENs) make up a variegated ensemble of malignancies encompassing typical carcinoid (TC) and atypical carcinoid (AC). These are low to intermediate grade neuroendocrine tumors (NETs), and large cell neuroendocrine carcinoma (LCNEC) and small cell lung carcinoma (SCLC), which are full-fledged high-grade neuroendocrine carcinomas (NECs) showing similar clinical outcomes. Through a peer interaction between oncologist and pathologist, we herein constructed a practical approach based on questioning and answering regarding 8 practical issues aimed to provide shared solutions for clinical decision-making. These issues were itemized as sequential steps guided by clinical reasoning and concerned differential diagnosis, combined subtypes, primary and metastatic tumors, small diagnostic material, predictive biomarkers, tumor staging and, lastly, standardizing terminology. This study takes advantage of the close interaction between oncologists and pathologists as a tool to better delineate the decision-making on lung NENs.
Introduction
Lung neuroendocrine neoplasms (NENs) account for about 20% of pulmonary malignancies and comprise neuroendocrine tumors (NETs) and neuroendocrine carcinomas (NECs)1, according to the current diagnostic criteria of 2021 World Health Organization (WHO) classification on thoracic tumors (Tab. I). NETs as a whole are considered relatively well-differentiated neoplasms and include low-grade typical carcinoid (TC)/G1-NET and intermediate-grade atypical carcinoid (AC)/G2-NET, whereas NECs are high-grade carcinomas composed of either small (small cell lung carcinoma, SCLC) or large cells (large cell neuroendocrine carcinoma, LCNEC) 1, 2. Such a distinction is not only clinically compelling – all the current guidelines and clinical trials always require the sharp separation of NETs and NECs 3-10- but also finds confirmation on molecular grounds, as NETs and NECs are currently perceived as distinct and separate malignancies with their own specific alterations and no appreciable pathogenetic link or continuum among them 1. Lung NENs make up a clinico-pathologic spectrum endowed by increasing biological aggressiveness where the traditional four-tier pathology approach (i.e., TC, AC, LCNEC, SCLC) conforms to a three-tier clinical outcome (i.e., low-, intermediate- and high-grade for TC, AC and NEC, respectively) and to a two-tier molecular interpretation with no pathogenetic continuum (i.e., NETs vs NECs) 1, 2, 11-15. Representative images of the above subtypes are depicted in Figure 1 A-G and Figure 2 A-E.
In this review article, a group of clinicians and pathologists experienced in lung NENs has explored 8 practical issues they considered important in the clinicopathologic handling of these patients by using a multidisciplinary approach and then summarizing their expert opinion on those topics.
Methods
An outlook of papers on the issues of clinical needs, pathology implications and molecular facts on lung NENs (not a systematic review) was carried out upon generating 8 key questions (Tab. II), which regard molecular alterations, diagnosis, prognosis, classification, treatment options and clinical facts in light of the 2021 WHO classification and the most updated international guidelines 1, 3-10. Only full-papers were considered along with smaller studies or single case reports, whenever appropriate, and the source was the English literature from PubMed/Medline until February 2025. The list of issues was reflecting the experience of oncologists and pathologists dealing with lung NENs with no intention to define diagnostic or therapeutical guidelines, but rather provide a subjective yet reasonable choice of practical issues useful in better characterizing these neoplasms. Methodologically, these 8 items were sequentially addressed, with the oncologist standpoints at first and the pathologist interpretations subsequently, with final shared comments at the end of each paragraph. As this is a review article on published literature data, no approval by Internal Review Board or Ethics Committee notification was needed.
Results
1. DISTINGUISHING AMONG HIGH-GRADE NECS AND CONVENTIONAL NSCCS/NON-NECS IN LOCAL/LOCALLY ADVANCED AND METASTATIC DISEASE
Oncologist.
Distinguishing high-grade NECs from conventional non-small cell carcinomas (NSCCs), non-NECs, is a crucial step in the therapeutic management of patients (https://www.iss.it/documents/20126/8403839/LG%20149_Polmone_agg2021) 6, 16. First of all, while surgery is the only curative intent for NSCCs/non-NECs, SCLCs or LCNECs are rarely treated with upfront surgery, inasmuch as just the uncommon very early stages (T1-2, N0/N1, M0, I-IIA stages) without evidence of mediastinal involvement are amenable of surgical resection. In contrast, treatment options for NSCCs/non-NECs include a broad range of systemic therapies 17-29 depending on the stage and the mutational patterns of disease, with neoadjuvant or adjuvant intents and may be further enhanced by the integration of local therapies 30-33. While etoposide and platinum salts remain the agents of choice for NECs as a whole, with no targeted therapies being approved for these tumors 34, other agents are preferred for conventional NSCCs/non-NECs 35. Therefore, distinguishing between NECs and conventional NSCCs/non-NECs is a crucial and essential step before exploring their mutational profile. In conventional NSCCs/non-NECs, the analysis of EGFR, ALK, KRAS, ROS1, BRAF, NTRK1/2/3, METexon14 skipping mutation, RET, ERBB2-HER2 and PD-L1 status is recommended to identify patients eligible for targeted therapies or immunotherapy. In some European countries, such as France, NGS and theranostic immunohistochemistry (including PD-L1 status) are recommended for RB1-proficient LCNECs on the basis of the Institute National du Cancer’s (INCA) guidelines (see http://www.e-cancer.fr/Patients-et-proches/Se-faire-soigner/Traitements/Therapies-ciblees-etimmunotherapie-specifique/Immunotherapie-mode-d-action) 36. Accordingly, LCNECs with retained RB1 expression are treated like other conventional NSCCs/non-NECs unless they harbor targetable fusions or mutations, while the RB1-negative ones are treated just like SCLCs. However, the prognostic role of RB-1 still remains debated 37. Additionally, while single-agent anti-PD-1 or PD-L1 therapies have shown significant effectiveness in NSCCs/non-NECs, especially in those with high PD-L1 expression, this is not the case for SCLCs, where these therapies have limited efficacy when used alone and are recommended only in combination with a platinum-etoposide regimen 38. As far as local treatments are concerned, while prophylactic cranial irradiation (PCI) is recommended for limited-stage SCLCs, no PCI is established for conventional NSCCs/non-NECs. Lastly, a distinction between conventional NSCCs/non-NECs and NECs is clinically of the upmost importance especially for patient enrollment in clinical trials. In fact, neoplasms identified as NECs or suspected to be NE due to expression of NE biomarkers are excluded from trials focusing on conventional NSCCs/non-NECs.
Pathologist.
The rule to approach lung cancer diagnosis always dictates subtyping according to three main differentiation lines, namely glandular for adenocarcinoma, keratinizing for squamous cell carcinoma and NE for NENs by combining morphology and appropriate panels of biomarkers 1, 39-41. The diagnosis of lung NENs is a stepwise process, where at first NE morphology needs to be recognized (organoid aggregates, ribbons, trabeculae, rosettes, stromal features such as high vascular density) and then subtyped according to cytologic (chromatin, nucleoli, cell size) and immunophenotypic features 1, 14. As recommended by the 2021 WHO classification 1, if a NE morphology is missing, NE biomarkers (synaptophysin, chromogranin A and INSM1) should not be performed, because the clinical relevance to NE differentiation in otherwise conventional adenocarcinomas or squamous carcinoma is limited and such an immunoprofile is not required 42. Therefore, the primacy of NE morphology over confirmatory immune markers has been reaffirmed as a guiding criterion for NEC definition by the current guidelines 1.
Shared solution.
Use NE markers as appropriate and limit the use of NSCC-NED in practice, because it is confounding with no established clinical counterpart of treatment thus far.
2. DISTINGUISHING AMONG HIGH-GRADE NECS (BIOPSY SETTING)
Oncologist.
As SCLCs and LCNECs are rarely treated by surgery, they are generally assessed through biopsy samples for the purpose of planning the therapeutic strategies 43, 44. In the metastatic setting, SCLCs and LCNECs differ in terms of systemic treatments and availability of clinical trials. There are 1248 registered studies for SCLCs compared to just five for LCNECs (see https://clinicaltrials.gov), highlighting the need for an accurate differential diagnosis. In this regard, LCNECs are no longer included in trials alongside SCLCs due to their differences in prognostic and predictive factor expression 44. Trials apart, according to the American Society of Clinical Oncology (ASCO) guidelines, platinum-etoposide regimen is deemed to be the most appropriate therapy for SCLCs and LCNECs, but the utility of NSCC regimens has been suggested in some LCNECs 36, 45-47. George et al., identifying two distinct subcategories of LCNECs according to different genomic profiles (SCLC-like or type II LCNEC and NSCC-like or type I LCNEC), suggested a better response to SCLC-regimens for SCLC-like LCNECs and a better response to NSCC-regimens for NSCC-like LCNECs 48. Conversely, Wang et al. reported no differences in terms of objective response rates, progression-free survival, and overall survival among LCNEC patients treated with SCLC- vs NSCC-regimens 49. However, when dividing LCNECs into the two molecular clusters 48, a better overall survival for patients with type II LCNEC (resembling SCLC) treated with SCLC regimens was reported as compared to other subcategories (37.7 vs. 10.5 months, p = 0.039). Since NEC patients are typically managed on the basis of small amounts of diagnostic material (e.g., cytology and biopsy samples), a reliable differential diagnosis is advocated through a strategic handling of lung tissue and appropriate immunohistochemistry adjuncts (e.g., RB1) 36 for the best clinical decision-making.
Pathologist.
Distinction between SCLC and LCNEC is accomplished by using a constellation of cytohistologic criteria (cell size and chromatin pattern) and immunohistochemistry details (Tab. I). SCLC is reliably diagnosed in resection, biopsy or cytology samples, whereas LCNEC may prove difficult to be recognized in small-sized material, where the term “NSCC with LCNEC features” is preferred, provided NE morphology and NE biomarkers are concurrently present 1. This task is demanding in smeared cytology samples 50, but the increasing use of cellblock preparations enables to readily appreciate the diagnostic criteria based on NE morphology and biomarkers 51. Separation of SCLC and LCNEC, however, may be poorly reproducible owing to the subjective interpretation of cell size and cytological features, also considering the continuum between small and large cells 52-55. As many as 10-15% of SCLC show larger cells with evident nucleoli and coarser chromatin, which are likely to correspond to the molecularly defined subtypes with low NE differentiation 56. In difficult instances, cytohistologic correlations are recommended, because cytology is considered the key tool to render an ultimate diagnosis of SCLC 57 or, alternatively, if some uncertainty should still persist, the noncommittal term high-grade NEC, not otherwise specified (NOS), is suggested by the 2021 WHO classification 1. Combined tumors are not infrequent in NECs 1, 58, 59 and they can be missed in biopsy or cytology (see below, chapter 3.1). More recently, artificial intelligence algorithms have been proposed to improve separation among SCLC and LCNEC, but these tools are not routinely used at preent 10, 60, 61. As RB1 expression is largely lost in SCLC but variably retained in LCNEC 48, the use of this simple immunostaining has been suggested in those RB1-proficient LCNEC, which could somewhat benefit from NSCC therapy protocols, whereas RB1-negative LCNEC would most benefit from drugs dedicated to SCLC 36.
Shared solution.
Try to distinguish SCLC and LCNEC as much as possible, by maximizing all available diagnostic material (e.g., by using core biopsies offering larger amounts of tissue), because such a distinction is clinically warranted for decision making.
3. DISTINGUISHING BETWEEN PURE AND COMBINED (3.1) OR PRIMARY AND METASTATIC (3.2) NECS
3.1 Between pure NECs and combined (NECs composed by neuroendocrine and non-neuroendocrine components) lung carcinomas
Oncologist.
International guidelines for cancer management underscore the relevance of distinguishing high-grade NECs of the lung, whether pure or combined, for effective therapeutic purposes (see also: https://www.iss.it/documents/20126/8403839/LG%20149_Polmone_agg2021). Firstly, combined and pure NECs present with different clinical patterns: combined NECs, especially LCNEC, mostly show peripheral locations 1, 62, 63, whereas pure NECs, particularly SCLCs, are more commonly central 64, 65, necessitating tailored locoregional approaches. Secondly, localized disease occurs in as many as 70% of combined NECs compared to 40% of pure NECs, with a mere 5% of pure NECs diagnosed in stage I-II. This disparity of clinico-pathologic presentation leads to markedly different resection rates: 45% of combined NECs vs just 3% of pure SCLCs, thus resulting in significantly variable clinical outcomes. Lastly, combined NECs have lower response rates to chemo-radiotherapy as compared to pure SCLCs, but show a greater sensitivity to NSCC regimens 36, 66. Consequently, all these differences result in 2.5 times higher survival rates for patients with combined NECs than those with pure forms of SCLCs 67.
Pathologist.
While most NECs, regardless of their subtyping, are histologically pure, up to 30%, whether SCLC or LCNEC, may combine with any other tumor type. Although a 10% threshold is required to define a combined SCLC with LCNEC due to the continuum between small and large cells and the purported greater biological aggressiveness of SCLC components, there is, however, no limitation regarding the percentage of NSCC or even sarcoma that may be recognized to classify the corresponding combined tumors 1, 58, 59, 68. Combined SCLCs are usually central lesions affecting major bronchi, whereas combined LCNECs prevail in peripheral location, thus reflecting possibly different pathogenesis mechanisms 1, 13, 59. Morphology and appropriate immunohistochemistry biomarkers are used to uncover such a type of tumors whose terminology remains a traditional one in the lung, while mixed neuroendocrine and nonneuroendocrine neoplasm (MINEN), as currently applied to the gastroenteropancreatic (GEP) tract 69, is discouraged. It is relevant to mention in the pathology report the various components of combined tumors for patient management and correct interpretation of metastatic deposits. In these instances, the finding of molecular traits common to NSCCs, such as EGFR mutation or ALK translocation, in otherwise classifiable NECs, especially if featuring SCLC, along with clinical correlations (e.g., nonsmokers), open up an avenue for de novo (in untreated patients) or transdifferentiated (in treated patients) SCLC developing from preexisting lung adenocarcinoma 70-72.
Shared solution.
Up to one-fourth of NECs are combined with NSCC components, whose recognition is clinically warranted due to different treatments and outcomes. Do not use the term MINEN in the lung. In small diagnostic material, combined NECs may be guessed by molecular profiling (e.g., EGFR mutations or ALK translocation).
3.2 Between primary and metastatic NECs
Oncologist.
