Efficacy of first-line immune checkpoint inhibitors in pulmonary sarcomatoid carcinoma
Original Article

Efficacy of first-line immune checkpoint inhibitors in pulmonary sarcomatoid carcinoma

Gary Birsen1 ORCID logo, Valérie Gounant2 ORCID logo, Nicolas Girard3 ORCID logo, Jacques Cadranel4 ORCID logo, Isabelle Monnet5 ORCID logo, Christine Raynaud-Donzel6 ORCID logo, Elizabeth Fabre7 ORCID logo, Etienne Giroux Leprieur8 ORCID logo, Karen Leroy9 ORCID logo, Diane Damotte10,11 ORCID logo, Marco Alifano12 ORCID logo, Jennifer Arrondeau13 ORCID logo, Isabelle Cremer11 ORCID logo, Jeanne Chapron1 ORCID logo, Marie Wislez1,11 ORCID logo

1Pneumology Department, Thoracic Oncology Unit, Cochin Hospital, AP-HP, Paris Cité University, Paris, France; 2Thoracic Oncology Department, Bichat Hospital, AP-HP, Paris Cité University, Paris, France; 3Medical Oncology Department, Curie Institute, Paris-Saclay University, Paris, France; 4Pneumology Department, Thoracic Oncology Unit, Tenon Hospital, AP-HP, Sorbonne University, Paris, France; 5Pneumology Department, Intercommunal Hospital of Créteil, Créteil, France; 6Pneumology Department, Argenteuil Hospital, Argenteuil, France; 7Thoracic Oncology Department, Georges Pompidou European Hospital, AP-HP, Paris Cité University, Paris, France; 8Pneumology Department, Thoracic Oncology Unit, Ambroise Paré Hospital, AP-HP, Versailles Saint-Quentin-en-Yvelines University, Boulogne-Billancourt, France; 9Biochemistry Department, Molecular Oncology and Pharmacogenetics Unit, Georges Pompidou European Hospital, AP-HP, Paris Cité University, Paris, France; 10Pathology Department, Cochin Hospital, AP-HP, Paris Cité University, Paris, France; 11Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, Paris, France; 12Thoracic Surgery Department, Cochin Hospital, AP-HP, Paris Cité University, Paris, France; 13Medical Oncology, Cochin Hospital, AP-HP, Paris Cité University, Paris, France

Contributions: (I) Conception and design: G Birsen, M Wislez; (II) Administrative support: G Birsen, M Wislez; (III) Provision of study materials or patients: G Birsen, M Wislez, V Gounant, N Girard, J Cadranel, I Monnet, C Raynaud-Donzel, E Fabre, E Giroux Leprieur, M Alifano, J Arrondeau, I Cremer, J Chapron; (IV) Collection and assembly of data: G Birsen, M Wislez, J Chapron; (V) Data analysis and interpretation: G Birsen, M Wislez, K Leroy, D Damotte; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Marie Wislez, MD, PhD. Pneumology Department, Thoracic Oncology Unit, Cochin Hospital, AP-HP, Paris Cité University, 27 Rue du Faubourg Saint-Jacques, 75014 Paris, France; Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, Paris, France. Email: marie.wislez@aphp.fr.

Background: Pulmonary sarcomatoid carcinomas (PSC) are notorious for their poor prognosis and resistance to chemotherapy. The literature suggests that immunotherapy might be effective against this aggressive tumor. This study aims to evaluate the efficacy of immunotherapy, either alone or combined with chemotherapy, as first-line treatment for PSC patients.

Methods: In a retrospective, multicentric, real-world study conducted between July 2017 and April 2021, patients with stage III (ineligible for surgery or radio-chemotherapy) or stage IV PSC were enrolled. These patients received their first-line treatment with immunotherapy and were categorized into two groups based on their treatment modality: the immuno-chemotherapy (IO CT) group or the immunotherapy-alone (IO) group.

Results: This study analyzed a population of 34 patients from eight different hospital centers. In this cohort, the objective response rate (ORR) was 56%, median duration of response was 20.5 months, median progression-free survival (PFS) was 5.11 months, and median overall survival (OS) 13.9 months. Demographic characteristics remained consistent among the treatment groups except for the age (54.0- and 71.0-year-old in the IO CT and IO group, respectively, P=0.02). The IO CT group demonstrated an ORR of 64.0%, a median PFS at 8.72 months, and a median OS of 16.08 months, while the IO group displayed respective values of 52.0%, 3.45 months, and 13.11 months.

