Published online May 24, 2025. doi: 10.5306/wjco.v16.i5.106630
Revised: March 21, 2025
Accepted: April 1, 2025
Published online: May 24, 2025
Processing time: 77 Days and 3.6 Hours
Neuroendocrine neoplasms are a group of tumors with heterogenous malignancy that evolve from neuroendocrine cells, most frequently in the gastrointestinal tract and in the lung. The latest 2021 World Health Organization (WHO) classification of lung tumors defines neuroendocrine neoplasms of the lung as an independent group of tumors, including typical and atypical neuroendocrine tumors and small cell and large cell neuroendocrine carcinomas. Although the overall nomenclature is essentially unchanged from the fourth WHO classification, there are several clinically relevant updates. In this review article, we discuss the epidemiological, clinical, diagnostic, therapeutic and prognostic features of these fascinating neoplasms, including the latest insights, current challenges and future pers
Core Tip: Neuroendocrine neoplasms (NENs) arise from neuroendocrine cells and vary in malignancy, commonly affecting the gastrointestinal tract and lungs. The 2021 World Health Organization classification of lung tumors categorizes lung NENs into typical and atypical neuroendocrine tumors and high-grade small cell and large cell neuroendocrine carcinomas. While the classification remains largely unchanged, key clinical updates have been introduced. This review explores the epidemiology, diagnosis, treatment, and prognosis of lung NENs, highlighting emerging insights, and future directions.
- Citation: Orlandi R. Neuroendocrine neoplasms of the lung: The latest updates. World J Clin Oncol 2025; 16(5): 106630
- URL: https://www.wjgnet.com/2218-4333/full/v16/i5/106630.htm
- DOI: https://dx.doi.org/10.5306/wjco.v16.i5.106630
Lung neuroendocrine neoplasms (LNNs) represent a group of uncommon tumors with multiple grades of malignancy apparently deriving from Kulchitsky cells, which are argentaffin amine precursor uptake and decarboxylation cells of the bronchial epithelium[1]. In 2021, the latest World Health Organization (WHO) classification of lung tumors affirmed LNNs as an independent group of tumors with heterogeneous malignancy[2], as previously established in 2015[3], including low-graded typical carcinoid (TC), intermediate-graded atypical carcinoids (AC), high-graded small cell lung cancer (SCLC) and large cell neuroendocrine carcinoma (LCNEC), as presented in Table 1. The distinction is based on histopathological aspects, the Ki-67 index, mitotic rate, and presence of necrosis. TC is characterized by carcinoid histology, < 2 mitosis/2 mm2, and lack of necrosis. AC has carcinoid histology but 2-10 mitosis/2 mm2 or spotted necrosis. SCLC has characteristic small cells, scant cytoplasm, granular chromatin without nucleoli, and a high mitotic rate > 11/2 mm2 (median 80) with extended necrosis. LCNEC has neuroendocrine morphology with palisades, trabecular cells and organoid clusters, a high mitotic rate > 11/2 mm2 (median 70), and diffuse necrosis. The mitotic count is suggested as the major criterion for discriminating among low-, intermediate-, and high-grade neuroendocrine neoplasms[4]. On the other hand, cautions must be adopted about the Ki-67 rate, which should be applied only for separating TC and AC from high-grade neoplasms[5], due to lack of validated scoring rules in lung tumors: Ki-67 rate is usually below 5% for TC, 10%-20% for AC, over 30% for SCLC or LCNEC. It must be emphasized that low and intermediate graded tumors are biologically distinct entities from high-grade cancers, having distinct genomic profiles[6]. Some LNNs, mainly carcinoids, are thought to have common precursor lesions: Diffuse idiopathic neuroendocrine cell hyperplasia (DIPNECH), which is a generalized proliferation of lung neuroendocrine cells associated with tumor lets and obliterative bronchiolitis[7]. On immunohistochemical evaluation, LNNs are typically positive for chromogranin A, Synaptophysin and cluster of differentiation (CD) 56 (NCAM1)[8]. Insulinoma-associated protein 1 (INSM1) has recently been claimed as useful neuroendocrine marker[9]: It is a nuclear transcription factor, considered a pan-neuroendocrine factor. ASCL1 is another neuroendocrine transcription factor, with high specificity and low sensibility since it is expressed only in a subgroup of LNNs[10]. LNNs represent about 20% of lung primary neoplasia: Carcinoids account for 4% (TC: AC = 10:1), SCLC for 15%, LCNEC for 3%. Since 1907, when Oberndorfer described a carcinoid tumor for the first time[11], several relevant updates have been established on LNNs. In this review article, we discuss the latest epidemiological, histological, clinical, diagnostic, therapeutic and prognostic features of these neoplasms.
Typical carcinoid | Atypical carcinoid | Carcinoid with high mitotic/proliferation index | LCNEC | SCLC | |
Definition | Well-differentiated neuroendocrine morphology, no necrosis, mitotic index < 2 mitoses/2 mm2 | Well-differentiated neuroendocrine morphology, necrosis (punctate) and/or mitotic index 2-10 mitoses/2 mm2 | Well-differentiated morphology, mitotic index > 10 mitoses/2 mm2 and/or Ki-67 > 30% | High-grade NSCLC with neuroendocrine morphology, cytology (prominent nucleoli and/or moderate to abundant cytoplasm) and a mitotic count of > 10 mitoses/2 mm2 | Small cells (usually less than the size of 3 resting lymphocytes) with scant cytoplasm, granular nuclear chromatin with absent or inconspicuous nucleoli and > 10 mitoses/2 mm2 |
Cytology | |||||
Cell size | Variable | Variable | Variable | Large | Small (less than 3 resting lymphocytes) |
Nuclear chromatin | Finely granular texture (salt and pepper) | Finely granular texture (salt and pepper) | Finely granular texture (salt and pepper) | Coarse to vesicular | Finely granular texture, evenly distributed |
Nucleoli | Occasional, small | Common, small | Common, small | Common, large | Absent or inconspicuous |
Cytoplasm | Variable | Variable | Variable | Abundant | Scarce |
Histology | |||||
Morphology | Organoid, trabecular, rosette formation, nested, insular | Organoid, trabecular, rosette formation, nested, insular | Organoid, trabecular, rosette formation, nested, insular | Organoid, trabecular, palisading | Sheet-like, diffuse |
Mitotic count/2 mm2 | 0-1 | 2-10 | > 10 | > 10 (50-60) | > 10 (70-80) |
Proliferation index (Ki-67) | < 10% | 10%-25% | > 30% | 25%-80% | 70%-100% |
Necrosis | Absent | Focal, punctate | Focal, punctate | Extensive | Geographic |
IHC | |||||
Synpatophysin | +++ | +++ | +++ | +++ | +/- |
CgA | +++ | ++ | + | + | +/- |
INSM1 | +++ | +++ | +++ | +++ | +++ |
RB1 | WT | WT | WT | WT/lost | Lost |
P53 | WT | WT | WT | Aberrant | Aberrant |
DIPNECH could be defined as pulmonary disseminated proliferation of neuroendocrine cells, linked to fibrosis and small nodular aggregates < 0.5 cm (tumorlets) without mitoses or necrosis. It is more frequently diagnosed in women, and it is linked to obliterative bronchiolitis. DIPNECH is thought to be the preinvasive form of carcinoid tumors, since disseminated hyperplasia of neuroendocrine cells can be found in up to 25% of resected carcinoids, in the adjacent pulmonary parenchyma[12]. Instead, its relationship with high-grade LNNs appears to be just random, corroborating the hypothesis that carcinoids and high-grade cancers are different entities. Affected patients can either present with chronic cough and shortness of breath, suggestive for constrictive bronchiolitis, or being asymptomatic. In the latest WHO classification, terms clinical and pathologic DIPNECH are introduced, to further define this condition depending on the presence or not of associated clinical manifestations[13]. Pathologic evaluation of lung sample allows for definite diagnosis, nevertheless its appearance on high-resolution computed tomography (CT) scan could be pathognomonic: Multiple punctate nodules, airway thickening and mosaic attenuation[14]. Management of DIPNECH is unclear, but considering the potential evolution to carcinoids, most authors suggest annual CT scan follow-up.