In case of suspected NEC of the lung, it is important for clinicians to distinguish between a primary lung neoplasm and a metastasis from other sites. In cases of suspected metastatic lung involvement from an unknown extra-pulmonary NEC, further radiologic and/or endoscopic examinations are necessary. For instance, if a lung metastasis from a small-cell NEC of the esophagus is suspected and the radiologic imaging is not conclusive, endoscopic evaluation with biopsy becomes mandatory 73. Similarly, if a small-cell NEC from the urinary bladder is suspected, a cystoscopy with a transurethral resection of the bladder may be indicated 74, 75. In all cases of unknown high-grade NEC primary, a 18Fluorodeoxyglucose (FDG) Positron Emission Tomography (PET)/Computed Tomography (CT) scan is recommended to identify any possible site of origin, regardless of the organ. While the staging differs among extrapulmonary NECs, it is noteworthy that, even in case of metastatic disease, the systemic approach is nonetheless quite similar and typically involves a combination of platinum-based polychemotherapy with or without immunotherapy 5, 76 and, regardless of the completion of staging procedures, treatment should begin as soon as the diagnosis is confirmed. Due to rapid progression of the disease, many patients are unable to receive even first-line treatment 77. Of course, various clinical trials are tailored to the specific site of origin, as extrapulmonary NECs may have different prognostic, mutational and survival features 78-81.
3.2 Pathologist.
The identification of metastatic NECs to the lung, whether SCLC or LCNEC, in pure or combined forms, may be challenging outside the proper clinical context and the personal history of patient 82 and morphology and immunohistochemistry may fail to a large extent in this task (for example, TTF1 is consistently positive in most pulmonary and extrapulmonary NECs) 83. Intra-intertumor heterogeneity has been described in both SCLC 84 and LCNEC 48 with different transcriptomic subsets governed by master regulators of neuroendocrine differentiation. Moreover, a branched evolution with a shift to ancestral clones has been proposed in SCLCs underlying tumor relapse upon therapy 85. Such molecular characterization seems to reflect intrinsic functional subtypes rather than specific traits of the organ of origin, which cannot reliably be used for differentiating primary or metastatic NECs at the level of the individual patient’s cancer 86-91. The switch among transcriptomic subtypes, with evolution of NEhigh to NElow histotypes via MYC amplification or overexpression in experimental mouse models 92, which in turn would cause a pro-tumorigenic microenvironment of therapy-resistant, stem cell-like non-NE cells to arise with feeding/trophic effects on metastatizing NE tumor cells 93, has weaker evidence in humans 85, 94, 95. While some molecular differences exist between pulmonary and extrapulmonary NECs 88, 89, many of the agreed-upon organ-specific transcription factors successfully used to create classification algorithms for unknown-primary NETs, such as OTP, TTF1, CDX2, PAX8, WT1 or ISLET-1, largely fail in NECs, due to either illegitimate transcription or alternative functions compared to other cancer models 83, 96. For example, NKX2.1/TTF1 gene expression (a marker of type II pneumocyte and club cells commonly observed in pulmonary adenocarcinoma) 97 is documentable in as many as 80-90% of NEhigh SCLCs according to the ASCL1/TTF-1/NFIB axis activation that potentially contributes to tumor development and metastatizing behavior 98. Furthermore, NKX2.1/TTF1 gene product is also expressed in many extrapulmonary NECs, making this marker particularly unreliable in recognizing a primary or metastatic tumor 83. An intestinal origin of NECs, however, could be envisaged by the preferential NKX2-5 expression (a gene involved in heart formation and development) observed in organoid models of digestive NEC compared to ASCL1 and POU2F3 expression that are more frequent in the lung 99 or by the fifth molecular subtype of NECs (beyond the classical ASCL1, NeuroD1, POU2F3 and YAP1 subtypes according to the Rudin and Gazdar’s scheme 84) dependent on HNF4-alpha transcriptional regulator (a key gene involved in the development of the liver, kidney, and intestine), which is hallmarked by RB1WT, GEP-like signature, NEhigh phenotype and LCNEC appearance 100. Lastly, while undifferentiated NSCC may express NE markers, such as SMARCA4- 101 or SMARCB1- 102, 103 deficient undifferentiated tumors or NUTM1-rearranged carcinoma 104, as many as 10-20% conventional NECs, both primary and metastatic, driven by YAP1 or POU2F3 84, 90, 101, 105, 106, lack NE differentiation and their recognition may be demanding in a diagnostic setting 88, 107, 108. A recent study by Zhang et al. compared by NGS 99 GEP NECs from the bile duct, colorectum, duodenum, esophagus, stomach, pancreas and unknown primary with 57 lung NEC of either small and large cells. It was found that TP53/KRAS/CTNNB1 mutation and TERT/IL7R amplification prevailed in the former, whereas GRIN2A/PTPRD/RB1 mutation and NKX2.1/PIK3CA amplification prevailed in the latter 109. These data suggest that NGS analysis may assist in the appropriate clinicopathologic context in the differential diagnosis between pulmonary and extrapulmonary NECs.
Shared solution.
Clinical history and presentation are useful to identify metastatic NECs to the lung, in that most site-specific immunohistochemistry biomarkers are largely shared by pulmonary and extra-pulmonary NECs and molecular characterization may be unpracticable in real life.
4. DISTINGUISHING AMONG WELL-DIFFERENTIATED NETS
Oncologist.
After the first histologic description of pulmonary carcinoids in the 1930s 110, diagnostic criteria for TC and AC were definitively established in the 1999 WHO classification of lung tumors 111 and subsequently maintained in the 2015 112 and 20211 WHO editions. As knowledge on lung carcinoids has continued to increase in the meantime, with distinct tumor properties and epidemiologic traits being uncovered (albeit ACs are by far the least frequent one) 113, the current outlook on a two-tier clinical and prognostic stratification of carcinoid patients has been definitively accepted. Therefore, the pathological diagnosis of NETs and their distinction from NECs has become an absolute precondition for the oncologist in every diagnostic context, in virtue of major differences in clinical outcomes, functional imaging and therapeutic approaches 7, 114.
From a clinical perspective, compared to TCs, ACs are associated with a higher risk of nodal involvement, metastasis, larger size, peripheral location, local recurrence and worse prognosis 115. With the replacement of conventional functional imaging based on 111In- or 99Tc-labelled radiotracers with 68Gallium (68Ga-) DOTA-peptide imaging, in addition to 18FDG-PET/CT scan, different functional patterns have been highlighted between ACs and TCs 116. In a recent analysis by Jiang et al., TCs seem to show a greater distribution of somatostatin receptors (SSTRs), and higher 68Ga-DOTA-peptide PET/CT positivity, compared to ACs, with SUVmax 36.5 ± 21.6 vs 9 ± 5.6 (p < 0.002). In turn, ACs seem to show a higher FDG uptake and thus a greater biologic aggressiveness in comparison with TCs (SUVmax 6.0 ± 3.4 vs 3.7 ± 2.6, p = 0.05) 117. Furthermore, as many as 11-22% of lung carcinoids are associated with multiple endocrine neoplasia type 1 (MEN-1) syndrome, which shows a higher prevalence of ACs over TCs 118, 119. All these traits imply different management strategies and reflect different outcomes 120, 121.
Recognition of metastatic NETs to the lung represents another crucial issue. Since the lung may be reached by extra-pulmonary NETs 122, 123, identifying the organ of origin is essential when well-differentiated NETs are detected in the absence of a primary site (CUP, carcinoma of unknown primary). In case of suspected GEP-NETs, 68Ga-DOTA-peptide PET/CT may effectively trace them for diagnosis and staging purposes, whereas NETs arising in hollow organs may be uncovered by endoscopy. For instance, in low-grade G2 GEP-NET, a further 18FDG-PET/CT scan may be unnecessary, while in the event of ACs, this procedure is mandatory especially when 68Ga-DOTA-peptide PET/CT scan is negative 3,124,125. In the choice of systemic treatment, identifying the primary is a key event according to international guidelines and nation-wide approvals. Beyond somatostatin analogues (SSA), the approved treatments for metastatic lung NETs include everolimus and chemotherapy 125. For all the other G1-G2 GEP-NETs, approved treatments include everolimus 126, chemotherapy 127, sunitinib (only for pancreatic NET) 128, peptide receptor radionuclide therapy (PRRT) 129 and, in some countries, surufatinib 130. Lastly, during follow-up and systemic treatment, knowing the origin of NETs helps guide imaging according to the natural history of disease. For instance, ileal NETs are mostly linked with liver and peritoneal metastases (67% when considering liver alone, peritoneum alone, and both) 131 and, rarely, with bone metastases (1.2%) 132. Conversely, lung NETs, beyond metastasizing to the liver, commonly spread, either synchronously or metachronously, to the lung, bone and brain, thus requiring different imaging approaches to accurately identify metastases at these sites 133.
Pathologist.
Diagnosis of NET is carried out by the criteria outlined in Table I, namely mitotic count per 2 mm2 and necrosis assessment as essential core criteria and Ki-67 labeling index (the product of the gene MKI67 mapping to 10q26.2 chromosome) 15, 134 as a nonessential (desirable) adjunct information1 (henceforth, simply Ki-67). The resulting terminology will be TC/NET G1 and AC/NET G2 on resection and carcinoid tumor NOS and metastatic carcinoid NOS in biopsy/cytology samples of primary and secondary tumors, respectively 1, 15, 135, 136. Comparison between biopsy samples and resection specimens may be unreliable as far as mitoses and necrosis identification is concerned 137-139. Of note, however, Reuling et al. have recently observed that overall size > 4 mm2 of rigid or flexible bronchoscopy biopsies rather than their number accurately predicted AC histology, while Ki-67 (threshold around 5%) showed good concordance but not discriminative power for definitive diagnosis 140. All these considerations may nonetheless have a minor clinical impact, once a NEC diagnosis has been excluded, because in most instances TCs and ACs are resectable and the final risk attribution 1 is left to the subsequent examination of the surgical specimen (see below for the category “carcinoid tumors with elevated proliferation”).
Regarding primary vs metastatic NETs, suffice it to say that, while pan-NE do not significantly contribute to this separation, specific panels of nuclear transcription factors are helpful to unveil the pulmonary or extra-pulmonary origin of NETs. In this regard, a combination of CDX2/Islet-1/PAX8 uncovers GEP NETs, of estrogen and progesterone receptors and TRPS1 141 breast NETs/NECs, and of TTF1 and OTP the pulmonary origin of NETs. TTF1 is found especially in peripheral and often spindle-cell carcinoids in the setting of diffuse idiopathic pulmonary NE cell hyperplasia (DIPNECH) 142, while OTP is a sensitive and purportedly specific marker of lung NET origin in cytologic and surgical specimens 143 (its specificity will hold until proven otherwise). Using a 40% OTP staining threshold in primary lung NENs, Viswanathan et al. have recently observed not only that 73% of TCs and 30% of ACs were positively stained for this marker (all NECs were instead negative), but also that this marker could operate discrimination among carcinoids 143, thus with potential diagnostic applications to bronchial biopsies 139,144,145. Immunohistochemistry for GATA binding protein 3 (GATA3) gene product (a transcription factor relatively unspecific that is expressed in many tumor types) 146 and estrogen/progesterone receptors have rarely been documented in single observations of lung NETs 147,148, thereby advising extreme caution to avoid the diagnostic trap of missing an unsuspected metastasis of breast NEN to the lung (Pelosi, personal unpublished observations). Notably, GATA3 is also expressed by paragangliomas/pheochromocytomas 149, thus making it important to verify keratin expression, which is usually lacking in these tumors.
Shared solution.
The correct splitting of NETs into TCs and ACs helps better stratify patients for outcomes and treatments, but is practicable only on resection specimens. However, the noncommittal terms of carcinoid tumor/NOS and metastatic tumor NOS are nonetheless adequate to assist clinical decision making, provided that mitoses, necrosis and, hopefully, Ki-67 index are contextually specified in the pathology report. Uncovering primary or metastatic lung NETs is feasible and clinically warranted by means of immunohistochemical markers, but diagnostic traps should be carefully avoided. Different terminologies have to be adopted according to the different NET origin, if known, with metastatic carcinoid NOS being used for lung primaries and metastatic NETG1 to G3 GEP for gastroenteropancreatic derivation.
5. DISTINGUISHING BETWEEN ACS AND LCNECS: MAY THE KI-67 PROLIFERATION INDEX BE THE SOLUTION?
Oncologist.
Since carcinoid-like LCNECs and highly proliferating carcinoids are on record among lung NENs and staging, imaging and therapeutic options may be different, distinguishing between carcinoids with elevated proliferation rates and LCNEC is becoming crucial to define the best therapeutic options 1, 47. This distinction, however, becomes less critical when comparing metastatic highly proliferating lung carcinoids with LCNECs showing lower proliferative indexes. In this scenario, the patient’s performance status, tumor burden and the biologic behavior of disease guide the choice of systemic therapy more than the initial diagnosis. This is particularly true since histologic diagnosis may be based on a biopsy that does not necessarily represent the entire tumor. In this regard, possible treatments include alkylating agents or platinum-based chemotherapy for either instance 150-156. Even if systemic therapies for lung carcinoids encompass everolimus and, in some trials or countries, PRRT and surufatinib 157, in cases with high tumor burden, aggressive biologic behavior and when tumor shrinkage is needed to improve symptoms, SCLC-like regimens may be more effective, regardless of histology. In these cases, more than apodictically distinguishing between high proliferative lung carcinoid and LCNEC, it becomes most important to pay attention to the tumor proliferation index, alongside clinical information (patient’s performance status, tumor burden, medical history and functional imaging results) 138.
Pathologist.
Although Ki-67 has been extensively studied over the years across the spectrum of lung NENs in virtue of its strong prognostic power 134, 158, 159, the 2021 WHO classification admits for this marker a nonessential, desirable or non-core role with two practical uses 1, 160: one for distinguishing NETs from NECs in biopsy or cytology samples, especially when occurring severe crush artifacts 15, 134, 138, 160, 161, the other for contributing to define primary or stage IV carcinoid tumors/NETs with elevated mitotic counts (> 10 mitoses per 2 mm2) and/or Ki-67 proliferation index (≥30%) 1, 12, 47, 160, 162. These tumors are currently classified as LCNECs in the pathology report, with the comment that morphology, expression of NE markers, retinoblastoma and wild-type pattern of p53, low tumor mutation burden, recurrently mutated genes (MEN1, ARID1A, ARID1B, KDM5C), lack of RB1 and TP53 inactivation and intermediate survival rates are more consistent with carcinoids than conventional LCNECs 1, 12, 101, 162-164. The identification of this provisional tumor entity dates back to previous genomic studies in which the term LCNEC-carcinoid like was introduced 47, characterized by recurrent MEN1 mutation and borderline Ki-67 values with well differentiated NETs 12, 101, 162. This entity is homologous to NET G3 of the GEP tract 163, although additional clinicopathologic and molecular investigations are needed to establish its proper position in the classification 1. Recently, attention has been drawn to lung carcinoids that are transcriptomically similar to NECs 135, 165, 166, termed by some authors as supra-carcinoids 167. Moreover, TP53 or RB1 alterations have been described in these proliferating carcinoids 165, 168-170, thereby expanding their spectrum of biological complexity. Conversely, the diagnosis of TC and AC is unpermitted by Ki-67 in any diagnostic context because of the imperfect biological correlation with mitotic count and necrosis 1, 15, 135, 136. However, different cut-off values ranging from 1.3 to 11.7% 157, 171-178 have been reported to separate TCs from ACs with progression-free survival information, thus leading to an increasing interest even in biopsies 137-139, 144. In the metastatic setting of NETs or low proliferating LCNECs, where different therapies (e.g., SSA, temozolomide - capecitabine duet, PRRT) are an option 135, a stepwise approach based on Ki-67 and tumor differentiation could help in decision making 15, 135, 158, 179, 180. Furthermore, Ki-67 may be desirable in diagnoses of carcinoid tumors/NETs NOS or metastatic carcinoids NOS to improve clinical decision making, especially in association with other markers (OTP, retinoblastoma, CD44, SSTR2A, ASCL1, p53) 138, 139, 144, 145, 172, inasmuch as TC and AC separation is unfeasible. Of note, purported limitations of quantification are not specific to Ki-67, because even mitotic count and necrosis are culprits of interobserver inconsistency 15, 181. Phosphohistone H3 (PHH3) immunohistochemistry (a surrogate marker of mitotic figures) has been proposed to ameliorate mitotic count in terms of scoring times, mitosis detection and reproducibility of results 182, 183, although it does not improve diagnosis and provides a similar prognostic value to the current morphologic criteria 181.
