Next Article in Journal
A Deep Learning Model Integrating Clinical and MRI Features Improves Risk Stratification and Reduces Unnecessary Biopsies in Men with Suspected Prostate Cancer
Previous Article in Journal
MYC Regulates a DNA Repair Gene Expression Program in Small Cell Carcinoma of the Ovary, Hypercalcemic Type
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Redefining the Fight Against SCLC: Standards, Innovations, and New Horizons

1
Department of Medicine A, University Hospital Muenster, 48149 Muenster, Germany
2
West German Cancer Center, University Hospital Muenster, 48149 Muenster, Germany
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(13), 2256; https://doi.org/10.3390/cancers17132256
Submission received: 26 May 2025 / Revised: 28 June 2025 / Accepted: 29 June 2025 / Published: 7 July 2025
(This article belongs to the Special Issue Advances in Targeted Therapies in Cancer (2nd Edition))

Simple Summary

Small cell lung cancer is an aggressive malignancy characterized by a poor prognosis. Standard treatment modalities include surgery, radiotherapy, chemotherapy, and immunotherapy; however, therapeutic responses are often limited in duration. Emerging therapeutic strategies involve the use of bispecific antibodies, antibody–drug conjugates, targeted therapies, and personalized treatment approaches guided by molecular biomarkers. Recent advancements in molecular subtyping and novel therapeutics hold promise for ultimately improving patient survival.

Abstract

Background: Small cell lung cancer (SCLC) remains a highly aggressive malignancy with a poor prognosis. Despite multimodal standard therapies, most patients relapse within months, and second-line treatment options such as topotecan offer only limited benefit. Novel therapeutic strategies are therefore urgently needed. Methods: This narrative review is based on a selective literature search conducted via PubMed and ClinicalTrials.gov (last updated June 2025). Results: Emerging treatment strategies include bispecific T-cell engagers (e.g., tarlatamab), antibody-drug conjugates (ADCs) such as sacituzumab govitecan, DS-7300, and ZL-1310, as well as targeted therapies. Among these, tarlatamab has demonstrated improved survival outcomes with an acceptable safety profile and is poised to become the new second-line standard. In contrast, ADCs and targeted agents have shown only modest efficacy and have yet to deliver meaningful survival benefits, often accompanied by increased toxicity. Additionally, the identification of molecular subtypes of SCLC has revealed subtype-specific differences in treatment response. However, clinical translation is challenged by intratumoral heterogeneity, plasticity, and the lack of standardized diagnostic assays. Conclusions: While tarlatamab represents a major therapeutic advancement, other agents remain in early clinical development and require validation in large, randomized trials. The clinical implementation of molecular subtyping remains limited, though it holds promise for future personalized treatment approaches. Despite recent progress, SCLC continues to pose substantial therapeutic challenges, emphasizing the need for improved treatment strategies and validated predictive biomarkers.

1. Introduction

1.1. Epidemiology

Lung cancer remains the leading cause of cancer-related mortality worldwide, accounting for approximately 1.8 million deaths in 2022 [1], with tobacco smoking as the most significant risk factor. In the United States (U.S.), the proportion of small cell lung cancer (SCLC) among newly diagnosed lung cancer cases declined from 14.5% in 2000 to 11.8% in 2019, with consistent reductions observed across all sex and racial groups, likely due to decreasing smoking rates and improved tobacco control measures [2]. Despite these trends, survival outcomes for SCLC have shown minimal improvement over the past two decades [2,3]. Depending on the disease stage at diagnosis, the 5-year relative survival rate for SCLC ranges from <5% to 25% [4]. Although global smoking rates are declining, some regions, including North Africa, the Middle East, and sub-Saharan Africa, continue to experience a rise in tobacco use [5]. In parallel, increased exposure to occupational carcinogens and air pollution has become a growing concern, particularly in emerging countries with limited healthcare infrastructure [6]. However, the specific contribution of these risk factors to SCLC remains unclear, as available data often do not differentiate between lung cancer subtypes. According to the American Cancer Society, an estimated 226,650 new cases of lung cancer are expected in the U.S. in 2025, with approximately 30,000 attributed to SCLC and a median age at diagnosis of approximately 69 years. At the time of diagnosis, approximately 70% of SCLC cases present with distant metastases, while only about 30% are diagnosed at a limited stage [2,7]. These numbers highlight the ongoing burden of SCLC and the need for better strategies in early detection and treatment.

1.2. Pathology

SCLC develops from neuroendocrine cells of the bronchi. Exposure to carcinogens induces mutations in tumor suppressor genes and/or proto-oncogenes. A hallmark of SCLC is the presence of biallelic inactivating mutations in the two tumor suppressor genes Tumor Protein p53 (TP53) and Retinoblastoma 1 (RB1) [8,9]. In addition, mutations in the Neurogenic Locus Notch Homolog (NOTCH) gene family or amplifications of the Myelocytomatosis (MYC) gene are frequently observed in SCLC [8,10].
Microscopically, SCLC typically presents as a small, blue, round cell tumor. The tumor cells are small and have a high nuclear-to-cytoplasmic ratio. The nuclei are hyperchromatic with finely granular chromatin (the so-called “salt and pepper” pattern). Due to the high mitotic activity, apoptosis is frequent, and extensive necrotic areas are commonly observed. To confirm the diagnosis, neuroendocrine markers are typically detected using immunohistochemistry. SCLC tumor cells are usually positive for CD56 (Neural Cell Adhesion Molecule [NCAM]), synaptophysin, chromogranin A, and Thyroid Transcription Factor-1 (TTF-1). A characteristically high Ki-67 proliferation index of over 80% is often observed. Due to the high proliferative activity, blood levels of neuron-specific enolase (NSE) and lactate dehydrogenase (LDH) are often elevated.

1.3. Diagnosis and Staging

In its early stages, SCLC often remains asymptomatic [11]. Nonspecific symptoms include fatigue, weight loss, loss of appetite, cough, and shortness of breath. As the disease progresses, patients may experience hemoptysis, chest pain, and symptoms caused by local tumor infiltration (e.g., hoarseness, stridor, dysphagia, superior vena cava syndrome). Symptoms related to metastases, such as bone pain or neurological abnormalities, can also occur [11]. Paraneoplastic syndromes are relatively common in SCLC. While neuroendocrine syndromes (e.g., Cushing’s syndrome, Syndrome of Inappropriate Antidiuretic Hormone Secretion [SIADH]) result from ectopic hormone production, neurological syndromes such as Lambert-Eaton myasthenic syndrome (present in 1–3% of cases) are immune-mediated [12,13,14]. Interestingly, patients with paraneoplastic syndromes have a better prognosis than those without, which might be due to enhanced efficacy of immunotherapy [15].
Diagnosis is based on a combination of medical history, physical examination, laboratory tests (such as NSE and LDH), imaging studies, and a biopsy, typically obtained through bronchoscopy or computed tomography (CT)-guided procedures. For SCLC in stages I–III, where surgery or radiotherapy may be an option, pulmonary function testing (including Forced Expiratory Volume in 1 Second [FEV1], Vital Capacity [VC], and Diffusing Capacity of the Lung for Carbon Monoxide [DLCO]) is also indicated [16]. The guidelines from the National Comprehensive Cancer Network (NCCN) recommend a diagnostic contrast-enhanced CT of the chest, abdomen, and pelvis for further evaluation [17]. If curative treatment is being considered, a fluorodeoxyglucose positron emission tomography (FDG-PET/CT) scan should be performed. As an alternative, bone scintigraphy may be used. Magnetic resonance imaging (MRI) of the brain is recommended for all patients and preferred over CT scans of the brain. Although staging evaluation is optional in patients with extensive stage and dependent on the clinical situation, patients who are not candidates for curative therapy should still receive appropriate staging, including imaging of the brain [17]. The guidelines from the European Society for Medical Oncology (ESMO) recommend a similar diagnostic work-up [16].
For many years, SCLC was classified into Limited Disease (LD-SCLC) and Extensive Disease (ED-SCLC) based on the Veterans Administration Lung Study in 1957. Limited Disease refers to stages where the primary tumor was confined to one hemithorax, with no evidence of tumor spread beyond potentially present ipsilateral pleural effusion or mediastinal lymph node involvement. All other disease scenarios were categorized as Extensive Disease and were associated with a correspondingly poorer prognosis. Today, SCLC is classified according to the TNM or Union for International Cancer Control (UICC) staging system, which allows for more precise categorization, treatment planning, and prognosis assessment [18,19,20]. In this context, staging follows the standard approach used for other tumors, based on the size and local extent of the primary tumor (T), as well as the presence of lymph node involvement (N) or distant metastases (M) (see Table 1).

2. Methods

This narrative review was conducted to provide a comprehensive and up-to-date synthesis of current knowledge on SCLC, with a focus on current standards, molecular subtypes, and emerging therapies (see Figure 1). A selective, non-systematic literature search was performed using PubMed to identify relevant peer-reviewed articles published up to June 2025. Search terms included combinations of keywords such as “SCLC”, “therapy”, “biomarker”, “clinical trial”, and “molecular subtyping”. Only articles published in English were considered. To enhance coverage of ongoing research, additional searches were conducted on ClinicalTrials.gov using similar terms to identify registered, ongoing, and completed clinical trials. Studies were selected based on their relevance to the review’s thematic focus, prioritizing original research, recent clinical trials, and high-quality reviews. In cases of conflicting evidence, we considered the methodological rigor, sample size, and recency of studies and aimed to reflect diverging findings in a balanced manner. Reference lists of key publications were screened for additional sources, and relevant abstracts from major oncology conferences were included when peer-reviewed data were not yet available. Furthermore, current national and international guidelines from the United States and Europe were reviewed and integrated. As this is a non-systematic narrative review, no formal protocol (e.g., PRISMA) was applied.