Lung carcinoids are a rare subset of lung tumors, accounting for about 2% of all lung tumors and 10%-20% of carcinoids found throughout the body[15]. Their incidence varies between 0.2 to 2 cases per 100000 individuals, and interestingly, their prevalence has been on the rise, showing an increase of up to 6% per year[16]. This increase is likely due to advancements in imaging and histological identification. Lung carcinoids are typically diagnosed in individuals aged 40 to 60, though they are the most common primary lung neoplasia in the first and second decades of life[17]. Most lung carcinoids are sporadic, with about 5% associated with multiple endocrine neoplasia type 1 (MEN1). Rarely, they occur in hereditary clusters known as familial pulmonary carcinoid tumors. Despite their slow-growing nature, lung carcinoids are not benign tumors. They are usually diagnosed at an early stage, with TC being more common than AC at a ratio of 10:1[18]. TCs rarely spread beyond the lungs, whereas ACs have a higher tendency to metastasize, particularly to the liver, bones, and brain.
Clinically, patients with lung carcinoids are often asymptomatic, with the tumors being discovered incidentally during chest imaging. When symptoms do occur, central carcinoids can cause cough, dyspnea, hemoptysis, and recurrent pulmonary infections[19]. In less than 10% of cases, symptoms may result from ectopic hormonal activity of the tumor, leading to syndromes such as the syndrome of inappropriate antidiuretic hormone secretion, Cushing syndrome due to adrenocorticotropic hormone hypersecretion, or carcinoid syndrome from serotonin hypersecretion, which causes skin flushing, diarrhea, dyspnea, and tachycardia[20].
The diagnostic workup typically begins with a routine chest radiograph, followed by contrast-enhanced CT scans and 18F-fluorodeoxyglucose positron emission tomography (PET)-CT to define the primary lesion and exclude metastasis[21]. Promising imaging methods include single-photon emission computed tomography-CT with 99mTc-Tektrotyd tracer for somatostatin receptors and 68Ga-Edotreotide PET, which is more sensitive for low-grade carcinoids[22]. Flexible bronchoscopy may be performed to assess bronchial involvement and collect samples for histopathological diagnosis.
Morphologically, carcinoids display uniform round or plasmacytoid cells in a nesting or organoid pattern, with granular or speckled chromatin and thin or absent nucleoli[23]. Immunohistochemistry aids diagnosis, with carcinoids typically testing positive for chromogranin A, synaptophysin, cluster of differentiation (CD) 56, and INSM1[24]. Thyroid transcription factor-1 (TTF-1) can be positive in fewer than half of cases. Orthopedia homeobox protein (OTP) is particularly useful for distinguishing primary lung carcinoids from metastatic ones as it is almost exclusively expressed in pulmonary carcinoids[25].
The mitotic count differentiates TCs from ACs: TCs have fewer than 2 mitoses/mm², while ACs have between 2 and 10 mitoses/mm². The role of the Ki67 rate is still debated, although it is considered a strong prognostic marker[26]. Generally, 10 mitoses/mm² and a Ki67 rate of 20%-30% are accepted thresholds for diagnosing carcinoids. Recently, cases of highly proliferative carcinoids with higher mitotic counts and Ki67 rates have been reported[27], classified as LCNEC with carcinoid morphology in the latest WHO classification[2]. The growing importance of Ki67 is evident in the latest WHO thoracic guidelines, which, while not requiring it for distinguishing typical from ACs, recommend its inclusion as a supplemental marker. Research confirms that, like mitotic counts, Ki67 is a strong prognostic indicator in carcinoids. Findings suggest that a Ki67 index above 5% in TCs and over 10% overall is linked to worse outcomes. Although classification relies on mitotic counts, Ki67 levels of 10% or higher are almost exclusively seen in ACs, warranting a careful mitotic recount in cases of discrepancy.
Surgical resection remains the treatment of choice for both TCs and ACs, ensuring a 5-year survival rate of 90% for TCs and 70% for ACs[28]. Due to their low proliferative index, standard chemotherapeutics are generally ineffective[29]. For advanced or metastatic carcinoids, especially those associated with carcinoid syndrome, somatostatin analogues are the first line of therapy[30]. In cases of disease progression, peptide receptor radionuclide therapy or targeted therapy with everolimus may be beneficial[31]. In localized carcinoids, radical surgical excision should always be pursued when feasible[32]. Other loco-regional treatments, such as endobronchial resection, stereotactic radiotherapy, radiofrequency, or microwave ablations, are reserved for patients with undue surgical risk, those who refuse surgery, or as ancillary perioperative methods. High rates of local relapse have been reported after ablation, making anatomical lung resection and systematic lymphadenectomy the preferred approach in patients with adequate cardio-respiratory reserve. Given the indolent behavior of TCs, some authors advocate for sub-lobar resections without radical lymphadenectomy, particularly for TCs[33]. For stage I TCs, radical anatomical resection (R0) through lobectomy is recommended. However, sublobar resection may be acceptable in patients with peripheral TCs smaller than 2 cm. While anatomical resections are still preferred by some surgeons due to the propensity of TCs to metastasize to lymph nodes (LNs), segmentectomy has been suggested over wedge resection for better oncological outcomes. Approximately two-thirds of all carcinoid tumors arise in the major airways, with about 20% presenting as purely intraluminal endobronchial lesions. These tumors can be managed with bronchoplastic resections to preserve lung function without compromising oncological results[34]. Endoscopic techniques such as Nd-YAG laser, diathermy, and cryosurgery have been largely abandoned due to high recurrence rates and the risk of extraluminal spread[35]. Bronchoplastic and parenchyma-saving procedures are viable options for highly selected patients with typical endobronchial carcinoids. The prognostic impact of LN involvement has been well documented, particularly for ACs[36]. Radical lymphadenectomy is highly recommended for ACs, while for TCs, LN sampling may be sufficient due to the lower incidence of nodal metastases. Current guidelines recommend at least LN sampling or dissection during resection for NETs.
While lung carcinoids have traditionally been considered a single pathological entity, recent multi-omic studies indicate they consist of three distinct biological subtypes[37]. These subtypes are defined by differences in gene expression, methylation, and mutation profiles. One subtype, characterized by MEN1 mutations and low OTP expression (Cluster B), is linked to poor prognosis, consistent with prior research on these markers[38]. Another small subset, termed “supra-carcinoids”, exhibits carcinoid-like morphology but shares molecular and clinical traits with neuroendocrine carcinomas, requiring further investigation[38]. While these findings offer insight into the biology and potential origins of lung carcinoids, their impact on diagnosis and clinical management remains uncertain.
Lung carcinoids, while rare, are a significant subset of lung tumors that require precise diagnostic and treatment strategies. Their slow-growing nature and indolent behavior do not preclude their potential for metastasis and recurrence. Surgical resection remains the cornerstone of treatment, with radical excision and systematic lymphadenectomy offering the best outcomes. Advances in imaging and histological techniques have improved the detection and management of these tumors, but ongoing research and clinical trials are essential to refine treatment protocols and improve patient survival rates.
SCLC is a highly aggressive form of lung cancer, representing more than 90% of small cell cancers in the body. It accounts for approximately 10%-15% of all lung cancers[39]. It is more prevalent among older adults, with the highest incidence occurring in individuals aged 65 years and older. Historically, SCLC has been more common in men than in women. However, this gap has been narrowing in recent years. The proportion of SCLC among newly diagnosed lung cancer cases dropped from 14.5% in 2000 to 11.8% in 2019. This decline occurred across all sex and racial groups but was more pronounced and began earlier in men than in women. As a result, the male-to-female ratio shifted from 1.14:1 in 2000 to 0.93:1 in 2019[40]. Smoking is the primary risk factor for SCLC[41]. Approximately 95% of cases are attributed to tobacco smoke, either through active smoking or exposure to secondhand smoke. The incidence of SCLC varies geographically, reflecting differences in smoking prevalence and exposure to environmental carcinogens[42]. Certain lung diseases, such as chronic obstructive pulmonary disease, are associated with an increased risk of developing SCLC[43]. There are some ethnic and racial disparities in the incidence of SCLC. For example, African American men have historically had higher rates of SCLC compared to other racial groups in the United States[44]. The overall incidence of SCLC has been declining over the past few decades, largely due to decreases in smoking rates and public health efforts aimed at tobacco control.