Shared solution.
Ki-67 should always be included in pathology reports of lung NETs, since it helps in several ways: differential diagnosis between NETs and NECs; identification of highly proliferating carcinoids (especially in metastases); and clinical decision-making process as a whole. Defining Ki-67 index in NECs, especially SCLC, is currently of no clinical help.
6. ARE THERE OTHER TREATMENT-RELATED BIOMARKERS?
Oncologist.
Over the years, various prognostic and predictive factors have been proposed for optimal lung NEN management. In metastatic TCs and ACs, after confirmation of SSTR expression, SSA have been approved as a first-line treatment 125. From this perspective, knowledge of clinical features has perfected the use of somatostatin receptors imaging (SRI) with an advancement in imaging techniques and radiotracers. Since 2010, the best functional imaging for studying NETs was OctreoScan (In-111 pentetreotide), which however is currently considered obsolete, with up to 75% of discrepancy in lung NETs detection between OctreoScan and the newer generation SRI techniques 184. When SRI are not available or outdated, detecting SSTR distribution through alternative methods becomes crucial for indicating systemic treatments with SSA and PRRT 185.
Regarding other predictive targets, Kunz et al. have recently demonstrated the role of methylguanine methyltransferase (MGMT) assessment by promoter methylation and/or immunohistochemistry in the treatment of low-to-intermediate GEP NETs with temozolomide-based chemotherapy 127. In this study, MGMT deficiency was associated with a better response to temozolomide/capecitabine treated GEP NETs, which could become a possibility even for advanced lung carcinoid 156.
Lastly, specific gene alterations variably reported have been documented in tumor series or single case reports across the spectrum of lung NENs, which opened up an avenue for targeted treatments with some dramatic responses, such as NTRK2-3 186 and EGFR 187 mutations in LCNECs or ALK translocation in ACs and LCNECs 188. Recently, Zhang et al. described ETV6-NTRK2 translocation in an AC patient who experienced complete response with entrectinib after several systemic efforts 189. Also, identifying carcinoid patients with high risk of progression 190 who could benefit from adjuvant treatment with chemotherapy or radiotherapy on the mediastinum as recommended by ESMO European Guidelines 125 is another clinical need. Lastly, detecting ACTH expression to predict potential endocrinologic paraneoplastic syndromes can help differentiate true Cushing syndrome from cushingoid changes due to concurrent steroid therapy 191. This is particularly important for patients with brain metastases who may be heavily treated with steroids.
Pathologist.
Most pathology laboratories are endowed by a vast armamentarium of immunohistochemical and molecular predictive assays 192, which can help in clinical decision making. For instance, SSTR2 and SSTR5 immunohistochemistry may replace the functional imaging in lung TCs and ACs 193, with a 69% correlation rate with in vivo OctreoScan determination 194, 195. In particular, given the varying affinities that different peptides (DOTA-NOC, DOTA-TOC, DOTA-TATE) 124 may show for SSTR2, SSTR3, and SSTR5, the immunohistochemical confirmation of SSTR expression can assist in managing patients 195.
Regarding MGMT scoring, Vatrano et al. demonstrated with different experimental approaches not only that a decreased activity in a large series of lung carcinoids correlated with traits of aggressiveness, namely AC histology, larger size and higher T-N stage, but also that this assay provided the rationale for using alkylating agents in the same tumors 196.
Next generation sequencing or other molecular techniques applied to the spectrum of lung NENs as a whole 197 may assist the pathologist in discovering actionable or prognostic gene alterations within specific study protocols or under clinical guidance for decision-making. These include NTRK2-3 186, EGFR 187, MEN1 165, 166, 198 or RB1/TP53 199 mutations, DLL3 200-202, EZH2 203 or c-MYC 204 overexpression, ETV6-NTRK2 189 or ALK translocation 188, and microsatellite instability/mismatch repair deficiency 205, as well as tumor mutation burden assessment for IO 206 or transdifferentiation mechanism identification in de novo or treated NECs 70, 71. Tumor mutation burden by NGS analysis is low in lung carcinoids and high in lung NECs, being correlated with Wnt, Hippo or Hedgehog signal activation 207, while TP53 mutations is related with higher stromal PD-L1 expression 208. Recently, Werr et al., in a large collaborative study on pulmonary NETs 190, have suggested that high TERT mRNA expression, leading to enhanced telomerase activity, identified clinically aggressive carcinoids, either TC or AC, with fatal outcome, thereby confirming our previous observations on TERT amplification (a surrogate for mRNA overexpression), which was shared by NETs and NECs and adversely affected survival 165, 209. Of note, TERT can also be assessed by immunohistochemistry, thus potentially supporting its introduction in the clinical management of lung cancer 210. Particularly interesting and easy to uncover by means of immunohistochemistry is the small subset (6%) of SCLC patients showing proficient RB1 expression and RB1WT status, who bear variable prognosis 199 but showed immune-enriched profile with increased response to checkpoint inhibitor blockage (nivolumab and ipilimumab) in the CheckMate 032 trial 211.
Potential biomarkers for prognostic stratification in lung carcinoids (Ki-67, OTP, retinoblastoma, CD44, SSTR2A, Ki-67, ASCL1, p53) can be readily performed by immunohistochemistry even in biopsy samples, thereby helping the oncologist to widen the decision-making options 138, 139, 144, 145, 172. Lastly, characterizing lung carcinoids for ACTH expression may be clinically warranted, inasmuch as these tumors are not so rare, do not always show overt Cushing syndrome and per se do not represent a clinically aggressive subtype 212.
Shared solution.
Molecular testing is not routinely mandatory in NETs/NECs and in NSCC/non-NEC, but it is up to the clinical judgment and the patients’ demography/clinical presentation, such as the lack of smoking history or increased likelihood of driver mutations. In this regard, the opportunity of molecular testing should be taken into account in those cases, including transformed NECs and de novo NECs/NETs with driver alterations. Therefore, local feasibility of these assays should be discussed thoroughly with the pathologist, who should make them available on demand for justified clinical reasons.
7. STAGING PROCEDURE
Oncologist.
According to the NCCN guidelines on NSCCs and NECs 6, 16 and the ESMO guidelines for the management of lung carcinoids and SCLC 5, 125, the TNM staging scheme overlaps the AJCC/UICC system in use for lung NSCCs/non-NECs as refined in the 9th edition TNM classification of lung cancer 213. Beyond prognostic significance, there are several therapeutic implications related to the different stages, especially in SCLC patients, in whom the classification of limited disease (i.e., cancer confined to the thorax in a single radiation field) or extensive disease (i.e., any T, any N, M1a/b/c) is clinically used 214. From a clinical perspective, in case of non-metastatic disease, the pivotal data an oncologist must consider, beyond histologic characterization and proliferation index, include the state of margins (R1 or R2 if microscopic and macroscopic residual disease, respectively) and the number and location of lymph node metastases relative to the total number of removed lymph nodes 215, 216. In the event of R1 or R2, a multidisciplinary evaluation is mandatory for determining the best course of action, in other words whether new surgery or, alternatively, radiotherapy with or without a systemic therapy should be planned (e.g., in N2 ACs) 125. This evaluation is crucial not only for immediate therapeutic decisions, but also a follow-up perspective. Given the lack of universally shared guidelines for follow-up timing and imaging, these specific details are critical for the NEN-dedicated multidisciplinary team to make informed decisions on a case-by-case basis. Moreover, in the setting of multifocal TCs and DIPNECH, underlying genetic syndromes should be ruled out 217
Pathologist.
Lung NENs are staged according to the pathologic classification of TNM scheme (pTNM) by the UICC and AJCC 213, including lung NENs. These proposals will account for residual tumor after resection (complete, uncertain or incomplete) 218, stage grouping 219, N descriptors 220, M descriptors 221 and spread through air spaces 222, in order to better meet the emerging needs of lung cancer patients. As the TNM system could not perfectly intercept the unique biologic properties of lung carcinoids, integrated staging/histology-based classification 223, 224, combined stages 225 or prognostic scores 226 have been proposed, but they still need confirmation. Bronchial, vascular and parenchymal resection should always be indicated in all lung NENs, including carcinoids 4, measuring the distance from tumor edges (especially in conservative resections) to guarantee radical excision (R0). Concomitant NE cell hyperplasia (NECH, i.e., limited to the bronchial epithelium) in pathologic/syndromic DIPNECH (generalized NECH with tumorlets and obliterative bronchiolitis) 1 should be recorded for staging purposes, especially in the event of multifocal/bilateral carcinoid nodules 142, 227, 228.
Shared solution.
Although TNM staging systems provide valuable prognostic and predictive information in lung cancer as a whole, it is more suited for NECs, while it should be perfected for NETs where an excess of low malignancy tumors is found. As long as new staging modalities will be made available, pathologic TNM and margin status should always be included in diagnostic reports.
8. STANDARDIZED REPORTING
Oncologist.
Standardizing the pathologic report is crucial for NEN-dedicated oncologists, thoracic surgeons and other members of the multidisciplinary team to ensure comprehensive patient management. To achieve adequate disease description and proper evaluation of all therapeutic options, requirements on reporting must be shared with the pathologist within the team of lung NEN experts. In April 2024, a consensus meeting of experts in endocrinology, oncology and endocrine pathology was held in Rome under the aegis of the Working Group of Endocrine Pathology (GIPE) of the Italian Society of Anatomic Pathology and Cytology (SIAPEC-IAP) to preliminarily discuss how to construct a standardized pathology report for endocrine and neuroendocrine neoplasms, including lung NENs, aiming to identify diagnostic criteria projected into clinical decision making (Pelosi et al., manuscript in preparation).
Pathologist.
In search of a shared standardized language, the 2021 WHO Classification on lung tumors offered practical solutions by devising essential (or core) and desirable (or non-core) criteria for both NETs and NECs 1. While a detailed analysis on the subject is beyond the scope of this review article, suffice it to say that the College of American Pathologists (CAP), the International Collaboration on Cancer Reporting (ICCR), the European Society of Medical Oncology (ESMO) and the European Neuroendocrine Tumor Society (ENETS) have repeatedly stated the need for standardized pathology reports, also proposing templates for examining specimens of primary lung NENs, including carcinoids and NECs. These templates are based on dedicated checklists that are useful for the sake of clarity, completeness of information and scientific soundness 215.
In lung NETs, essential or core criteria comprise histologic subtyping into low grade TC/NET G1 and intermediate grade AC/NET G2 on resection and carcinoid tumor NOS/metastatic carcinoid on scarce diagnostic material of primary and metastasis, respectively, always reporting the values of defining criteria (mitoses per 2 mm2 and necrosis and, desirably, Ki-67) (Tab. I). Immunohistochemistry for NE markers should be added to corroborate the pathology reporting. Pathologic TNM staging is also recommended after resection, with a comment on multiple NETs: pT factor is decided according to the highest lesion with “m” suffix for multicentricity (in the event of DIPNECH 142, 228, 229) or pT3/pT4/pM1a factors are chosen for intrapulmonary seeding, with single pN and pM factors for all lesions collectively considered 218. Although molecular testing is not routinely used in NETs/NECs and in NSCCs/non-NECs 1, 4, 6, it is left to the clinical judgment and the patients’ demography/clinical presentation, such as the lack of smoking history, increased likelihood of driver mutations or transdifferentiation of NECs (de novo or after therapy).
In NECs, essential or core criteria comprise subtyping into SCLC and LCNEC, in pure or combined subtypes (Tab. I). SCLC is diagnosed in any type of material; LCNEC only in surgical specimens, but is termed “NSCC possible LCNEC” in biopsy/cytology samples. NECs are staged according to the same TNM-R system as conventional lung cancer, whereas limited-extensive disease for SCLC is used in a clinical context. DIPNECH is unlikely to occur in NECs 229. A panel of immunohistochemical markers desirably improves differential diagnosis towards histologic mimickers or combined subtypes 230 and, additionally, Ki-67 avoids misdiagnosing NETs for NECs (usually 50 to 100% in SCLC and 40 to 80% in LCNEC) 1, 161.
Shared solution.
Standardizing the pathology report should become the gold standard and priority for clinical decision making for lung NEN patients. Adoption of shared checklists could improve consistency, comparability and cross-study evaluation. The world of predictive markers is rapidly expanding with data essential for the clinical practice.
Conclusions
This lung NEN review article was conducted by peer questioning and answering between oncologists and pathologists, which reflects their engagement in the clinical decision-making on the one hand and the diagnostic effort on the other. In particular, we aimed to present some practical and sequential steps that we adopt in clinical reasoning on both NETs and NECs, in order to elicit a critical and objective idea in the reader, who is hopefully capable of improving her/his own awareness and management skills. Our knowledge on lung NENs has moved from a generic tumor-centered vision to the current outlook focusing on the individual patient’s cancer, where modern classifications are largely integrated with pathogenetic mechanisms to guide therapeutic choices 1. In this scenario, standardizing pathology reports and shared languages of lung NENs matter for consistency/quality, efficacy/time saving, improved decision-making and compliance/regulations.
