1
|
Han X, Chen Y, Xie H, Zhang Y, Cui Y, Guan Y, Nie W, Xie Q, Li J, Wang B, Zhang B, Wang J. Organ-specific immune-related adverse events and prognosis in cancer patients receiving immune checkpoint inhibitors. BMC Cancer 2025; 25:139. [PMID: 39856626 PMCID: PMC11761211 DOI: 10.1186/s12885-025-13566-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Patients who developed immune-related adverse events (irAEs) could benefit more from treatment with immune checkpoint inhibitors (ICIs) than those who did not develop irAEs. This study was designed to assess whether the occurrence of irAEs or their characteristics are correlated with survival in advanced patients treated with ICIs. METHODS This retrospective cohort study enrolled a panel of cancer patients who received ICIs at a single institute. Kaplan‒Meier curves were generated to describe progression-free survival (PFS) and overall survival (OS) in patients with irAEs or specific irAE characteristics. RESULTS A total of 238 patients were enrolled, 83 (34.9%) of whom developed at least one irAE. Overall, irAE development was associated with prolonged OS (not reached vs. 17.8 months, P < 0.001), PFS (8.7 vs. 4.8 months, P = 0.003), and an improved objective response rate (24.1% vs. 10.3%, P = 0.005). Furthermore, only skin or endocrine toxicities were associated with improved OS and PFS. On the basis of the results from organ-specific irAEs, the first development of skin or endocrine toxicities as protective irAEs rather than other irAEs was an independent indicator for predicting OS (P < 0.001) and PFS (P < 0.001). A protective irAE burden score based on organ-specific irAEs was further developed to show the significant protective effect of total irAEs on patient outcomes. CONCLUSIONS Not all irAEs are associated with prolonged survival. The identification of organ-specific irAEs is useful for stratifying patients who actually respond to and benefit from ICIs across different cancer types.
Collapse
Affiliation(s)
- Xinyue Han
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yingcui Chen
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Hong Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yuekai Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yu Cui
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yaping Guan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Weiwei Nie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Jisheng Li
- Department of Medical Oncology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Baocheng Wang
- Department of Oncology, The 960 Hospital of the People's Liberation Army, Jinan, China
| | - Bicheng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan, 250014, China.
- Shandong Lung Cancer Institute, Jinan, China.
| |
Collapse
|
2
|
Wang Y, Zhou J, Peng S, Cui Z, Wang W, Zeng W, Qiu T, Liu Z. The effect of baseline versus early glucocorticoid use on immune checkpoint inhibitor efficacy in patients with advanced NSCLC. Front Oncol 2025; 15:1533556. [PMID: 39917167 PMCID: PMC11798793 DOI: 10.3389/fonc.2025.1533556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Purpose This study aims to investigate the specific effects of glucocorticoids (GC) on the efficacy of immune checkpoint inhibitors (ICIs), and whether this effect is influenced by the timing and dosage of GC administration. Changes in the neutrophil percentage and the helper/suppressor T lymphocyte ratio [NEUT %/(CD4+/CD8+)] during GC administration were monitored. Methods The clinical results of 130 patients with advanced non-small cell lung cancer (NSCLC) treated with ICIs were analyzed and compared with those of patients who did not use GC. Cox proportional hazards regression model and Logistic regression analysis were used to analyze the factors affecting ORR and PFS, and t test was used to analyze the changes of NEUT %/(CD4 +/CD8 +) during GC use. Results Multivariate Logistic analysis showed that GC use was associated with a higher ORR in 130 patients treated with ICIs [HR = 3.07,95% CI (1.31-7.21), P = 0.010]. Univariate Cox analysis showed that GC use was not significantly correlated with PFS [HR = 0.926,95% CI (0.603-1.420), P = 0.710]. Patients who used GC during the baseline period of ICIs treatment had a higher ORR than those who used GC at the early stage of ICIs treatment (65.4% vs 30.8%, p = 0.024). Multivariate Cox analysis showed that GC use had longer PFS [HR = 0.37,95% CI (0.17-0.78), p = 0.009]. The timing of GC use was different, and there was a difference in NEUT %/(CD4 +/CD8 +) levels before and after treatment. There was no significant difference in ORR and PFS between GC duration and dose. Conclusion The use of GC helps to enhance the efficacy of immunotherapy. In particular, GC use during the baseline period leads to higher ORR and PFS, regardless of the dose or duration of GC use. The levels of NEUT %/(CD4+/CD8+) varied depending on the timing of GC administration.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jianying Zhou
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
| | - Simin Peng
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhao Cui
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Weiqi Wang
