Download PDF
Review  |  Open Access  |  27 Dec 2024

Mechanisms of immunotherapy resistance in small cell lung cancer

Views: 74 |  Downloads: 10 |  Cited:  0
Cancer Drug Resist 2024;7:55.
10.20517/cdr.2024.154 |  © The Author(s) 2024.
Author Information
Article Notes
Cite This Article

Abstract

Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a poor prognosis. Although the addition of immunotherapy to chemotherapy has modestly improved outcomes, most patients rapidly develop resistance. Resistance to immunotherapy can be broadly categorized into primary resistance and acquired resistance, as proposed by the Society for Immunotherapy of Cancer (SITC) consensus definition. Primary resistance occurs in the setting of failure to respond to immune checkpoint inhibitors (ICIs), while acquired resistance develops after initial response. The mechanisms of acquired and primary resistance to ICI are not well understood in SCLC, denoting an area of critical unmet need.

Both intrinsic and extrinsic mechanisms play significant roles in immunotherapy resistance. Intrinsic mechanisms include defects in antigen presentation, mutations in key genes, reduced tumor immunogenicity, and epigenetic alterations. Extrinsic mechanisms involve the tumor microenvironment (TME), which is a complex interplay of both tumor- and immunosuppressive immune cells, vasculature, and microbiome.

An understanding of these resistance mechanisms is crucial for developing novel therapeutic strategies to advance effective immunotherapy in patients with SCLC, a critical area of unmet need.

Keywords

Immunotherapy resistance, small cell lung cancer, biomarker, primary resistance, acquired resistance

INTRODUCTION

Lung cancer remains the leading cause of cancer-related deaths worldwide, with an estimated 1.8 to 2 million deaths reported in 2022, a figure projected to exceed 3 million by 2045[1]. It is divided into two major histologic subtypes: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC), with SCLC comprising approximately 15% of all lung cancer cases[1,2]. SCLC is an aggressive high-grade neuroendocrine carcinoma that is initially chemo-sensitive but quickly becomes resistant and refractory even to second-line therapies. With current therapies, most patients with SCLC have an expected survival on the order of months, with a 5-year overall survival (OS) of 2%-4.5%[3]. While the addition of immunotherapy has improved outcomes, the lack of predictive biomarkers and an incomplete understanding of mechanisms of resistance highlight a need for further translational work.

Patients are commonly classified as having limited-stage SCLC (LS-SCLC), where involved areas are confined to the primary site and regional lymph nodes with the possibility of definitive therapy, and extensive-stage SCLC (ES-SCLC), where the disease is advanced and treated systemically with palliative intent[4]. The American Joint Committee on Cancer (AJCC) Staging System may also be used for SCLC staging to provide more information. SCLC is strongly associated with smoking, with approximately 85% of patients diagnosed with SCLC reporting a history of smoking[5,6]. While the incidence among non-smokers varies by region, there appears to be a greater proportion in East Asia[5].

Outcomes in SCLC have historically been and remain poor, with a median OS of 25 to 30 months for LS-SCLC and only 8 to 15 months for ES-SCLC[7-9]. For decades, the mainstay of therapy has been concurrent chemoradiotherapy (cCRT) for limited-stage disease and chemotherapy for extensive-stage disease with few options after progression[10]. Systemic treatment options for SCLC have been historically sparse, with topotecan approved in 1996 and no new drug approvals until 2019, with the approval of atezolizumab with chemotherapy as first-line therapy followed by durvalumab with chemotherapy in the first-line setting in 2020 and lurbinectedin as second-line therapy in 2020[11]. The last five years have seen more new drug approvals than in the 20 years prior due to the advent of immune checkpoint inhibitors (ICIs) and, most recently, tarlatamab, a bispecific T cell engager[12].

Several landmark trials of chemoimmunotherapy in ES-SCLC have been published within the last decade, with practice-changing results. In CASPIAN, durvalumab with or without tremilimumab plus platinum-etoposide was compared to platinum-etoposide, with significant improvements in OS with a hazard ratio (HR) of 0.71 and median OS of 12.9 months versus 10.5 months in the most recent 3-year OS update[13]. IMPower133 demonstrated that the addition of atezolizumab to first-line platinum-etoposide also conferred a survival benefit, with a HR of 0.76 and median OS of 12.3 vs. 10.3 months favoring the chemoimmunotherapy regimen[14,15]. These trials led to the approval of durvalumab or atezolizumab in combination with carboplatin and etoposide as frontline standard of care therapy for ES-SCLC.

While the addition of immunotherapy to chemotherapy in the first-line setting has clearly improved survival and clinical outcomes, median progression-free survival (PFS) and OS remain under 6 months and 13 months, respectively[13-21]. Other trials of immune checkpoint inhibitors (ICIs) in later-line therapy in SCLC include CheckMate-032, a phase 1/2 multi-arm trial that assessed nivolumab, a PD-1 inhibitor, with and without the addition of ipilimumab, a CTLA-4 inhibitor, and KEYNOTE 158, a phase 2 study of pembrolizumab, a PD-1 inhibitor, as third-line therapy[19,22]. These trials led to the United States Food and Drug Association (FDA)’s accelerated approval of nivolumab in 2018 and pembrolizumab in 2019 as a third-line treatment after progression on a platinum-containing chemotherapy regimen and at least one other line of treatment[23]. Pembrolizumab and nivolumab were withdrawn from the market for the treatment of SCLC as the Phase III trials CheckMate 451, CheckMate 331, and KEYNOTE 604 did not meet their primary endpoints of OS, although they did demonstrate a significant improvement in PFS, a secondary endpoint[24-26].

More recently, the first interim analysis of ADRIATIC, a Phase 3 trial of consolidation durvalumab compared to placebo after cCRT for LS-SCLC, demonstrated a PFS and OS benefit, with a HR of 0.73 for death and 55.9 months vs. 33.4 months for OS[27]. Patients were treated with up to 24 months of consolidation durvalumab after completion of cCRT. PFS was also significantly longer at 16.6 months vs. 9.2 months, with a HR for progression or death of 0.76. Consolidation durvalumab after cCRT is now included in the National Comprehensive Cancer Network (NCCN) guidelines for LS-SCLC and is the new standard of care. A summary of major trials of immunotherapy in SCLC is available in Table 1.

Table 1

Selected key trials of immunotherapy in LS-SCLC and ES-SCLC

StudyNo.Phase/SettingAuthorsYear of publicationImmune checkpoint inhibitor regimenComparison armPrimary endpointResultsFDA approved?NCCN guidelines inclusion
First line
IMPower133NCT02763579Phase 1/3, multinationalHorn et al.[14]; Liu et al.[15] (updated results)2018, 2021 (updated results)Atezolizumab + EPEP + placeboPFS, OSAtezolizumab + EP significantly improved PFS (HR 0.77; 95% CI 0.62-0.96; P = 0.02) and OS (HR 0.76; 95% CI 0.60-0.95, P = 0.0154)YesYes
CASPIANNCT03043872Phase 3, multinationalPaz-Ares et al.[13,16]2019, 2022 (updated results)Durvalumab (+/- tremilimumab) + EPEP + placeboOSDurvalumab + EP significantly improved OS (HR 0.71, 95% CI 0.60-0.86, P = 0.0003)YesYes
ASTRUM-005NCT04063163Phase 3, multinationalCheng et al.[17]2022Serplulimab + EPEP + placeboOSSerplulimab + EP significantly improved OS (HR 0.63, 95% CI 0.49-0.82, P < 0.001)No; NMPA approvedNo
CAPSTONE-1N/APhase 3, nationalWang et al.[28]2022Adebrelimab + EPEP + placeboOSAdebrelimab + EP significantly improved OS (HR 0.72, 95% CI 0.58-0.90, P = 0.0017)No; NMPA approvedNo
ETER701N/APhase 3, nationalCheng et al.[29]2024Benmelstobart + anolitinib + EPEP + double placebo; EP + anlotinib + placeboPFS, OSBenmelstobart + anlotinib + EP significantly improved OS (HR 0.61, 95% CI 0.45-0.79, P = 0.0003) and PFS (0.32, 95% CI 0.26-0.41, P < 0.0001)NoNo
KEYNOTE 604NCT03066778Phase 3, multinationalRudin et al.[26]2020Pembrolizumab + EPEP + placeboPFS, OS Pembrolizumab + EP significantly improved PFS (HR 0.75, 95% CI 0.61-0.91, P = 0.0023), significance threshold not met for OS (HR 0.80, 95% CI 0.64-0.98, P = 0.0164)NoNo
CheckMate 451NCT02538666Phase 3, multinationalOwonikoko et al.[24]2021Nivolumab +/- ipilimumab maintenance after EPEP + placebo maintenanceOSPrimary endpoint not reachedNoNo
CA184-156NCT01450761Phase 3, multinationalReck et al.[18]2016Ipilimumab + EPEP + placeboOSPrimary endpoint not reachedNoNo
Relapsed/Refractory
CheckMate 331NCT02481830Phase 3, multinationalSpigel et al.[25]2021NivolumabTopotecan or amrubicinOSPrimary endpoint not reachedNo (marketing authorization withdrawn)Yes
CheckMate 032NCT01928394Phase 1/2, multinationalAntonia et al.[19]; Ready et al.[21] (updated results)2016, 2020 (updated results)Nivolumab +/- ipilumumabN/AORRORR higher in nivolumab + ipilumumab group (odds ratio 2.12, 95% CI 1.06-4.25, P = 0.03), OS was similar
KEYNOTE 158NCT02628067Phase 2, multinationalChung et al.[22]ASCO 2018 Abstract2020 (pooled analysis of KEYNOTE 158 and phase 1b KEYNOTE 028)PembrolizumabN/AORRORR 18.7% overallPooled analysis of KEYNOTE-028 and KEYNOTE-158 with ORR 19.3%, 95% CI 11.4-29.4, with 61% of responders with responses > 18 monthsYes
KEYNOTE 028NCT02054806Phase 1b, multinationalOtt et al.[20]; Chung et al.[22]2017PembrolizumabN/ASafety, tolerability, ORRSafety consistent with known safety profile of pembrolizumab, ORR 33%, 95% CI 16%-55%
Limited stage
STIMULINCT02046733Phase 2, multinationalPeters et al.[10]2022Ipilimumab/nivolumab consolidation after cCRTPlaceboPFSClosed early due to slow accrualNoNo
ADRIATICNCT03703297Phase 3, multinationalCheng et al.[27]2024Durvalumab +/- tremilimumab consolidation after cCRTPlaceboPFS, OSDurvalumab consolidation significantly improved PFS (HR 0.76, 95% CI 0.59-0.98, P = 0.02) and OS (HR 0.73, 95% 0.54-0.98, P = 0.01)TBDYes

