Next Article in Journal
Deep Learning Improves Pancreatic Cancer Diagnosis Using RNA-Based Variants
Previous Article in Journal
Urine as a Source of Liquid Biopsy for Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular and Clinical Features of Hospital Admissions in Patients with Thoracic Malignancies on Immune Checkpoint Inhibitors

1
Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010-3000, USA
2
Integrative Genomics Core, Beckman Research Institute, City of Hope Medical Center, Duarte, CA 91010-3000, USA
3
Department of Computational & Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010-3000, USA
4
Applied AI and Data Science, City of Hope National Medical Center, Duarte, CA 91010-3000, USA
5
Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 91010-3000, USA
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(11), 2653; https://doi.org/10.3390/cancers13112653
Submission received: 30 April 2021 / Revised: 22 May 2021 / Accepted: 25 May 2021 / Published: 28 May 2021

Abstract

:

Simple Summary

Lung cancer immunotherapy has many complications and hospitalizations that often occur in non-small cell lung cancer (NSCLC) while on immunotherapy due to adverse events or other factors. The molecular and clinical profiles of these patients are often not well-defined, and the aim of our retrospective study is to better understand these clinical and molecular features. We evaluated a cohort of 90 stage IV thoracic malignancy patients who had hospital admissions after treatment with immune checkpoint inhibitors. We identified a relationship between immune-related adverse events (irAEs) and molecular markers that showed unique survival outcomes, as well as a significant overall survival improvement in patients who required discontinuation or interruption of immunotherapy due to irAEs.

Abstract

Lung cancer patients undergoing systemic treatment with immune checkpoint inhibitors (ICIs) can lead to severe immune-related adverse events (irAEs) that may warrant immediate hospitalization. Patients with thoracic malignancies hospitalized at City of Hope while undergoing treatment with ICIs were identified. Pathology and available next-generation sequencing (NGS) data, including the programmed death-ligand 1 (PD-L1) status and clinical information, including hospitalizations, invasive procedures, and the occurrence of irAEs, were collected. Unpaired T-tests, Chi-square/Fisher’s exact test, and logistic regression were used to analyze our cohort. The overall survival (OS) was calculated and compared using univariate and multivariate COX models. Ninety patients with stage IV lung cancer were admitted after ICI treatment. Of those patients, 28 (31.1%) had documented irAEs. Genomic analyses showed an enrichment of LRP1B mutations (n = 5/6 vs. n = 7/26, 83.3% vs. 26.9%; odds ratio (OR) (95% confidence interval (CI): 13.5 (1.7–166.1); p < 0.05) and MLL3 mutations (n = 4/6, 66.7% vs. n = 5/26, 19.2%; OR (95% CI): 8.4 (1.3–49.3), p < 0.05) in patients with irAE occurrences. Patients with somatic genomic alterations (GAs) in MET (median OS of 2.7 vs. 7.2 months; HR (95% CI): 3.1 (0.57–17.1); p < 0.05) or FANCA (median OS of 3.0 vs. 12.4 months; HR (95% CI): 3.1 (0.70–13.8); p < 0.05) demonstrated a significantly shorter OS. Patients with irAEs showed a trend toward improved OS (median OS 16.4 vs. 6.8 months, p = 0.19) compared to hospitalized patients without documented irAEs. Lung cancer patients who required treatment discontinuance or interruption due to irAEs (n = 19) had significantly longer OS (median OS 18.5 vs. 6.2 months; HR (95% CI): 0.47 (0.28–0.79); p < 0.05). Our results showed a significant survival benefit in lung cancer patients hospitalized due to irAEs that necessitated a treatment interruption. Patients with positive somatic GAs in MET and FANCA were associated with significantly worse OS compared to patients with negative GAs.

1. Introduction

Immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4) have transformed the landscape of lung cancer treatment. Pembrolizumab, an anti-PD-1 monoclonal antibody, was approved by the US Food and Drug Administration (FDA) as a monotherapy first-line treatment of metastatic non-small cell lung cancer (NSCLC) with PD-L1 expression ≥ 1, and in combination with chemotherapy regardless of PD-L1 status [1,2,3,4].
Atezolizumab and durvalumab, anti-PD-L1 monoclonal antibodies, were approved as a first-line treatment in combination with chemotherapy for small cell lung cancer (SCLC) [5,6,7]. Nivolumab, an anti-PD-1 monoclonal antibody, and ipilimumab, an anti-CTLA-4 monoclonal antibody, with or without chemotherapy, were also recently approved for first-line treatment of metastatic lung cancer [8,9,10,11,12]. Durvalumab was approved for consolidation therapy after chemoradiation in unresectable stage III NSCLC, leading to further investigation of ICIs in the neoadjuvant setting [13,14].
However, hospital admission during treatment is common in cancer patients undergoing systemic treatment. Distinct toxicity profiles and immune-related adverse events (irAEs) due to ICIs have been widely reported, including skin reactions, thyroid disorders, pneumonitis, colitis, hepatitis, hypophysitis, and myocarditis [15,16]. Other severe adverse events not related to ICIs can emerge as well during treatment and lead to hospitalization. In a meta-analysis of 35 clinical trials involving ICIs, irAEs of grade 3 and above were reported in 14% of patients treated with monotherapy ICIs, 34% with anti-CTLA-4 antibodies, 46% with combination ICI-chemotherapy, and 55% with ICIs combinations [17].
Fatality rates were observed in 0.36% of patients treated with PD-1 inhibitors, 0.38% with PD-L1 inhibitors, 1.08% with CTLA-4 inhibitors, and 1.23% with combination therapy of PD-1/PD-L1 and CTLA-4 inhibitors [18]. Patients who expired during PD-1/PD-L1 inhibitor treatment had severe complications, including pneumonitis (35%), hepatitis (22%), and neurotoxicities (15%); a majority of the deaths observed in the CTLA-4 treatment group were due to severe colitis (70%) [18].
Several prominent oncologic societies, such as the National Comprehensive Cancer Network (NCCN), American Society of Clinical Oncology (ASCO), and Society for Immunotherapy of Cancer (SITC), have published guidelines on the management of irAEs in the standard clinical setting. However, irAE management in patients who require hospitalization and are steroids-refractory remains problematic [19,20,21]. The characterization of clinical features regarding irAEs and non-irAEs in hospitalized patients may facilitate the understanding and management of toxicities in this setting.
Previous studies have described associations between several tumor genomic features and the tumor response to ICIs. Notably, a poor tumor response was reported in patients on ICIs with molecular alterations in EGFR or MET [22,23,24]. In addition to a poor response, the development of severe irAEs (especially within 3 months) has been described in a retrospective analysis of EGFR mutated NSCLC patients (15%; 6/41) treated with ICIs followed by osimertinib, although the underlying mechanisms are still poorly understood [25]. However, the development of irAEs has not been established as a predictive marker in measuring responsiveness to ICIs.
The clinical characterization of irAEs and full assessment of genomic data is necessary to optimize the patient selection criteria for ICI treatment, understand the underlying mechanisms of irAE development, and develop novel strategies to avoid irAEs while maintaining the anti-tumor efficacy [26]. In our retrospective analysis, we collected clinical and molecular information on 90 patients diagnosed with thoracic malignancies who received ICI treatment and were subsequently hospitalized in order to characterize irAE and non-irAE development, evaluate the management of irAEs, and analyze the survival outcomes.