The dramatic clinical presentation of lung NECs makes their distinction from NSCCs a clinical need. As this context is often mediated by small biopsies or cytology samples, distinguishing SCLC from LCNEC, as well as primaries from metastases, remains an essential one in light of non-overlapping therapeutic treatments. Moving on to the rarer NETs, the starting point, in our opinion, is the correct distinction between TCs and ACs or the use of a noncommittal terminology depending on the different diagnostic context (resection specimens and biopsy/cytology samples, respectively). The burden by the clinician will be to make appropriate material available by arranging all preoperative or pre-therapeutic investigations and drive molecular testing requests (if any), while the burden by the pathologist will be to strategically manage all tumor tissues for diagnostic and predictive/prognostic purposes 231.
Clinical management or accrual into specific study protocols rely on a standardized tumor nosology, which encourages diagnostic consistency, cross-study evaluation and determining the clinico-pathologic characteristics of the individual patient. In this regard, templates for a minimum pathology data set have been made available from international organizations, with essential or core and desirable or non-core criteria to promote a common language 232. A Delphi consensus approach has been used for a minimum data set collection in GEP NENs 233, and similar efforts should be implemented even in thoracic NENs for consistency 1, 163, 233. Recently, the special interest group on lung LCNEC of the British Thoracic Oncology Group (BTOG-LCNEC-SIG), which reunites oncologists, nurses, respiratory physicians, and pathologists in the UK, has been established to set care standards and improve outcomes for these patients 234. The aims are to facilitate a UK consensus on the definition/diagnosis of LCNEC, develop a UK clinical consensus around clinical management, transfer results to the international community and define clinical trial eligibility as a new setting for treatment progress 234. In fact, our understanding of lung NENs has significantly grown over time in terms of biologic, pathologic, clinical, imaging, oncologic and molecular characterization, especially for life-threatening NECs 13, many of which are presently given full consideration in daily practice for the treatment of the individual patient 235. Combined, integrated, and deepened clinical and molecular analyses 48, 70, 100, 167, 235 along with artificial intelligence tools 61, 181 probably represent the future in lung NEN management and the basis for the next WHO classification.
ACKNOWLEDMENTS
This work is dedicated to the memory of Carlotta, an extraordinarily lively girl who died an untimely death due to cancer in the prime of her life.
CONFLICT OF INTEREST STATEMENT
The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
FUNDING
There is no funding for this article.
ETHICAL CONSIDERATION
This is a review article on already existing literature data, for which no approval by Internal Review Board or Ethical Committee notification was needed.
AUTHORS CONTRIBUTION
Giuseppe Pelosi and Alice Laffi: conceptualization, data curation, literature investigation, methodology, supervision, validation, writing ug of the original draft, critical revision and finalization of the manuscript;
Mauro Papotti, Sylvie Lantuejoul, Jean-Yves Scoazec, Antonino Bruno, Fabrizio Bianchi, Barbara Bassani: data curation, methodology, validation, critical revision and finalization of the manuscript;
Maria Gemelli, Riccardo Ricotta, Sergio Harari, Eleonora Duregon, Riccardo Papa, Angelica Sonzogni, Silvia Uccella, Carlo Carnaghi, Alexia Francesca Bertuzzi: validation, critical revision and finalization of the manuscript.
ABBREVIATION LIST
History
Received: March 5, 2025
Accepted: March 25, 2025
Figures and tables
Figure 1. Histologic features of pulmonary neuroendocrine tumors. A spindle cell-shaped typical carcinoid (TC) features elongated tumor cells disposed in intertwining fascicles mimicking a mesenchymal neoplasm, with no necrosis or recognizable mitotic figures (A). Another TC example shows polygonal cells organized in trabecular and lobular structures of epithelial appearance, again with no necrosis or recognizable mitotic figures (B). INSM1 immunohistochemistry is shown to decorate tumor cell nuclei (B, inset). A case of atypical carcinoid (AC) presents with a solid histologic appearance, but superimposable cytologic characteristics (C), along with immunohistochemical membrane reactivity for somatostatin receptor type 2A (C, inset). Punctate necrosis is a histologic hallmark, which is diagnostic of AC (D). The new category of carcinoid tumor with elevated mitotic count and/or Ki-67 proliferation index exhibits increased mitotic activity (highlighted by yellow arrows) (E), punctate necrosis (F) and increased Ki-67 labeling index (G). Pictures taken at 20X or 40X magnification.
Figure 2. Histologic features of pulmonary neuroendocrine carcinomas. A large cell neuroendocrine carcinoma (LCNEC) is composed by organoid aggregates of tumor cells with peripheral palisading and abundant necrosis (A), with common TTF1 expression (A, inset). Tumor cells present with coarse nuclear chromatin and plentiful mitoses (B). Small cell lung carcinoma (SCLC) is characterized by small-sized tumor cells with extensive geographic-type necrosis (C) and faint immunohistochemical decoration for chromogranin A (C, inset). This case presents with variable immunoreactivity for the ASCL1 gene product (D). For comparison, another morphologically similar SCLC case expressed POU2F3 rather than ASCL1 (E). Pictures taken at 20X or 40X magnification.
Typical carcinoid/NET G1† | Atypical carcinoid/NET G2† | Carcinoid tumor/NET with elevated mitotic counts and/or Ki-67 proliferation index* | Large cell neuroendocrine carcinoma (LCNEC) | Small cell lung carcinoma (SCLC) | |
---|---|---|---|---|---|
Defining criteria | |||||
Neuroendocrine morphology | Yes | Yes | Yes | Yes | Yes§ |
Mitoses per 2 mm 2 | 0-1 | 2-10 | >10 | >10 (median: 70) | >10 (median: 80) |
Necrosis | No | Focal, if any | Focal | Frequent | Frequent |
Differentiation level | Well-differentiated | Well-differentiated | Well-differentiated | Poorly differentiated | Poorly differentiated |
Neoplasm grade | Low | Intermediate | Intermediate | High | High |
Ki-67 labeling index | Up to 5% | Up to 30% | > 30% | 40-80% | 50-100% |
Combined with NSCC component | Very rare | Very rare | Not reported, but possible | Up to 25% of resected LCNEC | Up to 25% of resected SCLC |
Cytology or biopsy samples | |||||
Primary | Carcinoid tumor/NET NOS | Carcinoid tumor/NET NOS | Carcinoid with elevated proliferation | NSCC, possible LCNEC | SCLC |
Metastasis | Metastatic carcinoid NOS | Metastatic carcinoid NOS | Carcinoid with elevated proliferation | NSCC, possible LCNEC | SCLC |
NSCC, non-small cell carcinoma; NOS, not otherwise specified. †: Diagnostic criteria for typical and atypical carcinoid are applicable just to lung resection specimens. *: provisional category included in the 2021 WHO classification (see text for more details). §: neuroendocrine morphology is less evident in biopsy/cytology samples, where diagnostic judgment mainly relies on the recognition of small-sized cells. |
1) Distinguishing among high-grade NECs and conventional NSCCs/non-NECs |
Oncologist. There are different systemic therapy options (and related clinical trials) to treat NECs and NSCCs. There is uncertainty about the treatment of NSCCs with NE differentiation (NSCCs-NED). |
Pathologist. The rule is that NE markers should be used only if there is NE morphology. |
2) Distinguishing among high-grade NECs (biopsy setting) |
Oncologist. There are different systemic therapy options (and related clinical trials) to treat small cell lung carcinoma (SCLC) and large cell NE carcinoma (LCNEC). However, diagnosis on biopsy may be demanding and combined tumors could be missed in small diagnostic material. |
Pathologist. It is possible to distinguish SCLC and LCNEC in most cases, provided that core biopsies with larger amounts of tissue are available. Some uncertainty, however, may persist while assessing cell size and cytological features reliably: in these instances, the noncommittal term NEC NOS can be used. While combined tumors are not infrequent and could be missed in small samples as a function of random sampling, clinical correlations (e.g., nonsmokers) and molecular investigation may prove useful for the relevant diagnosis. |
3) Distinguishing between pure and combined (3.1) or primary and metastatic (3.2) NECs |
Oncologist. There are different biologic features and sensitivity to chemo/radiotherapy between pure and combined NECs, as well as between pulmonary and extrapulmonary sites (urologic, head-neck, gynecologic), thereby realizing biologically, behaviorally and therapeutically different contexts. |
Pathologist. About 25-30% of NECs with either small or large cells are combined with any other histologic subtype, which may be readily recognized upon morphology and appropriate immunomarkers, especially in surgical specimens. Distinguishing pulmonary vs extrapulmonary NECs may prove difficult if not impossible using morphology or immunohistochemistry. Molecular assessment may be helpful, despite doubts about its reproducibility or large-scale applicability. |
4) Distinguishing among well-differentiated NETs |
Oncologist. Different clinical behavior, PET scan appearance and therapeutic approaches exist between typical (TC) and atypical (AC) carcinoids, thus necessitating punctual diagnosis and coherence of terminology in all diagnostic settings. Uncovering primary or metastatic lung NETs is clinically warranted. |
Pathologist. Classification of carcinoids differs according to the diagnostic context making precise comparison between biopsies and resection specimens or between primary and metastatic sites a demanding task. Separation requires mitotic count per 2 mm2 and necrosis assessment, which can be reliably carried out just in resection specimens of primary tumors. Uncovering primary or metastatic lung NETs is feasible by means of immunohistochemical markers (e.g., TTF1 or OTP). |
5) Distinguishing between ACs and LCNECs: is Ki-67 proliferation index the solution? |
Oncologist. Straddling ACs and NECs, could the Ki-67 proliferation index be the solution? It may help if low, but not high. |
Pathologist. Ki-67 proliferation marker has prognostic and diagnostic implications in lung NENs, bacause it reliably distinguishes NET from NEC in every context, identifies carcinoids with elevated proliferation rates and roughly correlates with carcinoid classification as desirable but not essential diagnostic tool. It is recommended to report the value of Ki-67 on diagnoses. |
6) Are there other treatment-related biomarkers? |
Oncologist. There are many biomarkers (SSTR, OTP, CD44, ASCL1, MGMT score, NTRK, ALK, TMB, MMR, p53, RB…) that can aid in stratification of lung NETs/NECs (to treat patients better) by offering further strategies of treatment. Even characterization for hormone production (e.g., ACTH for Cushing syndrome) may be clinically warranted. |
Pathologist. Many studies have used these biomarkers to stratify diversely behaving patient subsets, especially in the setting of NETs but even NECs. Molecular testing is not routinely used in NETs/NECs, but it is up to clinical judgment and patients’ demography/clinical presentation (e.g., lack of smoking history or increased likelihood of diverse mutations). Therefore, local feasibility of these assays should be discussed thoroughly with the pathologist, who should perform them on demand for justified clinical reasons. |
7) Staging procedure |
Oncologist. Information on state of margins, lymph node status (number of metastatic lymph node/number of total lymph nodes), presence of other neoplastic nodules and association with DIPNECH are clinically warranted, because one can best treat patient within a NEN-dedicated multidisciplinary team. |
Pathologist. pTNM-R classification according to UICC/AJCC scheme (currently the 8th edition, but the 9th is coming soon) is the required standard for staging all resection specimens, also for distinguishing multiple synchronous NETs (e.g., in the setting of DIPNECH) from intrapulmonary metastases. Future TNM classification should better address the issue of NET staging. |
8) Standardized reporting |
Oncologist. State of the art for clinical decision-making in the setting of lung NENs needs to develop shared language and clinical information integrated with pathology data, because specialists gravitating around these neoplasms are plentiful but the patient always is unique and at the center by definition. |
Pathologist. It is of the upmost relevance to standardize the pathology report by introducing essential or core and desirable or non-core criteria. Current schemes already exist, which could be further implemented in the near future by keeping in mind that pathology classification must be clinically oriented and clinically useful. |
NEN, neuroendocrine neoplasm; NEC, neuroendocrine carcinoma; NSCC, non-small cell carcinoma; NE, neuroendocrine; NED, neuroendocrine differentiation; SCLC, small cell lung carcinoma; LCNEC, large cell neuroendocrine carcinoma; NET, neuroendocrine tumor; PET, positron emission tomography; TC, typical carcinoid; AC, atypical carcinoid; SSTR, somatostatin receptor; OTP, orthopedia homeobox; CD44, CD44 molecule (IN Blood Group); ASCL1, achaete-scute family BHLH transcription factor 1; MGMT, O-6-methylguanine-DNA methyltransferase; NTRK, neurotrophic receptor tyrosine kinase; ALK, ALK receptor tyrosine kinase; TMB: tumor mutation burden; MMR: DNA mismatch repair; RB: retinoblastoma; ACTH, adrenocorticotropic hormone. |
References
- Borczuk C, Cooper W, Dacic S. WHO Classification of Tumours: Thoracic Tumours. International Agency for Research on Cancer; 2021.