There is significant variation in the duration and depth of response among patients with SCLC who receive immunotherapy, and the mechanism by which patients develop primary or acquired resistance to therapy is poorly understood. It is likely that heterogeneity within SCLC, driven by genomic and transcriptomic variation, is a driver of this differential response. As most patients with SCLC are diagnosed with advanced disease, surgical resections are rare and limited tissue availability has historically been a barrier for translational work[30]. In this review, we will discuss the clinical definitions of immunotherapy resistance and mechanisms of resistance to immunotherapy in patients with SCLC. As the molecular mechanisms of immunotherapy resistance in SCLC are understudied, we will review the evidence in SCLC in the context of mechanisms of resistance extrapolated from more extensively studied tumor types, such as NSCLC and melanoma.

DEFINITION OF IMMUNOTHERAPY RESISTANCE

A significant challenge in studying immunotherapy resistance, in general, is the lack of a uniform definition, which is made especially difficult in the context of combination therapies where immunotherapy is paired with other agents. The Society for Immunotherapy of Cancer (SITC) has published consensus definitions for resistance to single-agent immunotherapy, combination immunotherapies, chemoimmunotherapy, and adjuvant immunotherapy[31,32]. In general, while the taskforce felt 6-8 weeks (or 2 cycles) of the immunotherapy component was likely necessary in most circumstances, they also included the caveat that resistance may be determined sooner if there is clear rapidly progressing disease.

Here, we explore how SITC immunotherapy resistance definitions can be potentially applied to small-cell lung cancer, which has not previously been clearly defined.

Immunotherapy monotherapy resistance in ES-SCLC

In patients who receive single-agent immunotherapy, primary resistance was defined by SITC as disease progression after two cycles or at least 6 weeks of treatment and no more than 6 months of exposure to ICIs[31,32]. The patient must have received a dose of ICI within 12 weeks (or 3 months) of the evaluation for resistance [Figure 1]. Secondary or acquired resistance was defined as progression after a documented, confirmed objective response or prolonged stable disease for ≥ 6 months, independent of target lesion measurements. The patients were separated into adequate vs. inadequate exposure groups, with progression within 6-12 weeks after their last dose versus > 12 weeks after their last dose, respectively. An alternate classification of “early relapse” vs. “late relapse” was also proposed.

Mechanisms of immunotherapy resistance in small cell lung cancer

Figure 1. Immunotherapy monotherapy resistance in ES-SCLC. Schema of SITC immunotherapy monotherapy definitions, with a shorter timeframe for consideration of acquired resistance for ES-SCLC. SCLC: Small cell lung cancer; ES-SCLC: Extensive-stage; SITC: Society for immunotherapy of cancer.

In ES-SCLC, trials of single-agent immunotherapy include CheckMate 331 with nivolumab monotherapy, CheckMate 032 with a nivolumab monotherapy arm, and KEYNOTE 158 and 028 with pembrolizumab in relapsed/refractory disease [Table 1]. CheckMate 331 and CheckMate 032 demonstrated a median PFS of 1.4 months and 1.4 months, respectively[21,25]. The duration of response (DOR), defined as the time from the first confirmed complete response (CR) or partial response (PR) to documented progression, was 8.3 months (7.0-12.6) and 15.8 months (7.1-NR) in CheckMate 331 and CheckMate 032, respectively. The combined analysis of KEYNOTE 158 and KEYNOTE 028 demonstrated a median PFS of 2.0 months, with a DOR of 25.9 months (4.1-35.8)[22]. Although the median PFS was 2 months or less for the immunotherapy arm in these trials, the duration of response in the patients who did respond ranged from 4.1 months to not reached (NR), indicating that patients with a response had a longer PFS than would be suggested by the median PFS among all patients. In addition, the SITC discusses that for aggressive malignancies, “a shorter period of disease control may still indicate clinical benefit, and, as such, the consensus of the group was that a shorter duration of best response should be required for the definition for secondary resistance to ICI combinations for patient safety concerns”[31]. In light of these data, we suggest that the cut-off between primary and secondary resistance in single-agent immunotherapy for ES-SCLC also be set at a shorter time point than the 6 months published in the SITC consensus definitions, e.g., 4-6 months, as displayed in Figure 1.

Combination immunotherapy resistance in ES-SCLC

For combination ICI regimens, the taskforce recommended a confirmatory scan 4 weeks after progressive disease (PD), with primary resistance defined as a minimum of exposure to 2 cycles of both drugs (6-12 weeks) with best response PD or stable disease (SD) < 6 months. Similarly, secondary resistance was defined as progression after initial objective response or durable stable disease after > 6 months of drug exposure[31,32]. If induction dosing was utilized, where two ICIs were administered for a limited number of cycles followed by monotherapy maintenance, PD occurring after 6 months was defined as secondary resistance to the monotherapy for the drug that was continued. In CheckMate 032 which included ipilimumab/nivolumab arms, median PFS was 1.5 months with DOR of 10 months (6.7-NR)[21]. Given this PFS, we feel that it would again be reasonable to set the cut-off shorter than 6 months for primary versus secondary resistance in SCLC.

Chemoimmunotherapy resistance in ES-SCLC

Defining immunotherapy resistance in combined chemoimmunotherapy regimens poses a significant challenge due to the difficulty in determining the role of the ICI versus the chemotherapy in treatment response. In the case of chemoimmunotherapy combinations in the SITC taskforce consensus, primary resistance was defined as progression within the first 6 months of therapy, irrespective of the initial response[31]. Secondary or acquired resistance to chemoimmunotherapy was not defined due to the impossibility of establishing the contributions of the individual components.

Notably, patients with ES-SCLC are treated with a combination of chemoimmunotherapy and the vast majority of SCLC are platinum-sensitive at the initiation of therapy. Per NCCN guidelines, a chemotherapy-free interval (CTFI) of 3-6 months is used to determine candidacy for platinum rechallenge and patients who respond to rechallenge are considered platinum-sensitive[33]. Thus, if there is relapse/progression within 3 months of completing chemotherapy, it is impossible to determine primary ICI vs. chemotherapy resistance in ES-SCLC due to the definition of platinum sensitivity [Figure 2]. If there is progression within 6 months of starting maintenance immunotherapy, this may represent primary resistance to the immunotherapy component. Acquired resistance, therefore, could be considered if progression occurs after 6 months post-initiation of maintenance immunotherapy, but it remains difficult to parse out the contribution of the components without platinum rechallenge.

Mechanisms of immunotherapy resistance in small cell lung cancer

Figure 2. Chemoimmunotherapy resistance in ES-SCLC. Schema of SITC chemoimmunotherapy definition with a potential shorter cut-off for primary versus acquired resistance in ES-SCLC. SCLC: Small cell lung cancer; ES-SCLC: Extensive-stage; SITC: Society for immunotherapy of cancer.

Adjuvant/consolidation immunotherapy resistance in LS-SCLC

With the inclusion of consolidation durvalumab after cCRT in the NCCN guidelines v2.2025, the definition of adjuvant/consolidation immunotherapy resistance will need to be addressed[33]. In the adjuvant setting, primary resistance was defined as recurrence ≤ 12 weeks after completion/discontinuation of therapy or recurrence on therapy by SITC. For patients who progress after stopping therapy for any multitude of reasons, tumor regrowth ≤ 12 weeks after therapy was halted was deemed primary resistance. If recurrence/regrowth occurred after 12 weeks after treatment discontinuation, this could not be determined as resistance.

cCRT is the standard of care for LS-SCLC not amenable to surgical resection. The vast preponderance of patients receive both chemotherapy and radiation, which can make determination of resistance difficult[33]. In the CONVERT trial, which evaluated once-daily versus twice-daily radiation with concurrent chemotherapy, median PFS was 14.3 months and 15.4 months, respectively[34]. In STIMULI, a trial of consolidation ipilimumab/nivolumab that was closed early due to slow accrual, the median PFS in the control arm without immunotherapy was 14.5 months[10]. In ADRIATIC, the control arm demonstrated a median PFS of 9.2 months versus 16.6 months in the durvalumab arm which conferred a 7.4 month PFS benefit[27]. 46.0% of patients progressed and discontinued treatment before completing 2 years of treatment, with patients receiving a median of 12.9 infusions of durvalumab. Interestingly, the PFS in the control arm of STIMULI was notably longer than in the control arm of ADRIATIC. This difference may be due to the smaller study size of STIMULI compared to ADRIATIC, or variation in patient recruitment as ADRIATIC recruited from a global population while STIMULI recruited in Europe. Furthermore, while there was a higher proportion of patients in STIMULI who received once daily rather than twice daily Radiation Therapy (RT) than in ADRIATIC, the CONVERT trial, which addressed once-daily versus twice-daily RT, did not find a significant difference in outcomes.

Given these time points [Figure 3], it is not possible to determine the contribution of radiation, chemotherapy, or immunotherapy resistance if there is recurrence within 1 year of starting durvalumab consolidation. As almost half of patients progressed prior to completion of the 2 years of consolidation treatment and patients received a median of 12.9 infusions, it would also be difficult to define primary resistance as within 12 weeks of completing consolidation/adjuvant therapy. More data are needed to delineate primary versus acquired resistance in this population.