2. Materials and Methods

2.1. Patients

Patients with metastatic thoracic malignancies who were hospitalized after receiving ICI treatment (pembrolizumab, nivolumab, atezolizumab, and ipilimumab/nivolumab) in different treatment settings, including standard of care, compassionate use, and clinical trials at City of Hope were reviewed. Ninety patients with histologies, including SCLC, NSCLC, and other thoracic malignancies were identified. Demographic, clinical, and pathological information was collected with approval by the City of Hope institutional review board (IRB #18529). The overall survival (OS) was measured from the start of the ICI treatment to the date of death and calculated, if available, at the study time point. The data cutoff date was 8 November 2018.

2.2. Clinical and Molecular Information Collection

Tumor genomic alterations (GAs) were extracted from the available clinical data on next-generation sequencing (NGS) via several platforms, including FoundationOne (Foundation Medicine, Cambridge, MA, USA), Caris (Caris Life Sciences, Phoenix, AZ, USA), Paradigm (Paradigm Diagnostics, Phoenix, AZ, USA), Guardant360 (Guardant, Redwood City, CA, USA), NeoGenomics (NeoGenomics Laboratories, Fort Myers, FL, USA), and City of Hope gene sequencing panels. The PD-L1 (22C3) expression by immunohistochemistry was reported as the tumor proportion score (TPS), which is defined as the percentage of viable tumor cells showing partial or complete membrane staining of ≥1% relative to all viable tumor cells present in the sample.
Negative PD-L1 expression was defined as <1% of viable tumor cells showing membranous staining. The tumor mutational burden (TMB) was reported and categorized as low (≤5 Muts/Mb), intermediate (6–19 Muts/Mb), or high (≥20 Muts/Mb) by Foundation Medicine. Somatic GAs were sorted by the detected positive rate of GAs among all tested patients (the number of tested patients for each gene varied due to different gene panels in the testing platforms). IrAEs were defined as treatment-related toxicities documented by the admitting physician or primary oncologist and independently confirmed by another physician who reviewed the patient medical charts, including the laboratory, imaging, and pathological evidence.
The severity of irAEs was documented from grade 1 to 5 as per the National Institute of Health Common Terminology Criteria for Adverse Events (CTCAE), version 4.03. Clinical information, such as lines of therapy; the length of stay (LOS) in hospital; the status of metastatic disease in the brain; the therapy regimen of ICIs; the management of irAEs, including invasive, diagnostic, and therapeutic procedures; and any interruption or discontinuation of ICIs due to irAEs was also collected.

2.3. Statistical Analysis

First, the association of clinical and molecular features with the OS was analyzed using the univariate Cox proportional hazards model. Based on the results of the univariate analysis, clinically and biologically relevant features with statistical significance (cutoff p-value of 0.05 with the number of patients, n ≥ 5) were selected for the multivariate Cox proportional hazards model. TMB was categorized by Foundation Medicine molecular testing reports. PD-L1 expression was categorized as negative (<1%), and positive (grouped as 1–49% and ≥50%).
We used the Kaplan–Meier method and log-rank test to estimate the OS, and we compared the survival curves, respectively. The chi-square test, Fisher’s exact test, and logistic regression were used for comparison between patient groups (i.e., patients who had irAEs vs. patients who did not have irAEs). Statistical analyses and data visualization were performed using GraphPad Prism 8 (GraphPad Software, Version 8, Graphpad Holdings, LLC, San Diego, CA, USA) and R (version 3.6.2, R Foundation for Statistical Computing, Vienna, Austria) [27]. All tests were two-sided, and p < 0.05 was considered statistically significant.

3. Results

3.1. Patients Characteristics

Ninety patients with stage IV thoracic malignancies underwent admission to the City of Hope after ICI treatment. The dates of ICI treatment initiation were between 6 May 2015, and 6 August 2018. Of those admitted, 28 (31.1%) had documented irAEs, and 62 (68.9%) did not experience any irAEs (Table 1). The most common irAE was pneumonitis (n = 10, 11.1%) followed by adrenal insufficiency (n = 4, 4.4%), hypothyroidism (n = 4, 4.4%), colitis (n = 4, 4.4%), liver injury (n = 3, 3.3%), nephritis (n = 2, 2.2%), infection (n = 2, 2.2%), rash (n = 2, 2.2%), heart failure (n = 1, 1.1%), pancreatitis (n = 1, 1.1%), diabetic ketone acidosis (n = 1, 1.1%), and arthralgia (n = 1, 1.1%). Seven patients (7.8%) experienced multiple irAEs.
The baseline characteristics of the 90 patients are summarized in Table 2. Disease histologies included 63 patients (70%) with adenocarcinoma, 14 (15.6%) with squamous cell lung cancer, 5 (5.6%) with SCLC, and 8 (8.9%) with other types (1 poorly differentiated tumor including NSCLC, not otherwise specified (NSCLC-NOS), 1 large cell lung cancer, 1 lung atypical carcinoid, 1 adenosquamous tumor, 1 mixed large cell with neuroendocrine tumor, 1 small cell transformed lung adenocarcinoma, 1 mixed adenocarcinoma with large cell neuroendocrine tumor, and 1 mesothelioma). Thirty-five patients (38.9%) had documented brain metastases. The median age was 68.5 years (range 36–88), with 70.5 years in the irAEs group and 67.5 years in the non-irAEs group.
Gender was similarly divided in our patient population (41 women, 45.6%, and 49 men, 54.4%). Seventy-eight patients (86.7%) received ICIs as monotherapy and 12 (13.3%) received ICIs combined with chemotherapy. The median lines of therapy were two (range one to seven lines). The median LOS was 7 days (range 1–37 days). The smoking history of our cohort confirmed 29 never smokers (32.2%), 50 former smokers (55.6%), and 11 current smokers (12.2%). PD-L1 by IHC was reported in 45 patients: 16 (35.6%) were negative (TPS of <1%), 8 (17.8%) were positive (TPS of 1–49%), and 21 (46.7%) were highly positive (TPS of ≥50%). Seventy-seven patients underwent EGFR molecular testing, and 20.8% (n = 16) were EGFR positive. The mutational landscape of our patient population is shown in Figure 1.

