1
|
Chen X, Wu L, Lan G, Li X, Wang X, Zhang P, Huang W. Construction and validation of a risk prediction model for postoperative lung infection in elderly patients with lung cancer. Medicine (Baltimore) 2024; 103:e40337. [PMID: 39495987 PMCID: PMC11537623 DOI: 10.1097/md.0000000000040337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
This study aimed to analyze the risk factors for postoperative lung infection in elderly patients with lung cancer (LC) and construct a predictive model. A retrospective analysis was conducted on 192 elderly patients with LC who underwent surgical treatment in our hospital between February 2020 and May 2023. According to whether there is lung infection after surgery, they were divided into an infected group (n = 55) and a noninfected group (n = 137). Binary logistic regression was used to analyze factors influencing postoperative lung infection in elderly patients with LC. Based on the logistic regression results, a predictive model for postoperative lung infection in LC patients was constructed. The receiver operating characteristic curve was used to analyze C-reactive protein (CRP), interleukin-6 (IL-6), insulin-like growth factor-1 (IGF-1), and their combination in predicting postoperative lung infection in patients with LC. There were significant differences between the infected group and the noninfected group in age, smoking history, diabetes, and perioperative antibiotic use were significantly different between the infected and noninfected groups (P < .05). The postoperative CRP, IL-6, and IGF-1 levels in the infected group were higher than those in the noninfected group on the 1st day (P < .05). Logistic regression analysis showed that age > 70 years, history of smoking, history of diabetes, prolonged use of perioperative antibiotics, and elevated CRP, IL-6, and IGF-1 levels on the 1st day after surgery were risk factors for postoperative lung infection in elderly patients with LC (P < .05). Receiver operating characteristic curve analysis showed that the area under curve values of CRP, IL-6, IGF-1, and their combination in predicting postoperative lung infection in elderly patients with LC were 0.701, 0.806, 0.737, and 0.871, P < .05), with sensitivity values of 0.443, 0.987, 0.456, and 0.835, respectively; the specificity was 0.978, 0.525, 0.991, and 0.821, respectively. Age > 70 years, smoking history, diabetes history, prolonged use of perioperative antibiotics, and elevated CRP, IL-6, and IGF-1 levels on the 1st day after surgery have an impact on postoperative lung infection in elderly patients with LC. Early postoperative monitoring of changes in CRP, IL-6, and IGF-1 levels can provide an important reference for predicting the occurrence of postoperative lung infections.
Collapse
Affiliation(s)
- Xiaojie Chen
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Lixin Wu
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Gang Lan
- Department of Chest Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Xiaofeng Li
- Department of Chest Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Xuejing Wang
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Ping Zhang
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Weihu Huang
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| |
Collapse
|
2
|
Garaci E, Paci M, Matteucci C, Costantini C, Puccetti P, Romani L. Phenotypic drug discovery: a case for thymosin alpha-1. Front Med (Lausanne) 2024; 11:1388959. [PMID: 38903817 PMCID: PMC11187271 DOI: 10.3389/fmed.2024.1388959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
Phenotypic drug discovery (PDD) involves screening compounds for their effects on cells, tissues, or whole organisms without necessarily understanding the underlying molecular targets. PDD differs from target-based strategies as it does not require knowledge of a specific drug target or its role in the disease. This approach can lead to the discovery of drugs with unexpected therapeutic effects or applications and allows for the identification of drugs based on their functional effects, rather than through a predefined target-based approach. Ultimately, disease definitions are mostly symptom-based rather than mechanism-based, and the therapeutics should be likewise. In recent years, there has been a renewed interest in PDD due to its potential to address the complexity of human diseases, including the holistic picture of multiple metabolites engaging with multiple targets constituting the central hub of the metabolic host-microbe interactions. Although PDD presents challenges such as hit validation and target deconvolution, significant achievements have been reached in the era of big data. This article explores the experiences of researchers testing the effect of a thymic peptide hormone, thymosin alpha-1, in preclinical and clinical settings and discuss how its therapeutic utility in the precision medicine era can be accommodated within the PDD framework.