Mechanisms of immunotherapy resistance in small cell lung cancer

Figure 3. We summarize the median progression-free survival for cCRT and cCRT with consolidation immunotherapy through a timeline of median PFS after cCRT without consolidation immunotherapy vs. median PFS after cCRT with consolidation durvalumab as described in ADRIATIC[27]. cCRT: Concurrent chemoradiotherapy; PFS: Progression-free survival.

MECHANISMS OF IMMUNOTHERAPY RESISTANCE IN SCLC

Heterogeneity in SCLC

To frame the discussion of immunotherapy resistance mechanisms in SCLC and provide a plausible explanation for the significant variation in initial response and primary resistance, a brief discussion of SCLC genomic and transcriptional heterogeneity is essential. While SCLC has historically been treated as a homogenous disease, SCLC classification by both genomic and transcriptomic markers and the subsequent recognition of heterogeneity within this disease has emerged as a promising explanation for variability in immunotherapy response.

Genomically, the loss of the tumor suppressor genes RB1 and TP53 by deleterious mutations or deletions is near universal in SCLC[35]. A recent study, however, of twenty cases of RB1 and TP53 wild-type SCLC demonstrated that this subset of aggressive tumors exhibited genomic and pathologic features of pulmonary carcinoids and were associated with chromothripsis-massive, localized chromosome shattering[36]. This suggests that a small subset of SCLC may be due to transformation from carcinoids, a low-grade neuroendocrine tumor (NET), and may be differentially responsive to immunotherapy[37]. Mutations on other genes have been reported, including ARID1A and ARID1B, CREBBP, and FGFR amplification, as well as genes in the PIK3/mammalian target of rapamycin (mTOR) pathway (loss of phosphatase and tensin homolog (PTEN), rapamycin-insensitive companion of mTOR (RICTOR) amplification, regulatory associated protein of MTOR complex 1 (RPTOR), tuberous sclerosis complex 2 (TSC2)) that contribute to heterogeneity within this disease[38,39].

Transcriptomic subtypes have also emerged to support heterogeneity in SCLC, and are associated with differential response to immunotherapy. At least four SCLC subtypes, SCLC-A, SCLC-N, SCLC-P, and SCLC-I, have been described based on comprehensive transcriptional profiles[40,41]. These subtypes show prognostic associations and are based on the expression of three major transcription factors: achaete-scute homolog 1 (ASCL1), neurogenic differentiation factor 1 (NEUROD1), and POU class 2 homeobox 3 (POU2F3) in combination with the presence of a proinflammatory gene signature[40,41]. SCLC-A and SCLC-N are characterized by the expression of ASCL1 and NEUROD1, respectively, which are involved in neuroendocrine differentiation and characterized as “neuroendocrine subtypes.” SCLC-P is considered a “non-neuroendocrine subtype” as it is characterized by POU2F3 expression, which drives the expression of tuft cells in the mucosal epithelium[41]. SCLC-I is characterized by low levels of the transcriptional factors characteristic of the other three subtypes and demonstrates an inflammatory transcriptional profile. A subtype referred to as SCLC-Y has also previously been reported in the setting of elevated Yes-associated protein 1 (YAP1), which is sometimes described as a fourth subtype instead of SCLC-I[40,42]. A recent study identified further division of SCLC-I into SCLC-I-neuroendocrine (SCLC-I-NE) and SCLC-I-non-neuroendocrine (SCLC-I-nonNE) phenotypes[43]. Specific subtypes of SCLC, such as SCLC-I, may derive greater benefit from ICIs, indicating varied resistance mechanisms[43]. Non-neuroendocrine tumors have demonstrated significantly higher CD45+ leucocytes with increased CD8+ effector tumor-infiltrating lymphocytes (TILs) and higher MHC I expression, which likely contribute to variations in immunotherapy response[44-47]. For example, SCLC-I-NE, which was characterized by low tumor-associated macrophage (TAM) and high T-effector signal, displayed better clinical outcomes with atezolizumab and chemotherapy treatment[43].

Despite marked progress in the understanding of the molecular basis of SCLC, a universally accepted molecular classification is evolving. The clinical significance and therapeutic value of the reported genomic and transcriptomic subtypes are under investigation.

Overview of mechanisms of immunotherapy resistance

Broadly speaking, mechanisms of resistance can be characterized as tumor-intrinsic or tumor-extrinsic[48]. Tumor-intrinsic mechanisms are inherent to the malignant cells and include changes in the antigen presentation machinery or in the cancer cell immunogenicity. Tumor-extrinsic mechanisms, on the other hand, include the effect of other cell types within the tumor microenvironment (TME) or of other mechanisms outside the tumor bed. As the mechanisms of immunotherapy response and resistance in SCLC are relatively understudied, we will discuss SCLC-specific evidence in the context of broader research from other better-characterized solid tumors.

The antitumor response is dependent on effector T cell activation, which requires both an interaction between the T cell receptor (TCR) and surface antigens in malignant cells presented by the major histocompatibility complexes (MHC) and ligation of CD28, a T cell costimulatory receptor, to CD80 or CD86 [Figure 4][49]. Upon TCR engagement, phosphorylation of intracellular tyrosine residues leads to the recruitment of phosphatidylinositol 3-kinase (PI3K) and promotes activation of nuclear factor-kappa B (NF-kB) and protein kinase B/Ak strain transforming (PKB/AKT) to maintain T cell survival and activation. Tumor-specific naïve CD8+ T cells then differentiate into effector T cells and undergo clonal expansion to ultimately kill tumor cells expressing their cognate antigens. Co-inhibitory receptors such as programmed cell death 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) are expressed on most activated T cells to limit unchecked activation and prevent tissue damage but can also be co-opted into immune evasion by the tumor. Blockade of these immune checkpoint pathways through ICIs forms the foundation of the immunotherapy response. Alterations or deficiencies in any part of the above pathways can lead to immune evasion and immunotherapy resistance.

Mechanisms of immunotherapy resistance in small cell lung cancer

Figure 4. Representation of T cell activation in antitumor response and deactivation through immune checkpoints. Antigen-presenting cells (APCs) process and package tumor antigens into peptides to be displayed on its surface with major histocompatibility complex (MHC) molecules to T cells, accompanied by costimulatory signal binding of CD28 and CD80/86 to promote T cell activation. In addition, the presentation of tumor surface antigens via MHC activates T cell response and expansion. The binding of PD-L1/2 to PD-1 and CD80/86 to CTLA4 suppresses the antitumor immune response, allowing immune evasion by tumor cells. Blockade of these immune checkpoints through ICI restores T cell activity and reactivates the antitumor response. ICI: Immune checkpoint inhibitor.

APCs process and package tumor antigens into peptides to be displayed on its surface with MHC molecules to T cells, accompanied by costimulatory signal binding of CD28 and CD80/86 to promote T cell activation. In addition, the presentation of tumor surface antigens via MHC activates T cell response and expansion. The binding of PD-L1/2 to PD-1 and CD80/86 to CTLA4 suppresses the antitumor immune response, allowing immune evasion by tumor cells. Blockade of these immune checkpoints through ICI restores T cell activity and reactivates the antitumor response.

Intrinsic mechanisms of immunotherapy resistance

Tumor immunogenicity and antigen presentation

The availability of tumor neoantigens is expected to be a key feature in the immunogenicity of cancer cells and for activation of the adaptive immune response. Thus, low tumor mutation burden (TMB) is associated with decreased neoantigen load and decreased tumor immunogenicity, which is associated with limited immunotherapy responses in NSCLC[50]. Conversely, a high TMB is generally associated with increased sensitivity to ICIs. However, TMB is not an accurate predictor of immunotherapy benefits and could be affected by multiple factors, such as challenges in identifying optimal cut-points, differences across genomic testing methods and the impact of intratumor genetic heterogeneity and clonal diversity of malignant cells[51].

PD-L1 expression has been associated with clinical benefit to ICIs and outcomes can be stratified by PD-L1 expression in multiple solid tumors, including head and neck squamous cell cancers, non-small cell lung cancers, and breast carcinomas. Prevalence of PD-L1 expression in SCLC, however, is prominently lower at baseline, with one study finding less than 20% of the patients with positive expression as compared to NSCLC, where PD-L1 prevalence is much higher at 52%-67%[52-55]. Moreover, PD-L1 expression in SCLC does not consistently correlate with response to ICIs. Studies using small cohorts of SCLC have also found low expression of CTLA-4, T-cell immunoglobulin and ITIM domain (TIGIT), and other immunosuppressive molecules commonly targeted by ICIs, with increased expression of alternate checkpoints CD276 and CD200[56]. Another potential target for immunotherapy is B7-H3, a member of the same B7 ligand family as PD-L1, which has been found to be highly expressed in SCLC, with B7-H3 expressed in 64.9% of SCLC cases compared to PD-L1 in 7.3% of cases[57]. TMB, a commonly used marker of tumor immunogenicity, varies in SCLC, ranging from 0 to 276.3 mut/Mb in one large cohort with a median of 7.8 mut/Mb. Another study reported a median TMB of 12.7 mut/Mb[58]. The association of TMB with survival after ICIs has been mixed in SCLC, with findings of a significant association between high TMB and improved outcomes in the CheckMate 032 cohort but not in the IMPower 133 cohort, albeit using blood TMB[43,59]. These results suggest that a fraction of SCLC display relatively low TMB and, therefore, limited neoantigens that can be recognized as non-self and mediate robust antitumor adaptive immune rejection.