Conclusions: This study showed the potential efficacy of immunotherapy as a first-line treatment for PSC. While acknowledging the retrospective nature of the study, our findings suggest a trend favoring the combination of IO CT over IO alone in these patients.

Keywords: Pulmonary sarcomatoid carcinomas (PSC); immunotherapy; immunochemotherapy


Submitted Apr 11, 2024. Accepted for publication Jul 03, 2024. Published online Sep 12, 2024.

doi: 10.21037/tlcr-24-263


Highlight box

Key findings

• This manuscript confirms the efficacy of immunotherapy in the first-line treatment of metastatic pulmonary sarcomatoid carcinomas (PSC), as monotherapy or in combination with chemotherapy.

What is known and what is new?

• PSC have a poor prognosis and are associated with resistance to chemotherapy. Immunotherapy appears to be an effective treatment, but its use as monotherapy or in combination with chemotherapy has not been specifically evaluated for this type of tumor as first-line therapy.

• Our findings suggest a trend favoring the combination of immuno-chemotherapy (IO CT) over immunotherapy alone in these patients.

What is the implication, and what should change now?

• First-line treatment for metastatic PSC should incorporate immunotherapy. Further explorations in prospective studies will be necessary to determine the potential superiority of IO CT combinations.


Introduction

Pulmonary sarcomatoid carcinomas (PSC) are rare tumors, accounting for 0.4% of lung malignancies (1). PSC are associated with a shorter overall survival (OS) than conventional non-small cell lung cancer (NSCLC). Prior to the era of immunotherapy, metastatic PSC patients had a median OS ranging from 3.0 to 8.5 months (2-4). This poor prognosis can be attributed, at least in part, to the resistance of these tumors to standard chemotherapies (5). In 2020, our research group published a retrospective study involving 37 patients with PSC who received immunotherapy as a second-line treatment (6). The study reported an encouraging objective response rate (ORR) of 40.5%, a progression-free survival (PFS) of 4.9 months and an OS of 12.7 months. Based on these results, the primary objective of this retrospective study was to comprehensively assess the efficacy of immunotherapy, both as a monotherapy [immunotherapy alone (IO)] and in combination with chemotherapy (IO CT), as a first-line treatment of PSC. We present this article in accordance with the STROBE reporting checklist (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-263/rc).


Methods

Patients and study design

All patients with histologically proven, stage III (ineligible for surgery or radio-chemotherapy) or stage IV PSC, and who received a first line of treatment with immunotherapy (either in combination with chemotherapy or other immunotherapy, or as monotherapy) between July 2017 and April 2021 were included in this analysis. Patients who had received chemotherapy as part of curative treatment (surgery or radio-chemotherapy) prior to the metastatic relapse were eligible. Patient data were searched in the archives of the Pneumology, Thoracic Oncology and Medical Oncology departments of 13 hospitals in the Ile-de-France region and collected retrospectively and anonymously from the medical records. The detailed histological reports with programmed cell death 1 ligand 1 (PD-L1) status at diagnosis were retrieved with a centralized reviewing (D.D.). Molecular biology reports were retrieved, with a centralized reviewing (K.L.). The routine panels used have limited sensitivity for detecting STK11 (covering only 30% of anomalies) and do not include KEAP1. Imaging reports at diagnosis and at each evaluation were analyzed [computed tomography (CT) and positron emission tomography-CT (PET-CT)] (G.B.).

Evaluation criteria

Radiological responses were defined in accordance with the Response Evaluation Criteria In Solid Tumors (RECIST), version 1.1 (7). The ORR was defined as the proportion of patients achieving a partial or complete response according to RECIST. The disease control rate (DCR) was defined as the proportion of patients achieving stabilization, partial or complete response according to RECIST. PFS was defined as the interval between the start of a line of therapy to disease progression on that treatment. OS was defined as the time from initiation of treatment to death.

Data were updated 2 years after the last patient was included (April 2023).