Traditionally, SCLC has been a morphological diagnosis, but confirmatory immunohistochemistry has become more common to improve diagnostic accuracy[45]. The latest WHO classification recommends immunohistochemistry for excluding alternative diagnoses rather than as a requirement for SCLC diagnosis. SCLC is characterized by small cell size, typically smaller than three lymphocytes, with finely dispersed chromatin, scant cytoplasm, and fragile nuclei that exhibit molding. The presence of DNA streaming and encrustation of vessels (Azzopardi effect) are notable features[46]. Extensive necrosis and high mitotic and apoptotic rates are also common. While most SCLCs display a diffuse or sheet-like pattern, some may show a nested or organoid architecture. Concerning immunohistochemistry, CD56 is positive in most SCLC cases, though it can also be expressed in non-neuroendocrine neoplasms like hematolymphoid neoplasms[47]; Synaptophysin and chromogranin A are negative in up to 20% of SCLC cases; INSM1 is useful for diagnosing SCLC lacking other marker expressions but it is also present in non-neuroendocrine neoplasms such as sarcomas or adenoid cystic carcinomas[48]. Genomically, SCLC is relatively homogeneous, typically characterized by alterations in RB1 and TP53[49]. However, distinct subtypes exist based on global differences in gene expression and methylation profiles, primarily defined by master transcriptional regulators[50]: ASCL1 dominates in approximately 70% of SCLC cases, associated with the classic, chemosensitive type; NEUROD1 is linked with high levels of neuroendocrine marker expression; POU2F3 represents neuroendocrine-negative SCLC, accounting for about 10% of cases.
SCLC is typically treated with chemotherapy, with cisplatin and etoposide being the standard regimen[51]. Despite initial responsiveness, recurrence rates are high, up to 30%-50%. The prognosis for SCLC remains poor, with a 2-year survival rate of around 5% and median survival ranging from 15 to 20 months[52]. Surgery is considered only for early-stage SCLC (T1-2N0M0)[53]. Studies have shown 5-year survival rates of 50% for patients with stage I SCLC undergoing lobectomy. For non-metastatic SCLC, combined modality treatments, including chemotherapy and radiotherapy, can achieve 5-year survival rates of 25%-30%[54]. Prophylactic cranial irradiation is recommended for patients responding to initial treatment to reduce the risk of brain metastases[55]. Recent advancements include the addition of immune checkpoint inhibitors to first-line chemotherapy, improving progression-free and overall survival[56,57]. However, the response to T cell checkpoint blockade remains limited to about 15% of patients. Despite advances in treatment, SCLC continues to be associated with a poor prognosis due to its aggressive nature and high recurrence rates. There is a pressing need for novel therapeutic approaches and biomarkers to guide clinical decisions. Ongoing research into the molecular characteristics of SCLC and the development of targeted therapies hold promise for improving patient outcomes.
Significant advances in SCLC research are shaping new investigative paths and offering hope for patients. Genetically engineered mouse models, patient-derived models, and primary tumor studies have deepened understanding of SCLC biology. Transcriptomic and proteomic analyses have identified potential tumor-specific vulnerabilities and mechanisms of therapy resistance[58]. Defining transcriptional drivers has helped classify SCLC subtypes, paving the way for targeted therapies[59]. Technological improvements in imaging and radiotherapy have enhanced survival for localized cases while reducing side effects. Immunotherapy has modestly improved survival in metastatic SCLC, marking progress but highlighting the need for more effective treatments[59]. Screening for early-stage SCLC remains a critical unmet need, as most cases are diagnosed at an advanced stage. SCLC exhibits significant heterogeneity, with studies identifying distinct subtypes that may originate from different lung epithelial cells. Epigenetic mutations play a role in transcriptional shifts, but their impact on prognosis and therapy response remains unclear. Tumor heterogeneity, including a mix of neuroendocrine and non-neuroendocrine cells, may influence metastasis, but its role in human SCLC is not well defined. While chemo-immunotherapy is now the first-line standard for metastatic disease, its benefits are limited to a small subset of patients[60]. Research is focusing on enhancing immune response through alternative checkpoint inhibitors, bispecific T-cell engagers, and DNA damage response inhibitors. Efforts are also underway to integrate immunotherapy into early-stage treatment alongside chemoradiotherapy. New biomarker-driven approaches, such as circulating tumor DNA analysis, could help identify patients needing additional therapy vs those likely to be cured with current treatments[61]. Despite progress, clinical advances in SCLC lag behind non-small cell lung cancers (NSCLC), and many questions remain. However, continued translation of laboratory discoveries into clinical trials is expected to drive meaningful improvements in patient outcomes.
SCLC is a highly aggressive and challenging form of lung cancer with distinct morphological and genomic characteristics. Advances in diagnostic techniques and treatment approaches have improved patient management, but the prognosis remains poor. Continued research into the molecular underpinnings of SCLC and the development of targeted therapies are crucial for improving outcomes for patients with this devastating disease.
LCNEC is a rare but aggressive form of NSCLC that exhibits neuroendocrine differentiation, comprising approximately 3%-5% of all lung cancers[62]. It is less common than SCLC but more aggressive than typical NSCLC. LCNEC predominantly affects older adults, with a median age at diagnosis typically between 60 to 70 years. It shows a slight male predominance. Smoking is a significant risk factor for LCNEC. The majority of patients with LCNEC have a history of tobacco use[63]. There may be some geographic and ethnic variations in the incidence of LCNEC, but data specific to these variations are limited due to its rarity. Underlying lung diseases such as COPD may increase the risk of developing LCNEC.
LCNEC is defined by its unique blend of neuroendocrine morphology and non-small cell carcinoma cytologic features[64]. Key morphological characteristics include neuroendocrine architecture (organoid nesting with palisading, trabeculae, and rosettes), cytologic features (large cell size, prominent nucleoli, and abundant cytoplasm), high proliferation rate (greater than 10 mitoses per 2 mm², with median values often reaching around 70). The diagnosis of LCNEC requires the expression of at least one of the three standard neuroendocrine markers[65]: Synaptophysin, chromogranin A, or CD56. Newer markers such as INSM1 and ASCL1 show promise but their roles in diagnosing LCNEC remain to be fully clarified. Although not part of the WHO criteria for diagnosis, Ki67 is helpful in distinguishing LCNEC from carcinoids, particularly in biopsies with crush artifacts. Ki67 rates in LCNEC vary widely, typically between 70%-100%, but can overlap with highly proliferative carcinoids in the 20%-60% range[66].
LCNEC exhibits a wide morphological spectrum, spanning two relatively distinct extremes: NSCLC-like LCNEC and SCLC-like LCNEC[67]. The formers have large cells, abundant cytoplasm, and prominent nucleoli, combined with neuroendocrine features like nesting and rosettes. These tumors often resemble NSCLC entities such as adenocarcinoma with solid or cribriform patterns, large cell carcinoma, and basaloid squamous cell carcinoma. Characterized by alterations typical of smoking-associated adenocarcinomas, such as mutations in STK11, KEAP1, and KRAS, and the absence of RB1 alterations[68]. These tumors are also associated with chemoresistance and aggressive behavior. The latter’s exhibit nuclear features similar to SCLC, such as small or intermediate cell size and coarsely granular chromatin, but with more prominent nucleoli or more abundant cytoplasm. The presence of visible intercellular membranes is a useful criterion to support the diagnosis of LCNEC, as SCLC typically lacks these due to scant cytoplasm[69]. Characterized by RB1 and TP53 alterations, similar to SCLC. These tumors often exhibit aggressive and treatment-resistant behavior. Diagnosing LCNEC can be challenging, particularly in small or poorly preserved biopsy specimens. However, recent trends in obtaining larger tissue volumes for molecular testing have made biopsy diagnosis more feasible. A semiquantitative scoring system has been proposed to aid diagnosis in biopsies, considering factors like neuroendocrine differentiation, necrosis, Ki67 rate, and neuroendocrine marker positivity[70].