3. Molecular Subtyping

Based on the expression of specific transcription factors, SCLC can be classified into molecular subtypes. This molecular classification enhances our understanding of tumor heterogeneity and may help explain differences in prognosis and therapeutic responses among patients. The four major subtypes are as follows [21]:
-
SCLC-A (ASCL1-dominant): Characterized by high expression of Achaete-Scute Family BHLH Transcription Factor 1 (ASCL1), this subtype is associated with classic neuroendocrine features and may be sensitive to B-Cell Lymphoma 2 (BCL-2) inhibition due to its dependency on anti-apoptotic pathways.
-
SCLC-N (NEUROD1-dominant): Defined by predominant expression of Neurogenic Differentiation Factor 1 (NEUROD1), this subtype exhibits features of neuronal differentiation and often harbors MYC gene amplifications, suggesting potential vulnerability to aurora kinase inhibitors and other MYC-targeted therapies.
-
SCLC-P (POU2F3-dominant): Marked by high expression of POU Class 2 Homeobox 3 (POU2F3), this non-neuroendocrine subtype represents a distinct tuft-cell-like lineage and may be sensitive to Insulin-like Growth Factor 1 Receptor (IGF1R) inhibitors or other novel targeted agents.
-
SCLC-I (Inflamed subtype): Distinguished by low expression of traditional lineage-defining transcription factors but high levels of immune cell infiltration and the upregulation of immune response-related genes, SCLC-I tumors appear particularly sensitive to immunotherapy.
SCLC-Y refers to a proposed additional subtype of SCLC characterized by high expression of Yes-Associated Protein 1 (YAP1), a transcriptional co-activator involved in the Hippo signaling pathway. This subtype exhibits less neuroendocrine differentiation than SCLC-N or SCLC-P and is potentially associated with chemotherapy resistance and increased responsiveness to immune checkpoint blockade [22,23]. SCLC-Y also displays more mesenchymal and proliferative features and may possess distinct biological vulnerabilities compared to the other four established SCLC subtypes. However, SCLC-Y remains less well-defined and less widely accepted in the field [24,25]. Some experts suggest that SCLC-Y overlaps with or could be considered a subset of SCLC-I, given its non-neuroendocrine characteristics and stem-like traits.
These molecular subtypes differ not only in gene expression profiles but also in clinical behavior and therapeutic vulnerabilities. Retrospective analyses from the IMpower133 trial and preclinical studies suggest that the SCLC-I subtype may be more responsive to immunotherapy [21,26]. However, conflicting evidence also exists, with some studies reporting no clear association between the tumor immune microenvironment and molecular subtypes [27]. As such, these classifications remain under development and currently lack standardized, clinically validated assays, limiting their immediate clinical utility. Additionally, growing evidence highlights substantial intratumoral heterogeneity [28] and dynamic plasticity between subtypes [29], further challenging the stability and reproducibility of this framework. While molecular subtyping holds promise, prospective validation and translational research are needed to establish its role in clinical decision-making.

4. Treatment for Limited Disease (LD-SCLC)

4.1. Current Treatment Standards in LD-SCLC

A curative treatment approach is formally possible only for patients with LD-SCLC. The median overall survival (OS) in this stage (with treatment) is approximately 12–18 months, with a 2-year survival rate of about 41% [30]. For patients with LD-SCLC suitable for surgical resection (stage I-II, cT1-2 N0), the guidelines of the NCCN and ESMO recommend primary surgical resection followed by either four cycles of adjuvant platinum-based chemotherapy (in case of R0 resection) or concurrent chemoradiotherapy (cCRT) (in case of R1–2 resection). For patients not suitable for surgical resection (stages I–III, cT1-4 N0-3 M0), subsequent therapy depends on patient performance status (PS). While patients with good PS (0–1) are recommended to receive cCRT, patients with poor PS (≥2) can alternatively receive sequential CRT [16,17,31]. Studies have shown no clear significant difference between conventional fractionation (total dose of 60–66 Gy) and hyperfractionated radiotherapy (total dose of 45 Gy) [32,33,34]. After the completion of cCRT, prophylactic cranial irradiation (PCI) with a total dose of 25–30 Gy should be considered [30]. In LD-SCLC, PCI has prognostic relevance, as it can reduce the risk of central nervous system (CNS) metastases from 40% (without PCI) to 10% (with PCI), although the improvement in 5-year survival is only around 5% [35]. Patients who do not show disease progression following cCRT should also receive consolidation therapy with durvalumab according to the results from the phase III ADRIATIC trial (see Table 2). Here, durvalumab significantly improved both progression-free survival (PFS) and OS compared to placebo. The median OS was 55.9 months for durvalumab. The median PFS was 16.6 months vs. 9.2 months, respectively (Hazard Ratio [HR] = 0.76; 95% Confidence Interval [CI]: 0.61–0.95). Pneumonitis occurred more frequently with durvalumab (38% vs. 30%), though the overall safety profile remained acceptable. Based on these results, durvalumab has been approved as consolidation therapy following cCRT by both the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA). The results in the durvalumab–tremelimumab group remain blinded [36].

4.2. Novel Treatment Approaches in LD-SCLC

As only about 30% of SCLC cases are diagnosed at a limited disease stage, most research efforts have focused on optimizing treatment for extensive-stage disease and recurrence. Nevertheless, several agents under investigation in ED-SCLC are also being evaluated in LD-SCLC (see Figure 2 and Table 3). Drawing parallels to non-small cell lung cancer (NSCLC), neoadjuvant chemoimmunotherapy followed by surgery is currently being explored for LD-SCLC. Retrospective analyses suggest promising feasibility and efficacy, though no randomized phase III data are available to date. Ongoing phase II trials (NCT06911606, NCT04539977, NCT04542369) are evaluating this approach, but it cannot yet be considered an alternative treatment option. Whether pathological remission status can serve as a surrogate prognostic biomarker in SCLC, as it does in NSCLC, remains to be determined.
Another phase II study (NCT06719700) is investigating the combination of cCRT with immunotherapy and anti-angiogenic targeted therapy. In parallel, the phase III KEYLYNK-013 trial (NCT04624204) is assessing the addition of olaparib to immunotherapy as maintenance after cCRT. However, concerns over cumulative toxicity may limit the clinical applicability of these approaches, making it unlikely that they will redefine the current standard of care (SOC).
Although results from the durvalumab–tremelimumab arm of the ADRIATIC trial are still pending, targeting both Programmed Death (Ligand) 1 [PD-(L)1] and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4), has already been investigated in LD-SCLC. The STIMULI trial assessed consolidation therapy with nivolumab and ipilimumab following cCRT but failed to meet its primary endpoint of PFS, largely due to treatment discontinuations caused by immune-related adverse events (irAEs) [42]. Given these toxicity challenges, dual checkpoint inhibition in LD-SCLC appears limited, and attention has shifted towards more tolerable agents such as tarlatamab. Following the promising results from the DeLLphi-304 trial (see Section 5.2.3), tarlatamab is now also being evaluated in LD-SCLC. Therefore, the phase III DeLLphi-306 trial (NCT06117774) is currently investigating tarlatamab versus placebo as consolidation therapy following cCRT.
Table 3. Tabular overview of recent clinical trials on novel therapeutic approaches in small cell lung cancer (SCLC). This table summarizes key study data on the investigated therapeutics, including study design, patient cohorts, interventions/dosing, primary efficacy outcomes, safety profiles, and relevant references. Abbreviations used in this table include ADC (antibody-drug conjugate), AE (adverse events), BiTE (bispecific T-cell engager), cCRT (concurrent chemoradiotherapy), CRPC (castration-resistant prostate cancer), CRS (cytokine release syndrome), CTFI (chemotherapy-free interval), CTX (chemotherapy), DLT (dose-limiting toxicities), DCR (disease control rate), DOR (duration of response), ED (Extensive Disease), FL (follicular lymphoma), ICANS (Immune Effector Cell-Associated Neurotoxicity Syndrome), ILD (interstitial lung disease), LD (Limited Disease), mo (months), NEC (neuroendocrine carcinoma), ORR (objective response rate), OS (overall survival), PFS (progression-free survival), pts (patients), SD (stable disease), SOC (standard of care), TEAE (treatment-emergent adverse events), TRAE (treatment-related adverse events), and VOD (veno-occlusive disease).
Table 3. Tabular overview of recent clinical trials on novel therapeutic approaches in small cell lung cancer (SCLC). This table summarizes key study data on the investigated therapeutics, including study design, patient cohorts, interventions/dosing, primary efficacy outcomes, safety profiles, and relevant references. Abbreviations used in this table include ADC (antibody-drug conjugate), AE (adverse events), BiTE (bispecific T-cell engager), cCRT (concurrent chemoradiotherapy), CRPC (castration-resistant prostate cancer), CRS (cytokine release syndrome), CTFI (chemotherapy-free interval), CTX (chemotherapy), DLT (dose-limiting toxicities), DCR (disease control rate), DOR (duration of response), ED (Extensive Disease), FL (follicular lymphoma), ICANS (Immune Effector Cell-Associated Neurotoxicity Syndrome), ILD (interstitial lung disease), LD (Limited Disease), mo (months), NEC (neuroendocrine carcinoma), ORR (objective response rate), OS (overall survival), PFS (progression-free survival), pts (patients), SD (stable disease), SOC (standard of care), TEAE (treatment-emergent adverse events), TRAE (treatment-related adverse events), and VOD (veno-occlusive disease).
AgentTarget/
Mechanism
Trial
(Phase)
NPopulationKey ResultsSafety ProfileLimitationsClinical ImplicationsReference
TarlatamabDLL3/BiTEDeLLphi-301
(Phase II)
220≥2 L
ED-SCLC
ORR: 40%;
PFS 4.9 mo;
OS: 14.3 mo;
58% DOR ≥ 6 mo
CRS (51%), decreased appetite (29%), pyrexia (35%), ~3% treatment discontinuationNo control group; CRS/ICANS require careful monitoring; hospitalizationFavorable survival and tolerability,
FDA approval
[43]
DeLLphi-304
(Phase III)
~700Relapsed ED-SCLC after
platinum-based CTX
Improvement in OS compared to local SOC (13.6 vs. 8.3 mo)Lower grade ≥ 3 AEs (54% vs. 80%), fewer discontinuationsIndirect comparison to mix of chemo agents; CRS/ICANS monitoring not fully standardizedNew SOC for
second-line treatment in ED-SCLC
[44]
DeLLphi-305
(Phase III)
550Maintenance after 1 L induction in ED-SCLC without
tumor progression
recruiting
DeLLphi-306
(Phase III)
400Consolidation after cCRT
in LD-SCLC
recruiting
Rovalpituzumab Tesirine (Rova-T)DLL3/ADCNCT01901653
(Phase I/II)
82≥1 L
SCLC
ORR 18%
(38% in pts with high DLL3 expression)
Grade ≥ 3 TRAEs: thrombocytopenia (11%), pleural effusions (8%), elevated lipase (7%)No control, small sample size; modest efficacy; high-grade toxicitiesDLL3 expression linked to response, foundational for future ADCs[45]
TAHOE
(Phase III)
444≥1 L
Advanced/ metastatic SCLC with high DLL3 expression
Inferior OS compared to topotecan (6.3 vs. 8.6 mo)
→ trial discontinuation
serosal effusions, photosensitivity, peripheral edemaNo efficacy benefit; worse outcome vs. SOC; toxicity of ADC payloadFirst anti-DLL3 ADC tested in SCLC phase III; discontinuation of
Rova-T program
[46]
MERU
(Phase III)
748Maintenance after
1 L platinum-based CTX
in ED-SCLC
DLL3-high tumors: Significant improvement in PFS (4.0 vs. 1.4 mo) but not in OS (8.5 vs. 9.8 mo)
→ trial termination
≥20% AEs in the Rova-T arm (pleural effusion, decreased appetite, peripheral edema, photosensitivity, fatigue, nausea, dyspnea)
More grade ≥ 3 toxicities
No active comparator; high-grade AEs offset PFS gainsFirst phase III DLL3-targeted ADC in
maintenance;
discontinuation of
Rova-T program
[47]
ZL-1310NCT06179069
(Phase I)
112≥1 L
ED-SCLC;
±Atezolizumab
±Carboplatin
ORR 68%;
pts with brain metastases: ORR 80%, DCR 100%
39% grade ≥ 3 TRAEs (anemia, neutropenia, thrombocytopenia), one DLTEarly phase, small sample size; long-term safety and higher-dose toxicity unresolvedHighly promising early activity, rapid and intracranial responses; manageable early safety[48]
Sacituzumab GovitecanTrop2/ADCNCT03964727/
TROPiCS-03
(Phase II)
227
(43 SCLC)
Recurrent ED-SCLC after 1 L platinum-based CTX and PD(L)1 directed therapyORR: 41.9%
DOR: 4.7 mo
PFS: 4.4 mo
OS: 13.6 mo
74.4% grade ≥ 3 TEAEs; no TEAE led to treatment discontinuation; 1 TEAE (neutropenic sepsis) led to deathSingle-arm, small sample size; grade ≥ 3 events common; optimal patient selection unclearPromising second-line option in ED-SCLC[49]
Ifinatamab Deruxtecan (DS-7300)B7-H3/ADCNCT04145622
(Phase I/II);
250
(22 SCLC)
Advanced/unresectable solid tumor (incl. SCLC), that is refractory to or intolerable with standard treatment, or for which no standard treatment is available.ORR: 52.4%;
PFS: 5.6 mo;
OS: 12.2 mo
36.4% grade ≥ 3 TEAEs (nausea, decreased appetite, constipation); 22.7% treatment discontinuationSingle-arm; small sample size; ILD/pneumonitis risk unclear; Dose/regimen optimization pendingFirst-in-human for anti-B7-H3 ADC in SCLC; broad efficacy across SCLC patients[50]
NCT06203210/
IDeate-Lung02
(Phase III)
540Relapsed SCLC after 1 L
platinum-based CTX
recruiting
ABBV-011SEZ6/ADCNCT03639194
(Phase I)
99Relapsed/refractory ED-SCLC after 1 L platinum-based CTXAt 1.0 mg/kg:
ORR: 25%;
DOR: 4.2 mo;
PFS: 3.5 mo
Fatigue (50%), nausea (42%), thrombocytopenia (41%), increased ASAT (22%),
2 cases of VOD
No comparator; modest response in selected cohort; grade ≥ 3 AEs in ~48%; biomarker cutoff and expression variabilityFirst SEZ6-targeted ADC tested in SCLC; No further trials yet initiated[51]
OlaparibPARP inhibitorNCT04728230, PRIO
(Phase I/II)
631 L ED-SCLC;
+durvalumab
with carboplatin/etoposide and/or radiotherapy
recruiting
NCT04624204
KEYLYNK-013
(Phase III)
6721 L LD-SCLC;
+pembrolizumab
post cCRT
Active, not recruiting
TalazoparibSWOG S1929
(Phase II)
106Maintenance after 1 L in
SLFN11-positive ED-SCLC;
±atezolizumab
Improvement in PFS (2.9 mo vs. 2.4 mo), but no difference in OS17% grade ≥ 3 non-hematologic TRAEs, higher rate of hematologic TRAEs (50%)Phase II, small sample; No OS improvement; Hematologic toxicity in 50%; SLFN11 not validated as predictive for OSBiomarker-selected, first of its kind in SCLC; requires larger studies to confirm clinical value[52]
NiraparibNCT04701307
(Phase II)
48≥1 L SCLC or NECs;
+dostarlimab
Active, not recruiting
NavitoclaxBCL2 inhibitorNCT00445198
(Phase II)
39Recurrent/progressive SCLC
after ≥1 L
Early efficacy signs (PR 2.6%, SD 23%)
PFS 1.5 mo;
OS 3.2 mo
Dose-limiting thrombocytopenia (41% grades 3–4)Small, early-phase, no comparator; minimal reported efficacy; DLTs led to trial terminationIdentified thrombocytopenia as key toxicity[53]
VenetoclaxNCT0442221, NCT04543916
(Phase Ib/II)
N/A1 L ED-SCLC,
relapsed/refractory SCLC
terminated/withdrawn
TazemetostatEZH2 inhibitorNCT05353439
(Phase I)
60Relapsed/recurrent SCLC after platinum-based CTX;
+topotecan/pembrolizumab
recruiting
MevrometostatNCT03460977
(Phase I/II)
343Relapsed/refractory SCLC,
CRPC, and FL;
±SOC treatment
So far, no formal efficacy results have been reported for SCLC cohortSCLC-specific safety outcomes unreportedEarly-phase, small cohorts; unreported efficacy and safety dataFirst EZH1/2 inhibitor tested in SCLC[54]
ValemetostatEZH1/2 inhibitorNCT03879798
(Phase I/II)
22Recurrent SCLC after
platinum-based CTX
ORR: 21% (4/19);
DOR: 4.6 mo
PFS: 2.2 mo
OS: 6.6 mo
≥20% TRAEs were diarrhea, fatigue, nausea, and rash;
3 DLTs → early trial termination
Single-arm, small cohort; modest outcomes; DLTs occurredExplored SLFN11, epigenetic markers, subtype shifts[55]
LurbinectedinDNA damageNCT02611024
(Phase I/II)
320
(100 SCLC)
Relapsed/refractory solid tumors, including SCLC;
+irinotecan
ORR: 52.7%
DOR: 7.6 mo
PFS: 5.0 mo
OS: 12.7 mo
(in pts with CTFI > 20 days)
71.6% grade ≥ 3 TRAEs (neutropenia, anemia, diarrhea, fatigue); 31.1% serious AEs, 6.8% treatment discontinuationsSingle-arm, open-label;
high grade ≥ 3 AE rate
Proof-of-concept for the combination of lurbinectedin + irinotecan;
led to LAGOON phase III trial
[56]
NCT05153239/
LAGOON
(Phase III)
705Relapsed SCLC after platinum-based CTX with CTFI ≥ 30 days;
+irinotecan
Active, not recruiting
NCT05091567
IMforte
(Phase III)
660Maintenance after 1 L induction treatment in ED-SCLC without tumor progressionPFS: 5.4 mo
OS: 13.2 mo
25.6% grade 3/4 TRAEs; AEs led to treatment discontinuation in 6.2%Open-label; moderate absolute improvements; PFS still limited; increased serious AEs and some fatal eventsSignificant survival benefits; potential new option for maintenance therapy[57]
Despite ongoing trials exploring novel strategies such as neoadjuvant chemoimmunotherapy and consolidation with tarlatamab, their clinical value in LD-SCLC remains uncertain due to limited data and concerns regarding feasibility and toxicity.