Statistical analysis

The patients included were categorized according to their treatment modality into two groups: the IO CT group, comprising patients who received a combination of immunotherapy and chemotherapy, and the IO group, comprising patients who received either immunotherapy monotherapy or dual immunotherapy. Non-parametric tests (χ2, Fisher and Mann-Whitney) were used to compare these two treatment groups. OS and PFS were estimated using the Kaplan-Meier method. Univariate and multivariate analyses of clinical data were performed to identify prognostic variables. All statistical analyses were conducted using GraphPad Prism version 10.0.3. Univariate analysis was performed with the use of an unpaired Student’s t-test or fisher’s exact test, as appropriate. Multivariate logistic-regression analysis was then performed, with backward stepwise analysis, to identify independent prognostic variables. All comparisons of clinical variables with a P value of less than 0.20 by univariate analysis were entered into the model. A P value of less than 0.05 was considered to indicate statistical significance.

Ethical considerations

The study protocol and the patient information note were reviewed and approved by the “Committee for the Evaluation of Observational Research Protocols” of the “French Respiratory Society (SPLF)” (No. CEPRO 2021-016). The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). All participating hospitals/institutions were informed and agreed with this study. Patients under care at the investigational centers were informed of the option to withhold their medical data from being used for health research. None of the patients included in the study objected to the utilization of their medical records for research purposes. Patient data were anonymized at the point of collection and retained at Cochin Hospital, APHP, Paris.


Results

Patient characteristics

Thirteen centers agreed to participate in the study, with eight of them collectively enrolling a total of 34 patients (see Table 1). They were all non-irradiable stage III or stage IV. Among them, ten patients (29.4%) underwent surgery, and histological diagnosis was available for these patients on complete surgical specimens. For other patients, the diagnosis of PSC was made from small biopsies showing both an epithelial and sarcomatoid component. This diagnosis was supported using pan-cytokeratin (AE1/AE3, CAM5.2), epithelial membrane antigen (EMA), p40, and TTF-1 immunostaining (8). Most tumors (79.4%) were pleomorphic carcinomas. PD-L1 status was assessable for all patients, and all tumors exhibited positive PD-L1 expression (above the 1% threshold), with 31 tumors (91.2%) having a PD-L1 expression equal to or exceeding 50%. The clones used for PD-L1 status were QR1 (n=11), E1L3N (n=12), SP263 (n=1), and 22C3 (n=3) and not specified for seven patients. The mutational status of patients is summarized in Table S1. Next-generation sequencing analysis was available for 32 patients (94.1%). Among them, 20 patients (62.5%) carried at least one KRAS, MET, or BRAF oncogenic mutation. No patient was found to have an EGFR mutation, ALK or ROS1 fusion. No co-alterations of KEAP1 or STK11 were identified.

Table 1

Population characteristics by treatment

Characteristics Total (n=34) Immunotherapy + chemotherapy (n=11) Immunotherapy without chemotherapy (n=23)
Sex
   Male 29 (85.3) 10 (90.9) 19 (82.6)
   Female 5 (14.7) 1 (9.1) 4 (17.4)
Age (years) 67 [32–86] 54.0 [41–80] 71.0 [32–86]
Smoking status
   Non-smoker 2 (5.9) 0 2 (8.7)
   Former smoker 13 (38.2) 6 (54.5) 7 (30.4)
   Current smoker 19 (55.9) 5 (45.5) 14 (60.9)
   Pack years 35 [0–90] 35 [20–40] 35 [0–80]
Disease stage
   IIIA to IIIC 4 (11.8) 1 (9.1) 3 (13.0)
   IVA to IVB 30 (88.2) 10 (90.9) 20 (87.0)
Brain metastasis
   Yes 7 (20.6) 2 (18.2) 5 (21.7)
   No 27 (79.4) 9 (81.8) 18 (78.3)
ECOG PS score
   0 16 (47.1) 6 (54.5) 10 (43.5)
   1 12 (35.3) 4 (36.4) 8 (34.8)
   2 6 (17.6) 1 (9.1) 5 (21.7)
Histologic type of tumor
   Pleomorphic carcinoma 27 (79.4) 9 (81.8) 18 (78.3)
    With adenocarcinomatous component 10 (29.4) 3 (27.3) 7 (30.4)
    With squamous component 3 (8.8) 1 (9.1) 2 (8.7)
   Giant cell carcinoma 2 (5.9) 1 (9.1) 1 (4.3)
   Spindle cell carcinoma 5 (14.7) 1 (9.1) 4 (17.4)
Diagnostic technique
   Chest surgery 10 (29.4) 3 (27.3) 7 (30.4)
   Transthoracic puncture 18 (52.9) 6 (54.5) 12 (52.2)
   Bronchoscopy 1 (2.9) 0 1 (4.3)
   Pleuroscopy 2 (5.9) 0 2 (8.7)
   Other biopsy 3 (8.8) 2 (18.2) 1 (4.3)
PD-L1 status
   ≥75% 14 (41.2) 7 (63.6) 7 (30.4)
   ≥50%, <75% 17 (50.0) 1 (9.1) 16 (69.6)
   ≥1%, <50% 3 (8.8) 3 (27.3) 0
   <1% 0 0 0
Mutational status
   KRAS mutation 15 (44.1) 5 (45.5) 10 (43.5)
   MET ex.14 skipping mutation 5 (14.7) 1 (9.1) 4 (17.4)
   BRAF non-V600E 3 (8.8) 2 (18.2) 1 (4.3)
   EGFR 0 0 0
   TP53 11 (32.4) 5 (45.5) 6 (26.1)