The antigen presentation machinery is essential for the antitumor immune response. Multiple studies suggest that loss of MHC I presentation via genomic alterations or protein dysregulation, or expression of alternate MHC class IB molecules, such as human leukocyte antigen (HLA)-E and HLA-G, is associated with immune evasion and immunotherapy resistance across multiple solid tumors[60,61]. transporter associated with antigen processing (TAP), a peptide transporter that transfers proteasome and immunoproteasome peptide into the endoplasmic reticulum, is composed of two subunits, TAP 1 and TAP2. These peptides are then loaded onto newly assembled MHC I molecules[62,63]. Calreticulin, TAP-binding protein related (TAPBPR), endoplasmic reticulum aminopeptidase (ERAP) 1 and 2, endoplasmic reticulum protein 57 (ERP57) are components of the peptide loading complex, which are essential for the stabilization of MHC I peptide complexes. Mutations or downregulation of these pathways could result in immune evasion through decreased antigen presentation. Epigenetic silencing of TAP1 by EZH2 has been associated with low MHC I antigen presentation. β2 microglobulin (B2M) expression is well characterized as essential for MHC I expression and function. Deficiency of β2M by deleterious mutations or silencing can lead to defects in the antigen presentation mechanism and has been associated with resistance across multiple solid tumors[60]. Surface expression of MHC I is stimulated by interferons, particularly IFNγ (Type II IFN), which is secreted with the activation of the JAK/STAT pathway[64,65]. In a study of melanoma with acquired resistance to ICI, JAK1-mutated melanoma cell lines were resistant to both Type I and II IFN signaling, while JAK2-mutated cells were resistant to Type II IFN but responded to Type I IFN signaling. Mutations or loss of the components of this signaling cascade, particularly JAK1/JAK2, therefore, are associated with resistance[66].

Suppressed antigen presentation with low β2M and HLA I expression is seen more frequently in SCLC than in other lung malignancies, despite some of these tumors demonstrating high TMB and C:G to A:T transversions[47,67,68]. MHC I and MHC II expression are significantly lower in SCLC as compared to NSCLC, potentially leading to decreased CD8+ and CD4+ T cell activation[56,69]. DLL3, which is highly expressed in SCLC, can suppress the stimulator of IFN genes (STING) pathway and inhibit antigen presentation as well. The association of MHC I with clinical outcomes is supported by evidence that patients with SCLC that expressed low MHC I displayed less intratumoral infiltration of cytotoxic T cells and worse OS with immunotherapy than tumors with high MHC I expression[45]. In addition, differential expression of mitochondrial electron transport chain pathways in SCLC can also mediate antigen presentation and T cell-mediated killing[70,71].

Mutations and gene expression

Specific gene mutations have been associated with immunotherapy sensitivity and resistance, but a clear driver mutation for immunotherapy resistance has not yet been identified in SCLC. RB1 mutations are the second most frequent alteration in SCLC, and although it is most well-known for its role as a tumor suppressor gene, it is also implicated in immune function and response[72,73]. Dowlati et al. examined a cohort of SCLC patients who received ICI monotherapy and found that patients with RB1 wild-type tumors had significantly better outcomes[37,39]. Using RNAseq, they found that patients with RB1 mutations demonstrated downregulated immune-related genes and an immune exclusion phenotype[37]. Two well-known alterations associated with poor immunotherapy outcomes in NSCLC are STK11 gene inactivation and KEAP1 depletion, which are associated with epigenetic inhibition of STING important to the T cell activation pathway and reduced leukocyte infiltration with differential immune cell recruitment, respectively[74,75]. Of note, these mutations have also been associated with poor prognosis with chemotherapy and anti-angiogenesis agents, so the effect is not exclusive to immunotherapy and may represent an overall poor prognosis in general[76].

Approximately 3% of cases in one large cohort of SCLC patients had KEAP1 alterations; silencing of this gene has been shown to confer chemoresistance in vitro through deregulation of the nuclear factor erythroid 2-related factor 2 (NRF2) and neurogenic locus notch homolog protein 1 (NOTCH) pathways, making this an attractive target for further studies[77]. It is unclear, however, if KEAP1 alterations in SCLC confer the same association with decreased immunotherapy response as seen in NSCLC. There was also a subgroup of SCLC with STK11 mutations, though the association with decreased response to ICIs is again unclear. In addition, SCLC has a high prevalence of genetic alterations in the PI3K/AKT/mTOR pathway, found in approximately one-third of patient samples in one study[78]. Elevated myelocytomatosis oncogene (MYC) expression is common in SCLC, has been associated with resistance to platinum chemotherapies, and is likely involved in immunotherapy response given evidence that it is involved in IFN signaling[79]. Overexpression and deregulation of MYC have been implicated in resistance to immunotherapy in other cancers, including melanoma and head and neck squamous cell carcinoma, through decreased IFNγ via JAK2 downregulation[80]. Intrinsic expression of STING is also significantly reduced in SCLC, leading to decreased T cell activation[81].

Epigenetic regulators

Alterations in epigenetic regulators such as Enhancer of Zeste Homolog 2 (EZH2) and lysine-specific demethylase 1 (LSD1) have been implicated in downregulation of MHC I and loss of antigen presentation in SCLC[82-84], leading to decreased activation and recognition by the immune system. Epigenetic regulators have been implicated in the response to immunotherapy. EZH2 is an enzymatic subunit of the polycomb repressive complex 2, which catalyzes trimethylation of histone H3 at lysine 27, leading to gene silencing, and LSD1 is a class I demethylase that removes regulatory methyl groups from histone H3 at lysine positions 4 and 9. LSD1 gene expression is encoded by KMD1A, which is highly expressed in SCLC, and correlates with worse survival outcomes for patients treated on CheckMate 032[84,85]. Both EZH2 and LSD1 activation have been associated with the downregulation of the antigen presentation machinery in SCLC. Ongoing clinical studies of EZH2 and LSD1 inhibitors demonstrate promise for overcoming immunotherapy resistance.

Extrinsic mechanisms of immunotherapy resistance

The TME includes the extracellular matrix, vasculature, immune cells, stromal cells, and cytokines that surround tumor cells and plays an essential role in tumor proliferation and immunotherapy sensitivity and resistance. The TME has been implicated across tumor types in both anti- and pro-tumorigenic pathways.

The degree of TILs has been associated with immunotherapy response, while T regulatory cells (Tregs) promote an immunosuppressive TME that allows for cancer immune evasion and growth. TILs in SCLC tend to be both fewer in number and seen at the stroma rather than the tumor core, contributing to a landscape of immune exclusion and contributing to their classification as “immunologically cold” tumors[57]. Patients who develop primary resistance demonstrate minimal or no T cell infiltration into the tumor, and there is evidence that some SCLC can secrete cytokines such as IL-10, which can drive differentiation of Tregs, leading to blunting of the antitumor immune response[53,57,86]. There is also evidence that TIL infiltration may be dynamic in the context of immunotherapy treatment in SCLC based on unpublished data from correlative studies performed on paired biopsies obtained at baseline, on treatment, and at progression in patients treated with ipilimumab/nivolumab on a Phase 2 trial at our institution. These patient samples demonstrated changes in TILs and MHC-I at different time points in treatment, with increases in CD8+ effector T cells, CD4+ helper T cells, and MHC-I during treatment, followed by a subsequent decline back to baseline levels upon disease progression. Certain subtypes of SCLC that were characterized as “neuroendocrine (NE)-high” have also been found to have decreased TIL infiltration compared to “NE-low” patient samples[44].

Chemotactic cytokines (chemokines) play a significant role in immune cell migration and trafficking in tumors. IFN-γ inducible chemokines, including C-X-C motif ligand 9 (CXCL9), CXCL10, and CXCL11, have been associated with increased T-helper type 1 (Th1-type) immunity and antitumor response in multiple solid tumors, including melanoma, ovarian cancer, and squamous cell carcinoma[87-90]. CCL5 has been shown to be required for T cell infiltration and synergizes with CXCL9 to promote TIL infiltration, with high levels of these two chemokines associated with higher percentages of CD8+ and CD4+ T cells, as well as M1 macrophages[88]. Meanwhile, other cytokines, such as IL-10, TGF-β, and IL-35, lead to T cell inactivation and promote tumorigenesis[91]. While the cytotoxic CD8+ T cell is an essential component of the tumor-killing response, CD4+ T cell populations may support or hinder this process through the secretion of selected cytokines.

Although much of the work on immunotherapy resistance has been done on cells of the adaptive immune system, there is increasing evidence that cells of the innate immune system, including neutrophils and macrophages, also play an essential role in modulating the antitumor response. Myeloid-derived suppressor cells (MDSCs) include a heterogeneous population of immunosuppressive myeloid-lineage cells, and tumor recruitment of these cells has been associated with a worse prognosis[92,93]. Tumor-infiltrating neutrophils (TINs) and tumor-associated macrophages (TAMs) are also found in the milieu of immune cells that comprise the TME and have been found to create an immunosuppressed environment, which can promote tumor growth[94,95]. In addition, chemokines such as CCL7 and IL-8 recruit Tregs and myeloid-derived suppressor cells (MDSCs), while epigenetic modulators can silence chemokines involved in T cell migration, such as CXCL9 and CXCL10, to reduce TILs[89]. IDO1, for example, is an enzyme expressed in myeloid and cancer cells that converts tryptophan to an immunosuppressive metabolite, kynurenine, leading to decreased T cell clonal expansion[96]. Interestingly, there is evidence of higher levels of intra-tumoral IDO1 specifically in lymph nodes in the NE-low patient samples, which were densely infiltrated by immune cells; one hypothesis was that IDO overexpression was an escape mechanism in these patients that rendered TILs anergic, which is consistent with prior studies demonstrating an association between IDO1 and T cell inactivation[44]. Further supporting the role of the TME in immunotherapy response and the recognition of heterogeneity in SCLC, the recently identified SCLC-I-NE subtype is characterized by low TAM and high T-effector cells (lower TAM to T-effectors cell ratio) and demonstrates better outcomes with immunotherapy as compared to SCLC-I-nonNE, which displays higher immunosuppressive TAM levels[43].