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wenqin Zeng
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tingting Qiu
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
| | - Zhentian Liu
- Department of Thoracic Medical Oncology, Jiangxi Provincial Cancer Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Guan Y, Cui Y, Gong Y, Liang X, Han X, Chen Y, Xie H, Zhang Y, Wang B, Ye X, Wang J. Dissociated response and treatment outcome with immune checkpoint blockade in advanced cancer. Sci Rep 2024; 14:32147. [PMID: 39738789 PMCID: PMC11686300 DOI: 10.1038/s41598-024-84009-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025] Open
Abstract
Immune-related dissociated response (DR) has been recently recognized and have become a subject of ongoing interest. The purpose of the present study was to evaluate the frequency, treatment outcome, and predictors of DR in cancer patients with immune checkpoint inhibitors. We retrospectively collected clinicopathological data from a cohort of patients with cancer who received PD-1/PD-L1 inhibitor-based monotherapy or combination therapy at a single institution (developing cohort). An independent cohort of advanced non-small cell lung cancer (NSCLC) patients treated with immunotherapy at two institutions was used as the validating cohort. Progression-free survival (PFS) and overall survival (OS) were used as outcome measures. The pantumor cohort included 177 patients. DR were observed in 12 (6.8%) patients. The median PFS and OS were significantly longer in patients with atypical response versus nonresponse but shorter versus true response. Patients with DR had a longer median PFS and OS than those with true progressive disease (PD). Local treatment seemed to have a positive influence on DR patient outcomes, with a median OS of 32.3 months versus 21.9 months for no local treatment. No clinical characteristics remained significant predictors for DR. In the NSCLC cohort, DR was observed in 10 (12.5%) patients. Inferior PFS and OS were validated in patients with real PD when compared with patients with DR. Patients who experience DR exhibit a relatively favorable prognosis. Some patients with DR may benefit from the continuation of ICI administration and local treatment to the growing lesions and achieve a longer survival.
Collapse
Affiliation(s)
- Yaping Guan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Cui
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yanhong Gong
- Department of Stomatology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiuju Liang
- Department of Oncology, The 960 Hospital of the People's Liberation Army, Jinan, China
| | - Xinyue Han
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yingcui Chen
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Hong Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Yuekai Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
| | - Baocheng Wang
- Department of Oncology, The 960 Hospital of the People's Liberation Army, Jinan, China
| | - Xin Ye
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China.
- Shandong Lung Cancer Institute, Jinan, China.
- Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China.
- Shandong Lung Cancer Institute, Jinan, China.
| |
Collapse
|
4
|
Yu L, Li Y, Li C, Qi X, Lin Y, Li Y, Chen H, Lin L. Immunochemotherapy for small cell lung cancer with paraneoplastic Cushing syndrome: A case report and literature review. Medicine (Baltimore) 2024; 103:e41036. [PMID: 39705446 PMCID: PMC11666165 DOI: 10.1097/md.0000000000041036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 12/22/2024] Open
Abstract
RATIONALE Paraneoplastic Cushing syndrome (PCS) is an adverse prognostic factor for small cell lung cancer (SCLC) patients. Retrospective studies have shown that the median survival of SCLC complicated with PCS was <7 months. No immunochemotherapy has been recorded in the treatment of SCLC with PCS. Previous preclinical and clinical studies have suggested glucocorticoid exposure may affect the efficacy of immunotherapy. PATIENT CONCERNS AND DIAGNOSIS A 60-year-old man was admitted for his irritability and palpitation. During hospitalization, a chest computed tomography scan revealed a lobar soft tissue shadow in his left lower lung. He was diagnosed as limited-stage SCLC (T2bN1M0 IIB) with PCS, ultimately. INTERVENTIONS AND OUTCOME The patient received 4 courses of immunochemotherapy of etoposide plus platinum with durvalumab and 1 adjuvant radiotherapy alone in 2022 for his limited-stage SCLC, and underwent 5 courses of immunochemotherapy of irinotecan plus platinum with serplulimab in 2023 for his extensive-stage SCLC. The patient achieved a long survival of 20 months. LESSONS The case preliminarily demonstrated the efficacy of immunochemotherapy in the management of SCLC complicated with PCS. The regime of serplulimab with irinotecan-based chemotherapy also indicated its satisfactory efficacy as a second-line treatment for extensive-stage SCLC. Furthermore, the case has highlighted that the management of hypercortisolism, the improvement of myelosuppression, and the prophylaxis against infection were 3 hinges for the continuation of immunochemotherapy and the holistic management of SCLC with PCS.