LCNEC is associated with an extremely poor prognosis and high rates of brain metastases. The treatment approach to LCNEC has been uncertain, with both SCLC-type and NSCLC-type systemic therapies used in practice[71]. Recent studies suggest that molecular subtype, particularly the status of RB1, can influence treatment outcomes and prognosis. Surgery is the recommended treatment for early-stage, resectable LCNEC, achieving 5-year survival rates of 27%-67%[63]. However, a high incidence of recurrence, especially within the first 2 years, necessitates a multimodal approach, including adjuvant chemotherapy[72]. Lobar resection is preferred over sublobar resections for improved survival outcomes in stage I LCNEC. For advanced or metastatic LCNEC, the optimal systemic treatment remains debated. Platinum-based chemotherapy regimens used for SCLC (e.g., etoposide and cisplatin) are often recommended, although LCNEC generally shows lower chemosensitivity than SCLC[73]. Recent data suggest that RB1 expression could serve as a potential biomarker for selecting the appropriate treatment regimen[74]: RB1 loss indicates SCLC-type chemotherapy, while intact RB1 suggests NSCLC-type treatments. The role of immunotherapy and other targeted treatments in LCNEC remains under investigation[75-77].
The diagnosis of LCNEC requires differentiation from NSCLC and SCLC, given its molecular overlap with both. This continuum can create diagnostic challenges, particularly in tumors with ambiguous features. Neuroendocrine marker expression alone is insufficient for diagnosing LCNEC, as some NSCLCs also express these markers. However, most LCNECs are positive for two or more standard neuroendocrine markers, whereas conventional NSCLC typically expresses only one, often focally[78]. Certain poorly differentiated tumors, such as SMARCA4-deficient undifferentiated tumors, can also express NE markers, complicating the distinction. Other immunohistochemical (IHC) markers provide limited help. Strong, diffuse Napsin A expression favors adenocarcinoma, but weak/focal expression occurs in about 15% of LCNECs[79]. A TTF-1-positive/Napsin A-negative profile should raise suspicion for neuroendocrine carcinoma, as most TTF-1-positive adenocarcinomas also strongly express Napsin A. The distinction between SCLC and LCNEC is primarily morphologic, based on cell size, nucleoli prominence, and cytoplasmic volume. SCLC typically lacks visible cell membranes and often presents with crush artifacts and extensive necrosis, making diagnosis difficult on small biopsies[80]. In such cases, high-quality hematoxylin-eosin stains and cytologic preparations can be helpful. A tumor with any identifiable SCLC morphology qualifies as combined SCLC-LCNEC rather than pure LCNEC. No definitive IHC markers exist for this distinction, but Napsin A expression supports LCNEC, as it is consistently negative in SCLC, though with low sensitivity. Dot-like keratin reactivity, commonly seen in SCLC, can also occur in LCNEC. Rb protein retention is more frequent in LCNEC than SCLC but lacks absolute specificity.
LCNEC is a highly aggressive and challenging form of lung cancer with distinct morphological and molecular characteristics. Advances in diagnostic techniques and molecular profiling have improved our understanding of LCNEC and its subtypes. However, the prognosis for LCNEC remains poor, highlighting the need for continued research into novel therapeutic approaches and biomarkers to guide clinical decisions. Multimodal treatment strategies, including surgery and systemic therapies tailored to molecular subtypes, offer the best hope for improving outcomes in this rare and aggressive malignancy.
Lung neuroendocrine tumors (LNETs) exhibit a range of common characteristics yet remain heterogeneous in terms of aggressiveness, progression, and prognosis. This diversity underscores the ongoing controversies surrounding their diagnosis and treatment across different subtypes. Recent molecular advancements hold promise for the development of novel diagnostic, prognostic, and predictive tools soon. These advancements may also pave the way for innovative treatment strategies tailored to the specific molecular profiles of LNETs. SCLC continues to pose significant challenges with high mortality rates. Current treatments have demonstrated limited efficacy, reflecting gaps in our understanding of the disease’s underlying mechanisms. LCNEC lacks specific therapeutic guidelines supported by prospective clinical trials. Despite efforts to optimize chemotherapy regimens, prognosis remains unfavorable, particularly in comparison to NSCLC. A multidisciplinary approach is imperative for accurate diagnosis and effective multimodal treatment planning in LNETs, aiming to improve patient outcomes through integrated care strategies. The urgent need for paradigm shifts in the management of LNETs underscores the importance of ongoing research and collaborative efforts to address current limitations and improve therapeutic outcomes for affected patients.
1. | Yellin A, Benfield JR. The pulmonary Kulchitsky cell (neuroendocrine) cancers: from carcinoid to small cell carcinomas. Curr Probl Cancer. 1985;9:1-38. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 10] [Article Influence: 0.3] [Reference Citation Analysis (0)] |
2. | WHO Classification of Tumours Editorial Board. Thoracic Tumours, 5th ed. International Agency for Research on Cancer: Lyon, France, 2021. |
3. | Travis WD, Brambilla E, Nicholson AG, Yatabe Y, Austin JHM, Beasley MB, Chirieac LR, Dacic S, Duhig E, Flieder DB, Geisinger K, Hirsch FR, Ishikawa Y, Kerr KM, Noguchi M, Pelosi G, Powell CA, Tsao MS, Wistuba I; WHO Panel. The 2015 World Health Organization Classification of Lung Tumors: Impact of Genetic, Clinical and Radiologic Advances Since the 2004 Classification. J Thorac Oncol. 2015;10:1243-1260. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2160] [Cited by in RCA: 3058] [Article Influence: 339.8] [Reference Citation Analysis (0)] |
4. | Nicholson AG, Tsao MS, Beasley MB, Borczuk AC, Brambilla E, Cooper WA, Dacic S, Jain D, Kerr KM, Lantuejoul S, Noguchi M, Papotti M, Rekhtman N, Scagliotti G, van Schil P, Sholl L, Yatabe Y, Yoshida A, Travis WD. The 2021 WHO Classification of Lung Tumors: Impact of Advances Since 2015. J Thorac Oncol. 2022;17:362-387. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 57] [Cited by in RCA: 647] [Article Influence: 161.8] [Reference Citation Analysis (0)] |
5. | Dermawan JKT, Farver CF. The Role of Histologic Grading and Ki-67 Index in Predicting Outcomes in Pulmonary Carcinoid Tumors. Am J Surg Pathol. 2020;44:224-231. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 20] [Cited by in RCA: 21] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
6. | Clinical Lung Cancer Genome Project (CLCGP); Network Genomic Medicine (NGM). A genomics-based classification of human lung tumors. Sci Transl Med. 2013;5:209ra153. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 125] [Cited by in RCA: 242] [Article Influence: 22.0] [Reference Citation Analysis (0)] |
7. | Ramirez RA, Cass AS, Das S, Low SW, Mehrad M, Rickman OB, Scherer PM, Thomas KE, Gillaspie EA. 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. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 11] [Cited by in RCA: 10] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
8. | Rekhtman N. Lung neuroendocrine neoplasms: recent progress and persistent challenges. Mod Pathol. 2022;35:36-50. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 95] [Cited by in RCA: 110] [Article Influence: 36.7] [Reference Citation Analysis (0)] |