Upregulation of alternative immune checkpoint receptors including TIM-3, LAG-3, and TIGIT is also associated with T cell exhaustion/dysfunction[97-99]. Promising studies are underway to evaluate novel inhibitors for these alternative immune checkpoint receptors. Of note, however, the final PFS and OS analysis of the Phase 3 SKYSCRAPER-02 trial of tiragolumab, a TIGIT inhibitor, demonstrated no additional benefit with the addition of this drug to atezolizumab and platinum-etoposide[100]. These results suggest that additional blockade of these alternative immune checkpoints alone may not be sufficient and that the mechanism of resistance is more complex.

Other components of the TME, including the vasculature and the microbiome, have also been found to alter tumor response to ICIs through effects on immune cell populations[101-104]. Hypoxia and aberrant intratumoral vasculature are hallmarks of SCLC, with studies showing that hypoxia-inducible factor 1α (HIF-1α) is highly expressed in SCLC and elevated expression has been associated with a worse prognosis in these patients[105-107]. Hypoxia upregulates the expression of vascular endothelial growth factor (VEGF), an important pathway for increased tumor metastasis and growth, while aberrant vasculature can hinder ICI penetration as well as effector T cell trafficking and infiltration. In preclinical mouse models of SCLC, VEGFA could enhance the expression of the alternate inhibitory receptor TIM-3 in T cells from mice treated with anti-PD-L1 alone, leading to an exhausted T cell phenotype that was reversed with the addition of anti-VEGF[108].

The association between baseline differences in vasculature and the immune system with ICI response has been validated in several solid tumor types, including melanoma and gastric cancer, and the combination of anti-VEGF agents and immunotherapy has seen positive results in preclinical models of SCLC[101,108-110]. Prior clinical studies of anti-VEGF agents had demonstrated mixed efficacy in SCLC patients until the recent publication of ETER701, a Phase 3 trial for first-line treatment of ES-SCLC comparing benmelstobart, anlotinib, and platinum-etoposide or anlotinib and platinum-etoposide to platinum-etoposide (the standard of care in China at the time of trial design)[30,111-113]. Benmelstobart is a monoclonal antibody targeting PD-1, while anlotinib is a small molecule multi-target tyrosine kinase inhibitor with anti-angiogenesis effects[114,115]. The quadruplet arm of benmelstobart, anlotinib, and platinum etoposide demonstrated an impressive OS (19.3 months versus 11.9 months, HR 0.61, 95% CI 0.47-0.79, P = 0.0002) and PFS benefit (6.9 months vs. 4.2 months, 95% CI 0.0.26-0.41, P < 0.0001), while the triple arm of anlotinib with platinum-etoposide did not demonstrate an OS benefit, lending support for the role of VEGF with immunotherapy in SCLC, though further molecular studies are still needed.

Finally, the gut microbiome has also become an area of interest for ICI response given the role of the microbiome in host immunity, with patients exposed to antibiotics before or during ICI administration associated with worse outcomes, though most studies are retrospective and there are certainly many confounding factors in these studies[116]. In one small prospective study, patients with SCLC treated with chemoimmunotherapy who were administered oral probiotics did demonstrate an improvement in mPFS (11.1 months vs. 7.0 months, P = 0.049) which does warrant further study[117].

Primary resistance vs. acquired resistance in SCLC

The mechanisms of primary and acquired resistance to immunotherapy likely overlap to some extent and involve both intrinsic and extrinsic mechanisms of resistance. Although research is currently ongoing, it is difficult to fully identify which are involved in each type of resistance at this time, in part because studies of mechanisms of acquired resistance have historically been difficult due to the paucity of tissue samples from the time of progression and issues with sufficient quality of sample for sequencing. Particularly compared to other solid tumors, there are few studies that have been able to examine these mechanisms in patient samples and very few paired biopsies in clinical trials.

Almost all patients with SCLC develop acquired resistance to first-line chemoimmunotherapy, with 90% of patients in the atezolizumab arm and 89% of patients in the durvalumab arm experiencing RECIST-defined disease progression at the time of updated 2-year analysis in IMpower133 and CASPIAN, respectively[13,15]. Several studies examining radiologic patterns of progression in SCLC have demonstrated that the most common sites of progression are in the primary lung lesion, lymph nodes, liver, and brain[118].

Expansion of clonal populations may be a driver of acquired resistance. Subclonal expansion after cCRT in relapsed patients suggests that heterogeneous selection occurs, with the caveat that these were studied in patients who received chemoradiotherapy rather than chemoimmunotherapy or immunotherapy alone[119] Heterogeneity within the tumor is likely a contributor, given evidence to suggest that even among MHC I-low or -negative tumors, clusters of tumor cells express high MHC I that may drive initial response[45]. Immune infiltration into primary versus metastatic sites can differ, with increased infiltration into liver or brain metastases compared to primary lung tumors[120,121]. Interestingly, brain metastases and adrenal metastases had higher TMB than lung tumors, suggesting that there may be differences in the immunogenicity of primary lung lesions compared to metastatic sites. This may form the basis for oligoprogression, wherein resistance develops in areas of low tumor immunogenicity. In addition, brain tumors were significantly enriched for PTEN alterations compared to lung or liver metastases, which has also been implicated in the evasion of the immune response[58].

While it is certainly possible that oligoprogression occurs in a subclonal population, outcomes are mixed regarding the association of subtypes or the presence of heterogeneity in outcomes[71,85]. Dynamic changes in antigen presentation may also contribute to this response. Biopsies at the progression from a small cohort of patients treated with ipilimumab/nivolumab were found to have downregulation of β2M, suggesting a reduction in MHC I expression in response to immunotherapy[122]. One study found that patients at the time of acquired resistance to second-line ICI showed re-emergence of subclonal mutations that were present at diagnosis, prior to first-line chemotherapy[123]. These subclones were not seen prior to initiation of second-line ICIs and carried signs of platinum exposure from the patients’ first-line chemotherapies, suggesting that these cells expanded and seeded after ICI exposure. No specific mutations were shared among these patients to suggest a pathway for this re-emergence, although sample sizes were very small.

Subtype switching of molecular/transcriptomic subtypes has been hypothesized as a possible mechanism of resistance in SCLC, whereby non-neuroendocrine subtypes transition to neuroendocrine subtypes and vice versa. Although one study examined subtype switching after treatment, the cohort only included two patients treated with immunotherapy due to the timing of the study and neither patient demonstrated subtype switching[124]. Overall, our understanding of molecular SCLC subtypes and acquired resistance to immunotherapy is incomplete and will require concerted efforts to obtain and analyze samples at progression.

Several studies have analyzed transcriptomics in paired biopsies from patients who were treated with chemotherapy and have identified PI3K/AKT, HIF-1, MYC, and TGF-β pathways as potential mediators of acquired resistance, but similar studies in immunotherapy or chemoimmunotherapy are sparse in SCLC[82,125,126].

Immunotherapy in LS-SCLC

Consolidation durvalumab after concurrent chemoradiotherapy for LS-SCLC is already being adopted in clinical practice after inclusion in the NCCN guidelines, with the recent publication of the first interim analysis for ADRIATIC demonstrating significant PFS and OS benefit[27]. Of note, the addition of immunotherapy to concurrent chemoradiotherapy conferred a 22-month OS advantage for LS-SCLC, whereas, in ES-SCLC, the OS benefit is on the order of 2 months. At this time, it is unclear whether this difference represents differences in disease biology between limited-stage and advanced diseases or reflects the effects of radiation combined with immunotherapy.

It is not clear that the mutational landscape is significantly different between ES-SCLC and LS-SCLC. In one LS-SCLC cohort, the most frequently altered genes were TP53 (91%), RB1 (57%), KMT2D (18%), PTPRT (7%), NOTCH1 (5%), and STK11 (5%), which is similar to the alteration profiles seen in ES-SCLC[127]. Other studies have also remarked upon SETDB2, FAM135B, LRP1B, ZFX4, and LRRK2 as mutations of interest in LS-SCLC. PD-L1 expression prevalence appears similarly low between limited-stage and extensive-stage disease[52].

There is ample evidence that radiotherapy can induce tumor cell death, which increases the release of tumor antigens and could enrich immune activation-related pathways to enhance the effects of ICIs[119,128]. While this is a contributor, it is unlikely to be the sole driver of the difference in outcomes, as although consolidation radiation with immunotherapy in ES-SCLC has demonstrated efficacy, the improvement in outcomes is not nearly as dramatic[129]. With several other trials including consolidation immunotherapy or concurrent chemoimmunotherapy with radiotherapy, we anticipate that more data about these mechanisms of resistance will be forthcoming.

CONCLUDING REMARKS

A better understanding of the mechanisms of immunotherapy resistance will be essential for improving patient outcomes. With multiple therapeutics in development, the ability to predict which patients will derive benefit from immunotherapy alone will be critical for treatment sequencing. Meanwhile, a better understanding of the mechanisms of acquired resistance will drive the development of tailored treatment to bypass specific mechanisms, given the complexities of both extrinsic and intrinsic mechanisms of resistance [Figure 5]. As an example, bispecific T cell engagers can recruit T cells to the tumor if there are limited TILs and an “immune desert” is the driver of resistance. Epigenetic regulators such as EZH2 inhibition, LSD1 inhibition, and STING agonists represent research areas with the potential to overcome reduced MHC I expression. ADRIATIC suggests that the addition of radiation therapy to immunotherapy is well tolerated and improves outcomes, likely in the setting of release of tumor-associated antigens and increased immunogenicity. Although most patients with SCLC are diagnosed when the disease is advanced, there is a subset of LS-SCLC patients with node-negative disease for whom surgery may be an option. While perioperative and neoadjuvant immunotherapy has been studied in other solid tumors, large, randomized studies are still lacking for SCLC.