Data are presented as median [range] or n (%). ECOG PS, Eastern Cooperative Oncology Group performance status; PD-L1, programmed cell death 1 ligand 1.

Patients were categorized according to treatment modality. In the IO CT group, patients received immunotherapy with pembrolizumab combined with either paclitaxel (n=4) or pemetrexed (n=7). In the IO group, patients received either pembrolizumab (n=21) or nivolumab-ipilimumab (n=2). The criteria for selecting between ICI monotherapy and ICI combined with chemotherapy were based on the prevailing guidelines at the time of patient treatment. The characteristics of the patients are summarized in Table 1.

The clinical characteristics were similar in the two groups, except for age (54.0 years in the IO CT group vs. 71.0 years in the IO group, P=0.02). Only three patients had PD-L1 expression below the 50% threshold, and all of them were in the IO CT group. Molecular characteristics were comparable in both groups.

Efficiency data—overall population

In the overall population, the ORR was 56% (n=19/34) and the DCR was 68% (n=23/34) (Figure S1). Three patients had a complete response (8.8%), 16 had a partial response (47.1%), 4 were stabilized (11.8%) and 11 had progressive disease (32.4%). The median duration of response was 20.5 months. The median PFS was 5.11 months, and the median OS was 13.9 months (Figure S1). First-line treatment was discontinued in 30 patients (88.2%), mainly due to progression (47.0%), death (17.6%), but also prolonged response exceeding 2 years (11.7%) and less frequently treatment-related toxicity (8.8%). Four patients were on active surveillance following a complete response and five patients were still receiving treatment at the last data update. Among the patients who discontinued first-line treatment, 15 (50%) received a second line. Out of the five patients with MET exon 14 skipping mutation, one received targeted therapy as second-line (crizotinib), two died during the first-line treatment, one exhibited a prolonged response for first- and second-line treatment (Table S2). None of the patients with KRAS G12C mutations received targeted therapy due to the study’s inclusion period, during which no therapy targeting KRAS was available.

Efficiency data—according to treatment

Efficacy parameters were subsequently evaluated based on the treatment administered.

As shown in Figure 1, there was a trend although not statistically significant for a better ORR (P=0.71) and DCR (P=0.06) in the IO CT group compared to the IO group. A similar trend was noted for PFS (P=0.12) and OS (P=0.33). The median PFS and OS for the IO CT group were 8.72 and 16.08 months, respectively, while those for the IO group were 3.45 and 13.11 months (Figure 1).

Figure 1 Immunotherapy efficacy according to treatment. (A) ORR, DCR and progression. (B) Kaplan-Meier plot for PFS. (C) Kaplan-Meier plot for OS. ORR, objective response rate; DCR, disease control rate; OS, overall survival; PFS, progression-free survival; CI, confidence interval.

Factor predictive of treatment efficacy

Univariate and multivariate analyses were carried out to identify prognostic factors in the study population. Univariate analysis on OS showed that performance status (PS) was the sole significant prognostic factor. We did not find any statistical difference PFS between stage III and stage IV patients. The multivariate analysis incorporated clinical variables with a P value of less than 0.20 from the univariate analysis. Inclusion of variables such as presence of cerebral metastasis, PS and high expression of PD-L1 (≥75%) showed that PS remained the sole predictive factor for a better OS (Figure 2). High PD-L1 did not exhibit a significant association with improved OS in both univariate and multivariate analyses in this patient cohort.