Collapse
Affiliation(s)
- Ling Yu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanlong Li
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Caiyu Li
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiangjun Qi
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yeding Lin
- The First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanliang Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanrui Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lizhu Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Tong X, Jin M, Wang L, Zhang D, Yin Y, Shen Q. Prognostic biomarkers for immunotherapy in esophageal cancer. Front Immunol 2024; 15:1420399. [PMID: 39403382 PMCID: PMC11471503 DOI: 10.3389/fimmu.2024.1420399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 09/05/2024] [Indexed: 03/22/2025] Open
Abstract
Esophageal cancer (EC), a common type of malignant tumor, ranks as the sixth highest contributor to cancer-related mortality worldwide. Due to the condition that most patients with EC are diagnosed at advanced or metastatic status, the efficacy of conventional treatments including surgery, chemotherapy and radiotherapy is limited, resulting in a dismal 5-year overall survival rate. In recent years, the application of immune checkpoint inhibitors (ICIs) has presented a novel therapeutic avenue for EC patients. Both ICIs monotherapy and immunotherapy combined with chemotherapy or chemoradiotherapy (CRT) have demonstrated marked benefits for patients with advanced EC. Adjuvant or neoadjuvant therapy incorporating immunotherapy has also demonstrated promising prospects in the context of perioperative treatment. Nonetheless, due to the variable response observed among patients undergoing immunotherapy, it is of vital importance to identify predictive biomarkers for patient stratification, to facilitate identification of subgroups who may derive greater benefits from immunotherapy. In this review, we summarize validated or potential biomarkers for immunotherapy in EC in three dimensions: tumor-cell-associated biomarkers, tumor-immune microenvironment (TIME)-associated factors, and host-associated biomarkers, so as to provide a theoretical foundation to inform tailored therapy for individuals diagnosed with EC.
Collapse
Affiliation(s)
- Xu Tong
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meiyuan Jin
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lulu Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Dongli Zhang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yuping Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Shen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Zhang Y, Zhang C, Chen G, You H, Wang S, Wang X, Zhao P, Xu B, Gao Q, Yuan L. Subclone from CT26 resistant to anti-PD-1 therapy associated with increased expression of genes related to glucocorticoids. Transl Oncol 2024; 46:102031. [PMID: 38861853 PMCID: PMC11209639 DOI: 10.1016/j.tranon.2024.102031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Although the use of anti-PD-1 antibodies has fundamentally changed traditional cancer treatment, most patients are resistant to anti-PD-1 treatment. Glucocorticoids (GCs) play an important role in tumorigenesis and tumor progression, but the role of endogenous GCs in resistance to anti-PD-1 antibody therapy remains unclear. METHODS Single cell-derived cell lines (SCDCLs) were generated from a colorectal cancer cell line (CT26) using limiting dilution. We analyzed tumor tissues from anti-PD-1 antibody-treated and untreated mice inoculated with SCDCLs via transcriptome sequencing and flow cytometry to detect pathway activity and immune cell composition changes in the tumor microenvironment. RESULTS Five SCDCLs were inoculated into wild-type BALB/c mice (all tumorigenic). Single-cell clone (SCC)-2 exhibited the slowest growth rates both in vivo and in vitro compared to other single-cell clones, and better long-term survival than SCC1 and CT26. Flow cytometry showed that SCC2 tumor-bearing mice exhibited significantly higher infiltration of T cells within the tumor tissue, and higher expression of PD-1 on these T cells than the other groups in vivo. However, the SCC2 group showed no response to anti-PD-1 therapy. Transcriptome analysis revealed that the SCC2 group exhibited increased expression of genes related to GC (Hsd11b1, Sgk3, Tgfbr2, and Il7r) compared to SCC2-anti-PD-1 treated tumors. CONCLUSIONS GC pathway activation is related to resistance to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Yangyang Zhang
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China
| | - Chaoji Zhang
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Guangyu Chen
- Department of immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China
| | - Hongqin You
- Department of immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China
| | - Sen Wang
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China
| | - Xiaoming Wang
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China
| | - Peng Zhao
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China
| | - Benling Xu
- Department of immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China.
| | - Quanli Gao
- Department of immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China.