Mechanisms of immunotherapy resistance in small cell lung cancer

Figure 5. Summary of intrinsic and extrinsic mechanisms of resistance in SCLC and potential therapeutics to overcome resistance. Mechanism-directed strategies to overcome immunotherapy resistance and biomarker-driven therapies address the inherent heterogeneity in SCLC. The extrinsic mechanisms of resistance via interactions with the TME are comprised of a complex interplay of immune cells, vasculature, immune checkpoints, and microbiome. Therapies that may target aspects of the TME include cellular therapies, cytokine therapies, anti-angiogenesis agents, and immune checkpoint inhibitors. Intrinsic mechanisms of resistance, which include decreased tumor immunogenicity, altered antigen presentation machinery, immune checkpoints, and epigenetic modifications, may also be avenues for targeted therapies. SCLC: Small cell lung cancer; TME: Tumor microenvironment; CAR-T: Chimeric antigen receptor T-cell therapy; PARP: Poly-ADP ribose polymerase; STING: Stimulator of interferon genes.

Given the historical dearth of biopsy samples in SCLC, particularly at the time of progression and outside of clinical trials, the common presence of extensive necrosis in tumor samples, and the paucity of human-relevant mouse models to test immunotherapy effects, understanding of these resistance mechanisms has lagged behind that of other solid tumors. Efforts are currently underway to better characterize and overcome these resistance mechanisms; however, many studies do not include paired biopsies, and tissue samples on treatment and at the time of progression are still relatively rare. An upcoming trial, S2409 PRISM (Multicohort PRecIsion SCLC Subtype Maintenance Phase II Trial of Durvalumab Versus Biomarker-Directed Novel Agents in Combination with Durvalumab in Extensive Stage Small Cell Lung Cancer), will require testing of archival tissue for eligibility. While this trial will be foundational in identifying predictive markers and novel agents to overcome immunotherapy resistance, paired biopsies and biopsies upon progression will remain essential to studying mechanisms of ICI response and acquired resistance.

FUTURE DIRECTIONS

Overall, while patient selection for primary resistance can improve response rates, future directions should look toward methods to overcome resistance so that patients with SCLC can derive the durable benefit that immunotherapy brings. Given the recent advances in the treatment of SCLC and the widespread adoption of chemoimmunotherapy in the last 5 years, we are hopeful and expect significant progress in our understanding of and ability to bypass these resistance mechanisms.

DECLARATIONS

Acknowledgments

All figures were created using BioRender by Nie Y (2024).

Authors’ contributions

Made substantial contributions to the conception and design of the review: Chiang A, Nie Y, Schalper KA

Contributed to the writing and editing of the manuscript: Chiang A, Nie Y, Schalper, KA

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

Chiang A serves on advisory boards for AstraZeneca, GNE, Zai Lab, Amgen, Fosun, Janssen Daichi, and is a principal investigator on trials Amgen, AbbVie, AstraZeneca, Genentech, BMS. Schalper KA is a consultant, advisor, or speaker for Clinica Alemana Santiago, Shattuck Labs, AstraZeneca, EMD Serono, Takeda, Torque/Repertoire Therapeutics, CSRlife, Agenus, Genmab, OnCusp, Sanofi, Parthenon Therapeutics, Bristol-Myers Squibb, Roche, Molecular Templates, Merck, Dynamicure, Indaptus, Moderna Inc., Merus, PeerView, PER, Forefront Collaborative, and receives research funding from Tesaro/GSK, Takeda, Surface Oncology, Merck, Bristol-Myers Squibb, AstraZeneca, Ribon Therapeutics, Eli Lilly, Boehringer-Ingelheim, Roche, Akoya Biosciences, NextPoint Therapeutics.

The other author declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2024.

REFERENCES

1. Ferlay J, Ervik M, Lam F, et al. Global cancer observatory factsheet: lung. Global cancer observatory: cancer today. 2024. Lyon, France: International Agency for Research on Cancer. Available from: https://gco.iarc.who.int/today. [Last accessed on 25 Dec 2024].

2. Molina JR, Yang P, Cassivi SD, Schild SE, Adjei AA. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc 2008;83:584-94.

3. Cohen S, Brennan B, Banerjee M, Kalemkerian GP. Temporal trends in small cell lung cancer: analysis of the U.S. surveillance, epidemiology and end results (SEER) database. JCO 2023;41:e20641-e20641.

4. Micke P, Faldum A, Metz T, et al. Staging small cell lung cancer: veterans administration lung study group versus international association for the study of lung cancer-what limits limited disease? Lung Cancer 2002;37:271-6.

5. Tseng JS, Chiang CJ, Chen KC, et al. Association of smoking with patient characteristics and outcomes in small cell lung carcinoma, 2011-2018. JAMA Netw Open 2022;5:e224830.

6. Früh M, De Ruysscher D, Popat S, Crinò L, Peters S, Felip E. ESMO Guidelines Working Group. Small-cell lung cancer (SCLC): ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol 2013;24:vi99-105.

7. Steffens CC, Elender C, Hutzschenreuter U, et al. TLK-Group (Tumour Registry Lung Cancer). Treatment and outcome of 432 patients with extensive-stage small cell lung cancer in first, second and third line - Results from the prospective German TLK cohort study. Lung Cancer 2019;130:216-25.

8. Blackhall F, Girard N, Livartowski A, et al. Treatment patterns and outcomes among patients with small-cell lung cancer (SCLC) in Europe: a retrospective cohort study. BMJ Open 2023;13:e052556.

9. Fujimoto D, Morimoto T, Tamiya M, et al. Outcomes of chemoimmunotherapy among patients with extensive-stage small cell lung cancer according to potential clinical trial eligibility. JAMA Netw Open 2023;6:e230698.

10. Peters S, Pujol JL, Dafni U, et al. ETOP/IFCT 4-12 STIMULI Collaborators. 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.

11. Ardizzoni A, Hansen H, Dombernowsky P, et al. Topotecan, a new active drug in the second-line treatment of small-cell lung cancer: a phase II study in patients with refractory and sensitive disease. The European Organization for Research and Treatment of Cancer Early Clinical Studies Group and New Drug Development Office, and the Lung Cancer Cooperative Group. J Clin Oncol 1997;15:2090-6.

12. Ahn MJ, Cho BC, Felip E, et al. DeLLphi-301 Investigators. Tarlatamab for patients with previously treated small-cell lung cancer. N Engl J Med 2023;389:2063-75.

13. Paz-Ares L, Chen Y, Reinmuth N, 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.

14. Horn L, Mansfield AS, Szczęsna A, et al. IMpower133 Study Group. First-line atezolizumab plus chemotherapy in extensive-stage small-cell lung cancer. N Engl J Med 2018;379:2220-9.

15. Liu SV, Reck M, Mansfield AS, 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;39:619-30.

16. Paz-Ares L, Dvorkin M, Chen Y, et al. 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;394:1929-39.

17. Cheng Y, Han L, Wu L, et al. ASTRUM-005 Study Group. 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;328:1223-32.

18. Reck M, Luft A, Szczesna A, et al. Phase III randomized trial of ipilimumab plus etoposide and platinum versus placebo plus etoposide and platinum in extensive-stage small-cell lung cancer. J Clin Oncol 2016;34:3740-8.

19. Antonia SJ, López-Martin JA, Bendell J, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol 2016;17:883-95.

20. Ott PA, Elez E, Hiret S, et al. Pembrolizumab in patients with extensive-stage small-cell lung cancer: results from the phase Ib KEYNOTE-028 study. J Clin Oncol 2017;35:3823-9.

21. Ready NE, Ott PA, Hellmann MD, 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;15:426-35.

22. Chung HC, Piha-Paul SA, Lopez-Martin J, et al. Pembrolizumab after two or more lines of previous therapy in patients with recurrent or metastatic SCLC: results from the KEYNOTE-028 and KEYNOTE-158 studies. J Thorac Oncol 2020;15:618-27.

23. Vaddepally RK, Kharel P, Pandey R, Garje R, Chandra AB. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers 2020;12:738.

24. Owonikoko TK, Park K, Govindan R, et al. Nivolumab and ipilimumab as maintenance therapy in extensive-disease small-cell lung cancer: CheckMate 451. J Clin Oncol 2021;39:1349-59.

25. Spigel DR, Vicente D, Ciuleanu TE, et al. Second-line nivolumab in relapsed small-cell lung cancer: CheckMate 331. Ann Oncol 2021;32:631-41.

26. Rudin CM, Awad MM, Navarro A, et al. 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;38:2369-79.

27. Cheng Y, Spigel DR, Cho BC, et al. ADRIATIC Investigators. Durvalumab after chemoradiotherapy in limited-stage small-cell lung cancer. N Engl J Med 2024;391:1313-27.

28. Wang J, Zhou C, Yao W, et al. CAPSTONE-1 Study Group. Adebrelimab or placebo plus carboplatin and etoposide as first-line treatment for extensive-stage small-cell lung cancer (CAPSTONE-1): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:739-47.

29. Cheng Y, Chen J, Zhang W, et al. Benmelstobart, anlotinib and chemotherapy in extensive-stage small-cell lung cancer: a randomized phase 3 trial. Nat Med 2024;30:2967-76.

30. Thomas A, Pattanayak P, Szabo E, Pinsky P. Characteristics and outcomes of small cell lung cancer detected by CT screening. Chest 2018;154:1284-90.

31. Rizvi N, Ademuyiwa FO, Cao ZA, et al. Society for immunotherapy of cancer (SITC) consensus definitions for resistance to combinations of immune checkpoint inhibitors with chemotherapy. J Immunother Cancer 2023;11:e005920.

32. Kluger HM, Tawbi HA, Ascierto ML, et al. Defining tumor resistance to PD-1 pathway blockade: recommendations from the first meeting of the SITC immunotherapy resistance Taskforce. J Immunother Cancer 2020;8:e000398.

33. National Comprehensive Cancer Network, Inc. NCCN clinical practice guidelines in oncology (NCCN Guidelines®) for small cell lung cancer V.2.2025. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1462. [Last accessed on 25 Dec 2024].