Collapse
Affiliation(s)
| | - Maurizio Paci
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- San Raffaele Sulmona, L’Aquila, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
3
|
Deng J, Gong C, Xiao Q, Xu B, Li H, Wu Z, Xiao Q, Gu P, Li Q, Li B, Wang Y, Lin B, Xu K. Efficacy of Shenglin decoction in preventing acute severe lymphocytopenia in patients with non-small cell lung cancer undergoing concurrent chemoradiotherapy: a study protocol for a randomized controlled trial. Front Oncol 2024; 14:1378662. [PMID: 38779093 PMCID: PMC11109395 DOI: 10.3389/fonc.2024.1378662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Background Definitive concurrent chemoradiotherapy (CCRT) followed by maintenance therapy with immune checkpoint inhibitors offers the best chance of cure for patients with stage III non-small cell lung cancer (NSCLC). A significant challenge in this regimen is the occurrence of acute severe lymphopenia (ASL), which can compromise treatment efficacy. Currently, there are no effective strategies for preventing and treating ASL. Shenglin decoction (SLD), a traditional Chinese herbal medicine formulation, has demonstrated preliminary efficacy in mitigating ASL. However, robust evidence from clinical trials and a clear understanding of its mechanism of action are still needed. This study aims to comprehensively assess the efficacy, safety, and underlying mechanisms of SLD in the prevention of ASL. Methods This prospective, dual-center, open-label, randomized controlled trial will enroll 140 stage III NSCLC patients. Participants will be randomly allocated in a 1:1 ratio to a control group or an experimental group. Both groups will undergo definitive CCRT. Alongside the commencement of CCRT, the experimental group will receive an additional oral SLD intervention for a duration of three months. The primary outcome is the incidence rate of ASL, defined as the proportion of patients who experience at least one instance of a total lymphocyte count falling below 0.5 × 10^9 cells/L within 3 months of initiating CCRT treatment. Additionally, 16S rRNA gene sequencing analysis of fecal samples to assess gut microbiota, as well as metabolomic analysis of fecal/blood samples, will be conducted to explore potential mechanisms. Discussion This study protocol aims to rigorously evaluate the efficacy and safety of SLD, as well as elucidate its mechanism of action in preventing ASL. Successful outcomes could establish SLD as an evidence-based intervention for ASL prevention in NSCLC patients undergoing CCRT. Trial Registration The trial was registered at the Chinese Clinical Trials Registry (ChiCTR2300071788, https://www.chictr.org.cn/).
Collapse
Affiliation(s)
- Jiayao Deng
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cuicui Gong
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi Xiao
- Health Management Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bo Xu
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huakang Li
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ziliang Wu
- Health Management Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Xiao
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pengxuan Gu
- Clinical School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Li
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Bing Lin
- Health Management Center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Xu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
4
|
Sha L, Zhang H, Zhang X. Thymosin α1 combined with XELOX improves immune function and reduces serum tumor markers in colorectal cancer patients after radical surgery. Open Life Sci 2024; 19:20220793. [PMID: 38623586 PMCID: PMC11017181 DOI: 10.1515/biol-2022-0793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 04/17/2024] Open
Abstract
This study aimed to investigate the efficacy of thymosin α1 combined with XELOX in improving immune function and reducing serum tumor markers in patients with colorectal cancer (CRC) after radical surgery. A total of 180 patients who underwent radical surgery for CRC were divided into two groups: an observation group (n = 94) receiving thymosin α1 in combination with XELOX and a control group (n = 86) receiving XELOX alone. Immune function, inflammatory factor levels, serum tumor markers, and quality of life were assessed before and after treatment. Adverse reactions and recurrence rates were compared between the two groups in 1 and 3 years. Following therapy, there was a notable increase in the levels of CD3+, CD4+, and CD4+/CD8+ in all cohorts, particularly in the observation cohort, when compared to pre-therapy levels. Conversely, CD8+ levels decreased across all cohorts, especially in the observation cohort. Additionally, there was an increase in the levels of IL-2 and IFN-γ in the observation cohort, compared to both pre-therapy and control cohort levels, while IL-6 levels decreased. The presence of CEA, CA242, and CA724 reduced significantly across all cohorts following post-therapy, particularly in the observation cohort. Post-therapy, there was a significant increase in the scoring for role, cognitive, social, emotional, and somatic functions in all cohorts, with the most significant improvement observed in the observation cohort. There were no significant differences in the incidence of side effects across cohorts, while neutropenia events were significantly lower in the observation cohort (32.98%) compared to the control cohort (48.84%). The 12-month recurrence rate showed no statistical significance across cohorts, while the observation cohort had a significantly lower three-year recurrence rate (24.47%) compared to the control cohort (59.30%). Thymosin α1 combined with XELOX is effective in improving immune function, reducing serum tumor markers, and minimizing recurrence in CRC patients after radical surgery. This combination therapy may be a promising new direction for the treatment of CRC.