| | - Long Yuan
- Department of Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 127 Dongming Road, Jinshui District, Zhengzhou, Henan Province, China.
| |
Collapse
|
7
|
Liang X, Guan Y, Wang B, Liu X, Wang J. Histological sarcomatoid transformation in a lung adenocarcinoma patient following immune checkpoint blockade. Ther Adv Med Oncol 2024; 16:17588359241236450. [PMID: 38455710 PMCID: PMC10919128 DOI: 10.1177/17588359241236450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024] Open
Abstract
Histological transformation is a phenomenon that is well described as one of the causes of tyrosine kinase inhibitor resistance in oncogene-driven non-small-cell lung cancer (NSCLC). The use of immune checkpoint inhibitors (ICIs) as a potential mechanism of acquired resistance to immunotherapy in NSCLC to small-cell lung cancer was also recently found. Here, we report the histological transformation of sarcomatoid carcinoma and metastasis in a lung adenocarcinoma patient without targetable genetic alterations who experienced long-term disease remission after nivolumab therapy. The patient subsequently developed rapid progression in the mediastinal and retroperitoneal lymph nodes, bones, and small intestine. Surgical resection of the small intestine lesion due to acute small intestine bleeding revealed the transformation of NSCLC to sarcomatoid carcinoma. The patient died 3 months after sarcomatoid carcinoma transformation and extensive disease progression, although he was rechallenged with immunotherapy. Genomic and immunohistochemical analyses revealed a comparable abundance of gene mutations and a limited number of immune cells in the tumor microenvironment, with low infiltration of CD8+ T cells, CD4+ T cells, regulatory T cells, and PD-L1+ macrophages in metastatic tumors, revealing a noninflamed immune microenvironment for ICI-resistant tumors.
Collapse
Affiliation(s)
- Xiuju Liang
- Department of Oncology, 960th Hospital of the People’s Liberation Army, Jinan, China
| | - Yaping Guan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Baocheng Wang
- Department of Oncology, 960th Hospital of the People’s Liberation Army, Jinan, China
| | - Xiaohong Liu
- Department of Pathology, 960th Hospital of the People’s Liberation Army, No. 25, Shifan Road, Jinan 250031, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University, Shandong Provincial Qianfoshan Hospital, No. 16766, Jingshi Road, Jinan 250014, China
- Shandong Lung Cancer Institute, Jinan, China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| |
Collapse
|
8
|
Kochanek C, Gilde C, Zimmer L, Ugurel S, Meier F, Utikal J, Pföhler C, Herbst R, Haferkamp S, Welzel J, Dücker P, Leiter U, Weichenthal M, von Wasielewski I, Angela Y, Gutzmer R. Effects of an immunosuppressive therapy on the efficacy of immune checkpoint inhibition in metastatic melanoma - An analysis of the prospective skin cancer registry ADOREG. Eur J Cancer 2024; 198:113508. [PMID: 38183763 DOI: 10.1016/j.ejca.2023.113508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/19/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND The impact of immunosuppressive therapy (IST) on immune-checkpoint inhibition (ICI) is unclear. METHODS Patients with unresectable advanced melanoma (MM) treated with ICI in the years 2011-2020 were identified from the prospective multicenter German skin cancer registry ADOREG. Patients with IST within 60 days before, or within 30 days after start of ICI were compared to patients without IST. End points were disease control rate (DCR), overall survival (OS) and progression-free survival (PFS) determined by Kaplan-Meier method. Prognostic factors were evaluated in a Cox regression model. RESULTS Of 814 patients treated with ICI, 73 (9%) received concomitant IST, mainly steroids. Patients with brain metastases (BM) received IST more frequently (n = 34/130 patients; 26%), than patients without BM (39/684 patients; 6%). In patients without BM, IST initiated before, but not IST initiated after start of ICI was significantly associated with worse PFS (univariate hazard ratio (HR) 2.59, 95% confidence interval (95%-CI) 1.07-6.28, p = 0.035; multivariate HR 3.48, 95%-CI 1.26-9.6, p = 0.016). There was no association between IST and OS or DCR. In patients with BM, IST initiated before, but not after start of ICI was significantly associated with worse OS (univariate HR 2.06, 95%-CI 1.07-3.95, p = 0.031; multivariate HR 5.91, 95%-CI 1.74-20.14, p = 0.004). There was no association between IST and PFS or DCR. CONCLUSION Patients receiving IST 60 days before start of ICI showed a tendency to an impaired therapy outcome. IST initiated within 30 days after start of ICI, mainly due to early side effects, did not affect the efficacy of ICI therapy.