- Vocino Trucco G, Righi L, Volante M. Updates on lung neuroendocrine neoplasm classification. Histopathology. 2024;84:67-85. doi:https://doi.org/10.1111/his.15058
- Baudin E, Hayes A, Scoazec J. Unmet Medical Needs in Pulmonary Neuroendocrine (Carcinoid) Neoplasms. Neuroendocrinology. 2019;108:7-17. doi:https://doi.org/10.1159/000493980
- Caplin M, Baudin E, Ferolla P. Pulmonary neuroendocrine (carcinoid) tumors: European Neuroendocrine Tumor Society expert consensus and recommendations for best practice for typical and atypical pulmonary carcinoids. Ann Oncol. 2015;26:1604-1620. doi:https://doi.org/10.1093/annonc/mdv041
- Dingemans A, Fruh M, Ardizzoni A. Small-cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up(*). Ann Oncol. 2021;32:839-853. doi:https://doi.org/10.1016/j.annonc.2021.03.207
- Ganti A, Loo B, Bassetti M. Small Cell Lung Cancer, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2021;19:1441-1464. doi:https://doi.org/10.6004/jnccn.2021.0058
- Hendifar A, Marchevsky A, Tuli R. Neuroendocrine Tumors of the Lung: Current Challenges and Advances in the Diagnosis and Management of Well-Differentiated Disease. J Thorac Oncol. 2017;12:425-436. doi:https://doi.org/10.1016/j.jtho.2016.11.2222
- Shah M, Goldner W, Halfdanarson T. NCCN Guidelines Insights: Neuroendocrine and Adrenal Tumors, Version 2.2018. J Natl Compr Canc Netw. 2018;16:693-702. doi:https://doi.org/10.6004/jnccn.2018.0056
- Singh S, Bergsland E, Card C. Commonwealth Neuroendocrine Tumour Research Collaboration and the North American Neuroendocrine Tumor Society Guidelines for the Diagnosis and Management of Patients With Lung Neuroendocrine Tumors: An International Collaborative Endorsement and Update of the 2015 European Neuroendocrine Tumor Society Expert Consensus Guidelines. J Thorac Oncol. 2020;15:1577-1598. doi:https://doi.org/10.1016/j.jtho.2020.06.021
- Yang L, Fan Y, Lu H. Pulmonary Large Cell Neuroendocrine Carcinoma. Pathol Oncol Res. 2022;28. doi:https://doi.org/10.3389/pore.2022.1610730
- Metovic J, Barella M, Bianchi F. Morphologic and molecular classification of lung neuroendocrine neoplasms. Virchows Arch. 2021;478:5-19. doi:https://doi.org/10.1007/s00428-020-03015-z
- Metovic J, Bianchi F, Rossi G. Recent advances and current controversies in lung neuroendocrine neoplasms. Semin Diagn Pathol. 2021;38:90-97. doi:https://doi.org/10.1053/j.semdp.2021.03.002
- Pelosi G, Bianchi F, Hofman P. Recent advances in the molecular landscape of lung neuroendocrine tumors. Expert Rev Mol Diagn. 2019;19:281-297. doi:https://doi.org/10.1080/14737159.2019.1595593
- Pelosi G, Sonzogni A, Harari S. Classification of pulmonary neuroendocrine tumors: new insights. Transl Lung Cancer Res. 2017;6:513-529. doi:https://doi.org/10.21037/tlcr.2017.09.04
- Pelosi G, Travis W. Head-to-head: Should Ki67 proliferation index be included in the formal classification of pulmonary neuroendocrine neoplasms?. Histopathology. 2024;85:535-548. doi:https://doi.org/10.1111/his.15206
- Riely G, Wood D, Ettinger D. Non-Small Cell Lung Cancer, Version 4.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2024;22:249-274. doi:https://doi.org/10.6004/jnccn.2204.0023
- Lynch T, Bell D, Sordella R. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129-2139. doi:https://doi.org/10.1056/NEJMoa040938
- Mok T, Wu Y, Thongprasert S. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947-957. doi:https://doi.org/10.1056/NEJMoa0810699
- Yang J, Wu Y, Schuler M. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015;16:141-151. doi:https://doi.org/10.1016/S1470-2045(14)71173-8
- Rosell R, Carcereny E, Gervais R. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239-246. doi:https://doi.org/10.1016/S1470-2045(11)70393-X
- Soria J, Ohe Y, Vansteenkiste J. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med. 2018;378:113-125. doi:https://doi.org/10.1056/NEJMoa1713137
- Mok T, Cheng Y, Zhou X. Improvement in Overall Survival in a Randomized Study That Compared Dacomitinib With Gefitinib in Patients With Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. J Clin Oncol. 2018;36:2244-2250. doi:https://doi.org/10.1200/JCO.2018.78.7994
- Soda M, Choi Y, Enomoto M. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature. 2007;448:561-566. doi:https://doi.org/10.1038/nature05945
- Soria J, Tan D, Chiari R. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet. 2017;389:917-929. doi:https://doi.org/10.1016/S0140-6736(17)30123-X
- Peters S, Camidge D, Shaw A. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2017;377:829-838. doi:https://doi.org/10.1056/NEJMoa1704795
- Solomon B, Besse B, Bauer T. Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from a global phase 2 study. Lancet Oncol. 2018;19:1654-1667. doi:https://doi.org/10.1016/S1470-2045(18)30649-1
- Camidge D, Kim H, Ahn M. Brigatinib versus Crizotinib in ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med. 2018;379:2027-2039. doi:https://doi.org/10.1056/NEJMoa1810171
- Dziadziuszko R, Krebs M, De Braud F. Updated Integrated Analysis of the Efficacy and Safety of Entrectinib in Locally Advanced or Metastatic ROS1 Fusion-Positive Non-Small-Cell Lung Cancer. J Clin Oncol. 2021;39:1253-1263. doi:https://doi.org/10.1200/JCO.20.03025
- Doebele R, Drilon A, Paz-Ares L. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020;21:271-282. doi:https://doi.org/10.1016/S1470-2045(19)30691-6
- Cascone T, Awad M, Spicer J. Perioperative Nivolumab in Resectable Lung Cancer. N Engl J Med. 2024;390:1756-1769. doi:https://doi.org/10.1056/NEJMoa2311926
- Heymach J, Harpole D, Mitsudomi T. Perioperative Durvalumab for Resectable Non-Small-Cell Lung Cancer. N Engl J Med. 2023;389:1672-1684. doi:https://doi.org/10.1056/NEJMoa2304875
- Wakelee H, Liberman M, Kato T. Perioperative Pembrolizumab for Early-Stage Non-Small-Cell Lung Cancer. N Engl J Med. 2023;389:491-503. doi:https://doi.org/10.1056/NEJMoa2302983
- Antonia S, Villegas A, Daniel D. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med. 2017;377:1919-1929. doi:https://doi.org/10.1056/NEJMoa1709937
- Patel S, Das M. Small Cell Lung Cancer: Emerging Targets and Strategies for Precision Therapy. Cancers (Basel). Published online 2023. doi:https://doi.org/10.3390/cancers15164016
- Scagliotti G, Parikh P, von Pawel J. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol. 2008;26:3543-3551. doi:https://doi.org/10.1200/JCO.2007.15.0375
- Derks J, Leblay N, Thunnissen E. Molecular Subtypes of Pulmonary Large-cell Neuroendocrine Carcinoma Predict Chemotherapy Treatment Outcome. Clin Cancer Res. 2018;24:33-42. doi:https://doi.org/10.1158/1078-0432.CCR-17-1921
- Hadoux J, Kanaan C, Durand A. Prognostic factors of metastatic neuroendocrine carcinoma under first-line treatment with platinum etoposide with a focus on NEC score and Rb expression: Results from the multicentre RBNEC study of the Groupe d’Etude des Tumeurs Endocrines (GTE) and the ENDOCAN-RENATEN network. Eur J Cancer. 2021;152:100-115. doi:https://doi.org/10.1016/j.ejca.2021.04.030
- Nosaki K, Saka H, Hosomi Y. Safety and efficacy of pembrolizumab monotherapy in elderly patients with PD-L1-positive advanced non-small-cell lung cancer: Pooled analysis from the KEYNOTE-010, KEYNOTE-024, and KEYNOTE-042 studies. Lung Cancer. 2019;135:188-195. doi:https://doi.org/10.1016/j.lungcan.2019.07.004
- Pelosi G, Rossi G, Cavazza A. DeltaNp63 (p40) distribution inside lung cancer: a driver biomarker approach to tumor characterization. Int J Surg Pathol. 2013;21:229-239. doi:https://doi.org/10.1177/1066896913476750
- Yatabe Y, Dacic S, Borczuk A. Best Practices Recommendations for Diagnostic Immunohistochemistry in Lung Cancer. J Thorac Oncol. 2019;14:377-407. doi:https://doi.org/10.1016/j.jtho.2018.12.005
- Pelosi G, Scarpa A, Forest F. The impact of immunohistochemistry on the classification of lung tumors. Expert Rev Respir Med. 2016;10:1105-1121. doi:https://doi.org/10.1080/17476348.2017.1235975
- Travis W, Brambilla E, Noguchi M. International association for the study of lung cancer/american thoracic society/european respiratory society international multidisciplinary classification of lung adenocarcinoma. J Thorac Oncol. 2011;6:244-285.
- Kalemkerian G, Loo B, Akerley W. NCCN Guidelines Insights: Small Cell Lung Cancer, Version 2.2018. J Natl Compr Canc Netw. 2018;16:1171-1182. doi:https://doi.org/10.6004/jnccn.2018.0079
- Kalemkerian G, Schneider B. Advances in Small Cell Lung Cancer. Hematol Oncol Clin North Am. 2017;31:143-156. doi:https://doi.org/10.1016/j.hoc.2016.08.005
- Takahara Y, Tanaka T, Ishige Y. Differential response in patients with large cell neuroendocrine carcinoma of the lung to initial therapy: A case series. Cancer Rep (Hoboken). 2023;6. doi:https://doi.org/10.1002/cnr2.1754
- Derks J, van Suylen R, Thunnissen E. Chemotherapy for pulmonary large cell neuroendocrine carcinomas: does the regimen matter?. Eur Respir J. Published online 2017. doi:https://doi.org/10.1183/13993003.01838-2016
- Rekhtman N, Pietanza M, Hellmann M. 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. doi:https://doi.org/10.1158/1078-0432.CCR-15-2946
- George J, Walter V, Peifer M. Integrative genomic profiling of large-cell neuroendocrine carcinomas reveals distinct subtypes of high-grade neuroendocrine lung tumors. Nat Commun. 2018;9. doi:https://doi.org/10.1038/s41467-018-03099-x
- Wang Z, Wu Y, Lu T. The outcomes of different regimens depend on the molecular subtypes of pulmonary large-cell neuroendocrine carcinoma: A retrospective study in China. Cancer Med. 2024;13. doi:https://doi.org/10.1002/cam4.6834
- Jimenez-Heffernan J, Lopez-Ferrer P, Vicandi B. Fine-needle aspiration cytology of large cell neuroendocrine carcinoma of the lung: a cytohistologic correlation study of 11 cases. Cancer. 2008;114:180-186. doi:https://doi.org/10.1002/cncr.23539
- Rothrock A, Stewart J, Li F. Exploration of INSM1 and hASH1 as additional markers in lung cytology samples of high-grade neuroendocrine carcinoma with indeterminate neuroendocrine differentiation. Diagn Cytopathol. 2022;50:230-234. doi:https://doi.org/10.1002/dc.24938
- den Bakker M, Thunnissen F. Neuroendocrine tumours--challenges in the diagnosis and classification of pulmonary neuroendocrine tumours. J Clin Pathol. 2013;66:862-869. doi:https://doi.org/10.1136/jclinpath-2012-201310
- den Bakker M, Willemsen S, Grunberg K. Small cell carcinoma of the lung and large cell neuroendocrine carcinoma interobserver variability. Histopathology. 2010;56:356-363. doi:https://doi.org/10.1111/j.1365-2559.2010.03486.x
- Travis W, Gal A, Colby T. Reproducibility of neuroendocrine lung tumor classification. Hum Pathol. 1998;29:272-279. doi:https://doi.org/10.1016/s0046-8177(98)90047-8
- Marchevsky A, Gal A, Shah S. Morphometry confirms the presence of considerable nuclear size overlap between “small cells” and “large cells” in high-grade pulmonary neuroendocrine neoplasms. Am J Clin Pathol. 2001;116:466-472. doi:https://doi.org/10.1309/H40B-8W14-4Q47-03EP
- Qu S, Fetsch P, Thomas A. Molecular Subtypes of Primary SCLC Tumors and Their Associations With Neuroendocrine and Therapeutic Markers. J Thorac Oncol. 2022;17:141-153. doi:https://doi.org/10.1016/j.jtho.2021.08.763
- Righi L, Volante M, Papotti M. Small-Cell Carcinoma of the Lung: What We Learned about It?. Acta Cytol. 2022;66:257-268. doi:https://doi.org/10.1159/000519688
- Handa Y, Tsutani Y, Ito M. Clinical Behavior of Combined Versus Pure High-Grade Neuroendocrine Carcinoma. Clin Lung Cancer. 2022;23:e9-e16 e11. doi:https://doi.org/10.1016/j.cllc.2021.06.010
- Han Z, Yang F, Wang F. Advances in combined neuroendocrine carcinoma of lung cancer. Pathol Oncol Res. 2024;30. doi:https://doi.org/10.3389/pore.2024.1611693
- Gonzalez D, Dietz R, Pantanowitz L. Feasibility of a deep learning algorithm to distinguish large cell neuroendocrine from small cell lung carcinoma in cytology specimens. Cytopathology. 2020;31:426-431. doi:https://doi.org/10.1111/cyt.12829
- Ilie M, Benzaquen J, Tourniaire P. Deep Learning Facilitates Distinguishing Histologic Subtypes of Pulmonary Neuroendocrine Tumors on Digital Whole-Slide Images. Cancers (Basel). 2022;14. doi:https://doi.org/10.3390/cancers14071740
- Lee K, Lee Y, Oh S. Large cell neuroendocrine carcinoma of the lung: CT and FDG PET findings. Eur J Radiol. 2015;84:2332-2338. doi:https://doi.org/10.1016/j.ejrad.2015.07.033
- Oshiro Y, Kusumoto M, Matsuno Y. CT findings of surgically resected large cell neuroendocrine carcinoma of the lung in 38 patients. AJR Am J Roentgenol. 2004;182:87-91. doi:https://doi.org/10.2214/ajr.182.1.1820087
- Nicholson S, Beasley M, Brambilla E. Small cell lung carcinoma (SCLC): a clinicopathologic study of 100 cases with surgical specimens. Am J Surg Pathol. 2002;26:1184-1197. doi:https://doi.org/10.1097/00000478-200209000-00009
- Travis W. Update on small cell carcinoma and its differentiation from squamous cell carcinoma and other non-small cell carcinomas. Mod Pathol. 2012;25:S18-30. doi:https://doi.org/10.1038/modpathol.2011.150
- Lantuejoul S, Fernandez-Cuesta L, Damiola F. New molecular classification of large cell neuroendocrine carcinoma and small cell lung carcinoma with potential therapeutic impacts. Transl Lung Cancer Res. 2020;9:2233-2244. doi:https://doi.org/10.21037/tlcr-20-269
- Qin J, Lu H. Combined small-cell lung carcinoma. Onco Targets Ther. 2018;11:3505-3511. doi:https://doi.org/10.2147/OTT.S159057
- Pelosi G, Sonzogni A, Galetta D. Combined small-cell carcinoma of the lung with quadripartite differentiation of epithelial, neuroendocrine, skeletal muscle, and myofibroblastic type. Virchows Arch. 2011;458:497-503. doi:https://doi.org/10.1007/s00428-010-1011-8
- Board E. WHO Classification of Tumours: Digestive System Tumours. IARC; 2019.