3.2. Clinical Features in irAEs and Non-irAE Population

The irAE group comprised more male patients (n = 20, 71.4% vs. n = 29, 46.8%; p < 0.05; Figure 2A), and more current smokers (n = 6, 21.4% vs. n = 5, 8.1%; p < 0.01; Figure 2B) and former smokers (n = 19, 67.9% vs. n = 31, 50%; p < 0.01; Figure 2B). The non-irAE group comprised more never smokers (n = 26, 41.9% vs. n = 3, 4.8%; p < 0.01; Figure 2B). Seventeen (60.7%) patients in the irAE group underwent invasive diagnostic procedures during hospitalization, including bronchoscopy (n = 6, 21.4%), esophageal gastroscopy/colonoscopy (n = 5, 17.9%), thoracentesis (n = 2, 7.1%), liver biopsy (n = 1, 3.6%), skin biopsy (n = 1, 3.6%), kidney biopsy (n = 1, 3.6%), and brain surgery (n = 1, 3.6%) as shown in Table 3. In the non-irAE group, 25 (40.3%) patients underwent thoracentesis (n = 8, 12.9%), bronchoscopy (n = 6, 9.7%), EGD/colonoscopy (n = 6, 9.7%), liver biopsy (n = 3, 4.8%), brain surgery (n = 2, 3.2%), spine surgery (n = 1, 1.6%), and pericardium biopsy (n = 1, 1.6%).
IrAE and non-irAE development were not associated with statistically significant superior OS (Figure 3A). However, we observed a trend toward significance in OS with patients who experienced irAEs compared to those who did not experience irAEs (median 16.4 vs. 6.8 months, p = 0.19, Figure 3A). A significant OS benefit was confirmed by multivariate analysis for irAE patients (n = 19/28, 67.9%) who underwent ICI treatment interruption due to irAE occurrence (n = 19/90, 21.1% vs. n = 71/90, 78.9%; median 18.5 vs. 6.2 months; p < 0.05) and visualized on survival curves (HR with 95% CI: 0.47 (0.28–0.79); p < 0.05; Figure 3B). Patients on the first line of ICI therapy had significantly longer OS than those on second-line or greater ICI therapy (p < 0.01).

3.3. Molecular Features in irAE and Non-irAE Population

In the overall population, TP53 ranked as the most detected GA (n = 40/66, 60.6%) followed by LRP1B (n = 12/32, 37.5%), KRAS (n = 23/77, 29.9%), MLL3 (n = 9/32, 28.1%), EGFR (n = 16/77, 20.8%), and PIK3CA (n = 9/66, 13.6%). We analyzed the association between recurrent tumor mutations and irAE occurrence (Table 4). We observed the enrichment of LRP1B mutations (n = 5/6, 83.3% vs. n = 7/26, 26.9%; OR (95% CI) = 13.5 (1.7–166.1), p < 0.05; Figure 2C) and MLL3 mutations (n = 4/6, 66.7% vs. n = 5/26, 19.2%; OR (95% CI) = 8.4 (1.3–49.3), p < 0.05; Figure 2D) in irAE patients compared to non-irAE patients.
However, no statistically significant difference was found in the MLL3 or LRP1B mutation status corresponding with irAE occurrence in our multivariate logistic regression analysis (Table 5). The most frequent GAs and patient demographic information are visualized in the oncoplot in Figure 1. Patients with MET (n = 5/67, 7.5%) or FANCA GAs (n = 5/32, 15.6%) demonstrated shorter median OS compared to patients without MET (median 2.7 vs. 7.2 months; HR with 95% CI: 3.1 (0.57–17.1), p < 0.05) or FANCA GAs (median 3.0 vs. 12.4 months; HR with 95% CI: 3.1 (0.70–13.8); p < 0.05) (Figure 3D). This relationship between OS and GAs in MET (HR, 3.06; 95% CI, 1.08–8.65; p < 0.05) and FANCA (HR, 3.31; 95% CI, 1.22–9.04; p < 0.05) was retained in the multivariate Cox analysis (Table 6).