Collapse
Affiliation(s)
- Li Sha
- Department of General Surgery, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai200025, China
| | - Hao Zhang
- Department of General Surgery, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai200025, China
| | - Xiwei Zhang
- Department of General Surgery, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai200025, China
| |
Collapse
|
5
|
Kong Y, Chen R, Xu M, Zhang J, Chen G, Hong Z, Zhang H, Dai X, Ma Y, Zhao X, Peng Y, Zhang C, Xing P, Zhang L. Evaluation of the efficacy and safety of a precise thymalfasin-regulated PRaG regimen for advanced refractory solid tumours: protocol for the open-label, prospective, multicentre study (PRaG5.0 study). BMJ Open 2024; 14:e075642. [PMID: 38458816 DOI: 10.1136/bmjopen-2023-075642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION The PRaG regimen, which consists of hypofractionated radiotherapy combined with a programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitor and granulocyte-macrophage colony stimulating factor (GM-CSF), has been demonstrated to have a survival benefit in patients with advanced solid tumours who have failed at least two lines of treatment. Nonetheless, lymphopenia poses an impediment to the enduring efficacy of PD-1/PD-L1 inhibitor therapy. Adequate lymphocyte reserves are essential for the efficacy of immunotherapy. Coupling the PRaG regimen with immunomodulatory agents that augment the number and functionality of lymphocytes may yield further survival benefits in this cohort of patients. OBJECTIVE The aim of this study is to investigate the effectiveness and safety of a meticulously thymalfasin-controlled PRaG regimen in patients with advanced and chemotherapy-resistant solid tumours. METHODS AND ANALYSIS The study has a prospective, single-arm, open-label, multicentre design and aims to recruit up to 60 patients with histologically confirmed advanced solid tumours that have relapsed or metastasised. All eligible patients will receive a minimum of two cycles of the PRaG regimen comprising thymalfasin followed by maintenance treatment with a PD-1/PD-L1 inhibitor and thymalfasin for 1 year or until disease progression. Patients will be monitored according to the predetermined protocol for a year or until disease progression after initiation of radiotherapy. ETHICS AND DISSEMINATION The study protocol was approved by the Ethics Committee of the Second Affiliated Hospital of Soochow University, on 25 November 2022 (JD-LK-2022-151-01) and all other participating hospitals. Findings will be disseminated through national and international conferences. We also plan to publish our findings in high-impact peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT05790447.