Collapse
Affiliation(s)
- Corinna Kochanek
- Department of Dermatology, Allergology and Venerology, Skin Cancer Centre, Hannover Medical School, Hannover, Germany.
| | - Catharina Gilde
- Department of Dermatology, Allergology and Venerology, Skin Cancer Centre, Hannover Medical School, Hannover, Germany
| | - Lisa Zimmer
- Department of Dermatology, Venerology and Allergology, University Hospital Essen & Westdeutsches Tumorzentrum, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venerology and Allergology, University Hospital Essen & Westdeutsches Tumorzentrum, Essen, Germany; German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, Dresden, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venerology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector CancerInstitute at the University Medical Center Mannheim, Mannheim, Germany
| | - Claudia Pföhler
- Department of Dermatology, Venerology and Allergology, Skin Cancer Centre, Saarland University Medical School, Homburg, Saar, Germany
| | - Rudolf Herbst
- Department of Dermatology and Allergology, Skin Cancer Centre, Helios Clinic Erfurt, Erfurt, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, Skin Cancer Centre, University Hospital Regensburg, Regensburg, Germany
| | - Julia Welzel
- Department of Dermatology and Allergology, Skin Cancer Centre, Augsburg University Hospital, Augsburg, Germany
| | - Pia Dücker
- Department of Dermatology, Skin Cancer Centre, Hospital Dortmund, Dortmund, Germany
| | - Ulrike Leiter
- Centre of Dermatooncology, Department of Dermatology, Venerology and Allergology, University of Tuebingen, Germany
| | - Michael Weichenthal
- Department of Dermatology, Venerology and Allergology, Skin Cancer Centre, University Hospital of Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Imke von Wasielewski
- Department of Dermatology, Allergology and Venerology, Skin Cancer Centre, Hannover Medical School, Hannover, Germany
| | - Yenny Angela
- Department of Dermatology, Johannes Wesling Medical Center Minden, Ruhr University Bochum Campus Minden, Minden, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center Minden, Ruhr University Bochum Campus Minden, Minden, Germany
| |
Collapse
|
9
|
Gruden E, Kienzl M, Ristic D, Kindler O, Kaspret DM, Schmid ST, Kargl J, Sturm E, Doyle AD, Wright BL, Baumann-Durchschein F, Konrad J, Blesl A, Schlager H, Schicho R. Mononuclear cell composition and activation in blood and mucosal tissue of eosinophilic esophagitis. Front Immunol 2024; 15:1347259. [PMID: 38318168 PMCID: PMC10839056 DOI: 10.3389/fimmu.2024.1347259] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/09/2024] [Indexed: 02/07/2024] Open
Abstract
Introduction Eosinophilic esophagitis (EoE) is a chronic, inflammatory, antigen-driven disease of the esophagus. Tissue EoE pathology has previously been extensively characterized by novel transcriptomics and proteomic platforms, however the majority of surface marker determination and screening has been performed in blood due to mucosal tissue size limitations. While eosinophils, CD4+ T cells, mast cells and natural killer (NK) T cells were previously investigated in the context of EoE, an accurate picture of the composition of peripheral blood mononuclear cells (PBMC) and their activation is missing. Methods In this study, we aimed to comprehensively analyze the composition of peripheral blood mononuclear cells and their activation using surface marker measurements with multicolor flow cytometry simultaneously in both blood and mucosal tissue of patients with active EoE, inactive EoE, patients with gastroesophageal reflux disease (GERD) and controls. Moreover, we set out to validate our data in co-cultures of PBMC with human primary esophageal epithelial cells and in a novel inducible mouse model of eosinophilic esophagitis, characterized by extensive IL-33 secretion in the esophagus. Results Our results indicate that specific PBMC populations are enriched, and that they alter their surface expression of activation markers in mucosal tissue of active EoE. In particular, we observed upregulation of the immunomodulatory molecule CD38 on CD4+ T cells and on myeloid cells in biopsies of active EoE. Moreover, we observed significant upregulation of PD-1 on CD4+ and myeloid cells, which was even more prominent after corticosteroid treatment. With co-culture experiments we could demonstrate that direct cell contact is needed for PD-1 upregulation on CD4+ T cells. Finally, we validated our findings of PD-1 and CD38 upregulation in an inducible mouse model of EoE. Discussion Herein we show significant alterations in the PBMC activation profile of patients with active EoE in comparison to inactive EoE, GERD and controls, which could have potential implications for treatment. To our knowledge, this study is the first of its kind expanding the multi-color flow cytometry approach in different patient groups using in vitro and in vivo translational models.