4. Discussion

The use of ICIs in lung cancer treatment has drastically improved the outcomes of advanced NSCLC patients with an average five-year OS of 15.6% with nivolumab and 23.2% with pembrolizumab as a first-line therapy [28,29]. However, patients who undergo ICI treatment can experience hospital admissions due to severe irAEs and/or other comorbidities. As researchers continue to investigate ICI treatment in earlier-stage disease, it is necessary to explore strategies in minimizing toxicities and avoiding severe irAEs that could be long-lasting or fatal. In this study, we analyzed 90 patients with thoracic cancers who were hospitalized during ICI treatment. Of those, 28 patients (31.1%) experienced irAEs with the most common irAE being pneumonitis (n = 10/90, 11.1%).
This is consistent with other reports demonstrating that 12% of emergency room visits and inpatient care were associated with irAE development in metastatic solid tumor patients undergoing ICI treatment [30]. This result is also consistent with a previous study that reported immune-related interstitial pneumonia as the most common irAE in 13.2% (n = 5/38) of lung cancer patients treated with nivolumab [31].
We also reported that patients with documented irAEs underwent more invasive diagnostic procedures but with no observed difference in the hospital LOS. The severity of irAEs may have caused further intensive interventions due to the risk of long-lasting effects. Sattar et al. described a correlative study between irAEs and efficacy in an older patient population treated with ICIs, and patients age ≥75 years did not present with excess toxicities [32], consistent with our findings of no associations between irAE development and age.
However, we did not observe an OS benefit between our irAE and non-irAE populations. Previous studies have demonstrated superior progression-free survival (PFS) and OS in patients with irAEs, while our study only demonstrated a trend toward significance for OS in our irAE group [31,33,34,35]. In a large observational study, Grangeon et al. measured the survival outcomes in 270 patients with metastatic NSCLC treated with at least one dose of anti-PD-L1 or anti-PD-1 antibodies. The study stratified cohorts between patients who did and did not experience irAEs.
Correspondingly, superior PFS and OS were seen in the cohort who experienced irAEs compared to those who did not experience irAEs (OS: not reached (NR) versus (vs) 8.21 months (hazard ratio (HR) 0.29; 95% confidence interval (CI) 0.18–0.46; p = 0.001); PFS: 5.2 vs. 1.97 months (HR 0.42; 95% CI 0.32–0.57; p < 0.001)). Interestingly, other measures such as the overall response rate (ORR) (22.9% vs. 5.7%, p < 0.0001) and disease control rate (DCR) (76% vs. 58%, p < 0.001) were also lengthened in the irAE-positive vs. non-irAE cohorts [36]. In our cohort, we did not observe any survival benefit with the use of corticosteroids.
Interestingly, Haratani et al. showed that patients who required systemic corticosteroids for irAE management had superior survival outcomes, while Shafqat et al. [35]. demonstrated that irAEs were associated with improved PFS regardless of systemic corticosteroids use [35,37]. The 19 patients who had discontinuation or interruption of ICIs due to irAEs had significantly longer OS, which implied the positive correlations of irAEs with survival outcomes. However, in clinical practice, people might be more comfortable to stop treatment when their disease is better-controlled; therefore, this might be a highly selective patient subpopulation.
Our results demonstrated an OS benefit for patients who underwent ICI treatment as first-line compared to second-line or greater (p < 0.01). A study by Durbin et al. confirmed our results by showing a shorter OS in metastatic solid tumor patients who underwent ICI treatment as second-line or greater [38]. However, another study also analyzed the safety and efficacy of ICIs as second-line treatment in a real-world setting. Chen et al. described the association between the occurrence of irAEs and higher PFS in a patient population who received ICIs in the second-line setting and concluded that the presence of irAEs may act as a predictive marker for antitumor efficacy [39].
Next, our study revealed that patients who experienced an interruption of ICI treatment due to irAEs had significantly longer OS than those who continued treatment (p < 0.05), suggesting a positive correlation between irAE occurrence and survival outcomes. Conversely, Ksienski et al. showed that treatment interruptions in NSCLC patients undergoing treatment with pembrolizumab or nivolumab due to documented irAEs (n = 116/271, 42.8%) were associated with a worse OS [40].
A correlative study by Mouri et al. retrospectively analyzed 49 NSCLC patients treated with nivolumab that had treatment interruption due to a serious irAE. With patients stratified into a retreatment or discontinuation cohort, patients rechallenged with nivolumab displayed an ORR of 15%, without a significant increase in irAEs; however, the median OS and PFS did not differ significantly among the patient cohorts [41].
The difference among survival outcomes with varying ICIs used for treatment may also play a role in discontinuation if the patient experiences detrimental irAEs. Lastly, Jia et al. describe varying biomarkers that can predict irAEs based on specific and nonspecific symptoms. Due to irAE effects in every organ, ongoing investigation in regards to the application scope, benefit from treatment interruption, and selection of the treatment population for ICIs based on biomarkers is required [42].
Our analysis reported enrichment of somatic LRP1B and MLL3 mutations in patients with irAEs. Yet, it was not statistically significant in the multivariate analysis, likely due to the limited sample size and confounding factors of smoking and gender. LRP1B gene encodes for an LDL receptor and acts as a putative tumor suppressor in lung cancer whose function is only partially defined [43,44]. Chen et al. reported greater survival and higher TMB in melanoma and NSCLC patients with LRP1B mutations undergoing ICI treatment [45]. MLL3 gene encodes for histone 3 lysine 4 methyltransferases and acts as a tumor suppressor.
Mutated MLL3 (or KMT2C) proteins have been implicated in multiple cancers, including urothelial carcinoma, human lymphoid, and myeloid leukemia [46,47,48]. Further, we found that NSCLC patients with FANCA mutations had significantly worse OS compared to those without FANCA mutations. The FANCA gene is important for the repair of double-stranded DNA breaks and is involved in the cellular process known as the Fanconi anemia pathway. Outcomes have been assessed in patients treated with ICIs, and the results showed significantly higher objective response rate, longer median PFS, and longer median OS with patients on PD-(L)1 therapy [49].
The survival outcome that we observed was also previously confirmed in a larger cohort by our group, yet further investigation is required [50]. The MET gene encodes a transmembrane receptor tyrosine kinase, and its ligand hepatocyte growth factor (HGF) is involved in the MET/HGF signaling pathway. Patients in our cohort with MET GAs were associated with poor OS, and the statistical significance was retained in the multivariate analysis.
This is consistent with the previous findings of MET mutated lung cancer and worsened outcomes with immunotherapy treatment [24,51]. We did not observe a correlation between TMB with irAEs or OS, which may be explained by the lack of TMB information in our cohort as only 18 patients had TMB information available. It is unclear how somatic mutations in tumors contribute to the development of irAEs, and more research is warranted to examine the role of genomic mutations in lung cancer immunotherapy.

5. Conclusions

Our retrospective analysis investigated the clinical and molecular features of lung cancer patients undergoing ICI treatment who were hospitalized. We observed that patients with irAE occurrences who required treatment interruption had a significantly longer OS. Further, patients with somatic GAs in FANCA and MET had a worse OS, which is consistent with previously reported studies. A limitation of this study is that the patient cohort was of limited sample size and from a single institution. Further investigation is required to analyze a larger and diverse population set. Strikingly, our findings indicated that the majority of patients who were hospitalized on ICI treatment did not have an irAE. Therefore, future clinical studies should focus on identifying and cataloging the variables that may be associated with hospitalization due to ICI treatment.

Author Contributions

Conceptualization, D.Z., H.L., I.M., E.M., M.K., K.L.R., K.M., and R.S.; Data curation, D.Z., I.M., C.C., R.P., J.F., A.R.B., and Y.X.; Formal analysis, D.Z., H.L., I.M., C.C., E.M., M.K., K.L.R., K.M., and R.S.; Funding acquisition, D.Z. and R.S.; Investigation, D.Z., H.L., I.M., C.C., R.P., J.F., A.R.B., P.K., Y.X., E.M., M.K., K.L.R., K.M., and R.S.; Methodology, D.Z., H.L., I.M., C.C., R.P., J.F., A.R.B., P.K., Y.X., E.M., M.K., K.L.R., K.M., and R.S.; Project administration, P.K. and R.S.; Resources, R.S.; Software, H.L.; Supervision, P.K. and R.S.; Validation, D.Z., H.L., I.M., C.C., R.P., J.F., A.R.B., P.K., Y.X., E.M., M.K., K.L.R., K.M., and R.S.; Visualization, D.Z., H.L., I.M., and C.C.; Roles/Writing—original draft, D.Z., H.L., I.M., and R.S.; Writing—review & editing, D.Z., H.L., I.M., C.C., R.P., J.F., A.R.B., P.K., Y.X., E.M., M.K., K.L.R., K.M., and R.S.; All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Cancer Institute of the National Institutes of Health under awards numbers P30CA033572 and U54CA209978.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of City of Hope (IRB #18529; 12/20/2019).

Informed Consent Statement

Patient consent was waived per IRB requirements since it was a retrospective observational study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

We thank the Thoracic Disease Team, Department of Medical Oncology, and Center for Clinical Informatics at City of Hope for their help in this analysis.

Conflicts of Interest

The authors declare that they have no competing interest relevant to this work.