Collapse
Affiliation(s)
- Yuehong Kong
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Rongzheng Chen
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Meiling Xu
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Junjun Zhang
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Guangqiang Chen
- Department of Radiology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhihui Hong
- Department of Nuclear Medicine, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoxiao Dai
- Department of Pathology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yifu Ma
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Xiangrong Zhao
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Yong Peng
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Chenyang Zhang
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Pengfei Xing
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Liyuan Zhang
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
6
|
Nie T, Chen Z, Cai J, Ai S, Xue X, Yuan M, Li C, Shi L, Liu Y, Verma V, Bi J, Han G, Yuan Z. Integration of dosimetric parameters, clinical factors, and radiomics to predict symptomatic radiation pneumonitis in lung cancer patients undergoing combined immunotherapy and radiotherapy. Radiother Oncol 2024; 190:110047. [PMID: 38070685 DOI: 10.1016/j.radonc.2023.110047] [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: 05/10/2023] [Revised: 11/27/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023]
Abstract
PURPOSE This study aimed to combine clinical/dosimetric factors and handcrafted/deep learning radiomic features to establish a predictive model for symptomatic (grade ≥ 2) radiation pneumonitis (RP) in lung cancer patients who received immunotherapy followed by radiotherapy. MATERIALS AND METHODS This study retrospectively collected data of 73 lung cancer patients with prior receipt of ICIs who underwent thoracic radiotherapy (TRT). Of these 73 patients, 41 (56.2 %) developed symptomatic grade ≥ 2 RP. RP was defined per multidisciplinary clinician consensus using CTCAE v5.0. Regions of interest (ROIs) (from radiotherapy planning CT images) utilized herein were gross tumor volume (GTV), planning tumor volume (PTV), and PTV-GTV. Clinical/dosimetric (mean lung dose and V5-V30) parameters were collected, and 107 handcrafted radiomic (HCR) features were extracted from each ROI. Deep learning-based radiomic (DLR) features were also extracted based on pre-trained 3D residual network models. HCR models, Fusion HCR model, Fusion HCR + ResNet models, and Fusion HCR + ResNet + Clinical models were built and compared using the receiver operating characteristic (ROC) curve with measurement of the area under the curve (AUC). Five-fold cross-validation was performed to avoid model overfitting. RESULTS HCR models across various ROIs and the Fusion HCR model showed good predictive ability with AUCs from 0.740 to 0.808 and 0.740-0.802 in the training and testing cohorts, respectively. The addition of DLR features improved the effectiveness of HCR models (AUCs from 0.826 to 0.898 and 0.821-0.898 in both respective cohorts). The best performing prediction model (HCR + ResNet + Clinical) combined HCR & DLR features with 7 clinical/dosimetric characteristics and achieved an average AUC of 0.936 and 0.946 in both respective cohorts. CONCLUSIONS In patients undergoing combined immunotherapy/RT for lung cancer, integrating clinical/dosimetric factors and handcrafted/deep learning radiomic features can offer a high predictive capacity for RP, and merits further prospective validation.
Collapse
Affiliation(s)
- Tingting Nie
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zien Chen
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; School of Biomedical Engineering, South-Central Minzu University, Wuhan, PR China
| | - Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Nanhuan Road, Jingzhou, Hubei, PR China
| | - Shuangquan Ai
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; School of Biomedical Engineering, South-Central Minzu University, Wuhan, PR China
| | - Xudong Xue
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Mengting Yuan
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Chao Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Nanhuan Road, Jingzhou, Hubei, PR China
| | - Liting Shi
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, Jiangsu, China
| | - Yulin Liu
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Vivek Verma
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - Jianping Bi
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Guang Han
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Zilong Yuan
- Department of Radiology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
7
|
Liu Y, Lu J. Mechanism and clinical application of thymosin in the treatment of lung cancer. Front Immunol 2023; 14:1237978. [PMID: 37701432 PMCID: PMC10493777 DOI: 10.3389/fimmu.2023.1237978] [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: 06/12/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The burden of cancer on public health is becoming more widely acknowledged. Lung cancer has one of the highest incidence and mortality rates of all cancers. The prevalence of early screening, the emergence of targeted therapy, and the development of immunotherapy have all significantly improved the overall prognosis of lung cancer patients. The current state of affairs, however, is not encouraging, and there are issues like poor treatment outcomes for some patients and extremely poor prognoses for those with advanced lung cancer. Because of their potent immunomodulatory capabilities, thymosin drugs are frequently used in the treatment of tumors. The effectiveness of thymosin drugs in the treatment of lung cancer has been demonstrated in numerous studies, which amply demonstrates the potential and future of thymosin drugs for the treatment of lung cancer. The clinical research on thymosin peptide drugs in lung cancer and the basic research on the mechanism of thymosin drugs in anti-lung cancer are both systematically summarized and analyzed in this paper, along with future research directions.