9. | Yatabe Y, Dacic S, Borczuk AC, Warth A, Russell PA, Lantuejoul S, Beasley MB, Thunnissen E, Pelosi G, Rekhtman N, Bubendorf L, Mino-Kenudson M, Yoshida A, Geisinger KR, Noguchi M, Chirieac LR, Bolting J, Chung JH, Chou TY, Chen G, Poleri C, Lopez-Rios F, Papotti M, Sholl LM, Roden AC, Travis WD, Hirsch FR, Kerr KM, Tsao MS, Nicholson AG, Wistuba I, Moreira AL. Best Practices Recommendations for Diagnostic Immunohistochemistry in Lung Cancer. J Thorac Oncol. 2019;14:377-407. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 201] [Cited by in RCA: 209] [Article Influence: 34.8] [Reference Citation Analysis (0)] |
10. | Ye B, Cappel J, Findeis-Hosey J, McMahon L, Yang Q, Xiao GQ, Xu H, Li F. hASH1 is a specific immunohistochemical marker for lung neuroendocrine tumors. Hum Pathol. 2016;48:142-147. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 15] [Cited by in RCA: 15] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
11. | Modlin IM, Shapiro MD, Kidd M. Siegfried Oberndorfer: origins and perspectives of carcinoid tumors. Hum Pathol. 2004;35:1440-1451. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 68] [Cited by in RCA: 62] [Article Influence: 3.1] [Reference Citation Analysis (0)] |
12. | Miller RR, Müller NL. Neuroendocrine cell hyperplasia and obliterative bronchiolitis in patients with peripheral carcinoid tumors. Am J Surg Pathol. 1995;19:653-658. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 111] [Cited by in RCA: 119] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
13. | Vocino Trucco G, Righi L, Volante M, Papotti M. Updates on lung neuroendocrine neoplasm classification. Histopathology. 2024;84:67-85. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 1] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
14. | Foran PJ, Hayes SA, Blair DJ, Zakowski MF, Ginsberg MS. Imaging appearances of diffuse idiopathic pulmonary neuroendocrine cell hyperplasia. Clin Imaging. 2015;39:243-246. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 8] [Article Influence: 0.8] [Reference Citation Analysis (0)] |
15. | Swarts DR, Ramaekers FC, Speel EJ. Molecular and cellular biology of neuroendocrine lung tumors: evidence for separate biological entities. Biochim Biophys Acta. 2012;1826:255-271. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 31] [Cited by in RCA: 78] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
16. | Modlin IM, Lye KD, Kidd M. A 5-decade analysis of 13,715 carcinoid tumors. Cancer. 2003;97:934-959. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1848] [Cited by in RCA: 1844] [Article Influence: 83.8] [Reference Citation Analysis (1)] |
17. | Savu C, Melinte A, Diaconu C, Stiru O, Gherghiceanu F, Tudorica ȘDO, Dumitrașcu OC, Bratu A, Balescu I, Bacalbasa N. Lung neuroendocrine tumors: A systematic literature review (Review). Exp Ther Med. 2022;23:176. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 2] [Article Influence: 0.5] [Reference Citation Analysis (0)] |
18. | Caplin ME, Baudin E, Ferolla P, Filosso P, Garcia-Yuste M, Lim E, Oberg K, Pelosi G, Perren A, Rossi RE, Travis WD; ENETS consensus conference participants. 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 514] [Cited by in RCA: 453] [Article Influence: 45.3] [Reference Citation Analysis (0)] |
19. | Detterbeck FC. Clinical presentation and evaluation of neuroendocrine tumors of the lung. Thorac Surg Clin. 2014;24:267-276. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 25] [Cited by in RCA: 30] [Article Influence: 2.7] [Reference Citation Analysis (0)] |
20. | Savu C, Melinte A, Lukadi JL, Mirvald C, Savu C, Belu E, Diaconu C, Iliescu L, Balescu I, Stiru O, Bratu O, Gorecki G, Bacalbasa N. Neuroendocrine syndrome in bronchial carcinoid tumors. Exp Ther Med. 2020;20:200. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 3] [Article Influence: 0.6] [Reference Citation Analysis (0)] |
21. | Jeung MY, Gasser B, Gangi A, Charneau D, Ducroq X, Kessler R, Quoix E, Roy C. Bronchial carcinoid tumors of the thorax: spectrum of radiologic findings. Radiographics. 2002;22:351-365. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 143] [Cited by in RCA: 129] [Article Influence: 5.6] [Reference Citation Analysis (0)] |
22. | Tripathy S, Ballal S, Yadav MP, Joshi P, Bal C, Damle NA. Coexistence of Multiple Metastatic Lesions Showing Various Grades of Differentiation in a Single Patient with Neuroendocrine Tumor of Lung as Primary: A Combined Modality Approach of (68)Ga-DOTANOC Positron Emission Tomography/Computed Tomography and (18)F-Fluorodeoxyglucose Positron Emission Tomography/Computed Tomography Imaging. Indian J Nucl Med. 2019;34:260-262. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 2] [Article Influence: 0.3] [Reference Citation Analysis (0)] |
23. | Metovic J, Barella M, Bianchi F, Hofman P, Hofman V, Remmelink M, Kern I, Carvalho L, Pattini L, Sonzogni A, Veronesi G, Harari S, Forest F, Papotti M, Pelosi G. Morphologic and molecular classification of lung neuroendocrine neoplasms. Virchows Arch. 2021;478:5-19. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 20] [Cited by in RCA: 36] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
24. | Travis WD, Brambilla E, Nicholson AG. Testing for Neuroendocrine Immunohistochemical Markers Should Not Be Performed in Poorly Differentiated NSCCs in the Absence of Neuroendocrine Morphologic Features according to the 2015 WHO Classification. J Thorac Oncol. 2016;11:e26-e27. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 12] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
25. | Yoxtheimer LM, Heymann JJ, Cohen C, Rao RA, Goyal A, Siddiqui MT. Immunohistochemical analysis of OTP and NKX6.1 in neuroendocrine tumors of the lung and pancreas. Diagn Cytopathol. 2018;46:1010-1014. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 7] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
26. | Warth A, Fink L, Fisseler-Eckhoff A, Jonigk D, Keller M, Ott G, Rieker RJ, Sinn P, Söder S, Soltermann A, Willenbrock K, Weichert W; Pulmonary Pathology Working Group of the German Society of Pathology. Interobserver agreement of proliferation index (Ki-67) outperforms mitotic count in pulmonary carcinoids. Virchows Arch. 2013;462:507-513. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 51] [Cited by in RCA: 48] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
27. | Marchiò C, Gatti G, Massa F, Bertero L, Filosso P, Pelosi G, Cassoni P, Volante M, Papotti M. Distinctive pathological and clinical features of lung carcinoids with high proliferation index. Virchows Arch. 2017;471:713-720. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 52] [Cited by in RCA: 66] [Article Influence: 8.3] [Reference Citation Analysis (0)] |
28. | Soldath P, Petersen RH. The Surgical Management of Lung Neuroendocrine Neoplasms. Cancers (Basel). 2023;15. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
29. | Wirth LJ, Carter MR, Jänne PA, Johnson BE. Outcome of patients with pulmonary carcinoid tumors receiving chemotherapy or chemoradiotherapy. Lung Cancer. 2004;44:213-220. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 76] [Cited by in RCA: 73] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
30. | Baudin E, Capdevila J, Hörsch D, Singh S, Caplin ME, Wolin EM, Buikhuisen W, Raderer M, Dansin E, Grohe C, Ferone D, Houchard A, Truong-Thanh XM, Reidy-Lagunes D. Treatment of advanced BP-NETS with lanreotide autogel/depot vs placebo: the phase III SPINET study. Endocr Relat Cancer. 2024;31. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