Abbreviations

CIConfidence interval
CTLA-4Cytotoxic T-lymphocyte–associated antigen 4
DCRDisease control rate
EGDEsophagogastroduodenoscopy
FANCAFanconi anemia complementation group A
FDAFood and Drug Administration
GAsGenomic alterations
HGFHepatocyte growth factor
HRHazard ratio
ICIsImmune checkpoint inhibitors
IHCImmunohistochemistry
IrAEsImmune-related adverse events
IRBInstitutional review board
KMT2CLysine (K) methyltransferase 2C
LRP1BLow-density lipoprotein (LDL) receptor-related protein 1B
LOSLength of stay
MLL3Mixed-lineage leukemia protein 3
NGSNext-generation sequencing
NSCC-NOSNon-small cell carcinoma, not otherwise specified
NSCLCNon-small cell lung cancer
OROdds ratio
ORROverall response rate
OSOverall survival
PD-1Program death -1
PD-L1Programmed death-ligand 1
PFSProgression-free survival
SCLCSmall cell lung cancer
TMBTumor mutation burden
VSVersus

References

  1. Mok, T.S.K.; Wu, Y.L.; Kudaba, I.; Kowalski, D.M.; Cho, B.C.; Turna, H.Z.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830. [Google Scholar] [CrossRef]
  2. Gandhi, L.; Rodriguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
  3. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gumus, M.; Mazieres, J.; Hermes, B.; Cay Senler, F.; Csoszi, T.; Fulop, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef]
  4. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus chemotherapy for PD-L1–positive non–small-cell lung cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Horn, L.; Mansfield, A.S.; Szczesna, A.; Havel, L.; Krzakowski, M.; Hochmair, M.J.; Huemer, F.; Losonczy, G.; Johnson, M.L.; Nishio, M.; et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2220–2229. [Google Scholar] [CrossRef]
  6. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef]
  7. Paz-Ares, L.; Dvorkin, M.; Chen, Y.; Reinmuth, N.; Hotta, K.; Trukhin, D.; Statsenko, G.; Hochmair, M.J.; Özgüroğlu, M.; Ji, J.H.; et al. Durvalumab plus platinum–etoposide versus platinum–etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): A randomised, controlled, open-label, phase 3 trial. Lancet 2019, 394, 1929–1939. [Google Scholar] [CrossRef]
  8. Antonia, S.J.; Lopez-Martin, J.A.; Bendell, J.; Ott, P.A.; Taylor, M.; Eder, J.P.; Jager, D.; Pietanza, M.C.; Le, D.T.; de Braud, F.; 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, 883–895. [Google Scholar] [CrossRef] [Green Version]
  9. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crino, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [Green Version]
  10. Brahmer, J.R.; Govindan, R.; Anders, R.A.; Antonia, S.J.; Sagorsky, S.; Davies, M.J.; Dubinett, S.M.; Ferris, A.; Gandhi, L.; Garon, E.B.; et al. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of non-small cell lung cancer (NSCLC). J. Immunother. Cancer 2018, 6, 75. [Google Scholar] [CrossRef]
  11. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non‚ÄìSmall-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
  12. Reck, M.; Ciuleanu, T.-E.; Dols, M.C.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. Nivolumab (NIVO) + ipilimumab (IPI) + 2 cycles of platinum-doublet chemotherapy (chemo) vs. 4 cycles chemo as first-line (1L) treatment (tx) for stage IV/recurrent non-small cell lung cancer (NSCLC): CheckMate 9LA. J. Clin. Oncol. 2020, 38, 9501. [Google Scholar] [CrossRef]
  13. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef] [Green Version]
  14. Forde, P.M.; Chaft, J.E.; Smith, K.N.; Anagnostou, V.; Cottrell, T.R.; Hellmann, M.D.; Zahurak, M.; Yang, S.C.; Jones, D.R.; Broderick, S.; et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N. Engl. J. Med. 2018, 378, 1976–1986. [Google Scholar] [CrossRef] [PubMed]
  15. Postow, M.A.; Sidlow, R.; Hellmann, M.D. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N. Engl. J. Med. 2018, 378, 158–168. [Google Scholar] [CrossRef] [PubMed]
  16. Weber, J.S.; Kahler, K.C.; Hauschild, A. Management of immune-related adverse events and kinetics of response with ipilimumab. J. Clin. Oncol. 2012, 30, 2691–2697. [Google Scholar] [CrossRef]
  17. Arnaud-Coffin, P.; Maillet, D.; Gan, H.K.; Stelmes, J.J.; You, B.; Dalle, S.; Peron, J. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. Int. J. Cancer 2019, 145, 639–648. [Google Scholar] [CrossRef]
  18. Wang, D.Y.; Salem, J.E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 1721–1728. [Google Scholar] [CrossRef] [Green Version]
  19. Thompson, J.A.; Schneider, B.J.; Brahmer, J.; Andrews, S.; Armand, P.; Bhatia, S.; Budde, L.E.; Costa, L.; Davies, M.; Dunnington, D.; et al. Management of Immunotherapy-Related Toxicities, Version 1.2019. J. Natl. Compr. Cancer Netw. 2019, 17, 255–289. [Google Scholar] [CrossRef] [Green Version]
  20. Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O., 3rd; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. Immunother. Cancer 2017, 5, 95. [Google Scholar] [CrossRef] [Green Version]
  21. Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; Ernstoff, M.S.; Gardner, J.M.; Ginex, P.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018, 36, 1714–1768. [Google Scholar] [CrossRef]
  22. Hellmann, M.D.; Nathanson, T.; Rizvi, H.; Creelan, B.C.; Sanchez-Vega, F.; Ahuja, A.; Ni, A.; Novik, J.B.; Mangarin, L.M.B.; Abu-Akeel, M.; et al. Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. Cancer Cell 2018, 33, 843–852.e4. [Google Scholar] [CrossRef] [Green Version]