Collapse
Affiliation(s)
| | - Jibin Lu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
8
|
Wang S, Wei W, Yong H, Zhang Z, Zhang X, Zhang X, Wang S. Synergistic anti-cancer and attenuation effects of thymosin on chemotherapeutic drug vinorelbine in tumor-bearing zebrafish. Biomed Pharmacother 2023; 162:114633. [PMID: 37018994 DOI: 10.1016/j.biopha.2023.114633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/18/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Vinorelbine, the standard chemotherapy drug on advanced lung cancer, causes adverse events such as immunosuppression and bone marrow suppression. Thus, it is necessary to find drugs that could improve immune function and synergistically enhance the anti-tumor effect of vinorelbine. Thymosin is reported to inhibit tumor growth as an immunomodulator. Herein, to study the synergistic anti-cancer and attenuation effects of thymosin on vinorelbine, human lung cancer A549 cells that were labeled with CM-DiI were transplanted into zebrafish to establish the lung cancer xenotransplanted model. After treatment of vinorelbine and different concentrations of thymosin, the fluorescence intensity of CM-DiI-labeled A549 cells and the number of apoptotic muscle cells in the tumor-bearing zebrafish were detected. Besides, effects of thymosin on vinorelbine-reduced macrophages and T cells were identified in the transgenic zebrafish (Tg:zlyz-EGFP and Tg:rag2-DsRed). Then, the qRT-PCR was used to determine the alterations of the immune-related factors at the transcription level. Thymosin showed a marked synergistic anti-cancer effect with vinorelbine for the xenograft human lung cancer A549 cells, and the synergistic effect enhanced in a dose-dependent manner. Moreover, thymosin alleviated vinorelbine-induced muscle cell apoptosis, macrophage reduction, and T cell suppression. Compared with the vinorelbine group, co-administration with thymosin raised the mRNA levels of TNF-α, TNF-β, INF-γ, and GM-CSF. Thus, thymosin possesses synergistic anti-cancer effect on vinorelbine, and has protective effect on vinorelbine-induced immunosuppression. Thymosin, as an adjuvant immunomodulatory therapy, has great potential in enhancing the clinical application of vinorelbine.
Collapse
|
9
|
Cole K, Al-Kadhimi Z, Talmadge JE. Highlights into historical and current immune interventions for cancer. Int Immunopharmacol 2023; 117:109882. [PMID: 36848790 PMCID: PMC10355273 DOI: 10.1016/j.intimp.2023.109882] [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: 12/30/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 03/01/2023]
Abstract
Immunotherapy is an additional pillar when combined with traditional standards of care such as chemotherapy, radiotherapy, and surgery for cancer patients. It has revolutionized cancer treatment and rejuvenated the field of tumor immunology. Several types of immunotherapies, including adoptive cellular therapy (ACT) and checkpoint inhibitors (CPIs), can induce durable clinical responses. However, their efficacies vary, and only subsets of cancer patients benefit from their use. In this review, we address three goals: to provide insight into the history of these approaches, broaden our understanding of immune interventions, and discuss current and future approaches. We highlight how cancer immunotherapy has evolved and discuss how personalization of immune intervention may address present limitations. Cancer immunotherapy is considered a recent medical achievement and in 2013 was selected as the "Breakthrough of the Year" by Science. While the breadth of immunotherapeutics has been rapidly expanding, to include the use of chimeric antigen receptor (CAR) T-cell therapy and immune checkpoint inhibitor (ICI) therapy, immunotherapy dates back over 3000 years. The expansive history of immunotherapy, and related observations, have resulted in several approved immune therapeutics beyond the recent emphasis on CAR-T and ICI therapies. In addition to other classical forms of immune intervention, including human papillomavirus (HPV), hepatitis B, and the Mycobacterium bovis Bacillus Calmette-Guérin (BCG) tuberculosis vaccines, immunotherapies have had a broad and durable impact on cancer therapy and prevention. One classic example of immunotherapy was identified in 1976 with the use of intravesical administration of BCG in patients with bladder cancer; resulting in a 70 % eradication rate and is now standard of care. However, a greater impact from the use of immunotherapy is documented by the prevention of HPV infections that are responsible for 98 % of cervical cancer cases. In 2020, the World Health Organization (WHO) estimated that 