31. | Yao JC, Fazio N, Singh S, Buzzoni R, Carnaghi C, Wolin E, Tomasek J, Raderer M, Lahner H, Voi M, Pacaud LB, Rouyrre N, Sachs C, Valle JW, Fave GD, Van Cutsem E, Tesselaar M, Shimada Y, Oh DY, Strosberg J, Kulke MH, Pavel ME; RAD001 in Advanced Neuroendocrine Tumours, Fourth Trial (RADIANT-4) Study Group. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016;387:968-977. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 749] [Cited by in RCA: 884] [Article Influence: 98.2] [Reference Citation Analysis (0)] |
32. | Soldath P, Binderup T, Kjær A, Federspiel B, Langer SW, Knigge U, Petersen RH. Long-term survival and recurrence after resection of bronchopulmonary carcinoids: A single-center cohort study of 236 patients. Lung Cancer. 2021;156:109-116. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 13] [Article Influence: 3.3] [Reference Citation Analysis (0)] |
33. | Singh S, Bergsland EK, Card CM, Hope TA, Kunz PL, Laidley DT, Lawrence B, Leyden S, Metz DC, Michael M, Modahl LE, Myrehaug S, Padda SK, Pommier RF, Ramirez RA, Soulen M, Strosberg J, Sung A, Thawer A, Wei B, Xu B, Segelov E. 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 31] [Cited by in RCA: 61] [Article Influence: 12.2] [Reference Citation Analysis (0)] |
34. | Rizzardi G, Marulli G, Bortolotti L, Calabrese F, Sartori F, Rea F. Sleeve resections and bronchoplastic procedures in typical central carcinoid tumours. Thorac Cardiovasc Surg. 2008;56:42-45. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 24] [Cited by in RCA: 20] [Article Influence: 1.2] [Reference Citation Analysis (0)] |
35. | Luckraz H, Amer K, Thomas L, Gibbs A, Butchart EG. Long-term outcome of bronchoscopically resected endobronchial typical carcinoid tumors. J Thorac Cardiovasc Surg. 2006;132:113-115. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 46] [Cited by in RCA: 45] [Article Influence: 2.4] [Reference Citation Analysis (0)] |
36. | Brown LM, Cooke DT, Jett JR, David EA. Extent of Resection and Lymph Node Assessment for Clinical Stage T1aN0M0 Typical Carcinoid Tumors. Ann Thorac Surg. 2018;105:207-213. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 26] [Cited by in RCA: 27] [Article Influence: 3.9] [Reference Citation Analysis (0)] |
37. | Laddha SV, da Silva EM, Robzyk K, Untch BR, Ke H, Rekhtman N, Poirier JT, Travis WD, Tang LH, Chan CS. Integrative Genomic Characterization Identifies Molecular Subtypes of Lung Carcinoids. Cancer Res. 2019;79:4339-4347. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 27] [Cited by in RCA: 44] [Article Influence: 7.3] [Reference Citation Analysis (0)] |
38. | Alcala N, Leblay N, Gabriel AAG, Mangiante L, Hervas D, Giffon T, Sertier AS, Ferrari A, Derks J, Ghantous A, Delhomme TM, Chabrier A, Cuenin C, Abedi-Ardekani B, Boland A, Olaso R, Meyer V, Altmuller J, Le Calvez-Kelm F, Durand G, Voegele C, Boyault S, Moonen L, Lemaitre N, Lorimier P, Toffart AC, Soltermann A, Clement JH, Saenger J, Field JK, Brevet M, Blanc-Fournier C, Galateau-Salle F, Le Stang N, Russell PA, Wright G, Sozzi G, Pastorino U, Lacomme S, Vignaud JM, Hofman V, Hofman P, Brustugun OT, Lund-Iversen M, Thomas de Montpreville V, Muscarella LA, Graziano P, Popper H, Stojsic J, Deleuze JF, Herceg Z, Viari A, Nuernberg P, Pelosi G, Dingemans AMC, Milione M, Roz L, Brcic L, Volante M, Papotti MG, Caux C, Sandoval J, Hernandez-Vargas H, Brambilla E, Speel EJM, Girard N, Lantuejoul S, McKay JD, Foll M, Fernandez-Cuesta L. Integrative and comparative genomic analyses identify clinically relevant pulmonary carcinoid groups and unveil the supra-carcinoids. Nat Commun. 2019;10:3407. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 132] [Cited by in RCA: 120] [Article Influence: 20.0] [Reference Citation Analysis (0)] |
39. | Rekhtman N. Neuroendocrine tumors of the lung: an update. Arch Pathol Lab Med. 2010;134:1628-1638. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 247] [Cited by in RCA: 268] [Article Influence: 17.9] [Reference Citation Analysis (0)] |
40. | Wells LE, Cohen S, Brennan B, Banerjee M, Kalemkerian GP. Epidemiology of SCLC in the United States From 2000 to 2019: A Study Utilizing the Surveillance, Epidemiology, and End Results Registry. JTO Clin Res Rep. 2025;6:100799. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Reference Citation Analysis (0)] |
41. | Govindan R, Page N, Morgensztern D, Read W, Tierney R, Vlahiotis A, Spitznagel EL, Piccirillo J. Changing epidemiology of small-cell lung cancer in the United States over the last 30 years: analysis of the surveillance, epidemiologic, and end results database. J Clin Oncol. 2006;24:4539-4544. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1197] [Cited by in RCA: 1383] [Article Influence: 72.8] [Reference Citation Analysis (0)] |
42. | NIH. All Cancer Sites Combined Recent Trends in SEER Age-Adjusted Incidence Rates, 2000-2021. [cited March 25, 2025]. Available from: https://seer.cancer.gov/statistics-network/explorer/application.html?site=1&data_type=1&graph_type=2&compareBy=sex&chk_sex_3=3&chk_sex_2=2&rate_type=2&race=1&age_range=1&hdn_stage=101&advopt_precision=1&advopt_show_ci=on&hdn_view=0&advopt_show_apc=on&advopt_display=2#resultsRegion0. |
43. | Huang R, Wei Y, Hung RJ, Liu G, Su L, Zhang R, Zong X, Zhang ZF, Morgenstern H, Brüske I, Heinrich J, Hong YC, Kim JH, Cote M, Wenzlaff A, Schwartz AG, Stucker I, Mclaughlin J, Marcus MW, Davies MP, Liloglou T, Field JK, Matsuo K, Barnett M, Thornquist M, Goodman G, Wang Y, Chen S, Yang P, Duell EJ, Andrew AS, Lazarus P, Muscat J, Woll P, Horsman J, Teare MD, Flugelman A, Rennert G, Zhang Y, Brenner H, Stegmaier C, van der Heijden EH, Aben K, Kiemeney L, Barros-Dios J, Pérez-Ríos M, Ruano-Ravina A, Caporaso NE, Bertazzi PA, Landi MT, Dai J, Hongbing Shen H, Fernandez-Tardon G, Rodriguez-Suarez M, Tardon A, Christiani DC. Associated Links Among Smoking, Chronic Obstructive Pulmonary Disease, and Small Cell Lung Cancer: A Pooled Analysis in the International Lung Cancer Consortium. EBioMedicine. 2015;2:1677-1685. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 26] [Cited by in RCA: 48] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
44. | American Lung Association. Tobacco Use in Racial and Ethnic Populations. [cited March 25, 2025]. Available from: https://www.lung.org/quit-smoking/smoking-facts/impact-of-tobacco-use/tobacco-use-racial-and-ethnic. |
45. | Thunnissen E, Borczuk AC, Flieder DB, Witte B, Beasley MB, Chung JH, Dacic S, Lantuejoul S, Russell PA, den Bakker M, Botling J, Brambilla E, de Cuba E, Geisinger KR, Hiroshima K, Marchevsky AM, Minami Y, Moreira A, Nicholson AG, Yoshida A, Tsao MS, Warth A, Duhig E, Chen G, Matsuno Y, Travis WD, Butnor K, Cooper W, Mino-Kenudson M, Motoi N, Poleri C, Pelosi G, Kerr K, Aisner SC, Ishikawa Y, Buettner RH, Keino N, Yatabe Y, Noguchi M. 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 75] [Cited by in RCA: 86] [Article Influence: 9.6] [Reference Citation Analysis (0)] |
46. | Travis WD. Update on small cell carcinoma and its differentiation from squamous cell carcinoma and other non-small cell carcinomas. Mod Pathol. 2012;25 Suppl 1:S18-S30. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 174] [Cited by in RCA: 188] [Article Influence: 14.5] [Reference Citation Analysis (0)] |
47. | Pelosi G, Sonzogni A, Harari S, Albini A, Bresaola E, Marchiò C, Massa F, Righi L, Gatti G, Papanikolaou N, Vijayvergia N, Calabrese F, Papotti M. Classification of pulmonary neuroendocrine tumors: new insights. Transl Lung Cancer Res. 2017;6:513-529. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 89] [Cited by in RCA: 89] [Article Influence: 11.1] [Reference Citation Analysis (0)] |