  23. Skoulidis, F.; Goldberg, M.E.; Greenawalt, D.M.; Hellmann, M.D.; Awad, M.M.; Gainor, J.F.; Schrock, A.B.; Hartmaier, R.J.; Trabucco, S.E.; Gay, L.; et al. STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov. 2018, 8, 822–835. [Google Scholar] [CrossRef] [Green Version]
  24. Mazieres, J.; Drilon, A.; Lusque, A.; Mhanna, L.; Cortot, A.B.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  25. Schoenfeld, A.J.; Arbour, K.C.; Rizvi, H.; Iqbal, A.N.; Gadgeel, S.M.; Girshman, J.; Kris, M.G.; Riely, G.J.; Yu, H.A.; Hellmann, M.D. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann. Oncol. 2019, 30, 839–844. [Google Scholar] [CrossRef] [PubMed]
  26. June, C.H.; Warshauer, J.T.; Bluestone, J.A. Is autoimmunity the Achilles' heel of cancer immunotherapy? Nat. Med. 2017, 23, 540–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. R Core Team. R: A Language and Environment for Statistical Computing; R Core Team: Vienna, Austria, 2019. [Google Scholar]
  28. Garon, E.B.; Hellmann, M.D.; Rizvi, N.A.; Carcereny, E.; Leighl, N.B.; Ahn, M.J.; Eder, J.P.; Balmanoukian, A.S.; Aggarwal, C.; Horn, L.; et al. Five-Year Overall Survival for Patients With Advanced NonSmall-Cell Lung Cancer Treated With Pembrolizumab: Results From the Phase I KEYNOTE-001 Study. J. Clin. Oncol. 2019, 37, 2518. [Google Scholar] [CrossRef] [PubMed]
  29. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Sosman, J.A.; Atkins, M.B.; Leming, P.D.; et al. Five-Year Survival and Correlates Among Patients With Advanced Melanoma, Renal Cell Carcinoma, or Non-Small Cell Lung Cancer Treated With Nivolumab. JAMA Oncol. 2019, 5, 1411–1420. [Google Scholar] [CrossRef] [Green Version]
  30. Parikh, A.B.; Zhong, X.; Mellgard, G.; Qin, Q.; Patel, V.G.; Wang, B.; Alerasool, P.; Garcia, P.; Leiter, A.; Gallagher, E.J.; et al. Risk Factors for Emergency Room and Hospital Care Among Patients With Solid Tumors on Immune Checkpoint Inhibitor Therapy. Am. J. Clin. Oncol. 2021, 44, 114–120. [Google Scholar] [CrossRef]
  31. Sato, K.; Akamatsu, H.; Murakami, E.; Sasaki, S.; Kanai, K.; Hayata, A.; Tokudome, N.; Akamatsu, K.; Koh, Y.; Ueda, H.; et al. Correlation between immune-related adverse events and efficacy in non-small cell lung cancer treated with nivolumab. Lung Cancer 2018, 115, 71–74. [Google Scholar] [CrossRef] [Green Version]
  32. Sattar, J.; Kartolo, A.; Hopman, W.M.; Lakoff, J.M.; Baetz, T. The efficacy and toxicity of immune checkpoint inhibitors in a real-world older patient population. J. Geriatr. Oncol. 2018. [Google Scholar] [CrossRef]
  33. Ricciuti, B.; Genova, C.; De Giglio, A.; Bassanelli, M.; Dal Bello, M.G.; Metro, G.; Brambilla, M.; Baglivo, S.; Grossi, F.; Chiari, R. Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab: Long-term outcomes from a multi-institutional analysis. J. Cancer Res. Clin. Oncol. 2019, 145, 479–485. [Google Scholar] [CrossRef]
  34. Ahn, B.C.; Pyo, K.H.; Xin, C.F.; Jung, D.; Shim, H.S.; Lee, C.Y.; Park, S.Y.; Yoon, H.I.; Hong, M.H.; Cho, B.C.; et al. Comprehensive analysis of the characteristics and treatment outcomes of patients with non-small cell lung cancer treated with anti-PD-1 therapy in real-world practice. J. Cancer Res. Clin. Oncol. 2019, 145, 1613–1623. [Google Scholar] [CrossRef] [Green Version]
  35. Haratani, K.; Hayashi, H.; Chiba, Y.; Kudo, K.; Yonesaka, K.; Kato, R.; Kaneda, H.; Hasegawa, Y.; Tanaka, K.; Takeda, M.; et al. Association of Immune-Related Adverse Events With Nivolumab Efficacy in Non-Small-Cell Lung Cancer. JAMA Oncol. 2018, 4, 374–378. [Google Scholar] [CrossRef]
  36. Grangeon, M.; Tomasini, P.; Chaleat, S.; Jeanson, A.; Souquet-Bressand, M.; Khobta, N.; Bermudez, J.; Trigui, Y.; Greillier, L.; Blanchon, M.; et al. Association Between Immune-related Adverse Events and Efficacy of Immune Checkpoint Inhibitors in Non-small-cell Lung Cancer. Clin. Lung Cancer 2019, 20, 201–207. [Google Scholar] [CrossRef] [PubMed]
  37. Shafqat, H.; Gourdin, T.; Sion, A. Immune-related adverse events are linked with improved progression-free survival in patients receiving anti-PD-1/PD-L1 therapy. Semin. Oncol. 2018, 45, 156–163. [Google Scholar] [CrossRef] [PubMed]
  38. Durbin, S.M.; Zubiri, L.; Niemierko, A.; Bardia, A.; Sullivan, R.J.; McEwen, C.; Mulvey, T.M.; Allen, I.M.; Lawrence, D.P.; Cohen, J.V.; et al. Clinical Outcomes of Patients with Metastatic Cancer Receiving Immune Checkpoint Inhibitors in the Inpatient Setting. Oncologist 2021, 26, 49–55. [Google Scholar] [CrossRef]
  39. Chen, M.; Li, Q.; Xu, Y.; Zhao, J.; Zhang, L.; Wei, L.; Zhong, W.; Wang, M. Immunotherapy as second-line treatment and beyond for non-small cell lung cancer in a single center of China: Outcomes, toxicities, and clinical predictive factors from a real-world retrospective analysis. Thorac. Cancer 2020, 11, 1955–1962. [Google Scholar] [CrossRef]
  40. Ksienski, D.; Wai, E.S.; Croteau, N.; Fiorino, L.; Brooks, E.; Poonja, Z.; Fenton, D.; Geller, G.; Glick, D.; Lesperance, M. Efficacy of Nivolumab and Pembrolizumab in Patients With Advanced Non-Small-Cell Lung Cancer Needing Treatment Interruption Because of Adverse Events: A Retrospective Multicenter Analysis. Clin. Lung Cancer 2019, 20, e97–e106. [Google Scholar] [CrossRef] [PubMed]
  41. Mouri, A.; Kaira, K.; Yamaguchi, O.; Shiono, A.; Miura, Y.; Hashimoto, K.; Nishihara, F.; Murayama, Y.; Kobayashi, K.; Kagamu, H. Clinical difference between discontinuation and retreatment with nivolumab after immune-related adverse events in patients with lung cancer. Cancer Chemother. Pharmacol. 2019, 84, 873–880. [Google Scholar] [CrossRef] [PubMed]