- Balanis N, Sheu K, Esedebe F. Pan-cancer Convergence to a Small-Cell Neuroendocrine Phenotype that Shares Susceptibilities with Hematological Malignancies. Cancer Cell. 2019;36:17-34 e17. doi:https://doi.org/10.1016/j.ccell.2019.06.005
- Offin M, Chan J, Tenet M. Concurrent RB1 and TP53 Alterations Define a Subset of EGFR-Mutant Lung Cancers at risk for Histologic Transformation and Inferior Clinical Outcomes. J Thorac Oncol. 2019;14:1784-1793. doi:https://doi.org/10.1016/j.jtho.2019.06.002
- Yin X, Li Y, Wang H. Small cell lung cancer transformation: From pathogenesis to treatment. Semin Cancer Biol. 2022;86:595-606. doi:https://doi.org/10.1016/j.semcancer.2022.03.006
- Kim T, Kim H, Lee K. Multimodality assessment of esophageal cancer: preoperative staging and monitoring of response to therapy. Radiographics. 2009;29:403-421. doi:https://doi.org/10.1148/rg.292085106
- Fujimoto N, Harada S, Terado M. Multiple biopsies of normal-looking urothelium in patients with superficial bladder cancer: Are they necessary?. Int J Urol. 2003;10:631-635. doi:https://doi.org/10.1046/j.1442-2042.2003.00724.x
- Gudjonsson S, Blackberg M, Chebil G. The value of bladder mapping and prostatic urethra biopsies for detection of carcinoma in situ (CIS). BJU Int. 2012;110:E41-45. doi:https://doi.org/10.1111/j.1464-410X.2011.10654.x
- Baize N, Monnet I, Greillier L. Carboplatin plus etoposide versus topotecan as second-line treatment for patients with sensitive relapsed small-cell lung cancer: an open-label, multicentre, randomised, phase 3 trial. Lancet Oncol. 2020;21:1224-1233. doi:https://doi.org/10.1016/S1470-2045(20)30461-7
- Sorbye H, Welin S, Langer S. 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. doi:https://doi.org/10.1093/annonc/mds276
- Salvo G, Flores Legarreta A, Ramalingam P. Clinicopathologic characteristics, oncologic outcomes, and prognostic factors in neuroendocrine cervical carcinoma: a Neuroendocrine Cervical Tumor Registry study. Int J Gynecol Cancer. 2023;33:1359-1369. doi:https://doi.org/10.1136/ijgc-2023-004708
- Lin J, Chen X, Wang P. Conditional Survival of Patients with Limited-Stage Small Cell Lung Cancer After Surgery: A National Real-World Cohort Study. Ann Surg Oncol. 2024;31:4250-4260. doi:https://doi.org/10.1245/s10434-024-14983-w
- Jones G, Elimian K, Baldwin D. A systematic review of survival following anti-cancer treatment for small cell lung cancer. Lung Cancer. 2020;141:44-55. doi:https://doi.org/10.1016/j.lungcan.2019.12.015
- Mahdi H, Joehlin-Price A, Elishaev E. Genomic analyses of high-grade neuroendocrine gynecological malignancies reveal a unique mutational landscape and therapeutic vulnerabilities. Mol Oncol. 2021;15:3545-3558. doi:https://doi.org/10.1002/1878-0261.13057
- Olson A, Kammer J, Taher A. The inseparability of context and clinical reasoning. J Eval Clin Pract. 2024;30:533-538. doi:https://doi.org/10.1111/jep.13969
- Kaufmann O, Dietel M. Expression of thyroid transcription factor-1 in pulmonary and extrapulmonary small cell carcinomas and other neuroendocrine carcinomas of various primary sites. Histopathology. 2000;36:415-420. doi:https://doi.org/10.1046/j.1365-2559.2000.00890.x
- Rudin C, Poirier J, Byers L. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer. 2019;19:289-297. doi:https://doi.org/10.1038/s41568-019-0133-9
- George J, Maas L, Abedpour N. Evolutionary trajectories of small cell lung cancer under therapy. Nature. 2024;627:880-889. doi:https://doi.org/10.1038/s41586-024-07177-7
- Megyesfalvi Z, Barany N, Lantos A. Expression patterns and prognostic relevance of subtype-specific transcription factors in surgically resected small-cell lung cancer: an international multicenter study. J Pathol. 2022;257:674-686. doi:https://doi.org/10.1002/path.5922
- Popper H, Brcic L, Eidenhammer S. Does subtyping of high-grade pulmonary neuroendocrine carcinomas have an impact on therapy selection?. Transl Lung Cancer Res. 2023;12:2412-2426. doi:https://doi.org/10.21037/tlcr-23-505
- Koh J, Kim H, Moon K. Molecular Classification of Extrapulmonary Neuroendocrine Carcinomas With Emphasis on POU2F3-positive Tuft Cell Carcinoma. Am J Surg Pathol. 2023;47:183-193. doi:https://doi.org/10.1097/PAS.0000000000001977
- Metovic J, La Salvia A, Rapa I. Molecular Subtypes of Extra-pulmonary Neuroendocrine Carcinomas Identified by the Expression of Neuroendocrine Lineage-Specific Transcription Factors. Endocr Pathol. 2022;33:388-399. doi:https://doi.org/10.1007/s12022-022-09722-4
- Baine M, Hsieh M, Lai W. SCLC Subtypes Defined by ASCL1, NEUROD1, POU2F3, and YAP1: A Comprehensive Immunohistochemical and Histopathologic Characterization. J Thorac Oncol. 2020;15:1823-1835. doi:https://doi.org/10.1016/j.jtho.2020.09.009
- Yamada Y, Simon R, Iwane K. An exploratory study for tuft cells in the breast and their relevance in triple-negative breast cancer: the possible relationship of SOX9. BMC Cancer. 2023;23. doi:https://doi.org/10.1186/s12885-023-10949-5
- Ireland A, Micinski A, Kastner D. MYC Drives Temporal Evolution of Small Cell Lung Cancer Subtypes by Reprogramming Neuroendocrine Fate. Cancer Cell. 2020;38:60-78 e12. doi:https://doi.org/10.1016/j.ccell.2020.05.001
- Lim J, Ibaseta A, Fischer M. Intratumoural heterogeneity generated by Notch signalling promotes small-cell lung cancer. Nature. 2017;545:360-364. doi:https://doi.org/10.1038/nature22323
- Lo Y, Rivera-Concepcion J, Vasmatzis G. Subtype of SCLC Is an Intrinsic and Persistent Feature Through Systemic Treatment. JTO Clin Res Rep. 2023;4. doi:https://doi.org/10.1016/j.jtocrr.2023.100561
- Zhu Y, Li S, Wang H. Molecular subtypes, predictive markers and prognosis in small-cell lung carcinoma. J Clin Pathol. 2024;78:42-50. doi:https://doi.org/10.1136/jcp-2023-209109
- Roy M, Buehler D, Zhang R. Expression of Insulinoma-Associated Protein 1 (INSM1) and Orthopedia Homeobox (OTP) in Tumors with Neuroendocrine Differentiation at Rare Sites. Endocr Pathol. 2019;30:35-42. doi:https://doi.org/10.1007/s12022-018-9559-y
- Pelosi G, Fraggetta F, Pasini F. Immunoreactivity for thyroid transcription factor-1 in stage I non-small cell carcinomas of the lung. Am J Surg Pathol. 2001;25:363-372.
- Horie M, Miyashita N, Mattsson J. An integrative transcriptome analysis reveals a functional role for thyroid transcription factor-1 in small cell lung cancer. J Pathol. 2018;246:154-165. doi:https://doi.org/10.1002/path.5109
- Kawasaki K, Toshimitsu K, Matano M. An Organoid Biobank of Neuroendocrine Neoplasms Enables Genotype-Phenotype Mapping. Cell. 2020;183:1420-1435 e1421. doi:https://doi.org/10.1016/j.cell.2020.10.023
- Wang Z, Liu C, Zheng S. Molecular subtypes of neuroendocrine carcinomas: A cross-tissue classification framework based on five transcriptional regulators. Cancer Cell. 2024;42:1106-1125 e1108. doi:https://doi.org/10.1016/j.ccell.2024.05.002
- Rekhtman N. Lung neuroendocrine neoplasms: recent progress and persistent challenges. Mod Pathol. 2022;35:36-50. doi:https://doi.org/10.1038/s41379-021-00943-2
- Agaimy A, Jain D, Uddin N. SMARCA4-deficient Sinonasal Carcinoma: A Series of 10 Cases Expanding the Genetic Spectrum of SWI/SNF-driven Sinonasal Malignancies. Am J Surg Pathol. 2020;44:703-710. doi:https://doi.org/10.1097/PAS.0000000000001428
- Zagni M, Marando A, Negrelli M. SMARCB1/INI1-deficient undifferentiated tumour of the thorax: a case report and review of the literature. Pathologica. 2024;116:163-169. doi:https://doi.org/10.32074/1591-951X-955
- Pezzuto F, Fortarezza F, Mammana M. Immunohistochemical neuroendocrine marker expression in primary pulmonary NUT carcinoma: a diagnostic pitfall. Histopathology. 2020;77:508-510. doi:https://doi.org/10.1111/his.14166
- Wang Y, Jin Y, Shen X. POU2F3: A Sensitive and Specific Diagnostic Marker for Neuroendocrine-low/negative Small Cell Lung Cancer. Am J Surg Pathol. 2023;47:1059-1066. doi:https://doi.org/10.1097/PAS.0000000000002081
- Ng J, Cai L, Girard L. Molecular and Pathologic Characterization of YAP1-Expressing Small Cell Lung Cancer Cell Lines Leads to Reclassification as SMARCA4-Deficient Malignancies. Clin Cancer Res. 2024;30:1846-1858. doi:https://doi.org/10.1158/1078-0432.CCR-23-2360
- Akbulut D, Whiting K, Teo M. Differential NEUROD1, ASCL1, and POU2F3 Expression Defines Molecular Subsets of Bladder Small Cell/Neuroendocrine Carcinoma with Prognostic Implications. Mod Pathol. Published online 2024. doi:https://doi.org/10.1016/j.modpat.2024.100557
- Baine M, Febres-Aldana C, Chang J. POU2F3 in SCLC: Clinicopathologic and Genomic Analysis With a Focus on Its Diagnostic Utility in Neuroendocrine-Low SCLC. J Thorac Oncol. 2022;17:1109-1121. doi:https://doi.org/10.1016/j.jtho.2022.06.004
- Zhang J, Chen H, Zhang J. Molecular features of gastroenteropancreatic neuroendocrine carcinoma: A comparative analysis with lung neuroendocrine carcinoma and digestive adenocarcinomas. Chin J Cancer Res. 2024;36:90-102. doi:https://doi.org/10.21147/j.issn.1000-9604.2024.01.09
- Modlin I, Bodei L, Kidd M. A historical appreciation of bronchopulmonary neuroendocrine neoplasia: resolution of a carcinoid conundrum. Thorac Surg Clin. 2014;24:235-255. doi:https://doi.org/10.1016/j.thorsurg.2014.05.008
- Travis W, Colby T, Corrin B. Histological Typing of Lung and Pleural Tumours. Springer Verlag; 1999.
- Travis W, Brambilla E, Burke A. WHO Classification of Tumours of the Lung, Pleura, Thymus and Heart. IARC Press; 2015.
- Siegel R, Giaquinto A, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74:12-49. doi:https://doi.org/10.3322/caac.21820
- Wolin E. Advances in the Diagnosis and Management of Well-Differentiated and Intermediate-Differentiated Neuroendocrine Tumors of the Lung. Chest. 2017;151:1141-1146. doi:https://doi.org/10.1016/j.chest.2016.06.018
- Garcia-Yuste M, Matilla J, Cueto A. Typical and atypical carcinoid tumours: analysis of the experience of the Spanish Multi-centric Study of Neuroendocrine Tumours of the Lung. Eur J Cardiothorac Surg. 2007;31:192-197. doi:https://doi.org/10.1016/j.ejcts.2006.11.031
- Prosperi D, Carideo L, Russo V. A Systematic Review on Combined [(18)F]FDG and (68)Ga-SSA PET/CT in Pulmonary Carcinoid. J Clin Med. 2023;12. doi:https://doi.org/10.3390/jcm12113719
- Jiang Y, Hou G, Cheng W. The utility of 18F-FDG and 68Ga-DOTA-Peptide PET/CT in the evaluation of primary pulmonary carcinoid: A systematic review and meta-analysis. Medicine (Baltimore). 2019;98. doi:https://doi.org/10.1097/MD.0000000000014769
- Veschi S, Lattanzio R, Aceto G. Alterations of MEN1 and E-cadherin/beta-catenin complex in sporadic pulmonary carcinoids. Int J Oncol. 2012;41:1221-1228. doi:https://doi.org/10.3892/ijo.2012.1563
- Debelenko L, Brambilla E, Agarwal S. Identification of MEN1 gene mutations in sporadic carcinoid tumors of the lung. Hum Mol Genet. 1997;6:2285-2290. doi:https://doi.org/10.1093/hmg/6.13.2285
- Garcia-Yuste M, Matilla J. The significance of histology: typical and atypical bronchial carcinoids. Thorac Surg Clin. 2014;24:293-297. doi:https://doi.org/10.1016/j.thorsurg.2014.05.003
- Garcia-Yuste M, Matilla J, Alvarez-Gago T. Prognostic factors in neuroendocrine lung tumors: a Spanish Multicenter Study. Spanish Multicenter Study of Neuroendocrine Tumors of the Lung of the Spanish Society of Pneumonology and Thoracic Surgery (EMETNE-SEPAR). Ann Thorac Surg. 2000;70:258-263. doi:https://doi.org/10.1016/s0003-4975(00)01369-2
- Rinke A, Ambrosini V, Dromain C. European Neuroendocrine Tumor Society (ENETS) 2023 guidance paper for colorectal neuroendocrine tumours. J Neuroendocrinol. 2023;35. doi:https://doi.org/10.1111/jne.13309
- Kos-Kudla B, O’Toole D, Falconi M. ENETS consensus guidelines for the management of bone and lung metastases from neuroendocrine tumors. Neuroendocrinology. 2010;91:341-350. doi:https://doi.org/10.1159/000287255
- Bozkurt M, Virgolini I, Balogova S. Guideline for PET/CT imaging of neuroendocrine neoplasms with (68)Ga-DOTA-conjugated somatostatin receptor targeting peptides and (18)F-DOPA. Eur J Nucl Med Mol Imaging. 2017;44:1588-1601. doi:https://doi.org/10.1007/s00259-017-3728-y
- Baudin E, Caplin M, Garcia-Carbonero R. Lung and thymic carcinoids: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up(*). Ann Oncol. 2021;32:439-451. doi:https://doi.org/10.1016/j.annonc.2021.01.003
- Yao J, Shah M, Ito T. Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:514-523. doi:https://doi.org/10.1056/NEJMoa1009290
- Kunz P, Graham N, Catalano P. Randomized Study of Temozolomide or Temozolomide and Capecitabine in Patients With Advanced Pancreatic Neuroendocrine Tumors (ECOG-ACRIN E2211). J Clin Oncol. 2023;41:1359-1369. doi:https://doi.org/10.1200/JCO.22.01013