341,831 women died from cervical cancer [1]. However, administration of a single dose of a bivalent HPV vaccine was shown to be 97.5 % effective in preventing HPV infections. These vaccines not only prevent cervical squamous cell carcinoma and adenocarcinoma, but also oropharyngeal, anal, vulvar, vaginal, and penile squamous cell carcinomas. The breadth, response and durability of these vaccines can be contrasted with CAR-T-cell therapies, which have significant barriers to their widespread use including logistics, manufacturing limitations, toxicity concerns, financial burden and lasting remissions observed in only 30 to 40 % of responding patients. Another, recent immunotherapy focus are ICIs. ICIs are a class of antibodies that can increase the immune responses against cancer cells in patients. However, ICIs are only effective against tumors with a high mutational burden and are associated with a broad spectrum of toxicities requiring interruption of administration and/or administration corticosteroids; both of which limit immune therapy. In summary, immune therapeutics have a broad impact worldwide, utilizing numerous mechanisms of action and when considered in their totality are more effective against a broader range of tumors than initially considered. These new cancer interventions have tremendous potential notability when multiple mechanisms of immune intervention are combined as well as with standard of care modalities.
Collapse
Affiliation(s)
- Kathryn Cole
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zaid Al-Kadhimi
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5950, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
10
|
Mao L. Thymosin alpha 1 - Reimagine its broader applications in the immuno-oncology era. Int Immunopharmacol 2023; 117:109952. [PMID: 36871535 DOI: 10.1016/j.intimp.2023.109952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023]
Abstract
Thymosin alpha 1 (Tα1) is a highly conserved 28 amino-acid peptide naturally occurring in the thymus and plays critical roles in T cell maturity and differentiation. Its synthetic form, thymalfasin, has been approved by various regulatory agencies in the treatment of hepatitis B viral infection and as an enhancer of vaccine response in immune-compromised populations. In China, it has also widely utilized in patients with cancer and severe infections, as well as the emergency use during (Severe Acute Respiratory Syndrome)SARS and COVID-19 pandemic as an immune-regulator. Recent studies showed that Tα1 could significantly improve overall survival (OS) in patients with surgically resectable non-small cell lung cancer (NSCLC) and liver cancers in the adjuvant setting. For patients with locally advanced, unresectable NSCLC, Tα1 could significantly reduce chemoradiation-induced lymphopenia, pneumonia, and trending improvement of OS. Preclinical evidence are emerging to demonstrate that Tα1 may augment efficacy of cancer chemotherapy by reversing efferocytosis-induced M2 polarization of macrophages via activation of a TLR7/SHIP1 axis and enhancing anti-tumor immunity by turning "cold-tumors" to "hot-tumors"; a protective role in reducing colitis caused by immune check-point inhibitors (ICIs). Potential enhancement of ICIs' clinical efficacies has also been indicated. ICIs have transformed ways treating patients with cancer but limitations such as relatively low response rates and certain safety issues remains. Given the roles of Tα1 in regulating cellular immunities and exceptional safety profiles demonstrated in decades clinical uses, we believe that it is plausible to explore implications of Tα1 the immune-oncology setting by combining with ICI-based therapeutic strategies. Background Activities of Tα1. Tα1 is a biological response modifier which activates various cells in the immune system [1-3]. Tα1 is therefore expected to have clinical benefits in disorders where immune responses are impaired or ineffective. These disorders include acute and chronic infections, cancers, and vaccine non-responsiveness. In severe sepsis, for example, sepsis-induced immunosuppression is increasingly recognized as the overriding immune dysfunction in these vulnerable patients [4] and there is now agreement that many patients with severe sepsis survive the first critical hours of the syndrome but eventually die later due to patients' immunosuppression which make the system difficulty to fight the primary bacterial infection, decreased resistance to secondary nosocomial infections, and reactivation of viral infections [5]. Tα1 has been shown to restore immune functions and help to reduce mortality in patients with severe sepsis.