  42. Jia, X.-H.; Geng, L.-Y.; Jiang, P.-P.; Xu, H.; Nan, K.-J.; Yao, Y.; Jiang, L.-L.; Sun, H.; Qin, T.-J.; Guo, H. The biomarkers related to immune related adverse events caused by immune checkpoint inhibitors. J. Exp. Clin. Cancer Res. 2020, 39, 284. [Google Scholar] [CrossRef] [PubMed]
  43. Beer, A.G.; Zenzmaier, C.; Schreinlechner, M.; Haas, J.; Dietrich, M.F.; Herz, J.; Marschang, P. Expression of a recombinant full-length LRP1B receptor in human non-small cell lung cancer cells confirms the postulated growth-suppressing function of this large LDL receptor family member. Oncotarget 2016, 7, 68721–68733. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, C.X.; Musco, S.; Lisitsina, N.M.; Yaklichkin, S.Y.; Lisitsyn, N.A. Genomic organization of a new candidate tumor suppressor gene, LRP1B. Genomics 2000, 69, 271–274. [Google Scholar] [CrossRef]
  45. Chen, H.; Chong, W.; Wu, Q.; Yao, Y.; Mao, M.; Wang, X. Association of LRP1B Mutation With Tumor Mutation Burden and Outcomes in Melanoma and Non-small Cell Lung Cancer Patients Treated With Immune Check-Point Blockades. Front. Immunol. 2019, 10, 1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Fountzilas, E.; Kotoula, V.; Koliou, G.A.; Giannoulatou, E.; Gogas, H.; Papadimitriou, C.; Tikas, I.; Zhang, J.; Papadopoulou, K.; Zagouri, F.; et al. Pathogenic mutations and overall survival in 3,084 patients with cancer: The Hellenic Cooperative Oncology Group Precision Medicine Initiative. Oncotarget 2020, 11, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Chen, C.; Liu, Y.; Rappaport, A.R.; Kitzing, T.; Schultz, N.; Zhao, Z.; Shroff, A.S.; Dickins, R.A.; Vakoc, C.R.; Bradner, J.E.; et al. MLL3 is a haploinsufficient 7q tumor suppressor in acute myeloid leukemia. Cancer Cell 2014, 25, 652–665. [Google Scholar] [CrossRef] [Green Version]
  48. Rampias, T.; Karagiannis, D.; Avgeris, M.; Polyzos, A.; Kokkalis, A.; Kanaki, Z.; Kousidou, E.; Tzetis, M.; Kanavakis, E.; Stravodimos, K.; et al. The lysine-specific methyltransferase KMT2C/MLL3 regulates DNA repair components in cancer. EMBO Rep. 2019, 20. [Google Scholar] [CrossRef] [PubMed]
  49. Ricciuti, B.; Recondo, G.; Spurr, L.F.; Li, Y.Y.; Lamberti, G.; Venkatraman, D.; Umeton, R.; Cherniack, A.D.; Nishino, M.; Sholl, L.M.; et al. Impact of DNA Damage Response and Repair (DDR) Gene Mutations on Efficacy of PD-(L)1 Immune Checkpoint Inhibition in Non-Small Cell Lung Cancer. Clin. Cancer Res. 2020, 26, 4135–4142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Zhao, D.; Mambetsariev, I.; Li, H.; Chen, C.; Fricke, J.; Fann, P.; Kulkarni, P.; Xing, Y.; Lee, P.P.; Bild, A.; et al. Association of molecular characteristics with survival in advanced non-small cell lung cancer patients treated with checkpoint inhibitors. Lung Cancer 2020, 146, 174–181. [Google Scholar] [CrossRef]
  51. Sabari, J.K.; Leonardi, G.C.; Shu, C.A.; Umeton, R.; Montecalvo, J.; Ni, A.; Chen, R.; Dienstag, J.; Mrad, C.; Bergagnini, I.; et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann. Oncol. 2018, 29, 2085–2091. [Google Scholar] [CrossRef]
Figure 1. The top detected genomic alterations with OS and irAEs. Oncoplot demonstrating the patient demographic information and top detected genomic alterations in 90 lung cancer patients who were hospitalized during ICI treatment. Mutation rates are shown by patients with irAEs vs. no irAEs. The odds ratios were calculated using univariate logistic regression and hazard ratios with the univariate Cox model. Data visualization and statistical analysis were performed with R.
Figure 1. The top detected genomic alterations with OS and irAEs. Oncoplot demonstrating the patient demographic information and top detected genomic alterations in 90 lung cancer patients who were hospitalized during ICI treatment. Mutation rates are shown by patients with irAEs vs. no irAEs. The odds ratios were calculated using univariate logistic regression and hazard ratios with the univariate Cox model. Data visualization and statistical analysis were performed with R.
Cancers 13 02653 g001
Figure 2. Significant clinical and molecular features with irAEs. (A) Association of irAEs with gender, (B) smoking status, (C) genomic alterations in LRP1B, and (D) MLL3. * Fisher’s exact test by GraphPad.
Figure 2. Significant clinical and molecular features with irAEs. (A) Association of irAEs with gender, (B) smoking status, (C) genomic alterations in LRP1B, and (D) MLL3. * Fisher’s exact test by GraphPad.
Cancers 13 02653 g002
Figure 3. Clinical and molecular features with OS. (A) OS with irAEs, (B) interruption of ICIs due to irAEs, (C) genomic alterations in MET, and (D) FANCA. Log-rank (Mantel–Cox) tests were used to compare the survival curves. Data visualization and statistical analysis were performed with R.
Figure 3. Clinical and molecular features with OS. (A) OS with irAEs, (B) interruption of ICIs due to irAEs, (C) genomic alterations in MET, and (D) FANCA. Log-rank (Mantel–Cox) tests were used to compare the survival curves. Data visualization and statistical analysis were performed with R.
Cancers 13 02653 g003
Table 1. List of irAEs.
Table 1. List of irAEs.
IrAEsNo (%)
Pneumonitis10 (11.1%)
Adrenal insufficiency4 (4.4%)
Hypothyroidism4 (4.4%)