Figure 2 Prognostic factors in patients with pulmonary sarcomatoid carcinoma. (A) Univariate analysis on overall survival. (B) Multivariate analysis on overall survival. PD-L1, programmed cell death 1 ligand 1; PS, performance status; CI, confidence interval.

Discussion

In this cohort of 34 patients with metastatic PSC, first-line immunotherapy with or without chemotherapy was associated with an ORR of 56%, a PFS of 5.11 months and an OS of 13.9 months.

Phase III trials investigating immunotherapy with or without chemotherapy in non-sarcomatoid NSCLC have shown superior PFS and OS compared to our study (9-22) (Table S3). This discrepancy may be partially attributed to the poorer prognosis of PSCs and the more stringent eligibility criteria in phase III trials. Notably, these trials excluded patients with a PS greater than 1, whereas 17.6% of our study participants had a PS of 2. Nevertheless, when juxtaposed with historical data on PSC treatment before the immunotherapy era, where the ORR was below 20%, and OS ranged from 3.0 to 8.5 months, our findings underscore the benefits of immunotherapy for PSC (5,23).

Comparable findings have been reported by other teams in studies of PSCs treated with immunotherapy, either with or without chemotherapy (24-29) (Table S4). Across these studies, PFS ranged from 4.9 to 10.3 months, while OS ranged from 12.7 to 22.8 months. These studies are also retrospective, with sample sizes similar to our study. However, they differ in terms of treatment-line (first or subsequent), immunotherapy types (anti-PD-1, anti-PD-L1, anti-CTLA4), and whether combined with chemotherapy or anti-angiogenic agents, making direct comparisons challenging.

The choice among the two treatment options—immunotherapy with chemotherapy or immunotherapy alone—is particularly relevant for the management of PSCs, which are frequently associated with high PD-L1 expression on tumor cells (TC) (30): in our series, 91.2% of patients had PD-L1 TC expression greater than or equal to 50%. The limited response rate to conventional chemotherapies and the high PD-L1 TC expression of PSCs suggest that treatment with immunotherapy in monotherapy might be preferred in this situation. However, the frequent sarcopenia of PSC patients, the high tumor burden and the rapid progression of these cancers (2,5,31-33) could argue in favor of an approach combining immunotherapy and chemotherapy. By grouping patients based on their treatment, a trend emerged in favor of immunotherapy with chemotherapy over immunotherapy alone in terms of ORR, PFS, and OS.

Our study has several limitations, particularly a relatively small sample size and its retrospective nature. These aspects may raise questions about the comparability of patient characteristics between the two groups, notably regarding age. Although other variables such as PS did not exhibit a significant difference between the two groups, it is worth noting that 21.7% of patients in the IO group had a PS of 2, compared to 9.1% in the IO CT group, adding complexity to the interpretation of results.

The frequent occurrence of KRAS G12C mutations and MET exon 14 skipping in PSC patients emphasizes the need for a clearer understanding of the role of targeted therapies in managing these tumors (34). While studies have shown the effectiveness of immunotherapy for treating metastatic NSCLC with these mutations (35,36), caution is warranted due to increased toxicities observed in patients undergoing sequential immunotherapy followed by targeted therapy (37). In our cohort, 62.5% (n=20/32) had a driver mutation (Table S1), but only one received targeted therapy after immunotherapy, preventing a comprehensive assessment of toxicity in this context. In our cohort, the presence or absence of activating mutations (KRAS, BRAF, MET exon 14) did not predict immunotherapy efficacy. None of the patients had an EGFR mutation or ALK translocation, typically associated with a poorer prognosis under immunotherapy (38,39).


Conclusions

Our retrospective study of advanced or metastatic PSC patients treated with IO alone or in combination with chemotherapy demonstrated an improvement in clinical outcomes with a trend toward prolonged PFS and OS in patients treated with the combination of IO and chemotherapy. Prospective studies are needed to determine the optimal treatment modality for these patients.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-263/rc

Data Sharing Statement: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-263/dss