Collapse
Affiliation(s)
- Li Mao
- SciClone Pharmaceuticals, 381 Central Huaihai Road, Shanghai 311100, China.
| |
Collapse
|
11
|
Yamamoto T, Katsuta Y, Sato K, Tsukita Y, Umezawa R, Takahashi N, Suzuki Y, Takeda K, Kishida K, Omata S, Miyauchi E, Saito R, Kadoya N, Jingu K. Longitudinal analyses and predictive factors of radiation-induced lung toxicity-related parameters after stereotactic radiotherapy for lung cancer. PLoS One 2022; 17:e0278707. [PMID: 36459528 PMCID: PMC9718403 DOI: 10.1371/journal.pone.0278707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND PURPOSE The purpose of this prospective study was to investigate changes in longitudinal parameters after stereotactic radiotherapy for lung cancer and to identify possible pretreatment factors related to radiation-induced lung toxicity and the decline in pulmonary function after radiotherapy. MATERIALS AND METHODS Protocol-specified examinations, including 4-D CT, laboratory tests, pulmonary function tests (PFTs) and body composition measurements, were performed before SRT and at 1 month, 4 months and 12 months after stereotactic radiotherapy. Longitudinal differences were tested by using repeated-measures analysis of variance. Correlations were examined by using the Pearson product-moment correlation coefficient (r). RESULTS Sixteen patients were analyzed in this study. During a median follow-up period of 26.6 months, grade 1 and 2 lung toxicity occurred in 11 patients and 1 patient, respectively. The mean Hounsfield units (HU) and standard deviation (SD) of the whole lung, as well as sialylated carbohydrate antigen KL-6 (KL-6) and surfactant protein-D (SP-D), peaked at 4 months after radiotherapy (p = 0.11, p<0.01, p = 0.04 and p<0.01, respectively). At 4 months, lung V20 Gy (%) and V40 Gy (%) were correlated with changes in SP-D, whereas changes in the mean HU of the lung were related to body mass index and lean body mass index (r = 0.54, p = 0.02; r = 0.57, p = 0.01; r = 0.69, p<0.01; and r = 0.69, p<0.01, respectively). The parameters of PFTs gradually declined over time. When regarding the change in PFTs from pretreatment to 12 months, lung V5 Gy (cc) showed significant correlations with diffusion capacity for carbon monoxide (DLCO), DLCO/alveolar volume and the relative change in DLCO (r = -0.72, p<0.01; r = -0.73, p<0.01; and r = -0.63, p = 0.01, respectively). CONCLUSIONS The results indicated that some parameters peaked at 4 months, but PFTs were the lowest at 12 months. Significant correlations between lung V5 Gy (cc) and changes in DLCO and DLCO/alveolar volume were observed.
Collapse
Affiliation(s)
- Takaya Yamamoto
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| | - Yoshiyuki Katsuta
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kiyokazu Sato
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Rei Umezawa
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriyoshi Takahashi
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Suzuki
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazuya Takeda
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keita Kishida
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - So Omata
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryota Saito
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriyuki Kadoya
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keiichi Jingu
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|