Colitis4 (4.4%)
Liver injury3 (3.3%)
Nephritis2 (2.2%)
Heart failure 1 (1.1%)
Pancreatitis1 (1.1%)
Diabetic ketone acidosis1 (1.1%)
Arthralgia1 (1.1%)
Rash2 (2.2%)
Other/Infection2 (2.2%)
Multiple irAEs7 (7.8%)
Total patients with irAEs28 (31.1%) 1
1 90 patients had hospital admissions during ICI treatment.
Table 2. Baseline patient characteristics.
Table 2. Baseline patient characteristics.
Characteristicsn = 90 (%)IrAEs n = 28 (%)No irAEs n = 62 (%)p Values 1
Median age at ICI (range 36–88)68.570.567.5ns
Gender <0.05
Women41 (45.6%)8 (28.6%)33 (53.2%)
Men49 (54.4%)20 (71.4%)29 (46.8%)
Smoking status <0.01
Current11 (12.2%)6 (21.4%)5 (8.1%)
Former50 (55.6%) 19 (67.9%)31 (50.0%)
Never29 (32.2%)3 (10.7%)26 (41.9%)
Histology <0.01
Lung adenocarcinoma63 (70%)16 (57.1%)47 (75.8%)
Lung squamous 14 (15.6%)4 (14.3%)10 (16.1%)
SCLC5 (5.6%)5 (17.9%)0
Others 28 (8.9%)3 (10.7%)5 (8.1%)
ICIs with other therapy ns
Yes12 (13.3%)2 (7.1%)10 (16.1%)
No78 (86.7%)26 (92.9%)52 (83.9%)
PD-L1 ns
Negative16 (17.8%)5 (17.9%)11 (17.7%)
1% to <50% 8 (8.9%)1 (3.6%)7 (11.3%)
50% and above21 (23.3%)7 (25.0%)14 (22.6%)
Not tested45 (50.0%)15 (53.6%)30 (48.4%)
Median lines of therapy (range 1–7)222ns
Brain metastasis ns
Yes35 (38.9%) 10 (35.7%)25 (40.3%)
No55 (61.1%)18 (64.3%)37 (59.7%)
Median length of stay (range 1–37)776ns
1 Chi-square test and Fisher’s exact test. ns, not significant. 2 Others: 1 poorly differentiated tumor including NSCLC, not otherwise specified (NSCLC-NOS), 1 large cell lung cancer, 1 lung atypical carcinoid, 1 adenosquamous tumor, 1 mixed large cell with neuroendocrine tumor, 1 small cell transformed lung adenocarcinoma, 1 mixed adenocarcinoma with large cell neuroendocrine tumor, and 1 mesothelioma.
Table 3. Invasive procedures after ICIs.
Table 3. Invasive procedures after ICIs.
Invasive Procedures after ICIsirAEs (n = 28)No irAEs (n = 62)
Bronchoscopy/lung biopsy6 (21.4%)6 (9.7%)
EGD/Colonoscopy5 (17.9%)6 (9.7%)
Thoracentesis2 (7.1%)8 (12.9%)
Liver biopsy1 (3.6%)3 (4.8%)
Skin biopsy 1 (3.6%)0
Kidney biopsy1 (3.6%)0
Brain surgery1 (3.6%)2 (3.2%)
Spine surgery01 (1.6%)
Pericardium biopsy01 (1.6%)
Total 17 (60.7%)25 (40.3%) 1
1 Chi-square test p < 0.05. The total number of patients who had invasive procedures, including one patient who had a lung biopsy, thoracentesis, and pericardium biopsy.
Table 4. Mutations and their associations with irAEs.
Table 4. Mutations and their associations with irAEs.
GenomicsAll (%)IrAEs (%)No irAEs (%)OR (95% CI)p Values 1
TP53 ns
Positive4010 (66.7%)30 (58.8%)
Negative265 (33.3%)21 (41.2%)
Not tested241311
KRAS ns
Positive238 (40%)15 (26.3%)
Negative5412 (60%)42 (73.7%)
Not tested1385
EGFR ns
Positive162 (10%)14 (24.6%)
Negative6118 (90%)43 (75.4%)
Not tested1385
LRP1B 13.5 (1.7–166.1)<0.05
Positive125 (83.3%)7 (26.9%)
Negative201 (16.7%)19 (73.1%)
Not tested582236
PIK3CA ns
Positive93 (20%)6 (11.8%)
Negative5712 (80%)45 (88.2%)
Not tested241311
MLL3 8.4 (1.3–49.3)<0.05
Positive94 (66.7%)5 (19.2%)
Negative232 (33.3%)21 (80.8%)
Not tested582236
TMB ns
TMB-Low51 (33.3%)4 (26.7%)
TMB-Intermediate91 (33.3%)8 (53.3%)
TMB-High41 (33.3%)3 (20%)
Not tested722547
Table 5. Risk factors for irAEs by multivariate analysis.
Table 5. Risk factors for irAEs by multivariate analysis.
Risk FactorsOdds Ratio (95% CI)p Values 1
Gender
FemaleReferences
Male1.47 (0.43–4.99)0.5358
Smoking
NeverReferences
Current3.61 (0.43–30.11)0.2363
Former3.44 (0.76–15.45)0.1073
MLL3
NegativeReferences
Positive6.52 (0.70–60.62)0.0991
LRP1B
NegativeReferences
Positive8.00 (0.65–98.01)0.1037
1 Multivariate logistic regression for irAEs by R (excluding small cell lung cancer).
Table 6. Multivariate analysis for OS (n = 90).
Table 6. Multivariate analysis for OS (n = 90).
Risk FactorsHR (95%CI)p Values 1
Gender
FemaleReference
Male1.11 (0.57–2.15)ns
IrAEs
NoReference
Yes1.21 (0.50–2.92)ns
Interrupt ICIs due to irAEs
NoReference
Yes0.05 (0.01–0.19)<0.001
Lines of therapy
≥3 lines Reference
2nd line0.62 (0.25–1.49)
1st line0.21 (0.07–0.58)<0.01
EGFR
NegativeReference
Positive1.45 (0.55–3.77)ns
FANCA
NegativeReference
Positive11.30 (3.36–38.01)<0.001
MET
NegativeReference
Positive 11.17 (2.92–42.81)<0.001
1 Multivariate Cox proportional hazards model for OS (excluding small cell lung cancer).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhao, D.; Li, H.; Mambetsariev, I.; Chen, C.; Pharaon, R.; Fricke, J.; Baroz, A.R.; Kulkarni, P.; Xing, Y.; Massarelli, E.; et al. Molecular and Clinical Features of Hospital Admissions in Patients with Thoracic Malignancies on Immune Checkpoint Inhibitors. Cancers 2021, 13, 2653. https://doi.org/10.3390/cancers13112653

AMA Style

Zhao D, Li H, Mambetsariev I, Chen C, Pharaon R, Fricke J, Baroz AR, Kulkarni P, Xing Y, Massarelli E, et al. Molecular and Clinical Features of Hospital Admissions in Patients with Thoracic Malignancies on Immune Checkpoint Inhibitors. Cancers. 2021; 13(11):2653. https://doi.org/10.3390/cancers13112653

Chicago/Turabian Style

Zhao, Dan, Haiqing Li, Isa Mambetsariev, Chen Chen, Rebecca Pharaon, Jeremy Fricke, Angel R. Baroz, Prakash Kulkarni, Yan Xing, Erminia Massarelli, and et al. 2021. "Molecular and Clinical Features of Hospital Admissions in Patients with Thoracic Malignancies on Immune Checkpoint Inhibitors" Cancers 13, no. 11: 2653. https://doi.org/10.3390/cancers13112653

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

Article Metrics

Back to TopTop