5. Treatment for Extensive Disease (ED-SCLC)

5.1. Current Treatment Standards in ED-SCLC

In advanced-stage SCLC, the standard therapy according to NCCN and ESMO is platinum-based chemotherapy (carboplatin or cisplatin plus etoposide) combined with a PD-L1 inhibitor (atezolizumab or durvalumab) [16,17]. This combination has demonstrated a significant survival benefit in randomized phase III trials (IMpower133 and CASPIAN) and is now considered the international SOC [37,38]. The median OS under chemotherapy is just under 10 months, with a 2-year survival rate of approximately 9% [30]. The addition of immunotherapy has been shown to prolong OS by only about 2 months, from 10 to 12 months [58]. After successful first-line treatment, maintenance therapy with the checkpoint inhibitor is recommended until disease progression. PCI is only considered for selected ED-SCLC patients with a high risk of brain metastases [16]. Since February 2025, a new approval has been granted for the combination of the PD-1-targeting monoclonal antibody serplulimab with cis-/carboplatin and etoposide. This combination demonstrated a significant improvement in OS compared to chemotherapy alone in the randomized phase III ASTRUM-005 trial [39] (see Table 2).
Topotecan is the recommended second-line treatment according to ESMO guidelines [16], though it shows limited efficacy. Alternatively, lurbinectedin can be used [17], based on data from a single-arm phase II basket trial (NCT02454972) published in 2020 that showed promising efficacy [40]. However, lurbinectedin has only been approved by the FDA, while approval by the EMA is still pending.

5.2. Novel Treatment Approaches in ED-SCLC

Despite modest improvements with the addition of immunotherapy to chemotherapy, the overall prognosis of SCLC remains poor, particularly in cases of recurrence or disease progression. This underscores the urgent need for novel therapeutic strategies. The following section provides a concise overview of current research directions and recent clinical trial results in this field (see Figure 2 and Table 3).

5.2.1. Lurbinectedin

Lurbinectedin is a synthetic alkaloid analogue with cytotoxic activity that functions as a DNA-binding inhibitor of transcription [59,60]. In relapsed SCLC, lurbinectedin showed activity and had an acceptable safety profile [40]. In addition to its direct cytotoxic effects, lurbinectedin exhibits immunomodulatory properties that further enhance its antitumor activity [61]. Very recently, the results of the phase III, randomized, open-label IMforte trial have been released, which evaluated the efficacy of maintenance therapy with lurbinectedin plus atezolizumab in patients with ED-SCLC who had not progressed after first-line chemoimmunotherapy. The trial demonstrated a significant improvement in PFS, with a median PFS of 5.4 months in the combination arm versus 2.1 months in the atezolizumab monotherapy arm (HR 0.54; p < 0.0001). The median OS also improved from 10.6 months to 13.2 months (HR 0.73; p = 0.0174), respectively. Although treatment-related adverse events (TRAEs) were more common in the combination arm (83.5% vs. 40%), the regimen was overall well tolerated, with a low rate of treatment discontinuation. These findings suggest that lurbinectedin combined with atezolizumab may represent a promising new maintenance strategy for patients with ED-SCLC [57]. While lurbinectedin is approved by the FDA but not yet by the EMA, for second-line treatment of SCLC, the phase III ATLANTIS trial failed to show an OS benefit for the combination of lurbinectedin and doxorubicin compared to standard regimens [41]. Building on phase II results [56], an ongoing phase III trial (LAGOON) is currently evaluating lurbinectedin with or without irinotecan as second-line treatment for SCLC (NCT05153239). The first results are expected in the first quarter of 2026. Overall, lurbinectedin appears promising for use in maintenance and second-line settings; however, its long-term applicability may be restricted by toxicity, particularly given the high burden of comorbidities in SCLC patients.

5.2.2. Dual Checkpoint Inhibition in SCLC

In the phase I/II CheckMate 032 trial, combining nivolumab (anti-PD-1) with ipilimumab (anti-CTLA-4) led to a modest increase in objective response rate (ORR) compared to nivolumab alone (21% vs. 10%) in patients with recurrent SCLC, albeit with higher toxicity [62]. In the CASPIAN trial, the addition of tremelimumab (anti-CTLA-4) to durvalumab and chemotherapy did not result in a significant survival advantage and led to a higher incidence of serious adverse events (AEs) [63]. These findings suggest that while dual checkpoint blockade may potentiate immune activity against SCLC, its clinical benefit remains modest and is counterbalanced by substantial toxicity. Therefore, these regimens may be appropriate only for a small subset of patients with minimal comorbidities.