48. | Kriegsmann K, Zgorzelski C, Kazdal D, Cremer M, Muley T, Winter H, Longuespée R, Kriegsmann J, Warth A, Kriegsmann M. Insulinoma-associated Protein 1 (INSM1) in Thoracic Tumors is Less Sensitive but More Specific Compared With Synaptophysin, Chromogranin A, and CD56. Appl Immunohistochem Mol Morphol. 2020;28:237-242. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 24] [Cited by in RCA: 32] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
49. | George J, Lim JS, Jang SJ, Cun Y, Ozretić L, Kong G, Leenders F, Lu X, Fernández-Cuesta L, Bosco G, Müller C, Dahmen I, Jahchan NS, Park KS, Yang D, Karnezis AN, Vaka D, Torres A, Wang MS, Korbel JO, Menon R, Chun SM, Kim D, Wilkerson M, Hayes N, Engelmann D, Pützer B, Bos M, Michels S, Vlasic I, Seidel D, Pinther B, Schaub P, Becker C, Altmüller J, Yokota J, Kohno T, Iwakawa R, Tsuta K, Noguchi M, Muley T, Hoffmann H, Schnabel PA, Petersen I, Chen Y, Soltermann A, Tischler V, Choi CM, Kim YH, Massion PP, Zou Y, Jovanovic D, Kontic M, Wright GM, Russell PA, Solomon B, Koch I, Lindner M, Muscarella LA, la Torre A, Field JK, Jakopovic M, Knezevic J, Castaños-Vélez E, Roz L, Pastorino U, Brustugun OT, Lund-Iversen M, Thunnissen E, Köhler J, Schuler M, Botling J, Sandelin M, Sanchez-Cespedes M, Salvesen HB, Achter V, Lang U, Bogus M, Schneider PM, Zander T, Ansén S, Hallek M, Wolf J, Vingron M, Yatabe Y, Travis WD, Nürnberg P, Reinhardt C, Perner S, Heukamp L, Büttner R, Haas SA, Brambilla E, Peifer M, Sage J, Thomas RK. Comprehensive genomic profiles of small cell lung cancer. Nature. 2015;524:47-53. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1125] [Cited by in RCA: 1645] [Article Influence: 164.5] [Reference Citation Analysis (0)] |
50. | Rudin CM, Poirier JT, Byers LA, Dive C, Dowlati A, George J, Heymach JV, Johnson JE, Lehman JM, MacPherson D, Massion PP, Minna JD, Oliver TG, Quaranta V, Sage J, Thomas RK, Vakoc CR, Gazdar AF. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer. 2019;19:289-297. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 651] [Cited by in RCA: 780] [Article Influence: 130.0] [Reference Citation Analysis (0)] |
51. | Hann CL, Wu MA, Rekhtman N, Rudin CM. In: Cancer Principles & Practice of Oncology DeVita VT, Lawrence TS, Rosenberg SA, editor. Wolters Kluwer, 2019: 671-700. |
52. | Rudin CM, Brambilla E, Faivre-Finn C, Sage J. Small-cell lung cancer. Nat Rev Dis Primers. 2021;7:3. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 546] [Cited by in RCA: 806] [Article Influence: 201.5] [Reference Citation Analysis (0)] |
53. | Barnes H, See K, Barnett S, Manser R. Surgery for limited-stage small-cell lung cancer. Cochrane Database Syst Rev. 2017;4:CD011917. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 38] [Article Influence: 4.8] [Reference Citation Analysis (0)] |
54. | Brock MV, Hooker CM, Syphard JE, Westra W, Xu L, Alberg AJ, Mason D, Baylin SB, Herman JG, Yung RC, Brahmer J, Rudin CM, Ettinger DS, Yang SC. Surgical resection of limited disease small cell lung cancer in the new era of platinum chemotherapy: Its time has come. J Thorac Cardiovasc Surg. 2005;129:64-72. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 107] [Cited by in RCA: 110] [Article Influence: 5.5] [Reference Citation Analysis (0)] |
55. | Peng H, Hao J, Dong B, Chen M, Li Z, Chen C, Liu L. Prophylactic cranial irradiation in patients with resected small-cell lung cancer: A systematic review and meta-analysis. Thorac Cancer. 2024;15:2309-2318. [RCA] [PubMed] [DOI] [Full Text] [Reference Citation Analysis (0)] |
56. | Horn L, Mansfield AS, Szczęsna A, Havel L, Krzakowski M, Hochmair MJ, Huemer F, Losonczy G, Johnson ML, Nishio M, Reck M, Mok T, Lam S, Shames DS, Liu J, Ding B, Lopez-Chavez A, Kabbinavar F, Lin W, Sandler A, Liu SV; IMpower133 Study Group. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N Engl J Med. 2018;379:2220-2229. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1567] [Cited by in RCA: 2285] [Article Influence: 326.4] [Reference Citation Analysis (0)] |
57. | Rudin CM, Awad MM, Navarro A, Gottfried M, Peters S, Csőszi T, Cheema PK, Rodriguez-Abreu D, Wollner M, Yang JC, Mazieres J, Orlandi FJ, Luft A, Gümüş M, Kato T, Kalemkerian GP, Luo Y, Ebiana V, Pietanza MC, Kim HR; KEYNOTE-604 Investigators. Pembrolizumab or Placebo Plus Etoposide and Platinum as First-Line Therapy for Extensive-Stage Small-Cell Lung Cancer: Randomized, Double-Blind, Phase III KEYNOTE-604 Study. J Clin Oncol. 2020;38:2369-2379. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 203] [Cited by in RCA: 502] [Article Influence: 100.4] [Reference Citation Analysis (0)] |
58. | Coles GL, Cristea S, Webber JT, Levin RS, Moss SM, He A, Sangodkar J, Hwang YC, Arand J, Drainas AP, Mooney NA, Demeter J, Spradlin JN, Mauch B, Le V, Shue YT, Ko JH, Lee MC, Kong C, Nomura DK, Ohlmeyer M, Swaney DL, Krogan NJ, Jackson PK, Narla G, Gordan JD, Shokat KM, Sage J. Unbiased Proteomic Profiling Uncovers a Targetable GNAS/PKA/PP2A Axis in Small Cell Lung Cancer Stem Cells. Cancer Cell. 2020;38:129-143.e7. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 44] [Cited by in RCA: 58] [Article Influence: 11.6] [Reference Citation Analysis (0)] |
59. | Gardner EE, Lok BH, Schneeberger VE, Desmeules P, Miles LA, Arnold PK, Ni A, Khodos I, de Stanchina E, Nguyen T, Sage J, Campbell JE, Ribich S, Rekhtman N, Dowlati A, Massion PP, Rudin CM, Poirier JT. Chemosensitive Relapse in Small Cell Lung Cancer Proceeds through an EZH2-SLFN11 Axis. Cancer Cell. 2017;31:286-299. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 290] [Cited by in RCA: 366] [Article Influence: 45.8] [Reference Citation Analysis (0)] |
60. | Paz-Ares L, Dvorkin M, Chen Y, Reinmuth N, Hotta K, Trukhin D, Statsenko G, Hochmair MJ, Özgüroğlu M, Ji JH, Voitko O, Poltoratskiy A, Ponce S, Verderame F, Havel L, Bondarenko I, Kazarnowicz A, Losonczy G, Conev NV, Armstrong J, Byrne N, Shire N, Jiang H, Goldman JW; CASPIAN investigators. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): a randomised, controlled, open-label, phase 3 trial. Lancet. 2019;394:1929-1939. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 831] [Cited by in RCA: 1359] [Article Influence: 226.5] [Reference Citation Analysis (0)] |
61. | Faivre-Finn C, Snee M, Ashcroft L, Appel W, Barlesi F, Bhatnagar A, Bezjak A, Cardenal F, Fournel P, Harden S, Le Pechoux C, McMenemin R, Mohammed N, O'Brien M, Pantarotto J, Surmont V, Van Meerbeeck JP, Woll PJ, Lorigan P, Blackhall F; CONVERT Study Team. Concurrent once-daily versus twice-daily chemoradiotherapy in patients with limited-stage small-cell lung cancer (CONVERT): an open-label, phase 3, randomised, superiority trial. Lancet Oncol. 2017;18:1116-1125. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 350] [Cited by in RCA: 388] [Article Influence: 48.5] [Reference Citation Analysis (0)] |
62. | Iyoda A, Hiroshima K, Toyozaki T, Haga Y, Fujisawa T, Ohwada H. Clinical characterization of pulmonary large cell neuroendocrine carcinoma and large cell carcinoma with neuroendocrine morphology. Cancer. 2001;91:1992-2000. [PubMed] [DOI] [Full Text] |
63. | Ferrara MG, Stefani A, Simbolo M, Pilotto S, Martini M, Lococo F, Vita E, Chiappetta M, Cancellieri A, D'Argento E, Trisolini R, Rindi G, Scarpa A, Margaritora S, Milella M, Tortora G, Bria E. Large Cell Neuro-Endocrine Carcinoma of the Lung: Current Treatment Options and Potential Future Opportunities. Front Oncol. 2021;11:650293. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 29] [Cited by in RCA: 34] [Article Influence: 8.5] [Reference Citation Analysis (0)] |