Peer Review File: Available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-263/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tlcr.amegroups.com/article/view/10.21037/tlcr-24-263/coif). N.G. received research grants/support from Abbvie, Amgen, AstraZeneca, Beigene, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi-Sankyo, Gilead, Hoffmann-La Roche, Janssen, LeoPharma, Lilly, Merck Serono, Merck Sharp & Dohme, Novartis, Sanofi, Sivan; provided consultative services for Abbvie, Amgen, AstraZeneca, Beigene, Bristol Myers Squibb, Daiichi-Sankyo, Gilead, Ipsen, Hoffmann-La Roche, Janssen, LeoPharma, Lilly, Merck Sharp & Dohme, Mirati, Novartis, Pfizer, Pierre Fabre, Sanofi, Takeda; participated on a data safety monitoring board for Hoffmann-La Roche; employment of a family member with AstraZeneca. Jacques Cadranel received consulting fees for participation to boards of experts from AMGEN, AZ, Daiichi (and travelling, BMS, MSD, Roche, Sanofi, Janssen (and travelling), Takeda, Pfizer (and travelling). I.M. received honoraria from Regeneron and support from Takeda, MSD, Pfizer, Oxyvie. E.G.L. received honoraria from AMGEN, AstraZeneca, Ipsen, Janssen, Lilly, MSD, Novartis, Pfizer, Sanofi, Takeda and support for attending meetings and/or travel from Takeda, MSD, AstraZeneca. K.L. received honoraria from AstraZeneca, MSD, Janssen, GSK, Lilly, AMGEN, Roche for lectures, presentations or educational events and support for attending meetings and/or travel from AMGEN. Jeanne Chapron received honoraria from BMS, AstraZeneca, Pfizer, Sanofi, MSD and support for attending meetings and/or travel from Sanofi and Regeneron. M.W. received payment or honoraria for lectures and presentations from AMGEN, AstraZeneca, Bristol Myers Squibb, F. Hoffmann-La Roche, Janssen, MSD Oncology, Lilly, Merck KGaA; grants from AstraZeneca; support for attending meetings and/or travel from Janssen, Amgen, MSD and F. Hoffmann-La Roche; participated on a data safety monitoring board or advisory board for AMGEN, AstraZeneca, Bristol Myers Squibb, F. Hoffmann-La Roche, Janssen, MSD Oncology, Lilly and Merck KGaA. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study protocol and the patient information note were reviewed and approved by the “Committee for the Evaluation of Observational Research Protocols” of the “French Respiratory Society (SPLF)” (No. CEPRO 2021-016). All participating hospitals/institutions were informed and agreed with this study. Patients under care at the investigational centers were informed of the option to withhold their medical data from being used for health research. None of the patients included in the study objected to the utilization of their medical records for research purposes.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Wu W, Zheng L, Li F, et al. Survival analysis and nomogram for pulmonary sarcomatoid carcinoma: an SEER analysis and external validation. BMJ Open 2023;13:e072260. [Crossref] [PubMed]
  2. Ito K, Oizumi S, Fukumoto S, et al. Clinical characteristics of pleomorphic carcinoma of the lung. Lung Cancer 2010;68:204-10. [Crossref] [PubMed]
  3. Bae HM, Min HS, Lee SH, et al. Palliative chemotherapy for pulmonary pleomorphic carcinoma. Lung Cancer 2007;58:112-5. [Crossref] [PubMed]
  4. Giroux Leprieur E, Antoine M, Vieira T, et al. Clinical and molecular features in patients with advanced non-small-cell lung carcinoma refractory to first-line platinum-based chemotherapy. Lung Cancer 2013;79:167-72. [Crossref] [PubMed]
  5. Vieira T, Girard N, Ung M, et al. Efficacy of first-line chemotherapy in patients with advanced lung sarcomatoid carcinoma. J Thorac Oncol 2013;8:1574-7. [Crossref] [PubMed]