5.2.3. Tarlatamab

Tarlatamab is a bispecific T-cell engager (BiTE) that simultaneously binds Delta-like protein 3 (DLL3) on tumor cells and CD3 on T-cells. DLL3 is overexpressed in >80% of SCLC and minimally present in normal tissues, making it an attractive therapeutic target. In the phase II DeLLphi-301 study, tarlatamab (10 mg every two weeks) was administered to patients with advanced, previously treated SCLC and showed notable activity, achieving an ORR of 40%, with 58% of responders maintaining their response for at least six months. AEs such as cytokine release syndrome (CRS), fatigue, and fever were reported, with severe events leading to treatment discontinuation in approximately 3% [43]. The FDA approved tarlatamab in May 2024 for adults with ED-SCLC after progression on platinum-based chemotherapy. These findings were confirmed in the phase III DeLLphi-304 trial, where tarlatamab significantly improved OS compared to chemotherapy (13.6 vs. 8.3 months; HR 0.60; 95% CI, 0.47–0.77; p < 0.001). Tarlatamab was also associated with fewer grade ≥ 3 AEs (54% vs. 80%) and lower rates of treatment discontinuations (5% vs. 12%) [44]. An ongoing phase III trial is also evaluating tarlatamab as maintenance therapy after first-line treatment (DeLLphi-305). In summary, tarlatamab is expected to establish itself as the new SOC for second-line SCLC treatment, with EMA approval eagerly anticipated.

5.2.4. Rovalpituzumab Tesirine (Rova-T)

Rova-T is an anti-DLL3 ADC. Early trials showed promising results, with a phase I study reporting an ORR of 18%, and 38% in the subgroup of patients with high DLL3 expression [45]. However, subsequent larger trials were disappointing. The phase III TAHOE study comparing Rova-T to topotecan in second-line therapy was terminated early due to inferior survival in the Rova-T group [46]. The MERU trial demonstrated that Rova-T provided no benefit as a maintenance therapy [47]. Therefore, AbbVie discontinued Rova-T development in August 2019.

5.2.5. ZL-1310

ZL-1310 is a next-generation anti-DLL3 ADC linked to a novel camptothecin derivative (a topoisomerase I inhibitor). In an ongoing phase Ia/b trial (NCT06179069), preliminary results from the dose-escalation phase showed promising activity in previously treated ED-SCLC patients. Objective responses were observed in 19 of 28 patients (68%). Patients with baseline brain metastases had an 80% response rate and 100% disease control rate (DCR) [48]. ZL-1310 was generally well tolerated, with grade ≥ 3 TRAEs reported in 39% of patients, most commonly anemia (21%), neutropenia (18%), thrombocytopenia (11%), and one case of dose-limiting toxicity (DLT). An ongoing phase I trial (NCT06179069) is investigating ZL-1310 as monotherapy and in combination with atezolizumab, with or without carboplatin, for the treatment of ED-SCLC. The agent has received orphan drug designation from the FDA, highlighting its early potential; however, available data remain preliminary and require further validation.

5.2.6. Sacituzumab Govitecan (SG)

This anti-Trop-2 (Tumor-associated calcium signal transducer 2) ADC was evaluated in the phase II TROPiCS-03 trial in patients with recurrent ED-SCLC, administered at 10 mg/kg on days 1 and 8 of a 21-day cycle. ORR was 41.9%, with a median duration of response (DOR) of 4.7 months. The safety profile was manageable. Based on these results, the FDA granted Breakthrough Therapy Designation in December 2024 [49]. The phase III EVOKE-SCLC-04 trial (NCT06801834) is currently evaluating SG as a second-line treatment compared to topotecan; however, enrollment only began in April 2025. It remains unclear how these results will compare once tarlatamab becomes the new SOC in the second-line setting. Additionally, another phase II trial (NCT06667167) is investigating SG as a maintenance therapy in combination with pembrolizumab following first-line induction therapy. In summary, the role of SG in the treatment landscape will need to be further defined.

5.2.7. Ifinatamab Deruxtecan (DS-7300)

This novel ADC targets B7-H3 (B7 Homolog 3, CD276), a transmembrane protein associated with poor prognosis and overexpressed in SCLC. Preclinical data showed strong antitumor activity. In a phase I/II study (NCT04145622), DS-7300 achieved a 52.4% ORR in evaluable SCLC patients and was well tolerated. A phase III trial (IDeate-Lung02, NCT06203210) is now underway to further evaluate its efficacy in relapsed SCLC [50,64].

5.2.8. ABBV-011

ABBV-011 targets Seizure-Related 6 Homolog (SEZ6), a protein overexpressed in SCLC. This ADC combines a SEZ6-specific monoclonal antibody with calicheamicin, linked via a stable, non-cleavable linker. Preclinical studies demonstrated potent activity. In a phase I trial (NCT03639194), ABBV-011 was well tolerated with dose-dependent side effects (fatigue, nausea, thrombocytopenia). At 1.0 mg/kg, the ORR was 25%, with a median DOR of 4.2 months and PFS of 3.5 months. These results support further investigation of SEZ6 as a therapeutic target [51,65].

5.2.9. PARP Inhibitors

Poly ADP-ribose polymerase (PARP) inhibitors are being actively explored in clinical trials for their potential to enhance therapeutic outcomes in SCLC. Building upon preclinical evidence suggesting that PARP inhibition can sensitize SCLC cells to DNA-damaging agents, several studies are assessing combinations of PARP inhibitors with chemotherapy, immunotherapy, and radiation [66]. For example, the ongoing phase I/II PRIO trial (NCT04728230) is evaluating the combination of olaparib with durvalumab, carboplatin, etoposide, and/or radiotherapy in patients with ED-SCLC. Additionally, the SWOG S1929 trial showed that maintenance therapy with talazoparib and atezolizumab improved PFS in patients with Schlafen 11 (SLFN11)-expressing tumors, highlighting the importance of biomarker-driven approaches [52]. Another phase II trial (NCT04701307) is evaluating the combination of niraparib and dostarlimab. Furthermore, Allarity Therapeutics has initiated a phase II trial examining the efficacy of combining stenoparib, a dual PARP and tankyrase inhibitor, with temozolomide in patients with recurrent SCLC, aiming to improve tolerability and therapeutic synergy. However, a key limitation of these approaches is the lack of robust, prospective validation for predictive biomarkers like SLFN11, as well as the need to better define patient subgroups that will truly benefit from PARP inhibitor–based combinations, given the heterogeneous responses and potential for added hematologic and gastrointestinal toxicity in heavily pretreated populations.

5.2.10. BCL2 Inhibitors

BCL-2 inhibitors, including navitoclax and venetoclax, are currently under investigation in SCLC due to the frequent overexpression of BCL-2 in these tumors. Navitoclax (ABT-263) demonstrated limited efficacy as a single agent in a phase II study involving patients with relapsed SCLC, with a partial response (PR) observed in 2.6% of patients and stable disease (SD) in 23%. The median PFS was 1.5 months, and the median OS was 3.2 months. Thrombocytopenia was the most common toxicity, reaching grades 3–4 in 41% of patients [53]. Preclinical studies have shown that venetoclax effectively induces apoptosis in SCLC cells with high BCL-2 expression, indicating potential therapeutic benefit [67]. A phase Ib/II clinical trial aimed to evaluate the safety and efficacy of venetoclax in combination with chemoimmunotherapy in patients with untreated ED-SCLC (NCT04422210), whereas another phase I/II trial aimed to establish the recommended phase II dose for venetoclax when given in combination with irinotecan in patients with relapsed or refractory SCLC (NCT04543916). A major limitation is the limited clinical efficacy and high toxicity observed, along with early trial terminations, which have hindered further evaluation despite promising preclinical data.

5.2.11. Epigenetic Modulators (EZH2 Inhibitors)

In SCLC, EZH2 (Enhancer of Zeste Homolog 2), a histone methyltransferase, is commonly overexpressed and plays a key role in driving tumor progression and resistance to chemotherapy. Ongoing clinical trials are investigating EZH2 inhibitors as potential therapeutic agents in SCLC. A phase I trial (NCT05353439) is investigating whether adding tazemetostat to topotecan and pembrolizumab can improve efficacy in recurrent SCLC. Another investigational agent, mevrometostat (PF-06821497), is being assessed in a phase I trial (NCT03460977) involving patients with SCLC, castration-resistant prostate cancer (CRPC), and follicular lymphoma (FL). The preliminary results indicate that while some patients with FL achieved a PR, SCLC patients experienced disease progression, highlighting the need for further research to optimize treatment strategies [54]. Additionally, valemetostat (DS-3201b) is being evaluated in a phase I/II study for its potential to overcome chemoresistance in SCLC by targeting the EZH2–SLFN11 pathway. Although the combination of valemetostat with irinotecan demonstrated clinical activity, the regimen was not well tolerated due to overlapping toxicities, limiting its further development [55]. Preclinical studies have also explored the use of proteolysis-targeting chimeras (PROTACs) to degrade EZH2, demonstrating the potential to overcome chemo-resistance [68,69]. Collectively, these ongoing trials highlight the potential of EZH2 inhibitors as components of combination therapies to improve outcomes in SCLC. However, in the absence of phase III data, these approaches remain experimental and are currently limited by toxicity and modest efficacy.

6. Recommendations and Future Perspectives

Despite recent advances, SCLC continues to present substantial clinical and translational challenges:
  • A major obstacle remains the lack of durable treatment responses, particularly in the relapsed or refractory setting, where most available therapies offer only modest survival benefits. Rapid disease progression and the early development of treatment resistance further complicate clinical management.
  • The absence of reliable predictive biomarkers significantly limits effective patient selection for emerging therapies. While molecular subtyping shows promise, its clinical implementation is hindered by inconsistent classification systems and the lack of standardized assays.
  • Treatment-related toxicity remains a significant barrier, especially in combination regimens involving chemotherapy, immunotherapy, or targeted agents. Such regimens are often poorly tolerated by patients with limited performance status or significant comorbidities.
  • Clinical trial development is also hampered by slow patient accrual, insufficient biomarker stratification, and early treatment discontinuations, collectively contributing to the slow pace of progress in this aggressive malignancy.
Nevertheless, recent therapeutic developments have reinvigorated the treatment landscape, with novel immunotherapies and ADCs showing particular promise:
  • The bispecific T-cell engager tarlatamab has emerged as a leading candidate, demonstrating improved survival and a favorable safety profile in a recent phase III trial. It is anticipated to become a new standard of care in the second-line setting, with potential use in earlier stages, including consolidation therapy for LD-SCLC, currently under investigation.
  • The addition of lurbinectedin to immunotherapy as maintenance therapy in ED-SCLC remains under evaluation; however, its potential to become a new standard is uncertain given concerns that toxicity may outweigh any survival benefit.
  • ADCs such as sacituzumab govitecan and ZL-1310 have shown promising activity in heavily pretreated populations, including patients with brain metastases. Newer-generation ADCs may offer effective alternatives for patients ineligible for more intensive treatment.
  • By contrast, the future of targeted therapies such as PARP-, BCL-2-, and EZH2-inhibitors remains uncertain, due to modest clinical activity, toxicity concerns, and the early termination of several trials.
  • Future efforts should prioritize the validation of predictive biomarkers (e.g., SLFN11, DLL3, BCL-2), development of rational combination regimens, expansion into earlier treatment settings, and long-term safety assessments of novel agents.
In summary, while targeted therapies remain largely investigational, tarlatamab and ADCs represent the most promising avenues for improving outcomes in SCLC. Sustained investment in biomarker research and innovative trial designs will be essential to achieving meaningful clinical advances.