Collapse
Affiliation(s)
- Eva Gruden
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Melanie Kienzl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Dusica Ristic
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Oliver Kindler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - David Markus Kaspret
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Sophie Theresa Schmid
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Eva Sturm
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Alfred D. Doyle
- Division of Allergy, Asthma, and Clinical Immunology, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Benjamin L. Wright
- Division of Allergy, Asthma, and Clinical Immunology, Mayo Clinic Arizona, Scottsdale, AZ, United States
| | - Franziska Baumann-Durchschein
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Julia Konrad
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Blesl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Hansjörg Schlager
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Rudolf Schicho
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
10
|
Lei Y, Liao F, Tian Y, Wang Y, Xia F, Wang J. Investigating the crosstalk between chronic stress and immune cells: implications for enhanced cancer therapy. Front Neurosci 2023; 17:1321176. [PMID: 38089966 PMCID: PMC10713832 DOI: 10.3389/fnins.2023.1321176] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/13/2023] [Indexed: 06/04/2025] Open
Abstract
Chronic stress has a substantial influence on the tumor microenvironment (TME), leading to compromised effectiveness of anti-cancer therapies through diverse mechanisms. It disrupts vital functions of immune cells that play a critical role in anti-tumor immunity, such as the inhibition of dendritic cells (DCs) and lymphocytes, while simultaneously enhancing the activity of immune cells that support tumor growth, such as myeloid-derived suppressor cells and tumor-associated macrophages. Furthermore, chronic stress exerts a significant impact on crucial mechanisms within the TME, including angiogenesis, DNA repair, hypoxia, extracellular matrix deposition, and tumor metabolism. These alterations in the TME, induced by stress, result from the activation of the hypothalamic-pituitary-adrenal axis and sympathetic nervous system, in conjunction with epigenetic modifications. In conclusion, chronic stress significantly influences the TME and impedes the efficacy of anti-cancer treatments, underscoring the importance of targeting stress pathways to improve therapeutic results.
Collapse
Affiliation(s)
- YongRong Lei
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), College of Bioengineering, Chongqing University, Chongqing, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fenghui Liao
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), College of Bioengineering, Chongqing University, Chongqing, China
| | - YiChen Tian
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), College of Bioengineering, Chongqing University, Chongqing, China
| | - YaNi Wang
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), College of Bioengineering, Chongqing University, Chongqing, China
| | - Feng Xia
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - JianHua Wang
- Key Laboratory of Biorheological Science and Technology (Ministry of Education), College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
11
|
Papavassiliou KA, Anagnostopoulos N, Papavassiliou AG. Glucocorticoid Receptor Signaling in NSCLC: Mechanistic Aspects and Therapeutic Perspectives. Biomolecules 2023; 13:1286. [PMID: 37759686 PMCID: PMC10526876 DOI: 10.3390/biom13091286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Recent advances in non-small cell lung cancer (NSCLC) biology and the discovery of novel therapeutic targets have led to the development of new pharmacological agents that may improve the clinical outcome of patients with NSCLC. The glucocorticoid receptor (GR) is an evolutionarily conserved protein belonging to the nuclear receptor superfamily of transcription factors and mediates the diverse actions of glucocorticoids in cells. Data suggest that the GR may play a relevant role in the molecular mechanisms of NSCLC tumorigenesis and malignant progression. Additionally, evidence indicates that glucocorticoids may affect the efficacy of standard treatment, including chemotherapy, immune checkpoint inhibitors, and targeted therapy. Furthermore, several findings show that GR expression may probably be associated with NSCLC patient survival. Finally, glucocorticoids may be used as therapeutic agents for the clinical management of NSCLC patients. Here, we briefly review the latest advances on the biological role of GR signaling in NSCLC and discuss the potential use of the GR as a prognostic and predictive biomarker. Importantly, we explore the therapeutic potential of glucocorticoids and the effect of adding such drugs to standard therapies for NSCLC.
Collapse
Affiliation(s)
- Kostas A. Papavassiliou
- First Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (N.A.)
| | - Nektarios Anagnostopoulos
- First Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (K.A.P.); (N.A.)
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|