  6. Domblides C, Leroy K, Monnet I, et al. Efficacy of Immune Checkpoint Inhibitors in Lung Sarcomatoid Carcinoma. J Thorac Oncol 2020;15:860-6. [Crossref] [PubMed]
  7. Seymour L, Bogaerts J, Perrone A, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol 2017;18:e143-52. [Crossref] [PubMed]
  8. Pelosi G, Sonzogni A, De Pas T, et al. Review article: pulmonary sarcomatoid carcinomas: a practical overview. Int J Surg Pathol 2010;18:103-20. [Crossref] [PubMed]
  9. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50. J Clin Oncol 2021;39:2339-49. [Crossref] [PubMed]
  10. Carbone DP, Reck M, Paz-Ares L, et al. First-Line Nivolumab in Stage IV or Recurrent Non-Small-Cell Lung Cancer. N Engl J Med 2017;376:2415-26. [Crossref] [PubMed]
  11. Jassem J, de Marinis F, Giaccone G, et al. Updated Overall Survival Analysis From IMpower110: Atezolizumab Versus Platinum-Based Chemotherapy in Treatment-Naive Programmed Death-Ligand 1-Selected NSCLC. J Thorac Oncol 2021;16:1872-82. [Crossref] [PubMed]
  12. Sezer A, Kilickap S, Gümüş M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet 2021;397:592-604. [Crossref] [PubMed]
  13. Garassino MC, Gadgeel S, Speranza G, et al. Pembrolizumab Plus Pemetrexed and Platinum in Nonsquamous Non-Small-Cell Lung Cancer: 5-Year Outcomes From the Phase 3 KEYNOTE-189 Study. J Clin Oncol 2023;41:1992-8. [Crossref] [PubMed]
  14. Nishio M, Barlesi F, West H, et al. Atezolizumab Plus Chemotherapy for First-Line Treatment of Nonsquamous NSCLC: Results From the Randomized Phase 3 IMpower132 Trial. J Thorac Oncol 2021;16:653-64. [Crossref] [PubMed]
  15. Zhou C, Wu L, Fan Y, et al. Sintilimab Plus Platinum and Gemcitabine as First-Line Treatment for Advanced or Metastatic Squamous NSCLC: Results From a Randomized, Double-Blind, Phase 3 Trial (ORIENT-12). J Thorac Oncol 2021;16:1501-11. [Crossref] [PubMed]
  16. Lu S, Wang J, Yu Y, et al. Tislelizumab Plus Chemotherapy as First-Line Treatment for Locally Advanced or Metastatic Nonsquamous NSCLC (RATIONALE 304): A Randomized Phase 3 Trial. J Thorac Oncol 2021;16:1512-22. [Crossref] [PubMed]
  17. Zhou C, Chen G, Huang Y, et al. Camrelizumab Plus Carboplatin and Pemetrexed as First-Line Treatment for Advanced Nonsquamous NSCLC: Extended Follow-Up of CameL Phase 3 Trial. J Thorac Oncol 2023;18:628-39. [Crossref] [PubMed]
  18. Novello S, Kowalski DM, Luft A, et al. Pembrolizumab Plus Chemotherapy in Squamous Non-Small-Cell Lung Cancer: 5-Year Update of the Phase III KEYNOTE-407 Study. J Clin Oncol 2023;41:1999-2006. [Crossref] [PubMed]
  19. Wang J, Lu S, Yu X, et al. Tislelizumab Plus Chemotherapy vs Chemotherapy Alone as First-line Treatment for Advanced Squamous Non-Small-Cell Lung Cancer: A Phase 3 Randomized Clinical Trial. JAMA Oncol 2021;7:709-17. [Crossref] [PubMed]
  20. Ren S, Chen J, Xu X, et al. Camrelizumab Plus Carboplatin and Paclitaxel as First-Line Treatment for Advanced Squamous NSCLC (CameL-Sq): A Phase 3 Trial. J Thorac Oncol 2022;17:544-57. [Crossref] [PubMed]
  21. Paz-Ares LG, Ciuleanu TE, Cobo M, et al. First-Line Nivolumab Plus Ipilimumab With Chemotherapy Versus Chemotherapy Alone for Metastatic NSCLC in CheckMate 9LA: 3-Year Clinical Update and Outcomes in Patients With Brain Metastases or Select Somatic Mutations. J Thorac Oncol 2023;18:204-22. [Crossref]
  22. Brahmer JR, Lee JS, Ciuleanu TE, et al. Five-Year Survival Outcomes With Nivolumab Plus Ipilimumab Versus Chemotherapy as First-Line Treatment for Metastatic Non-Small-Cell Lung Cancer in CheckMate 227. J Clin Oncol 2023;41:1200-12. [Crossref] [PubMed]
  23. Yendamuri S, Caty L, Pine M, et al. Outcomes of sarcomatoid carcinoma of the lung: a Surveillance, Epidemiology, and End Results Database analysis. Surgery 2012;152:397-402. [Crossref] [PubMed]