7. Conclusions

SCLC remains a highly aggressive malignancy with limited therapeutic progress over recent decades. While conventional treatment is still dominated by chemoimmunotherapy, novel agents such as tarlatamab and ADCs are beginning to redefine second-line options, offering improved outcomes and manageable toxicity. However, challenges persist, including rapid disease progression, limited biomarker integration, and toxicity concerns with targeted therapies. Future advances will depend on biomarker-driven strategies, refined combination regimens, and rational trial design. With ongoing research, particularly in molecular profiling and immunotherapeutic innovation, the outlook for patients with SCLC may improve meaningfully in the years ahead.

Author Contributions

Conceptualization, M.K. and L.E.R.; methodology, M.K. and L.E.R.; data curation, M.K. and L.E.R.; writing—original draft preparation, M.K. and L.E.R.; writing—review and editing, G.L., G.E. and A.B.; visualization, M.K. and L.E.R.; supervision, G.E. and A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This project was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under grant number 493624047 (Clinician Scientist CareerS Münster) and the Open Access Publication Fund of the University of Muenster.

Conflicts of Interest

M.K. received honoraria or travel grants not related to this manuscript from Pierre Fabre, Amgen, AstraZeneca, Boehringer Ingelheim, Daiichi Sankyo, Janssen-Cilag, Novartis, Roche Pharma, Takeda Pharma. L.R. received travel grants not related to this manuscript from Daiichi Sankyo. G.E. received honoraria or travel grants not related to this manuscript from BMS, MSD, Pfizer, Bayer, AstraZeneca, Boehringer, Roche, Merck, Takeda, Novocure, Pierre Fabre, Janssen-Cilag, Lilly, Servier, Roche, Celgene, Actelion, CLS Behring, Fresenius, AOP Health, AMGEN, Daiichi-Sankyo. G.L. received research grants not related to this manuscript from AGIOS, AQUINOX, AstraZeneca, Bayer, Celgene, Gilead, Janssen, MorphoSys, Novartis, F. Hoffmann-La Roche Ltd., and Verastem. G.L. received honoraria from ADC Therapeutics, AbbVie, Amgen, AstraZeneca, Bayer, BeiGene, BMS, Celgene, Constellation, Genase, Genmab, Gilead, Hexal/Sandoz, Immagene, Incyte, Janssen, Karyopharm, Lilly, Miltenyi, MorphoSys, MSD, NanoString, Novartis, PentixaPharm, Pierre Fabre, F. Hoffmann-La Roche Ltd., and Sobi. A.B. received honoraria or travel grants not related to this manuscript from Bayer, BMS, Takeda, MSD, Boehringer, AstraZeneca, Sanofi, Pfizer, Lilly, Amgen, RG GmbH, Roche, Novartis, Janssen, Daiichi.

Nomenclature

ADCAntibody-Drug Conjugate
AEAdverse Event
ASCL1Achaete-Scute Family BHLH Transcription Factor 1
BCL-2B-cell lymphoma 2
BiTEBispecific T-cell Engager
B7-H3B7 Homolog 3/CD276
cCRTConcurrent Chemoradiotherapy
CIConfidence Interval
CNSCentral Nervous System
CRPCCastration-Resistant Prostate Cancer
CRSCytokine-Release Syndrome
CRTChemoradiotherapy
CTComputed Tomography
CTFIChemotherapy-Free Interval
CTLA-4Cytotoxic T-Lymphocyte Antigen 4
CTXChemotherapy
DLCODiffusing Capacity of the Lung for Carbon Monoxide
DLL3Delta-like protein 3
DLTDose-Limiting Toxicity
DCRDisease Control Rate
DORDuration Of Response
ED-SCLCExtensive-Disease small cell lung cancer
EMANational Comprehensive Cancer Network
ESMOEuropean Society For Medical Oncology
EZH2Enhancer of Zeste Homolog 2
FDAUnited States Food and Drug Administration
FDG-PETFluorodeoxyglucose Positron Emission Tomography
FEV1Forced Expiratory Volume in 1 Second
FLFollicular Lymphoma
HRHazard Ratio
ICANS Immune Effector Cell-Associated Neurotoxicity Syndrome
IGFR1Insulin-like Growth Factor 1 Receptor
ILDInterstitial Lung Disease
IOImmunotherapy
irAEImmune-Related Adverse Events
LD-SCLCLimited-Disease small cell lung cancer
LDHLactate Dehydrogenase
MRIMagnetic Resonance Imaging
MYCMyelocytomatosis oncogene
NCAMNeural Cell Adhesion Molecule, also known as CD56
NCCNNational Comprehensive Cancer Network
NECNeuroendocrine Carcinoma
NEURO1Neurogenic Differentiation Factor 1
NOTCHNeurogenic locus notch homolog (NOTCH) signaling pathway gene
NSCLCNon-Small Cell Lung Cancer
NSENeuron-Specific Enolase
ORRObjective Response Rate
OSOverall Survival
PARPPoly ADP-ribose Polymerase
PARPiPoly ADP-ribose Polymerase inhibitor
PCIProphylactic Cranial Irradiation
PD-1Programmed Cell Death Protein 1
PD-L1Programmed Death-Ligand 1
PFSProgression-Free Survival
POU2F3POU Class 2 Homeobox 3
PRPartial Response
PROTACProteolysis-Targeting Chimera
PSPerformance Status
RB1Retinoblastoma 1
SCLCSmall cell lung cancer
SDStable Disease
SEZ6Seizure-Related 6 Homolog
SGSacituzumab Govitecan
SIADHSyndrome of Inappropriate Antidiuretic Hormone Secretion
SLFN11Schlafen Family Member 11
SOCStandarf Of Care
TEAETreatment-Emergent Adverse Event
TKITyrosine Kinase Inhibitor
TP53Tumor protein p53
TRAETreatment-Related Adverse Events
Trop-2Tumor-associated calcium signal transducer 2
TTF-1Thyroid Transcription Factor-1
UICCUnion for International Cancer Control
VCVital Capacity
VEGFVascular Endothelial Growth Factor
VODVeno-Occlusive Disease
YAP1Yes-associated protein 1