- Raymond E, Dahan L, Raoul J. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med. 2011;364:501-513. doi:https://doi.org/10.1056/NEJMoa1003825
- Zandee W, Brabander T, Blazevic A. Symptomatic and Radiological Response to 177Lu-DOTATATE for the Treatment of Functioning Pancreatic Neuroendocrine Tumors. J Clin Endocrinol Metab. 2019;104:1336-1344. doi:https://doi.org/10.1210/jc.2018-01991
- Xu J, Shen L, Zhou Z. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020;21:1500-1512. doi:https://doi.org/10.1016/S1470-2045(20)30496-4
- Niederle B, Pape U, Costa F. ENETS Consensus Guidelines Update for Neuroendocrine Neoplasms of the Jejunum and Ileum. Neuroendocrinology. 2016;103:125-138. doi:https://doi.org/10.1159/000443170
- Clift A, Faiz O, Al-Nahhas A. Role of Staging in Patients with Small Intestinal Neuroendocrine Tumours. J Gastrointest Surg. 2016;20:180-188. doi:https://doi.org/10.1007/s11605-015-2953-6
- Hermans B, de Vos-Geelen J, Derks J. Unique Metastatic Patterns in Neuroendocrine Neoplasms of Different Primary Origin. Neuroendocrinology. 2021;111:1111-1120. doi:https://doi.org/10.1159/000513249
- Pelosi G, Rindi G, Travis W. Ki-67 antigen in lung neuroendocrine tumors: unraveling a role in clinical practice. J Thorac Oncol. 2014;9:273-284. doi:https://doi.org/10.1097/JTO.0000000000000092
- Pelosi G, Massa F, Gatti G. Ki-67 Evaluation for Clinical Decision in Metastatic Lung Carcinoids: A Proof of Concept. Clin Pathol. 2019;12. doi:https://doi.org/10.1177/2632010X19829259
- Pelosi G, Papotti M, Rindi G. Unraveling tumor grading and genomic landscape in lung neuroendocrine tumors. Endocr Pathol. 2014;25:151-164. doi:https://doi.org/10.1007/s12022-014-9320-0
- Boland J, Kroneman T, Jenkins S. Ki-67 Labeling Index in Pulmonary Carcinoid Tumors: Comparison Between Small Biopsy and Resection Using Tumor Tracing and Hot Spot Methods. Arch Pathol Lab Med. 2020;144:982-990. doi:https://doi.org/10.5858/arpa.2019-0374-OA
- Fabbri A, Cossa M, Sonzogni A. Ki-67 labeling index of neuroendocrine tumors of the lung has a high level of correspondence between biopsy samples and surgical specimens when strict counting guidelines are applied. Virchows Arch. 2017;470:153-164. doi:https://doi.org/10.1007/s00428-016-2062-2
- Naso J, Jenkins S, Roden A. Prognostic Immunohistochemistry for Ki-67 and OTP on Small Biopsies of Pulmonary Carcinoid Tumors: Ki-67 Index Predicts Progression-free Survival and Atypical Histology. Am J Surg Pathol. 2024;48:742-750. doi:https://doi.org/10.1097/PAS.0000000000002227
- Reuling E, Naves D, Daniels J. Diagnosis of atypical carcinoid can be made on biopsies > 4 mm(2) and is accurate. Virchows Arch. 2022;480:587-593. doi:https://doi.org/10.1007/s00428-022-03279-7
- Ai D, Yao J, Yang F. TRPS1: a highly sensitive and specific marker for breast carcinoma, especially for triple-negative breast cancer. Mod Pathol. 2021;34:710-719. doi:https://doi.org/10.1038/s41379-020-00692-8
- Mengoli M, Rossi G, Cavazza A. Diffuse Idiopathic Pulmonary Neuroendocrine Cell Hyperplasia (DIPNECH) Syndrome and Carcinoid Tumors With/Without NECH: A Clinicopathologic, Radiologic, and Immunomolecular Comparison Study. Am J Surg Pathol. 2018;42:646-655. doi:https://doi.org/10.1097/PAS.0000000000001033
- Viswanathan K, Borczuk A, Siddiqui M. Orthopedia homeobox protein (OTP) is a sensitive and specific marker for primary pulmonary carcinoid tumors in cytologic and surgical specimens. J Am Soc Cytopathol. 2019;8:39-46. doi:https://doi.org/10.1016/j.jasc.2018.09.004
- Naves D, Reuling E, Dickhoff C. In-depth analysis of immunohistochemistry concordance in biopsy-resection pairs of bronchial carcinoids. Ann Diagn Pathol. 2023;67. doi:https://doi.org/10.1016/j.anndiagpath.2023.152181
- Reuling E, Naves D, Kortman P. A Multimodal Biomarker Predicts Dissemination of Bronchial Carcinoid. Cancers (Basel). 2022;14. doi:https://doi.org/10.3390/cancers14133234
- Khazaeli Najafabadi M, Mirzaeian E, Memar Montazerin S. Role of GATA3 in tumor diagnosis: A review. Pathol Res Pract. 2021;226. doi:https://doi.org/10.1016/j.prp.2021.153611
- Xu L, Yu G, Jiang L. HepPar1 and GATA-3 Expression in Neuroendocrine Neoplasms: A Potential Trap for Pathologic Diagnosis. Appl Immunohistochem Mol Morphol. 2023;31:668-672. doi:https://doi.org/10.1097/PAI.0000000000001160
- Sica G, Wagner P, Altorki N. Immunohistochemical expression of estrogen and progesterone receptors in primary pulmonary neuroendocrine tumors. Arch Pathol Lab Med. 2008;132:1889-1895. doi:https://doi.org/10.5858/132.12.1889
- Kimura N, Shiga K, Kaneko K. The Diagnostic Dilemma of GATA3 Immunohistochemistry in Pheochromocytoma and Paraganglioma. Endocr Pathol. 2020;31:95-100. doi:https://doi.org/10.1007/s12022-020-09618-1
- Spada F, Antonuzzo L, Marconcini R. Oxaliplatin-Based Chemotherapy in Advanced Neuroendocrine Tumors: Clinical Outcomes and Preliminary Correlation with Biological Factors. Neuroendocrinology. 2016;103:806-814. doi:https://doi.org/10.1159/000444087
- Engstrom P, Lavin P, Moertel C. Streptozocin plus fluorouracil versus doxorubicin therapy for metastatic carcinoid tumor. J Clin Oncol. 1984;2:1255-1259. doi:https://doi.org/10.1200/JCO.1984.2.11.1255
- Moertel C, Hanley J. Combination chemotherapy trials in metastatic carcinoid tumor and the malignant carcinoid syndrome. Cancer Clin Trials. 1979;2:327-334.
- Sun W, Lipsitz S, Catalano P. Phase II/III study of doxorubicin with fluorouracil compared with streptozocin with fluorouracil or dacarbazine in the treatment of advanced carcinoid tumors: Eastern Cooperative Oncology Group Study E1281. J Clin Oncol. 2005;23:4897-4904. doi:https://doi.org/10.1200/JCO.2005.03.616
- Ferolla P, Berruti A, Spada F. Efficacy and Safety of Lanreotide Autogel and Temozolomide Combination Therapy in Progressive Thoracic Neuroendocrine Tumors (Carcinoid): Results from the Phase 2 ATLANT Study. Neuroendocrinology. 2023;113:332-342. doi:https://doi.org/10.1159/000526811
- Saranga-Perry V, Morse B, Centeno B. Treatment of metastatic neuroendocrine tumors of the thymus with capecitabine and temozolomide: a case series. Neuroendocrinology. 2013;97:318-321. doi:https://doi.org/10.1159/000345938
- Al-Toubah T, Morse B, Strosberg J. Capecitabine and Temozolomide in Advanced Lung Neuroendocrine Neoplasms. Oncologist. 2020;25:e48-e52. doi:https://doi.org/10.1634/theoncologist.2019-0361
- Rubino M, Scoazec J, Pisa E. Lung carcinoids with high proliferative activity: Further support for the identification of a new tumor category in the classification of lung neuroendocrine neoplasms. Lung Cancer. 2020;148:149-158. doi:https://doi.org/10.1016/j.lungcan.2020.08.001
- Pelosi G, Pattini L, Morana G. Grading lung neuroendocrine tumors: Controversies in search of a solution. Histol Histopathol. 2017;32:223-241. doi:https://doi.org/10.14670/HH-11-822
- Rindi G, Klersy C, Inzani F. Grading the neuroendocrine tumors of the lung: an evidence-based proposal. Endocr Relat Cancer. 2014;21:1-16. doi:https://doi.org/10.1530/ERC-13-0246
- Pelosi G, Travis W. The Ki-67 antigen in the new 2021 World Health Organization classification of lung neuroendocrine neoplasms. Pathologica. 2021;113:377-387. doi:https://doi.org/10.32074/1591-951X-542
- Pelosi G, Rodriguez J, Viale G. Typical and atypical pulmonary carcinoid tumor overdiagnosed as small-cell carcinoma on biopsy specimens: a major pitfall in the management of lung cancer patients. Am J Surg Pathol. 2005;29:179-187. doi:https://doi.org/10.1097/01.pas.0000149690.75462.29
- Rekhtman N, Desmeules P, Litvak A. Stage IV lung carcinoids: spectrum and evolution of proliferation rate, focusing on variants with elevated proliferation indices. Mod Pathol. 2019;32:1106-1122. doi:https://doi.org/10.1038/s41379-019-0248-2
- Kasajima A, Kloppel G. Neuroendocrine tumor G3 of bronchopulmonary origin and its classification. Pathol Int. 2022;72:488-495. doi:https://doi.org/10.1111/pin.13266
- Kasajima A, Konukiewitz B, Oka N. Clinicopathological Profiling of Lung Carcinoids with a Ki67 Index > 20. Neuroendocrinology. 2019;108:109-120. doi:https://doi.org/10.1159/000495806
- Pelosi G, Bianchi F, Dama E. Most high-grade neuroendocrine tumours of the lung are likely to secondarily develop from pre-existing carcinoids: innovative findings skipping the current pathogenesis paradigm. Virchows Arch. 2018;472:567-577. doi:https://doi.org/10.1007/s00428-018-2307-3
- Pelosi G, Melocchi V, Dama E. An in-silico analysis reveals further evidence of an aggressive subset of lung carcinoids sharing molecular features of high-grade neuroendocrine neoplasms. Exp Mol Pathol. 2024;135. doi:https://doi.org/10.1016/j.yexmp.2024.104882
- Alcala N, Leblay N, Gabriel A. Integrative and comparative genomic analyses identify clinically relevant pulmonary carcinoid groups and unveil the supra-carcinoids. Nat Commun. 2019;10. doi:https://doi.org/10.1038/s41467-019-11276-9
- Cros J, Theou-Anton N, Gounant V. Specific Genomic Alterations in High-Grade Pulmonary Neuroendocrine Tumours with Carcinoid Morphology. Neuroendocrinology. 2021;111:158-169. doi:https://doi.org/10.1159/000506292
- Simbolo M, Barbi S, Fassan M. Gene Expression Profiling of Lung Atypical Carcinoids and Large Cell Neuroendocrine Carcinomas Identifies Three Transcriptomic Subtypes with Specific Genomic Alterations. J Thorac Oncol. 2019;14:1651-1661. doi:https://doi.org/10.1016/j.jtho.2019.05.003
- Kasajima A, Konukiewitz B, Schlitter A. An analysis of 130 neuroendocrine tumors G3 regarding prevalence, origin, metastasis, and diagnostic features. Virchows Arch. 2022;480:359-368. doi:https://doi.org/10.1007/s00428-021-03202-6
- Centonze G, Maisonneuve P, Simbolo M. Lung carcinoid tumours: histology and Ki-67, the eternal rivalry. Histopathology. 2023;82:324-339. doi:https://doi.org/10.1111/his.14819
- Centonze G, Maisonneuve P, Simbolo M. Ascl1 and OTP tumour expressions are associated with disease-free survival in lung atypical carcinoids. Histopathology. 2023;82:870-884. doi:https://doi.org/10.1111/his.14873
- Clay V, Papaxoinis G, Sanderson B. Evaluation of diagnostic and prognostic significance of Ki-67 index in pulmonary carcinoid tumours. Clin Transl Oncol. 2017;19:579-586. doi:https://doi.org/10.1007/s12094-016-1568-z
- Dermawan J, Farver C. The Role of Histologic Grading and Ki-67 Index in Predicting Outcomes in Pulmonary Carcinoid Tumors. Am J Surg Pathol. 2020;44:224-231. doi:https://doi.org/10.1097/PAS.0000000000001358
- Zahel T, Krysa S, Herpel E. Phenotyping of pulmonary carcinoids and a Ki-67-based grading approach. Virchows Arch. 2012;460:299-308. doi:https://doi.org/10.1007/s00428-012-1194-2
- Oka N, Kasajima A, Konukiewitz B. Classification and Prognostic Stratification of Bronchopulmonary Neuroendocrine Neoplasms. Neuroendocrinology. 2020;110:393-403. doi:https://doi.org/10.1159/000502776
- Swarts D, van Suylen R, den Bakker M. Interobserver variability for the WHO classification of pulmonary carcinoids. Am J Surg Pathol. 2014;38:1429-1436. doi:https://doi.org/10.1097/PAS.0000000000000300
- Manem V, Sazonova O, Gagne A. Unravelling actionable biology using transcriptomic data to integrate mitotic index and Ki-67 in the management of lung neuroendocrine tumors. Oncotarget. 2021;12:209-220. doi:https://doi.org/10.18632/oncotarget.27874
- Pelosi G. Labeling Lung Neuroendocrine Neoplasms for Ki-67 Antigen to Score a Bull’s-Eye, Not Shoot in the Dark. J Thorac Oncol. 2022;17:e41-e44. doi:https://doi.org/10.1016/j.jtho.2021.10.018
- Pelosi G. The Natural History in Lung Neuroendocrine Neoplasms: The Stone Guest Who Matters. J Thorac Oncol. 2022;17:e5-e8. doi:https://doi.org/10.1016/j.jtho.2021.09.018
- Mathian E, Drouet Y, Sexton-Oates A. Assessment of the current and emerging criteria for the histopathological classification of lung neuroendocrine tumours in the lungNENomics project. ESMO Open. 2024;9. doi:https://doi.org/10.1016/j.esmoop.2024.103591
- Laflamme P, Mansoori B, Sazanova O. Phospho-histone-H3 immunostaining for pulmonary carcinoids: impact on clinical appraisal, interobserver correlation, and diagnostic processing efficiency. Hum Pathol. 2020;106:74-81. doi:https://doi.org/10.1016/j.humpath.2020.09.009
- Tsuta K, Liu D, Kalhor N. Using the mitosis-specific marker anti-phosphohistone H3 to assess mitosis in pulmonary neuroendocrine carcinomas. Am J Clin Pathol. 2011;136:252-259. doi:https://doi.org/10.1309/AJCPDXFOPXGEF0RPP
- Buchmann I, Henze M, Engelbrecht S. Comparison of 68Ga-DOTATOC PET and 111In-DTPAOC (Octreoscan) SPECT in patients with neuroendocrine tumours. Eur J Nucl Med Mol Imaging. 2007;34:1617-1626. doi:https://doi.org/10.1007/s00259-007-0450-1