  24. Inomata M, Tsuda T, Ichikawa T, et al. Efficacy of immune checkpoint inhibitor therapy in patients with pulmonary sarcomatoid carcinoma in clinical practice. Thorac Cancer 2023;14:1618-23. [Crossref] [PubMed]
  25. Kim M, Keam B, Ock CY, et al. Phase II study of durvalumab and tremelimumab in pulmonary sarcomatoid carcinoma: KCSG-LU16-07. Thorac Cancer 2020;11:3482-9. [Crossref] [PubMed]
  26. Lee J, Choi Y, Jung HA, et al. Outstanding clinical efficacy of PD-1/PD-L1 inhibitors for pulmonary pleomorphic carcinoma. Eur J Cancer 2020;132:150-8. [Crossref] [PubMed]
  27. Qian X, Wang Y, Liu F, et al. The efficacy and safety analysis of first-line immune checkpoint inhibitors in pulmonary sarcomatoid carcinoma. Front Immunol 2022;13:956982. [Crossref] [PubMed]
  28. Zhou F, Guo H, Zhou X, et al. Immune checkpoint inhibitors plus chemotherapy in patients with locally advanced or metastatic pulmonary sarcomatoid carcinoma: a multicentric real-world study. Ther Adv Med Oncol 2022;14:17588359221136759. [Crossref] [PubMed]
  29. Chu Q, Jiang J, Yang B, et al. 73P Camrelizumab plus famitinib as first-line treatment in patient with locally advanced or metastatic pulmonary sarcomatoid carcinomas (CAPSTONE): A multi-center, single-arm, phase II study. Ann Oncol 2023;20:100535.
  30. Kim S, Kim MY, Koh J, et al. Programmed death-1 ligand 1 and 2 are highly expressed in pleomorphic carcinomas of the lung: Comparison of sarcomatous and carcinomatous areas. Eur J Cancer 2015;51:2698-707. [Crossref] [PubMed]
  31. Bolte FJ, McTavish S, Wakefield N, et al. Association of sarcopenia with survival in advanced NSCLC patients receiving concurrent immunotherapy and chemotherapy. Front Oncol 2022;12:986236. [Crossref] [PubMed]
  32. Parikh RB, Min EJ, Wileyto EP, et al. Uptake and Survival Outcomes Following Immune Checkpoint Inhibitor Therapy Among Trial-Ineligible Patients With Advanced Solid Cancers. JAMA Oncol 2021;7:1843-50. [Crossref] [PubMed]
  33. Goldberg SB, Herbst RS. Should chemotherapy plus immune checkpoint inhibition be the standard front-line therapy for patients with metastatic non-small cell lung cancer? Cancer 2018;124:4592-6. [Crossref] [PubMed]
  34. Nagano M, Kohsaka S, Hayashi T, et al. Comprehensive molecular profiling of pulmonary pleomorphic carcinoma. NPJ Precis Oncol 2021;5:57. [Crossref] [PubMed]
  35. Kauffmann-Guerrero D, Tufman A, Kahnert K, et al. Response to Checkpoint Inhibition in Non-Small Cell Lung Cancer with Molecular Driver Alterations. Oncol Res Treat 2020;43:289-98. [Crossref] [PubMed]
  36. Mayenga M, Assié JB, Monnet I, et al. Durable responses to immunotherapy of non-small cell lung cancers harboring MET exon-14-skipping mutation: A series of 6 cases. Lung Cancer 2020;150:21-5. [Crossref] [PubMed]
  37. Kalra A, Rashdan S. The toxicity associated with combining immune check point inhibitors with tyrosine kinase inhibitors in patients with non-small cell lung cancer. Front Oncol 2023;13:1158417. [Crossref] [PubMed]
  38. Mazieres J, Drilon A, Lusque A, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 2019;30:1321-8. [Crossref] [PubMed]
  39. Lee CK, Man J, Lord S, et al. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non-Small Cell Lung Cancer-A Meta-Analysis. J Thorac Oncol 2017;12:403-7. [Crossref] [PubMed]
Cite this article as: Birsen G, Gounant V, Girard N, Cadranel J, Monnet I, Raynaud-Donzel C, Fabre E, Giroux Leprieur E, Leroy K, Damotte D, Alifano M, Arrondeau J, Cremer I, Chapron J, Wislez M. Efficacy of first-line immune checkpoint inhibitors in pulmonary sarcomatoid carcinoma. Transl Lung Cancer Res 2024;13(9):2212-2221. doi: 10.21037/tlcr-24-263

Download Citation