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  2. Wells, L.E.; Cohen, S.; Brennan, B.; Banerjee, M.; Kalemkerian, G.P. 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. [Google Scholar] [CrossRef]
  3. Uprety, D.; Seaton, R.; Niroula, A.; Hadid, T.; Parikh, K.; Ruterbusch, J.J. Trends in the Incidence and Survival Outcomes in Patients with Small Cell Lung Cancer in the United States: An Analysis of the SEER Database. Cancer Med. 2025, 14, e70608. [Google Scholar] [CrossRef] [PubMed]
  4. Chansky, K.; Detterbeck, F.C.; Nicholson, A.G.; Rusch, V.W.; Vallières, E.; Groome, P.; Kennedy, C.; Krasnik, M.; Peake, M.; Shemanski, L.; et al. The IASLC Lung Cancer Staging Project: External Validation of the Revision of the TNM Stage Groupings in the Eighth Edition of the TNM Classification of Lung Cancer. J. Thorac. Oncol. 2017, 12, 1109–1121. [Google Scholar] [CrossRef] [PubMed]
  5. GBD 2019 Tobacco Collaborators. Spatial, Temporal, and Demographic Patterns in Prevalence of Smoking Tobacco Use and Attributable Disease Burden in 204 Countries and Territories, 1990–2019: A Systematic Analysis from the Global Burden of Disease Study 2019. Lancet 2021, 397, 2337–2360. [Google Scholar] [CrossRef]
  6. Wang, Q.; Gümüş, Z.H.; Colarossi, C.; Memeo, L.; Wang, X.; Kong, C.Y.; Boffetta, P. SCLC: Epidemiology, Risk Factors, Genetic Susceptibility, Molecular Pathology, Screening, and Early Detection. J. Thorac. Oncol. 2023, 18, 31–46. [Google Scholar] [CrossRef]
  7. National Cancer Institute, Surveillance Research Program. SEER*Explorer: An Interactive Website for SEER Cancer Statistics [Internet]. 16 April 2025. Data Source (s): SEER Incidence Data, November 2024 Submission (1975–2022), SEER 21 Registries. 2025. Available online: https://seer.cancer.gov/statistics-network/explorer/application.html (accessed on 25 May 2025).
  8. George, J.; Lim, J.S.; Jang, S.J.; Cun, Y.; Ozretić, L.; Kong, G.; Leenders, F.; Lu, X.; Fernández-Cuesta, L.; Bosco, G.; et al. Comprehensive Genomic Profiles of Small Cell Lung Cancer. Nature 2015, 524, 47–53. [Google Scholar] [CrossRef]
  9. Peifer, M.; Fernández-Cuesta, L.; Sos, M.L.; George, J.; Seidel, D.; Kasper, L.H.; Plenker, D.; Leenders, F.; Sun, R.; Zander, T.; et al. Integrative Genome Analyses Identify Key Somatic Driver Mutations of Small-Cell Lung Cancer. Nat. Genet. 2012, 44, 1104–1110. [Google Scholar] [CrossRef]
  10. Mollaoglu, G.; Guthrie, M.R.; Böhm, S.; Brägelmann, J.; Can, I.; Ballieu, P.M.; Marx, A.; George, J.; Heinen, C.; Chalishazar, M.D.; et al. MYC Drives Progression of Small Cell Lung Cancer to a Variant Neuroendocrine Subtype with Vulnerability to Aurora Kinase Inhibition. Cancer Cell 2017, 31, 270–285. [Google Scholar] [CrossRef]
  11. Ruano-Raviña, A.; Provencio, M.; Calvo de Juan, V.; Carcereny, E.; Moran, T.; Rodriguez-Abreu, D.; López-Castro, R.; Cuadrado Albite, E.; Guirado, M.; Gómez González, L.; et al. Lung Cancer Symptoms at Diagnosis: Results of a Nationwide Registry Study. ESMO Open 2020, 5, e001021. [Google Scholar] [CrossRef]
  12. Giometto, B.; Grisold, W.; Vitaliani, R.; Graus, F.; Honnorat, J.; Bertolini, G. PNS Euronetwork Paraneoplastic Neurologic Syndrome in the PNS Euronetwork Database: A European Study from 20 Centers. Arch. Neurol. 2010, 67, 330–335. [Google Scholar] [CrossRef] [PubMed]
  13. Gozzard, P.; Woodhall, M.; Chapman, C.; Nibber, A.; Waters, P.; Vincent, A.; Lang, B.; Maddison, P. Paraneoplastic Neurologic Disorders in Small Cell Lung Carcinoma: A Prospective Study. Neurology 2015, 85, 235–239. [Google Scholar] [CrossRef]
  14. Soomro, Z.; Youssef, M.; Yust-Katz, S.; Jalali, A.; Patel, A.J.; Mandel, J. Paraneoplastic Syndromes in Small Cell Lung Cancer. J. Thorac. Dis. 2020, 12, 6253–6263. [Google Scholar] [CrossRef] [PubMed]
  15. Iams, W.T.; Shiuan, E.; Meador, C.B.; Roth, M.; Bordeaux, J.; Vaupel, C.; Boyd, K.L.; Summitt, I.B.; Wang, L.L.; Schneider, J.T.; et al. Improved Prognosis and Increased Tumor-Infiltrating Lymphocytes in Patients Who Have SCLC with Neurologic Paraneoplastic Syndromes. J. Thorac. Oncol. 2019, 14, 1970–1981. [Google Scholar] [CrossRef]
  16. Dingemans, A.-M.C.; Früh, M.; Ardizzoni, A.; Besse, B.; Faivre-Finn, C.; Hendriks, L.E.; Lantuejoul, S.; Peters, S.; Reguart, N.; Rudin, C.M.; et al. Small-Cell Lung Cancer: ESMO Clinical Practice Guidelines for Diagnosis, Treatment and Follow-Up. Ann. Oncol. 2021, 32, 839–853. [Google Scholar] [CrossRef]
  17. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®): Small Cell Lung Cancer [v.3.2025]. 2025. Available online: https://www.nccn.org/Guidelines/Guidelines-Detail?Category=1&id=1462 (accessed on 25 May 2025).
  18. Arriola, E.; Trigo, J.M.; Sánchez-Gastaldo, A.; Navarro, A.; Perez, C.; Crama, L.; Ponce-Aix, S. Prognostic Value of Clinical Staging According to TNM in Patients with SCLC: A Real-World Surveillance Epidemiology and End-Results Database Analysis. JTO Clin. Res. Rep. 2022, 3, 100266. [Google Scholar] [CrossRef] [PubMed]
  19. Shepherd, F.A.; Crowley, J.; Van Houtte, P.; Postmus, P.E.; Carney, D.; Chansky, K.; Shaikh, Z.; Goldstraw, P. International Association for the Study of Lung Cancer International Staging Committee and Participating Institutions The International Association for the Study of Lung Cancer Lung Cancer Staging Project: Proposals Regarding the Clinical Staging of Small Cell Lung Cancer in the Forthcoming (Seventh) Edition of the Tumor, Node, Metastasis Classification for Lung Cancer. J. Thorac. Oncol. 2007, 2, 1067–1077. [Google Scholar] [CrossRef]
  20. Nicholson, A.G.; Chansky, K.; Crowley, J.; Beyruti, R.; Kubota, K.; Turrisi, A.; Eberhardt, W.E.E.; van Meerbeeck, J.; Rami-Porta, R.; Staging and Prognostic Factors Committee, Advisory Boards, and Participating Institutions; et al. The International Association for the Study of Lung Cancer Lung Cancer Staging Project: Proposals for the Revision of the Clinical and Pathologic Staging of Small Cell Lung Cancer in the Forthcoming Eighth Edition of the TNM Classification for Lung Cancer. J. Thorac. Oncol. 2016, 11, 300–311. [Google Scholar] [CrossRef]
  21. Gay, C.M.; Stewart, C.A.; Park, E.M.; Diao, L.; Groves, S.M.; Heeke, S.; Nabet, B.Y.; Fujimoto, J.; Solis, L.M.; Lu, W.; et al. Patterns of Transcription Factor Programs and Immune Pathway Activation Define Four Major Subtypes of SCLC with Distinct Therapeutic Vulnerabilities. Cancer Cell 2021, 39, 346–360.e7. [Google Scholar] [CrossRef]
  22. Chen, P.; Sun, C.; Wang, H.; Zhao, W.; Wu, Y.; Guo, H.; Zhou, C.; He, Y. YAP1 Expression Is Associated with Survival and Immunosuppression in Small Cell Lung Cancer. Cell Death Dis. 2023, 14, 636. [Google Scholar] [CrossRef]
  23. Owonikoko, T.K.; Dwivedi, B.; Chen, Z.; Zhang, C.; Barwick, B.; Ernani, V.; Zhang, G.; Gilbert-Ross, M.; Carlisle, J.; Khuri, F.R.; et al. YAP1 Expression in SCLC Defines a Distinct Subtype with T-Cell-Inflamed Phenotype. J. Thorac. Oncol. 2021, 16, 464–476. [Google Scholar] [CrossRef] [PubMed]
  24. Ng, J.; Cai, L.; Girard, L.; Prall, O.W.J.; Rajan, N.; Khoo, C.; Batrouney, A.; Byrne, D.J.; Boyd, D.K.; Kersbergen, A.J.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  25. Baine, M.K.; Hsieh, M.-S.; Lai, W.V.; Egger, J.V.; Jungbluth, A.A.; Daneshbod, Y.; Beras, A.; Spencer, R.; Lopardo, J.; Bodd, F.; et al. SCLC Subtypes Defined by ASCL1, NEUROD1, POU2F3, and YAP1: A Comprehensive Immunohistochemical and Histopathologic Characterization. J. Thorac. Oncol. 2020, 15, 1823–1835. [Google Scholar] [CrossRef]
  26. Mahadevan, N.R.; Knelson, E.H.; Wolff, J.O.; Vajdi, A.; Saigí, M.; Campisi, M.; Hong, D.; Thai, T.C.; Piel, B.; Han, S.; et al. Intrinsic Immunogenicity of Small Cell Lung Carcinoma Revealed by Its Cellular Plasticity. Cancer Discov. 2021, 11, 1952–1969. [Google Scholar] [CrossRef]
  27. Velut, Y.; Arqué, B.; Wislez, M.; Blons, H.; Burroni, B.; Prieto, M.; Beau, S.; Fournel, L.; Birsen, G.; Cremer, I.; et al. The Tumor Immune Microenvironment of SCLC Is Not Associated with Its Molecular Subtypes. Eur. J. Cancer 2024, 212, 115067. [Google Scholar] [CrossRef]
  28. Peressini, M.; Garcia-Campelo, R.; Massuti, B.; Martí, C.; Cobo, M.; Gutiérrez, V.; Dómine, M.; Fuentes, J.; Majem, M.; de Castro, J.; et al. Spatially Preserved Multi-Region Transcriptomic Subtyping and Biomarkers of Chemoimmunotherapy Outcome in Extensive-Stage Small Cell Lung Cancer. Clin. Cancer Res. 2024, 30, 3036–3049. [Google Scholar] [CrossRef]
  29. Duplaquet, L.; Li, Y.; Booker, M.A.; Xie, Y.; Olsen, S.N.; Patel, R.A.; Hong, D.; Hatton, C.; Denize, T.; Walton, E.; et al. KDM6A Epigenetically Regulates Subtype Plasticity in Small Cell Lung Cancer. Nat. Cell Biol. 2023, 25, 1346–1358. [Google Scholar] [CrossRef]
  30. Jones, G.S.; Elimian, K.; Baldwin, D.R.; Hubbard, R.B.; McKeever, T.M. A Systematic Review of Survival Following Anti-Cancer Treatment for Small Cell Lung Cancer. Lung Cancer 2020, 141, 44–55. [Google Scholar] [CrossRef] [PubMed]
  31. Shiono, A.; Imai, H.; Endo, S.; Katayama, K.; Sato, H.; Hashimoto, K.; Miura, Y.; Okazaki, S.; Abe, T.; Mouri, A.; et al. A Retrospective Evaluation of Therapeutic Efficacy and Safety of Chemoradiotherapy in Older Patients (Aged ≥ 75 Years) with Limited-Disease Small Cell Lung Cancer: Insights from Two Institutions and Review of the Literature. Radiol. Oncol. 2024, 58, 432–443. [Google Scholar] [CrossRef]
  32. Faivre-Finn, C.; Snee, M.; Ashcroft, L.; Appel, W.; Barlesi, F.; Bhatnagar, A.; Bezjak, A.; Cardenal, F.; Fournel, P.; Harden, S.; et al. 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. [Google Scholar] [CrossRef]
  33. Grønberg, B.H.; Killingberg, K.T.; Fløtten, Ø.; Brustugun, O.T.; Hornslien, K.; Madebo, T.; Langer, S.W.; Schytte, T.; Nyman, J.; Risum, S.; et al. High-Dose versus Standard-Dose Twice-Daily Thoracic Radiotherapy for Patients with Limited Stage Small-Cell Lung Cancer: An Open-Label, Randomised, Phase 2 Trial. Lancet Oncol. 2021, 22, 321–331. [Google Scholar] [CrossRef] [PubMed]