64. | Travis WD, Linnoila RI, Tsokos MG, Hitchcock CL, Cutler GB Jr, Nieman L, Chrousos G, Pass H, Doppman J. Neuroendocrine tumors of the lung with proposed criteria for large-cell neuroendocrine carcinoma. An ultrastructural, immunohistochemical, and flow cytometric study of 35 cases. Am J Surg Pathol. 1991;15:529-553. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 653] [Cited by in RCA: 579] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
65. | Baine MK, Rekhtman N. Multiple faces of pulmonary large cell neuroendocrine carcinoma: update with a focus on practical approach to diagnosis. Transl Lung Cancer Res. 2020;9:860-878. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 18] [Cited by in RCA: 27] [Article Influence: 5.4] [Reference Citation Analysis (0)] |
66. | Milione M, Maisonneuve P, Grillo F, Mangogna A, Centonze G, Prinzi N, Pusceddu S, Garzone G, Cattaneo L, Busico A, Bossi P, Spaggiari P, Pellegrinelli A, Del Gobbo A, Ferrero S, Kankava K, Pruneri G, Rolli L, Roca E, Bercich L, Tironi A, Benvenuti MR, Gallazzi MS, Romano R, Berruti A, Pastorino U, Capella C. Ki-67 Index of 55% Distinguishes Two Groups of Bronchopulmonary Pure and Composite Large Cell Neuroendocrine Carcinomas with Distinct Prognosis. Neuroendocrinology. 2021;111:475-489. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 25] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
67. | Rekhtman N, Pietanza MC, Hellmann MD, Naidoo J, Arora A, Won H, Halpenny DF, Wang H, Tian SK, Litvak AM, Paik PK, Drilon AE, Socci N, Poirier JT, Shen R, Berger MF, Moreira AL, Travis WD, Rudin CM, Ladanyi 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. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 248] [Cited by in RCA: 330] [Article Influence: 36.7] [Reference Citation Analysis (0)] |
68. | George J, Walter V, Peifer M, Alexandrov LB, Seidel D, Leenders F, Maas L, Müller C, Dahmen I, Delhomme TM, Ardin M, Leblay N, Byrnes G, Sun R, De Reynies A, McLeer-Florin A, Bosco G, Malchers F, Menon R, Altmüller J, Becker C, Nürnberg P, Achter V, Lang U, Schneider PM, Bogus M, Soloway MG, Wilkerson MD, Cun Y, McKay JD, Moro-Sibilot D, Brambilla CG, Lantuejoul S, Lemaitre N, Soltermann A, Weder W, Tischler V, Brustugun OT, Lund-Iversen M, Helland Å, Solberg S, Ansén S, Wright G, Solomon B, Roz L, Pastorino U, Petersen I, Clement JH, Sänger J, Wolf J, Vingron M, Zander T, Perner S, Travis WD, Haas SA, Olivier M, Foll M, Büttner R, Hayes DN, Brambilla E, Fernandez-Cuesta L, Thomas RK. Integrative genomic profiling of large-cell neuroendocrine carcinomas reveals distinct subtypes of high-grade neuroendocrine lung tumors. Nat Commun. 2018;9:1048. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 249] [Cited by in RCA: 246] [Article Influence: 35.1] [Reference Citation Analysis (0)] |
69. | den Bakker MA, Willemsen S, Grünberg K, Noorduijn LA, van Oosterhout MF, van Suylen RJ, Timens W, Vrugt B, Wiersma-van Tilburg A, Thunnissen FB. Small cell carcinoma of the lung and large cell neuroendocrine carcinoma interobserver variability. Histopathology. 2010;56:356-363. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 72] [Cited by in RCA: 75] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
70. | Baine MK, Sinard JH, Cai G, Homer RJ. A Semiquantitative Scoring System May Allow Biopsy Diagnosis of Pulmonary Large Cell Neuroendocrine Carcinoma. Am J Clin Pathol. 2020;153:165-174. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 19] [Cited by in RCA: 15] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
71. | Gu J, Gong D, Wang Y, Chi B, Zhang J, Hu S, Min L. The demographic and treatment options for patients with large cell neuroendocrine carcinoma of the lung. Cancer Med. 2019;8:2979-2993. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 15] [Cited by in RCA: 33] [Article Influence: 5.5] [Reference Citation Analysis (0)] |
72. | Lowczak A, Kolasinska-Cwikla A, Osowiecka K, Glinka L, Palucki J, Rzepko R, Doboszynska A, Cwikla JB. Outcomes of Patients with Pulmonary Large Cell Neuroendocrine Carcinoma in I-IV Stage. Medicina (Kaunas). 2021;57. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 10] [Article Influence: 2.5] [Reference Citation Analysis (3)] |
73. | Niho S, Kenmotsu H, Sekine I, Ishii G, Ishikawa Y, Noguchi M, Oshita F, Watanabe S, Nakajima R, Tada H, Nagai K. Combination chemotherapy with irinotecan and cisplatin for large-cell neuroendocrine carcinoma of the lung: a multicenter phase II study. J Thorac Oncol. 2013;8:980-984. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 95] [Cited by in RCA: 102] [Article Influence: 9.3] [Reference Citation Analysis (0)] |
74. | Derks JL, Leblay N, Thunnissen E, van Suylen RJ, den Bakker M, Groen HJM, Smit EF, Damhuis R, van den Broek EC, Charbrier A, Foll M, McKay JD, Fernandez-Cuesta L, Speel EM, Dingemans AC; PALGA-Group. Molecular Subtypes of Pulmonary Large-cell Neuroendocrine Carcinoma Predict Chemotherapy Treatment Outcome. Clin Cancer Res. 2018;24:33-42. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 149] [Cited by in RCA: 140] [Article Influence: 20.0] [Reference Citation Analysis (0)] |
75. | Wang VE, Urisman A, Albacker L, Ali S, Miller V, Aggarwal R, Jablons D. Checkpoint inhibitor is active against large cell neuroendocrine carcinoma with high tumor mutation burden. J Immunother Cancer. 2017;5:75. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 39] [Cited by in RCA: 52] [Article Influence: 6.5] [Reference Citation Analysis (0)] |
76. | Iyoda A, Travis WD, Sarkaria IS, Jiang SX, Amano H, Sato Y, Saegusa M, Rusch VW, Satoh Y. Expression profiling and identification of potential molecular targets for therapy in pulmonary large-cell neuroendocrine carcinoma. Exp Ther Med. 2011;2:1041-1045. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 41] [Cited by in RCA: 44] [Article Influence: 3.1] [Reference Citation Analysis (0)] |
77. | Hermans BCM, Derks JL, Thunnissen E, van Suylen RJ, den Bakker MA, Groen HJM, Smit EF, Damhuis RA, van den Broek EC, Stallinga CM, Roemen GM, Speel EJM, Dingemans AC; PALGA-group. Prevalence and prognostic value of PD-L1 expression in molecular subtypes of metastatic large cell neuroendocrine carcinoma (LCNEC). Lung Cancer. 2019;130:179-186. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 22] [Cited by in RCA: 26] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
78. | Kriegsmann K, Zgorzelski C, Muley T, Christopoulos P, Thomas M, Winter H, Eichhorn M, Eichhorn F, von Winterfeld M, Herpel E, Goeppert B, Stenzinger A, Herth FJF, Warth A, Kriegsmann M. Role of Synaptophysin, Chromogranin and CD56 in adenocarcinoma and squamous cell carcinoma of the lung lacking morphological features of neuroendocrine differentiation: a retrospective large-scale study on 1170 tissue samples. BMC Cancer. 2021;21:486. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 10] [Cited by in RCA: 20] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
79. | Rekhtman N, Pietanza CM, Sabari J, Montecalvo J, Wang H, Habeeb O, Kadota K, Adusumilli P, Rudin CM, Ladanyi M, Travis WD, Joubert P. Pulmonary large cell neuroendocrine carcinoma with adenocarcinoma-like features: napsin A expression and genomic alterations. Mod Pathol. 2018;31:111-121. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 45] [Cited by in RCA: 42] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
80. | Dowlati A, Lipka MB, McColl K, Dabir S, Behtaj M, Kresak A, Miron A, Yang M, Sharma N, Fu P, Wildey G. Clinical correlation of extensive-stage small-cell lung cancer genomics. Ann Oncol. 2016;27:642-647. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 61] [Cited by in RCA: 55] [Article Influence: 6.1] [Reference Citation Analysis (0)] |