34. Faivre-Finn C, Snee M, Ashcroft L, et al. 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;18:1116-25.

35. Sutherland KD, Proost N, Brouns I, Adriaensen D, Song JY, Berns A. Cell of origin of small cell lung cancer: inactivation of Trp53 and Rb1 in distinct cell types of adult mouse lung. Cancer Cell 2011;19:754-64.

36. Rekhtman N, Tischfield SE, Febres-Aldana CA, et al. Chromothripsis-mediated small cell lung carcinoma. Cancer Discov 2024; doi: 10.1158/2159-8290.cd-24-0286.

37. Dowlati A, Abbas A, Chan T, et al. Immune checkpoint blockade outcome in small-cell lung cancer and its relationship with retinoblastoma mutation status and function. JCO Precis Oncol 2022;6:e2200257.

38. Poirier JT, Gardner EE, Connis N, et al. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene 2015;34:5869-78.

39. Dowlati A, Lipka MB, McColl K, et al. Clinical correlation of extensive-stage small-cell lung cancer genomics. Ann Oncol 2016;27:642-7.

40. Gay CM, Stewart CA, Park EM, 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.

41. Rudin CM, Poirier JT, Byers LA, et al. Molecular subtypes of small cell lung cancer: a synthesis of human and mouse model data. Nat Rev Cancer 2019;19:289-97.

42. Baine MK, Hsieh MS, Lai WV, et al. SCLC subtypes defined by ASCL1, NEUROD1, POU2F3, and YAP1: a comprehensive immunohistochemical and histopathologic characterization. J Thorac Oncol 2020;15:1823-35.

43. Nabet BY, Hamidi H, Lee MC, et al. Immune heterogeneity in small-cell lung cancer and vulnerability to immune checkpoint blockade. Cancer Cell 2024;42:429-443.e4.

44. Dora D, Rivard C, Yu H, et al. Neuroendocrine subtypes of small cell lung cancer differ in terms of immune microenvironment and checkpoint molecule distribution. Mol Oncol 2020;14:1947-65.

45. Mahadevan NR, Knelson EH, Wolff JO, et al. Intrinsic immunogenicity of small cell lung carcinoma revealed by its cellular plasticity. Cancer Discov 2021;11:1952-69.

46. Chan JM, Quintanal-Villalonga Á, Gao VR, et al. Signatures of plasticity, metastasis, and immunosuppression in an atlas of human small cell lung cancer. Cancer Cell 2021;39:1479-1496.e18.

47. George J, Lim JS, Jang SJ, et al. Comprehensive genomic profiles of small cell lung cancer. Nature 2015;524:47-53.

48. Wang F, Wang S, Zhou Q. The resistance mechanisms of lung cancer immunotherapy. Front Oncol 2020;10:568059.

49. O'Donnell JS, Long GV, Scolyer RA, Teng MW, Smyth MJ. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat Rev 2017;52:71-81.

50. Ricciuti B, Wang X, Alessi JV, et al. Association of high tumor mutation burden in non-small cell lung cancers with increased immune infiltration and improved clinical outcomes of PD-L1 blockade across PD-L1 expression levels. JAMA Oncol 2022;8:1160-8.

51. McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016;351:1463-9.

52. Yu H, Batenchuk C, Badzio A, et al. PD-L1 Expression by two complementary diagnostic assays and mRNA in situ hybridization in small cell lung cancer. J Thorac Oncol 2017;12:110-20.

53. Thomas A, Vilimas R, Trindade C, et al. Durvalumab in combination with olaparib in patients with relapsed SCLC: results from a phase II study. J Thorac Oncol 2019;14:1447-57.

54. Dietel M, Savelov N, Salanova R, et al. Real-world prevalence of programmed death ligand 1 expression in locally advanced or metastatic non-small-cell lung cancer: the global, multicenter EXPRESS study. Lung Cancer 2019;134:174-9.

55. Aggarwal C, Abreu DR, Felip E, et al. Prevalence of PD-L1 expression in patients with non-small cell lung cancer screened for enrollment in KEYNOTE-001, -010, and -024. Ann Oncol 2016;27:vi363.

56. Chen HZ, Bonneville R, Paruchuri A, et al. Genomic and transcriptomic characterization of relapsed SCLC through rapid research autopsy. JTO Clin Res Rep 2021;2:100164.

57. Carvajal-Hausdorf D, Altan M, Velcheti V, et al. Expression and clinical significance of PD-L1, B7-H3, B7-H4 and TILs in human small cell lung cancer (SCLC). J Immunother Cancer 2019;7:65.

58. Sivakumar S, Moore JA, Montesion M, et al. Integrative analysis of a large real-world cohort of small cell lung cancer identifies distinct genetic subtypes and insights into histologic transformation. Cancer Discov 2023;13:1572-91.

59. Hellmann MD, Callahan MK, Awad MM, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell 2018;33:853-861.e4.

60. Gettinger S, Choi J, Hastings K, et al. Impaired HLA class i antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov 2017;7:1420-35.

61. Dhatchinamoorthy K, Colbert JD, Rock KL. Cancer immune evasion through loss of MHC class i antigen presentation. Front Immunol 2021;12:636568.

62. Ritz U, Seliger B. The Transporter associated with antigen processing (TAP): structural integrity, expression, function, and its clinical relevance. Mol Med 2001;7:149-58.

63. Garbi N, Tan P, Diehl AD, et al. Impaired immune responses and altered peptide repertoire in tapasin-deficient mice. Nat Immunol 2000;1:234-8.

64. Liu X, Ye L, Bai Y, Mojidi H, Simister NE, Zhu X. Activation of the JAK/STAT-1 signaling pathway by IFN-gamma can down-regulate functional expression of the MHC class I-related neonatal Fc receptor for IgG. J Immunol 2008;181:449-63.

65. Shin DS, Zaretsky JM, Escuin-Ordinas H, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov 2017;7:188-201.

66. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016;375:819-29.

67. Funa K, Gazdar AF, Minna JD, Linnoila RI. Paucity of beta 2-microglobulin expression on small cell lung cancer, bronchial carcinoids and certain other neuroendocrine tumors. Lab Invest 1986;55:186-93.

68. Doyle A, Martin WJ, Funa K, et al. Markedly decreased expression of class I histocompatibility antigens, protein, and mRNA in human small-cell lung cancer. J Exp Med 1985;161:1135-51.

69. He Y, Rozeboom L, Rivard CJ, et al. MHC class II expression in lung cancer. Lung Cancer 2017;112:75-80.

70. Mangalhara KC, Varanasi SK, Johnson MA, et al. Manipulating mitochondrial electron flow enhances tumor immunogenicity. Science 2023;381:1316-23.

71. Peressini M, Garcia-Campelo R, Massuti B, et al. Spatially preserved multi-region transcriptomic subtyping and biomarkers of chemoimmunotherapy outcome in extensive-stage small cell lung cancer. Clin Cancer Res 2024;30:3036-49.

72. Taura M, Suico MA, Koyama K, et al. Rb/E2F1 regulates the innate immune receptor toll-like receptor 3 in epithelial cells. Mol Cell Biol 2012;32:1581-90.

73. Markey MP, Bergseid J, Bosco EE, et al. Loss of the retinoblastoma tumor suppressor: differential action on transcriptional programs related to cell cycle control and immune function. Oncogene 2007;26:6307-18.

74. Lenahan SM, Sarausky HM, Deming P, Seward DJ. STK11 loss leads to YAP1-mediated transcriptional activation in human KRAS-driven lung adenocarcinoma cell lines. Cancer Gene Ther 2024;31:1-8.

75. Zavitsanou AM, Pillai R, Hao Y, et al. KEAP1 mutation in lung adenocarcinoma promotes immune evasion and immunotherapy resistance. Cell Rep 2023;42:113295.

76. Papillon-Cavanagh S, Doshi P, Dobrin R, Szustakowski J, Walsh AM. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort. ESMO Open 2020;5:e000706.

77. Fabrizio FP, Sparaneo A, Gorgoglione G, et al. Effects of KEAP1 silencing on NRF2 and NOTCH Pathways in SCLC cell lines. Cancers 2024;16:1885.

78. Umemura S, Mimaki S, Makinoshima H, et al. Therapeutic priority of the PI3K/AKT/mTOR pathway in small cell lung cancers as revealed by a comprehensive genomic analysis. J Thorac Oncol 2014;9:1324-31.

79. Chen J, Guanizo AC, Jakasekara WSN, et al. MYC drives platinum resistant SCLC that is overcome by the dual PI3K-HDAC inhibitor fimepinostat. J Exp Clin Cancer Res 2023;42:100.

80. Markovits E, Harush O, Baruch EN, et al. MYC induces immunotherapy and IFNγ resistance through downregulation of JAK2. Cancer Immunol Res 2023;11:909-24.

81. Li L, Liang Y, Yu M, et al. Advances in immune checkpoint inhibitors therapy for small cell lung cancer. Cancer Med 2023;12:11097-106.

82. Burr ML, Sparbier CE, Chan KL, et al. An evolutionarily conserved function of rolycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell 2019;36:385-401.e8.

83. Augert A, Eastwood E, Ibrahim AH, et al. Targeting NOTCH activation in small cell lung cancer through LSD1 inhibition. Sci Signal 2019:12.

84. Nguyen EM, Taniguchi H, Chan JM, et al. Targeting lysine-specific demethylase 1 rescues major histocompatibility complex class I antigen oresentation and overcomes programmed death-ligand 1 blockade resistance in SCLC. J Thorac Oncol 2022;17:1014-31.

85. Rudin CM, Balli D, Lai WV, et al. Clinical benefit from immunotherapy in patients with SCLC is associated with tumor capacity for antigen presentation. J Thorac Oncol 2023;18:1222-32.

86. Wang W, Hodkinson P, McLaren F, et al. Small cell lung cancer tumour cells induce regulatory T lymphocytes, and patient survival correlates negatively with FOXP3+ cells in tumour infiltrate. Int J Cancer 2012;131:E928-37.