  34. Grønberg, B.H.; Halvorsen, T.O.; Fløtten, Ø.; Brustugun, O.T.; Brunsvig, P.F.; Aasebø, U.; Bremnes, R.M.; Tollåli, T.; Hornslien, K.; Aksnessæther, B.Y.; et al. Randomized Phase II Trial Comparing Twice Daily Hyperfractionated with Once Daily Hypofractionated Thoracic Radiotherapy in Limited Disease Small Cell Lung Cancer. Acta Oncol. 2016, 55, 591–597. [Google Scholar] [CrossRef] [PubMed]
  35. Aupérin, A.; Arriagada, R.; Pignon, J.P.; Le Péchoux, C.; Gregor, A.; Stephens, R.J.; Kristjansen, P.E.; Johnson, B.E.; Ueoka, H.; Wagner, H.; et al. Prophylactic Cranial Irradiation for Patients with Small-Cell Lung Cancer in Complete Remission. Prophylactic Cranial Irradiation Overview Collaborative Group. N. Engl. J. Med. 1999, 341, 476–484. [Google Scholar] [CrossRef]
  36. Cheng, Y.; Spigel, D.R.; Cho, B.C.; Laktionov, K.K.; Fang, J.; Chen, Y.; Zenke, Y.; Lee, K.H.; Wang, Q.; Navarro, A.; et al. Durvalumab after Chemoradiotherapy in Limited-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 2024, 391, 1313–1327. [Google Scholar] [CrossRef] [PubMed]
  37. Horn, L.; Mansfield, A.S.; Szczęsna, A.; Havel, L.; Krzakowski, M.; Hochmair, M.J.; Huemer, F.; Losonczy, G.; Johnson, M.L.; Nishio, M.; et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2220–2229. [Google Scholar] [CrossRef]
  38. Paz-Ares, L.; Dvorkin, M.; Chen, Y.; Reinmuth, N.; Hotta, K.; Trukhin, D.; Statsenko, G.; Hochmair, M.J.; Özgüroğlu, M.; Ji, J.H.; et al. 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. [Google Scholar] [CrossRef]
  39. Cheng, Y.; Han, L.; Wu, L.; Chen, J.; Sun, H.; Wen, G.; Ji, Y.; Dvorkin, M.; Shi, J.; Pan, Z.; et al. Effect of First-Line Serplulimab vs Placebo Added to Chemotherapy on Survival in Patients with Extensive-Stage Small Cell Lung Cancer: The ASTRUM-005 Randomized Clinical Trial. JAMA 2022, 328, 1223–1232. [Google Scholar] [CrossRef] [PubMed]
  40. Trigo, J.; Subbiah, V.; Besse, B.; Moreno, V.; López, R.; Sala, M.A.; Peters, S.; Ponce, S.; Fernández, C.; Alfaro, V.; et al. Lurbinectedin as Second-Line Treatment for Patients with Small-Cell Lung Cancer: A Single-Arm, Open-Label, Phase 2 Basket Trial. Lancet Oncol. 2020, 21, 645–654. [Google Scholar] [CrossRef] [PubMed]
  41. Aix, S.P.; Ciuleanu, T.E.; Navarro, A.; Cousin, S.; Bonanno, L.; Smit, E.F.; Chiappori, A.; Olmedo, M.E.; Horvath, I.; Grohé, C.; et al. Combination Lurbinectedin and Doxorubicin versus Physician’s Choice of Chemotherapy in Patients with Relapsed Small-Cell Lung Cancer (ATLANTIS): A Multicentre, Randomised, Open-Label, Phase 3 Trial. Lancet Respir. Med. 2023, 11, 74–86. [Google Scholar] [CrossRef]
  42. Peters, S.; Pujol, J.-L.; Dafni, U.; Dómine, M.; Popat, S.; Reck, M.; Andrade, J.; Becker, A.; Moro-Sibilot, D.; Curioni-Fontecedro, A.; et al. Consolidation Nivolumab and Ipilimumab versus Observation in Limited-Disease Small-Cell Lung Cancer after Chemo-Radiotherapy—Results from the Randomised Phase II ETOP/IFCT 4-12 STIMULI Trial. Ann. Oncol. 2022, 33, 67–79. [Google Scholar] [CrossRef]
  43. Ahn, M.-J.; Cho, B.C.; Felip, E.; Korantzis, I.; Ohashi, K.; Majem, M.; Juan-Vidal, O.; Handzhiev, S.; Izumi, H.; Lee, J.-S.; et al. Tarlatamab for Patients with Previously Treated Small-Cell Lung Cancer. N. Engl. J. Med. 2023, 389, 2063–2075. [Google Scholar] [CrossRef] [PubMed]
  44. Mountzios, G.; Sun, L.; Cho, B.C.; Demirci, U.; Baka, S.; Gümüş, M.; Lugini, A.; Zhu, B.; Yu, Y.; Korantzis, I.; et al. Tarlatamab in Small-Cell Lung Cancer after Platinum-Based Chemotherapy. N. Engl. J. Med. 2025. [Google Scholar] [CrossRef]
  45. Rudin, C.M.; Pietanza, M.C.; Bauer, T.M.; Ready, N.; Morgensztern, D.; Glisson, B.S.; Byers, L.A.; Johnson, M.L.; Burris, H.A.; Robert, F.; et al. Rovalpituzumab Tesirine, a DLL3-Targeted Antibody-Drug Conjugate, in Recurrent Small-Cell Lung Cancer: A First-in-Human, First-in-Class, Open-Label, Phase 1 Study. Lancet Oncol. 2017, 18, 42–51. [Google Scholar] [CrossRef]
  46. Blackhall, F.; Jao, K.; Greillier, L.; Cho, B.C.; Penkov, K.; Reguart, N.; Majem, M.; Nackaerts, K.; Syrigos, K.; Hansen, K.; et al. Efficacy and Safety of Rovalpituzumab Tesirine Compared with Topotecan as Second-Line Therapy in DLL3-High SCLC: Results From the Phase 3 TAHOE Study. J. Thorac. Oncol. 2021, 16, 1547–1558. [Google Scholar] [CrossRef]
  47. Johnson, M.L.; Zvirbule, Z.; Laktionov, K.; Helland, A.; Cho, B.C.; Gutierrez, V.; Colinet, B.; Lena, H.; Wolf, M.; Gottfried, M.; et al. Rovalpituzumab Tesirine as a Maintenance Therapy After First-Line Platinum-Based Chemotherapy in Patients with Extensive-Stage-SCLC: Results from the Phase 3 MERU Study. J. Thorac. Oncol. 2021, 16, 1570–1581. [Google Scholar] [CrossRef] [PubMed]
  48. Patel, M.R.; Wu, Y.-L.L.C.; Wang, Z.; Rocha, P.; Wang, Q.; Du, Y.; Dy, G.K.; Dowlati, A.; Spira, A.; Dong, X.; et al. ZL-1310, a DLL3 ADC, in Patients with Extensive Stage Small Cell Lung Cancer: Ph1 Trial Update. JCO 2025, 43, 3041. [Google Scholar] [CrossRef]
  49. Dowlati, A.; Chiang, A.C.; Cervantes, A.; Babu, S.; Hamilton, E.; Wong, S.F.; Tazbirkova, A.; Sullivan, I.G.; van Marcke, C.; Italiano, A.; et al. Phase 2 Open-Label Study of Sacituzumab Govitecan as Second-Line Therapy in Patients with Extensive-Stage SCLC: Results From TROPiCS-03. J. Thorac. Oncol. 2025, 20, 799–808. [Google Scholar] [CrossRef] [PubMed]
  50. Johnson, M.; Awad, M.; Koyama, T.; Gutierrez, M.; Falchook, G.S.; Piha-Paul, S.A.; Doi, T.; Satoh, T.; Okamoto, N.; Singh, J.; et al. OA05.05 Ifinatamab Deruxtecan (I-DXd; DS-7300) in Patients with Refractory SCLC: A Subgroup Analysis of a Phase 1/2 Study. J. Thorac. Oncol. 2023, 18, S54–S55. [Google Scholar] [CrossRef]
  51. Morgensztern, D.; Ready, N.; Johnson, M.L.; Dowlati, A.; Choudhury, N.; Carbone, D.P.; Schaefer, E.; Arnold, S.M.; Puri, S.; Piotrowska, Z.; et al. A Phase I First-in-Human Study of ABBV-011, a Seizure-Related Homolog Protein 6-Targeting Antibody-Drug Conjugate, in Patients with Small Cell Lung Cancer. Clin. Cancer Res. 2024, 30, 5042–5052. [Google Scholar] [CrossRef]
  52. Karim, N.A.; Miao, J.; Reckamp, K.L.; Gay, C.M.; Byers, L.A.; Zhao, Y.-Q.; Redman, M.W.; Carrizosa, D.R.; Wang, W.-L.; Petty, W.J.; et al. Phase II Randomized Study of Maintenance Atezolizumab Versus Atezolizumab Plus Talazoparib in Patients with SLFN11 Positive Extensive-Stage SCLC: S1929. J. Thorac. Oncol. 2025, 20, 383–394. [Google Scholar] [CrossRef]
  53. Rudin, C.M.; Hann, C.L.; Garon, E.B.; Ribeiro de Oliveira, M.; Bonomi, P.D.; Camidge, D.R.; Chu, Q.; Giaccone, G.; Khaira, D.; Ramalingam, S.S.; et al. Phase II Study of Single-Agent Navitoclax (ABT-263) and Biomarker Correlates in Patients with Relapsed Small Cell Lung Cancer. Clin. Cancer Res. 2012, 18, 3163–3169. [Google Scholar] [CrossRef] [PubMed]
  54. Schweizer, M.; Penkov, K.D.; Tolcher, A.W.; Choudhury, A.D.; Doronin, V.; Aljumaily, R.; Calvo, E.; Frank, R.C.; Hamm, J.T.; Moreno Garcia, V.; et al. 488P Phase I Trial of PF-06821497, a Potent and Selective Inhibitor of Enhancer of Zeste Homolog 2 (EZH2), in Follicular Lymphoma (FL), Small Cell Lung Cancer (SCLC) and Castration-Resistant Prostate Cancer (CRPC). Ann. Oncol. 2022, 33, S763–S764. [Google Scholar] [CrossRef]
  55. Choudhury, N.J.; Lai, W.V.; Makhnin, A.; Heller, G.; Eng, J.; Li, B.; Preeshagul, I.; Santini, F.C.; Offin, M.; Ng, K.; et al. 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. 2024, 30, 3697–3703. [Google Scholar] [CrossRef]
  56. Zugazagoitia, J.; Falcon Gonzalez, A.; Navarro Mendivil, A.F.; Sanchez Gastaldo, A.; Simoes da Rocha, P.F.; Cote, G.M.; Bockorny, B.; Molina Cerrillo, J.; Artal, A.; Baena Espinar, J.; et al. 1790P Phase II Data of Lurbinectedin (LUR) and Irinotecan (IRI) in Relapsed Small Cell Lung Cancer (SCLC) Patients (Pts) with Chemotherapy-Free Interval (CTFI)>30 Days (d). Ann. Oncol. 2024, 35, S1064–S1065. [Google Scholar] [CrossRef]
  57. Paz-Ares, L.G.; Borghaei, H.; Liu, S.V.; Peters, S.; Herbst, R.S.; Stencel, K.M.; Majem, M.; Czyz, G.; Caro, R.B.; Lee, K.H.; et al. Lurbinectedin (Lurbi) + Atezolizumab (Atezo) as First-Line (1 L) Maintenance Treatment (Tx) in Patients (Pts) with Extensive-Stage Small Cell Lung Cancer (ES-SCLC): Primary Results of the Phase 3 IMforte Trial. In Proceedings of the 2025 American Society of Clinical Oncology Annual Meeting, Chicago, IL, USA, 31 May–5 June 2025. Abstract 8006. 2025. [Google Scholar]
  58. Liu, S.V.; Reck, M.; Mansfield, A.S.; Mok, T.; Scherpereel, A.; Reinmuth, N.; Garassino, M.C.; De Castro Carpeno, J.; Califano, R.; Nishio, M.; et al. Updated Overall Survival and PD-L1 Subgroup Analysis of Patients with Extensive-Stage Small-Cell Lung Cancer Treated with Atezolizumab, Carboplatin, and Etoposide (IMpower133). J. Clin. Oncol. 2021, 39, 619–630. [Google Scholar] [CrossRef]
  59. Leal, J.F.M.; Martínez-Díez, M.; García-Hernández, V.; Moneo, V.; Domingo, A.; Bueren-Calabuig, J.A.; Negri, A.; Gago, F.; Guillén-Navarro, M.J.; Avilés, P.; et al. PM01183, a New DNA Minor Groove Covalent Binder with Potent in Vitro and in Vivo Anti-Tumour Activity. Br. J. Pharmacol. 2010, 161, 1099–1110. [Google Scholar] [CrossRef] [PubMed]
  60. Santamaría Nuñez, G.; Robles, C.M.G.; Giraudon, C.; Martínez-Leal, J.F.; Compe, E.; Coin, F.; Aviles, P.; Galmarini, C.M.; Egly, J.-M. Lurbinectedin Specifically Triggers the Degradation of Phosphorylated RNA Polymerase II and the Formation of DNA Breaks in Cancer Cells. Mol. Cancer Ther. 2016, 15, 2399–2412. [Google Scholar] [CrossRef]
  61. Allavena, P.; Belgiovine, C.; Digifico, E.; Frapolli, R.; D’Incalci, M. Effects of the Anti-Tumor Agents Trabectedin and Lurbinectedin on Immune Cells of the Tumor Microenvironment. Front. Oncol. 2022, 12, 851790. [Google Scholar] [CrossRef]