- Zidan L, Iravani A, Kong G. Theranostic implications of molecular imaging phenotype of well-differentiated pulmonary carcinoid based on (68)Ga-DOTATATE PET/CT and (18)F-FDG PET/CT. Eur J Nucl Med Mol Imaging. 2021;48:204-216. doi:https://doi.org/10.1007/s00259-020-04915-7
- Marchetti A, Felicioni L, Pelosi G. Frequent mutations in the neurotrophic tyrosine receptor kinase gene family in large cell neuroendocrine carcinoma of the lung. Hum Mutat. 2008;29:609-616. doi:https://doi.org/10.1002/humu.20707
- De Pas T, Giovannini M, Manzotti M. Large-cell neuroendocrine carcinoma of the lung harboring EGFR mutation and responding to gefitinib. J Clin Oncol. 2011;29:e819-822. doi:https://doi.org/10.1200/JCO.2011.36.2251
- Akhoundova D, Haberecker M, Fritsch R. Targeting ALK in Neuroendocrine Tumors of the Lung. Front Oncol. 2022;12. doi:https://doi.org/10.3389/fonc.2022.911294
- Zhang W, Tian S, Li X. ETV6-NTRK2 Fusion in a Patient With Metastatic Pulmonary Atypical Carcinoid Successfully Treated With Entrectinib: A Case Report and Review of the Literature. Clin Lung Cancer. 2024;25:215-224 e213. doi:https://doi.org/10.1016/j.cllc.2024.03.005
- Werr L, Bartenhagen C, Rosswog C. TERT Expression and Clinical Outcome in Pulmonary Carcinoids. J Clin Oncol. 2025;43:214-225. doi:https://doi.org/10.1200/JCO.23.02708
- Lacroix A, Feelders R, Stratakis C. Cushing’s syndrome. Lancet. 2015;386:913-927. doi:https://doi.org/10.1016/S0140-6736(14)61375-1
- de Jager V, Timens W, Bayle A. Developments in predictive biomarker testing and targeted therapy in advanced stage non-small cell lung cancer and their application across European countries. Lancet Reg Health Eur. 2024;38. doi:https://doi.org/10.1016/j.lanepe.2024.100838
- Volante M, Brizzi M, Faggiano A. Somatostatin receptor type 2A immunohistochemistry in neuroendocrine tumors: a proposal of scoring system correlated with somatostatin receptor scintigraphy. Mod Pathol. 2007;20:1172-1182. doi:https://doi.org/10.1038/modpathol.3800954
- Righi L, Volante M, Tavaglione V. Somatostatin receptor tissue distribution in lung neuroendocrine tumours: a clinicopathologic and immunohistochemical study of 218 ‘clinically aggressive’ cases. Ann Oncol. 2010;21:548-555. doi:https://doi.org/10.1093/annonc/mdp334
- Rufini V, Lorusso M, Inzani F. Correlation of somatostatin receptor PET/CT imaging features and immunohistochemistry in neuroendocrine tumors of the lung: a retrospective observational study. Eur J Nucl Med Mol Imaging. 2022;49:4182-4193. doi:https://doi.org/10.1007/s00259-022-05848-z
- Vatrano S, Giorcelli J, Votta A. Multiple Assays to Determine Methylguanine-Methyltransferase Status in Lung Carcinoids and Correlation with Clinical and Pathological Features. Neuroendocrinology. 2020;110:1-9. doi:https://doi.org/10.1159/000500158
- Kawasaki K, Rekhtman N, Quintanal-Villalonga A. Neuroendocrine neoplasms of the lung and gastrointestinal system: convergent biology and a path to better therapies. Nat Rev Clin Oncol. 2023;20:16-32. doi:https://doi.org/10.1038/s41571-022-00696-0
- Swarts D, Scarpa A, Corbo V. MEN1 Gene Mutation and Reduced Expression Are Associated With Poor Prognosis in Pulmonary Carcinoids. J Clin Endocrinol Metab. 2014;99:E374-378. doi:https://doi.org/10.1210/jc.2013-2782
- Febres-Aldana C, Chang J, Ptashkin R. Rb Tumor Suppressor in Small Cell Lung Cancer: Combined Genomic and IHC Analysis with a Description of a Distinct Rb-Proficient Subset. Clin Cancer Res. 2022;28:4702-4713. doi:https://doi.org/10.1158/1078-0432.CCR-22-1115
- Su P, Chakravarthy K, Furuya N. DLL3-guided therapies in small-cell lung cancer: from antibody-drug conjugate to precision immunotherapy and radioimmunotherapy. Mol Cancer. 2024;23. doi:https://doi.org/10.1186/s12943-024-02012-z
- Patel S, Whang Y, Medley C. Tarlatamab for Large Cell Neuroendocrine Carcinoma in a Young Adult: A Case Report. JTO Clin Res Rep. 2024;5. doi:https://doi.org/10.1016/j.jtocrr.2024.100712
- Silva S, Sousa J, Nogueira C. Relationship between the expressions of DLL3, ASC1, TTF-1 and Ki-67: First steps of precision medicine at SCLC. Oncotarget. 2024;15:750-763. doi:https://doi.org/10.18632/oncotarget.28660
- Choudhury N, Lai W, Makhnin A. A phase I/II study of valemetostat (DS-3201b), an EZH1/2 inhibitor, in combination with irinotecan in patients with recurrent small cell lung cancer. Clin Cancer Res. Published online 2024. doi:https://doi.org/10.1158/1078-0432.CCR-23-3383
- Miyagawa-Hayashino A, Okada S, Takeda-Miyata N. TTF-1 and c-MYC-defined Phenotypes of Large Cell Neuroendocrine Carcinoma and Delta-like Protein 3 Expression for Treatment Selection. Appl Immunohistochem Mol Morphol. 2021;29:313-320. doi:https://doi.org/10.1097/PAI.0000000000000875
- Yang S, Gedvilaite E, Ptashkin R. Microsatellite Instability and Mismatch Repair Deficiency Define a Distinct Subset of Lung Cancers Characterized by Smoking Exposure, High Tumor Mutational Burden, and Recurrent Somatic MLH1 Inactivation. J Thorac Oncol. 2024;19:409-424. doi:https://doi.org/10.1016/j.jtho.2023.10.004
- Sung S, Heymann J, Politis M. Small Biopsy and Cytology of Pulmonary Neuroendocrine Neoplasms: Brief Overview of Classification, Immunohistochemistry, Molecular Profiles, and World Health Organization Updates. Adv Anat Pathol. 2022;29:329-336. doi:https://doi.org/10.1097/PAP.0000000000000360
- Ishiwata T, Iwasawa S, Ebata T. Inhibition of Gli leads to antitumor growth and enhancement of cisplatin-induced cytotoxicity in large cell neuroendocrine carcinoma of the lung. Oncol Rep. 2018;39:1148-1154. doi:https://doi.org/10.3892/or.2018.6183
- Li Y, Shi X, Mao B. The genomic mutational landscape and its correlation with TMB, PD-L1 expression and CD8(+) T cell infiltration in Chinese lung large cell neuroendocrine carcinoma. Lung Cancer. 2022;166:161-169. doi:https://doi.org/10.1016/j.lungcan.2022.01.007
- Simbolo M, Mafficini A, Sikora K. Lung neuroendocrine tumours: deep sequencing of the four World Health Organization histotypes reveals chromatin-remodelling genes as major players and a prognostic role for TERT, RB1, MEN1 and KMT2D. J Pathol. 2017;241:488-500. doi:https://doi.org/10.1002/path.4853
- Gomatou G, Masaoutis C, Vamvakaris I. Differential immunohistochemical expression of hTERT in lung cancer patients with and without idiopathic pulmonary fibrosis. Pulmonology. 2024;30:214-221. doi:https://doi.org/10.1016/j.pulmoe.2021.12.001
- Dowlati A, Abbas A, Chan T. Immune Checkpoint Blockade Outcome in Small-Cell Lung Cancer and Its Relationship With Retinoblastoma Mutation Status and Function. JCO Precis Oncol. 2022;6. doi:https://doi.org/10.1200/PO.22.00257
- La Rosa S, Volante M, Uccella S. ACTH-producing tumorlets and carcinoids of the lung: clinico-pathologic study of 63 cases and review of the literature. Virchows Arch. 2019;475:587-597. doi:https://doi.org/10.1007/s00428-019-02612-x
- Detterbeck F, Woodard G, Bader A. The Proposed 9th Edition TNM Classification of Lung Cancer. Chest. 2024;166:882-895. doi:https://doi.org/10.1016/j.chest.2024.05.026
- Kalemkerian G. Staging and imaging of small cell lung cancer. Cancer Imaging. 2012;11:253-258. doi:https://doi.org/10.1102/1470-7330.2011.0036
- Butnor K, Beasley M, Cagle P. Protocol for the examination of specimens from patients with primary non-small cell carcinoma, small cell carcinoma, or carcinoid tumor of the lung. Arch Pathol Lab Med. 2009;133:1552-1559. doi:https://doi.org/10.5858/133.10.1552
- Nicholson A, Tsao M, Beasley M. The 2021 WHO Classification of Lung Tumors: Impact of Advances Since 2015. J Thorac Oncol. 2022;17:362-387. doi:https://doi.org/10.1016/j.jtho.2021.11.003
- Volante M, Pelosi G. The Spectrum of Neuroendocrine Neoplasia. (Asa S, La Rosa S, Mete O, eds.). Springer; 2021.
- Detterbeck F, Ostrowski M, Hoffmann H. The International Association for the Study of Lung Cancer Lung Cancer Staging Project: Proposals for Revision of the Classification of Residual Tumor After Resection for the Forthcoming (Ninth) Edition of the TNM Classification of Lung Cancer. J Thorac Oncol. 2024;19:1052-1072. doi:https://doi.org/10.1016/j.jtho.2024.03.021
- Rami-Porta R, Nishimura K, Giroux D. The International Association for the Study of Lung Cancer Lung Cancer Staging Project: Proposals for Revision of the TNM Stage Groups in the Forthcoming (Ninth) Edition of the TNM Classification for Lung Cancer. J Thorac Oncol. 2024;19:1007-1027. doi:https://doi.org/10.1016/j.jtho.2024.02.011
- Kim I, Lee G, Choi S. Validation Study for the N Descriptor of the Newly Proposed Ninth Edition of the TNM Staging System Proposed by the International Association for the Study of Lung Cancer. J Thorac Oncol. Published online 2024. doi:https://doi.org/10.1016/j.jtho.2024.04.002
- Fong K, Rosenthal A, Giroux D. The International Association for the Study of Lung Cancer Staging Project for Lung Cancer: Proposals for the Revision of the M Descriptors in the Forthcoming Ninth Edition of the TNM Classification for Lung Cancer. J Thorac Oncol. 2024;19:786-802. doi:https://doi.org/10.1016/j.jtho.2024.01.019
- Travis W, Eisele M, Nishimura K. The International Association for the Study of Lung Cancer (IASLC) Staging Project for Lung Cancer: Recommendation to Introduce Spread Through Air Spaces as a Histologic Descriptor in the Ninth Edition of the TNM Classification of Lung Cancer. Analysis of 4061 Pathologic Stage I NSCLC. J Thorac Oncol. 2024;19:1028-1051. doi:https://doi.org/10.1016/j.jtho.2024.03.015
- Yi C, Dai J, Song N. Improvement of pathological staging system for neuroendocrine tumors of the lung. Ann Transl Med. 2021;9. doi:https://doi.org/10.21037/atm-20-5910
- Liu Y, Yang M, Pang Z. Incidence and prognostic nomogram for resected non-small cell neuroendocrine tumor: A population-based respective study in China and the SEER database. Heliyon. 2023;9. doi:https://doi.org/10.1016/j.heliyon.2023.e15319
- Yoon J, Sigel K, Martin J. Evaluation of the Prognostic Significance of TNM Staging Guidelines in Lung Carcinoid Tumors. J Thorac Oncol. 2019;14:184-192. doi:https://doi.org/10.1016/j.jtho.2018.10.166
- La Salvia A, Marcozzi B, Manai C. Rachel score: a nomogram model for predicting the prognosis of lung neuroendocrine tumors. J Endocrinol Invest. Published online 2024. doi:https://doi.org/10.1007/s40618-024-02346-x
- Hayes A, Luong T, Banks J. Diffuse idiopathic pulmonary neuroendocrine cell hyperplasia (DIPNECH): Prevalence, clinicopathological characteristics and survival outcome in a cohort of 311 patients with well-differentiated lung neuroendocrine tumours. J Neuroendocrinol. 2022;34. doi:https://doi.org/10.1111/jne.13184
- Rossi G, Cavazza A, Spagnolo P. Diffuse idiopathic pulmonary neuroendocrine cell hyperplasia syndrome. Eur Respir J. 2016;47:1829-1841. doi:https://doi.org/10.1183/13993003.01954-2015
- Ramirez R, Cass A, Das S. A multidisciplinary approach to the work up and management of pulmonary carcinoid tumors and DIPNECH: a narrative review. Transl Lung Cancer Res. 2022;11:2567-2587. doi:https://doi.org/10.21037/tlcr-22-415
- Thunnissen E, Borczuk A, Flieder D. The Use of Immunohistochemistry Improves the Diagnosis of Small Cell Lung Cancer and Its Differential Diagnosis. An International Reproducibility Study in a Demanding Set of Cases. J Thorac Oncol. 2017;12:334-346. doi:https://doi.org/10.1016/j.jtho.2016.12.004
- Roy-Chowdhuri S. Molecular Pathology of Lung Cancer. Clin Lab Med. 2024;44:137-147. doi:https://doi.org/10.1016/j.cll.2023.08.002
- Jones K, Churg A, Henderson D. Data set for reporting of lung carcinomas: recommendations from International Collaboration on Cancer Reporting. Arch Pathol Lab Med. 2013;137:1054-1062. doi:https://doi.org/10.5858/arpa.2012-0511-OA
- Klimstra D, Beltran H, Lilenbaum R. The spectrum of neuroendocrine tumors: histologic classification, unique features and areas of overlap. Am Soc Clin Oncol Educ Book. Published online 2015:92-103. doi:https://doi.org/10.14694/EdBook_AM.2015.35.92
- Shackcloth K, Bury D, Dorward D. 20 LCNEC Foundations: Defining Care and Improving UK Outcomes for High Grade Pulmonary Large Cell NeuroEndocrine Carcinomas (LCNEC). Lung Cancer. 2025;200.
- Wang X, Chiang A. Big Decisions on Small Cell Lung Cancer: A Focus on Clinical Care Updates and Patient Perspectives. Am Soc Clin Oncol Educ Book. 2024;44. doi:https://doi.org/10.1200/EDBK_432520
Downloads
License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.
Copyright
Copyright (c) 2024 Società Italiana di Anatomia Patologica e Citopatologia Diagnostica, Divisione Italiana della International Academy of Pathology
How to Cite
- Abstract viewed - 459 times
- PDF downloaded - 115 times