87. Matsumoto A, Hiroi M, Mori K, Yamamoto N, Ohmori Y. Differential anti-tumor effects of IFN-inducible chemokines CXCL9, CXCL10, and CXCL11 on a mouse squamous cell carcinoma cell line. Med Sci 2023;11:31.

88. Dangaj D, Bruand M, Grimm AJ, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell 2019;35:885-900.e10.

89. Peng D, Kryczek I, Nagarsheth N, et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 2015;527:249-53.

90. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568-71.

91. Mirlekar B. Tumor promoting roles of IL-10, TGF-β, IL-4, and IL-35: its implications in cancer immunotherapy. SAGE Open Med 2022;10:20503121211069012.

92. Zhou J, Nefedova Y, Lei A, Gabrilovich D. Neutrophils and PMN-MDSC: their biological role and interaction with stromal cells. Semin Immunol 2018;35:19-28.

93. Umansky V, Blattner C, Gebhardt C, Utikal J. The role of myeloid-derived suppressor cells (MDSC) in cancer progression. Vaccines 2016;4:36.

94. Wu Y, Yi M, Niu M, Mei Q, Wu K. Myeloid-derived suppressor cells: an emerging target for anticancer immunotherapy. Mol Cancer 2022;21:184.

95. Akbay EA, Koyama S, Liu Y, et al. Interleukin-17A promotes lung tumor progression through neutrophil attraction to tumor sites and mediating resistance to PD-1 blockade. J Thorac Oncol 2017;12:1268-79.

96. Dutta R, Rathor A, Sharma HP, et al. Mapping the immune landscape in small cell lung cancer by analysing expression of immuno-modulators in tissue biopsies and paired blood samples. Sci Rep 2023;13:3739.

97. Wen J, Mao X, Cheng Q, Liu Z, Liu F. A pan-cancer analysis revealing the role of TIGIT in tumor microenvironment. Sci Rep 2021;11:22502.

98. Fourcade J, Sun Z, Benallaoua M, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med 2010;207:2175-86.

99. Li R, Qiu J, Zhang Z, et al. Prognostic significance of lymphocyte-activation gene 3 (LAG3) in patients with solid tumors: a systematic review, meta-analysis and pan-cancer analysis. Cancer Cell Int 2023;23:306.

100. Rudin CM, Liu SV, Soo RA, et al. SKYSCRAPER-02: tiragolumab in combination with atezolizumab plus chemotherapy in untreated extensive-stage small-cell lung cancer. J Clin Oncol 2024;42:324-35.

101. Subramanian M, Kabir AU, Barisas D, Krchma K, Choi K. Conserved angio-immune subtypes of the tumor microenvironment predict response to immune checkpoint blockade therapy. Cell Rep Med 2023;4:100896.

102. Ren S, Feng L, Liu H, Mao Y, Yu Z. Gut microbiome affects the response to immunotherapy in non-small cell lung cancer. Thorac Cancer 2024;15:1149-63.

103. Feig C, Jones JO, Kraman M, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A 2013;110:20212-7.

104. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359:91-7.

105. Lin Q, Choyke PL, Sato N. Visualizing vasculature and its response to therapy in the tumor microenvironment. Theranostics 2023;13:5223-46.

106. Lin CS, Liu TC, Lee MT, Yang SF, Tsao TC. Independent prognostic value of hypoxia-inducible factor 1-alpha expression in small cell lung cancer. Int J Med Sci 2017;14:785-90.

107. Munksgaard Persson M, Johansson ME, Monsef N, et al. HIF-2α expression is suppressed in SCLC cells, which survive in moderate and severe hypoxia when HIF-1α is repressed. Am J Pathol 2012;180:494-504.

108. Meder L, Schuldt P, Thelen M, et al. Combined VEGF and PD-L1 blockade displays synergistic treatment effects in an autochthonous mouse model of small cell lung cancer. Cancer Res 2018;78:4270-81.

109. Fan Y, Zhao J, Wang Q, et al. Camrelizumab plus apatinib in extensive-stage SCLC (PASSION): a multicenter, two-stage, phase 2 trial. J Thorac Oncol 2021;16:299-309.

110. Stefanou D, Batistatou A, Arkoumani E, Ntzani E, Agnantis NJ. Expression of vascular endothelial growth factor (VEGF) and association with microvessel density in small-cell and non-small-cell lung carcinomas. Histol Histopathol 2004;19:37-42.

111. Han B, Ohe Y, Nishio M, et al. 1789P second OS interim analysis from BEAT-SC: a randomized phase III study of bevacizumab (bev) or placebo in combination with atezolizumab and platinum-based chemotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC). Annals of Oncology 2024;35:S1063-4.

112. Ohe Y, Han B, Nishio M, et al. BEAT-SC: A randomized phase III study of bevacizumab or placebo in combination with atezolizumab and platinum-based chemotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC). JCO 2024;42:8001-8001.

113. Ma S, He Z, Liu Y, et al. Sintilimab plus anlotinib as second or further-line therapy for extensive disease small cell lung cancer: a phase 2 investigator-initiated non-randomized controlled trial. EClinicalMedicine 2024;70:102543.

114. Shen G, Zheng F, Ren D, et al. Anlotinib: a novel multi-targeting tyrosine kinase inhibitor in clinical development. J Hematol Oncol 2018;11:120.

115. Wei Z, Yang L, Li Y, et al. Abstract A200: CBT-502 (TQB2450), a novel anti-PD-L1 antibody, demonstrates favorable activity in MC-38/H-11 murine colon and A375 human melanoma animal models. Molecular Cancer Therapeutics 2018;17:A200-A200.

116. Zhong J, Xiong D, Liu Y, Yuan S. Association of antibiotic exposure with survival in patients with extensive-stage small cell lung cancer receiving immune checkpoint inhibitor therapy. Thorac Cancer 2024;15:152-62.

117. Tong L, Wan Y, Shi X, et al. Evaluating oral probiotic supplements as complementary treatment in advanced lung cancer patients receiving ICIs: a prospective real-world study. Cancer Control 2024;31:10732748241253959.

118. Liu L, Liu T, Wang X, et al. Patterns of treatment failure for PD-(L)1 refractory extensive-stage small cell lung cancer in continued PD-(L)1 treatment. Transl Oncol 2023;33:101687.

119. Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014;3:e28518.

120. Berghoff AS, Ricken G, Wilhelm D, et al. Tumor infiltrating lymphocytes and PD-L1 expression in brain metastases of small cell lung cancer (SCLC). J Neurooncol 2016;130:19-29.

121. Best SA, Hess JB, Souza-Fonseca-Guimaraes F, et al. Harnessing natural killer immunity in metastatic SCLC. J Thorac Oncol 2020;15:1507-21.

122. Chiang AC, Asghari H, Ashley K, et al. Molecular predictors and immunomodulatory role of dual checkpoint inhibitor blockade using ipilimumab/nivolumab in patients with extensive stage small cell lung cancer. JCO 2023;41:8597-8597.

123. George J, Maas L, Abedpour N, et al. Evolutionary trajectories of small cell lung cancer under therapy. Nature 2024;627:880-9.

124. Lo YC, Rivera-Concepcion J, Vasmatzis G, Aubry MC, Leventakos K. Subtype of SCLC is an intrinsic and persistent feature through systemic treatment. JTO Clin Res Rep 2023;4:100561.

125. Yang F, Fan J, Yang R, Cun Y. Integrative analysis of blood transcriptome profiles in small-cell lung cancer patients for identification of novel chemotherapy resistance-related biomarkers. Front Immunol 2024;15:1338162.

126. Jin Y, Chen Y, Qin Z, Hu L, Guo C, Ji H. Understanding SCLC heterogeneity and plasticity in cancer metastasis and chemotherapy resistance. Acta Biochim Biophys Sin 2023;55:948-55.

127. Shirasawa M, Yoshida T, Shiraishi K, et al. Tumor microenvironment-mediated immune profiles and efficacy of anti-PD-L1 antibody plus chemotherapy stratified by DLL3 expression in small-cell lung cancer. Br J Cancer 2023;129:2003-13.

128. Ji D, Yi H, Zhang D, et al. Somatic mutations and immune alternation in rectal cancer following neoadjuvant chemoradiotherapy. Cancer Immunol Res 2018;6:1401-16.

129. Li L, Yang D, Min Y, et al. First-line atezolizumab/durvalumab plus platinum-etoposide combined with radiotherapy in extensive-stage small-cell lung cancer. BMC Cancer 2023;23:318.

Cite This Article

Review
Open Access
Mechanisms of immunotherapy resistance in small cell lung cancer
Yunan NieYunan Nie, ... Anne ChiangAnne Chiang

How to Cite

Nie, Y.; Schalper, K. A.; Chiang, A. Mechanisms of immunotherapy resistance in small cell lung cancer. Cancer. Drug. Resist. 2024, 7, 55. http://dx.doi.org/10.20517/cdr.2024.154

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click on download.

Export Citation File:

Type of Import

Tips on Downloading Citation

This feature enables you to download the bibliographic information (also called citation data, header data, or metadata) for the articles on our site.

Citation Manager File Format

Use the radio buttons to choose how to format the bibliographic data you're harvesting. Several citation manager formats are available, including EndNote and BibTex.

Type of Import

If you have citation management software installed on your computer your Web browser should be able to import metadata directly into your reference database.

Direct Import: When the Direct Import option is selected (the default state), a dialogue box will give you the option to Save or Open the downloaded citation data. Choosing Open will either launch your citation manager or give you a choice of applications with which to use the metadata. The Save option saves the file locally for later use.

Indirect Import: When the Indirect Import option is selected, the metadata is displayed and may be copied and pasted as needed.

About This Article

Special Issue

This article belongs to the Special Issue Tackling the Challenge of Drug Resistance in Lung Cancer
© The Author(s) 2024. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
74
Downloads
10
Citations
0
Comments
0
0

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles will preserved here permanently:

https://www.portico.org/publishers/oae/