  62. Ready, N.E.; Ott, P.A.; Hellmann, M.D.; Zugazagoitia, J.; Hann, C.L.; de Braud, F.; Antonia, S.J.; Ascierto, P.A.; Moreno, V.; Atmaca, A.; et al. Nivolumab Monotherapy and Nivolumab Plus Ipilimumab in Recurrent Small Cell Lung Cancer: Results From the CheckMate 032 Randomized Cohort. J. Thorac. Oncol. 2020, 15, 426–435. [Google Scholar] [CrossRef]
  63. Paz-Ares, L.; Chen, Y.; Reinmuth, N.; Hotta, K.; Trukhin, D.; Statsenko, G.; Hochmair, M.J.; Özgüroğlu, M.; Ji, J.H.; Garassino, M.C.; et al. Durvalumab, with or without Tremelimumab, plus Platinum-Etoposide in First-Line Treatment of Extensive-Stage Small-Cell Lung Cancer: 3-Year Overall Survival Update from CASPIAN. ESMO Open 2022, 7, 100408. [Google Scholar] [CrossRef]
  64. Yamato, M.; Hasegawa, J.; Maejima, T.; Hattori, C.; Kumagai, K.; Watanabe, A.; Nishiya, Y.; Shibutani, T.; Aida, T.; Hayakawa, I.; et al. DS-7300a, a DNA Topoisomerase I Inhibitor, DXd-Based Antibody-Drug Conjugate Targeting B7-H3, Exerts Potent Antitumor Activities in Preclinical Models. Mol. Cancer Ther. 2022, 21, 635–646. [Google Scholar] [CrossRef] [PubMed]
  65. Wiedemeyer, W.R.; Gavrilyuk, J.; Schammel, A.; Zhao, X.; Sarvaiya, H.; Pysz, M.; Gu, C.; You, M.; Isse, K.; Sullivan, T.; et al. ABBV-011, A Novel, Calicheamicin-Based Antibody-Drug Conjugate, Targets SEZ6 to Eradicate Small Cell Lung Cancer Tumors. Mol. Cancer Ther. 2022, 21, 986–998. [Google Scholar] [CrossRef]
  66. Barayan, R.; Ran, X.; Lok, B.H. PARP Inhibitors for Small Cell Lung Cancer and Their Potential for Integration into Current Treatment Approaches. J. Thorac. Dis. 2020, 12, 6240–6252. [Google Scholar] [CrossRef] [PubMed]
  67. Lochmann, T.L.; Floros, K.V.; Naseri, M.; Powell, K.M.; Cook, W.; March, R.J.; Stein, G.T.; Greninger, P.; Maves, Y.K.; Saunders, L.R.; et al. Venetoclax Is Effective in Small-Cell Lung Cancers with High BCL-2 Expression. Clin. Cancer Res. 2018, 24, 360–369. [Google Scholar] [CrossRef] [PubMed]
  68. Khan, S.; Cao, L.; Wiegand, J.; Zhang, P.; Zajac-Kaye, M.; Kaye, F.J.; Zheng, G.; Zhou, D. PROTAC-Mediated Dual Degradation of BCL-xL and BCL-2 Is a Highly Effective Therapeutic Strategy in Small-Cell Lung Cancer. Cells 2024, 13, 528. [Google Scholar] [CrossRef]
  69. Shi, M.-X.; Ding, X.; Tang, L.; Cao, W.-J.; Su, B.; Zhang, J. PROTAC EZH2 Degrader-1 Overcomes the Resistance of Podophyllotoxin Derivatives in Refractory Small Cell Lung Cancer with Leptomeningeal Metastasis. BMC Cancer 2024, 24, 504. [Google Scholar] [CrossRef]
Figure 1. Flowchart illustrating the thematic structure of this review. Abbreviations used in this figure include ADCs (antibody-drug conjugates), BCL-2 (B-cell lymphoma 2), B7-H3 (B7 Homolog 3, CD276), cCRT (concurrent chemoradiotherapy), CTX (chemotherapy), DLL3 (Delta-like protein 3), ED (Extensive Disease), EZH2 (enhancer of zeste homolog 2), IO (immunotherapy), LD (Limited Disease), PARPi (poly ADP-ribose polymerase inhibitor), SEZ6 (seizure protein 6 homolog), SG (sacituzumab govitecan), SOC (standard of care), TKI (Tyrosine Kinase Inhibitor), Trop-2 (Tumor-associated calcium signal transducer 2), and VEGF (Vascular Endothelial Growth Factor).
Figure 1. Flowchart illustrating the thematic structure of this review. Abbreviations used in this figure include ADCs (antibody-drug conjugates), BCL-2 (B-cell lymphoma 2), B7-H3 (B7 Homolog 3, CD276), cCRT (concurrent chemoradiotherapy), CTX (chemotherapy), DLL3 (Delta-like protein 3), ED (Extensive Disease), EZH2 (enhancer of zeste homolog 2), IO (immunotherapy), LD (Limited Disease), PARPi (poly ADP-ribose polymerase inhibitor), SEZ6 (seizure protein 6 homolog), SG (sacituzumab govitecan), SOC (standard of care), TKI (Tyrosine Kinase Inhibitor), Trop-2 (Tumor-associated calcium signal transducer 2), and VEGF (Vascular Endothelial Growth Factor).
Cancers 17 02256 g001
Figure 2. Graphical overview of emerging therapeutic strategies in small cell lung cancer (SCLC). This schematic illustrates key classes of novel agents under investigation for the treatment of SCLC. Antibody-drug conjugates (ADCs) such as Rova-T, ZL-1310, sacituzumab govitecan, infinatamab-deruxtecan, and ABBV-011 deliver targeted cytotoxic payloads to tumor-associated antigens. Tarlatamab represents a novel bispecific T-cell engager targeting DLL3. Additional compounds include inhibitors of anti-apoptotic BCL2 proteins (venetoclax, navitoclax), epigenetic regulators EZH1/2 (tazemetostat, merumetostat, valemetostat), and DNA repair enzymes PARP (olaparib, talazoparib, niraparib). Lurbinectedin, a cytotoxic agent, induces DNA damage independently of targeted pathways. Image created in BioRender. Reitnauer, L. (2025) https://BioRender.com/miyx8lj (accessed on 25 May 2025).
Figure 2. Graphical overview of emerging therapeutic strategies in small cell lung cancer (SCLC). This schematic illustrates key classes of novel agents under investigation for the treatment of SCLC. Antibody-drug conjugates (ADCs) such as Rova-T, ZL-1310, sacituzumab govitecan, infinatamab-deruxtecan, and ABBV-011 deliver targeted cytotoxic payloads to tumor-associated antigens. Tarlatamab represents a novel bispecific T-cell engager targeting DLL3. Additional compounds include inhibitors of anti-apoptotic BCL2 proteins (venetoclax, navitoclax), epigenetic regulators EZH1/2 (tazemetostat, merumetostat, valemetostat), and DNA repair enzymes PARP (olaparib, talazoparib, niraparib). Lurbinectedin, a cytotoxic agent, induces DNA damage independently of targeted pathways. Image created in BioRender. Reitnauer, L. (2025) https://BioRender.com/miyx8lj (accessed on 25 May 2025).
Cancers 17 02256 g002
Table 1. Overview of the TNM classification and UICC 8th edition staging system for small cell lung cancer (SCLC), including 5-year survival rates based on data from the National Cancer Database (NCDB) [4].
Table 1. Overview of the TNM classification and UICC 8th edition staging system for small cell lung cancer (SCLC), including 5-year survival rates based on data from the National Cancer Database (NCDB) [4].
TNM/UICC StageTumor Characteristics5-Year Survival Rate
Limited Disease
Stage I (IA, IB)Early-stage tumor: T1–T2 (≤5 cm or >5 cm but confined to one lobe), N0, M0~13–25%
Stage II (IIA, IIB)Locally advanced tumor: T2–T3 (infiltrating adjacent structures), N0 or N1, M0~17–21%
Stage III (IIIA, IIIB, IIIC)Advanced local tumor: T3–T4 (e.g., invasion of chest wall, vessels, or other lobes), N1–N3, M0~9–13%
Extensive Disease
Stage IVPresence of distant metastases (M1)<5%
Table 2. Overview of clinical trials and recent approvals in the treatment of small cell lung cancer (SCLC). Abbreviations used in this table include cCRT (concurrent chemoradiotherapy), CTFI (chemotherapy free interval), ED (Extensive Disease), LD (Limited Disease), OS (overall survival), PD1 (programmed death 1 protein) PD-L1 (programmed death ligand 1 protein), and SOC (standard of care).
Table 2. Overview of clinical trials and recent approvals in the treatment of small cell lung cancer (SCLC). Abbreviations used in this table include cCRT (concurrent chemoradiotherapy), CTFI (chemotherapy free interval), ED (Extensive Disease), LD (Limited Disease), OS (overall survival), PD1 (programmed death 1 protein) PD-L1 (programmed death ligand 1 protein), and SOC (standard of care).
TrialNInterventional ArmControl ArmIndicationKey ResultsLimitationsClinical
Implications
Reference
IMpower133
(phase I/III)
403Atezolizumab +
carboplatin/
etoposide
Placebo +
carboplatin/
etoposide
First-line,
ED-SCLC
Significant improvement in OS: 12.3 vs. 10.3 monthsLimited biomarker stratificationFirst-line anti-PD-L1 treatment option in ED-SCLC (SOC)[37]
CASPIAN
(phase III)
805Durvalumab +
platinum/etoposide
(±tremelimumab)
Platinum/
etoposide
First-line,
ED-SCLC
Significant improvement in OS: 13.0 vs. 10.5 monthsAddition of tremelimumab without extra benefitFirst-line anti-PD-L1 treatment option in ED-SCLC (SOC)[38]
ASTRUM-005
(phase III)
585Serplulimab +
carboplatin/
etoposide
Placebo +
carboplatin/
etoposide
First-line,
ED-SCLC
Significant improvement in OS: 15.4 vs. 10.9 monthsNo direct comparison to PD-L1 inhibitorsNovel anti-PD1 first-line treatment option in ED-SCLC[39]
ADRIATIC
(phase III)
600Durvalumab
(±tremelimumab)
PlaceboConsolidation post-cCRT,
LD-SCLC
Significant improvement in OS: 55.9 vs. 33.4 monthsData on dual checkpoint inhibition still pendingSOC for consolidation therapy post-cCRT in LD-SCLC[36]
ATLANTIS
(phase III)
613Lurbinectedin +
doxorubicin
Topotecan or
CAV (cyclophosphamide, doxorubicin, vincristine)
Second-line SCLCNo significant improvement in OS: 8.6 vs. 7.6 monthsNo survival benefit; heterogeneity in patient selection; no approvalSigns of better tolerability; CTFI proved as prognostic/predictive[40,41]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kemper, M.; Reitnauer, L.E.; Lenz, G.; Evers, G.; Bleckmann, A. Redefining the Fight Against SCLC: Standards, Innovations, and New Horizons. Cancers 2025, 17, 2256. https://doi.org/10.3390/cancers17132256

AMA Style

Kemper M, Reitnauer LE, Lenz G, Evers G, Bleckmann A. Redefining the Fight Against SCLC: Standards, Innovations, and New Horizons. Cancers. 2025; 17(13):2256. https://doi.org/10.3390/cancers17132256

Chicago/Turabian Style

Kemper, Marcel, Lea Elisabeth Reitnauer, Georg Lenz, Georg Evers, and Annalen Bleckmann. 2025. "Redefining the Fight Against SCLC: Standards, Innovations, and New Horizons" Cancers 17, no. 13: 2256. https://doi.org/10.3390/cancers17132256

APA Style

Kemper, M., Reitnauer, L. E., Lenz, G., Evers, G., & Bleckmann, A. (2025). Redefining the Fight Against SCLC: Standards, Innovations, and New Horizons. Cancers, 17(13), 2256. https://doi.org/10.3390/cancers17132256

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

Article Metrics

Back to TopTop