1
|
Mahendra I, Kurniawan A, Febrian MB, Halimah I, Rizaludin A, Syarif DG. Cell-Derived Allograft Models as a Solution to the Obstacles of Preclinical Studies under Limited Resources: A Systematic Review on Experimental Lung Cancer Animal Models. Curr Rev Clin Exp Pharmacol 2025; 20:49-59. [PMID: 38659262 DOI: 10.2174/0127724328295592240419064719] [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: 12/30/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND The use of appropriate animal models for cancer studies is a major challenge, particularly for investigators who lack the resources to maintain and use xenograft animals or genetically engineered mouse models (GEMM). In addition, several countries intending to incorporate these models must conduct importation procedures, posing an additional challenge. OBJECTIVE This review aimed to explore the use of cell-derived allograft or syngeneic models under limited resources. The results can be used by investigators, specifically from low-middle-income countries, to contribute to lung cancer eradication. METHODS A literature search was carried out on various databases, including PubMed, Web of Science, and Scopus. In addition, the publication year of the selected articles was set between 2013 and 2023 with different search components (SC), namely lung cancer (SC1), animal models (SC2), and preclinical studies (SC3). RESULTS This systematic review focused on selecting animals, cells, and methods that could be applied to generating allograft-type lung cancer animal models from 101 included articles. CONCLUSION Based on the results, the use of cell-derived allograft models in cancer studies is feasible and relevant, and it provides valuable insights regarding the conditions with limited resources.
Collapse
Affiliation(s)
- Isa Mahendra
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Ahmad Kurniawan
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Muhamad Basit Febrian
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Iim Halimah
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Asep Rizaludin
- Research Center for Radioisotope, Radiopharmaceuticals and Biodosimetry Technology, National Research and Innovation Agency, Serpong, Indonesia
| | - Dani Gustaman Syarif
- Research Center for Radiation Process Technology, National Research and Innovation Agency, Serpong, Indonesia
| |
Collapse
|
2
|
Fan Y, Chen A, Zhu J, Liu R, Mei Y, Li L, Sha X, Wang X, Ren W, Wang L, Liu B. Engineered lactococcus lactis intrapleural therapy promotes regression of malignant pleural effusion by enhancing antitumor immunity. Cancer Lett 2024; 588:216777. [PMID: 38432582 DOI: 10.1016/j.canlet.2024.216777] [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: 12/05/2023] [Revised: 02/14/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Intrapleural immunotherapies have emerged as a prominent field in treating malignant pleural effusion (MPE). Among these, bacteria-based intrapleural therapy has exerted an anti-MPE effect by immuno-stimulating or cytotoxic properties. We previously engineered a probiotic Lactococcus lactis (FOLactis) expressing a fusion protein of Fms-like tyrosine kinase 3 and co-stimulator OX40 ligands. FOLactis activates tumor antigen-specific immune responses and displays systemic antitumor efficacy via intratumoral delivery. However, no available lesions exist in the pleural cavity of patients with MPE for intratumoral administration. Therefore, we further optimize FOLactis to treat MPE through intrapleural injection. Intrapleural administration of FOLactis (I-Pl FOLactis) not only distinctly suppresses MPE and pleural tumor nodules, but also significantly extends noticeable survival in MPE-bearing murine models. The proportion of CD103+ dendritic cells (DCs) in tumor-draining lymph nodes increases three-fold in FOLactis group, compared to the wild-type bacteria group. The enhanced DCs recruitment promotes the infiltration of effector memory T and CD8+ T cells, as well as the activation of NK cells and the polarization of macrophages to M1. Programmed death 1 blockade antibody combination further enhances the antitumor efficacy of I-Pl FOLactis. In summary, we first develop an innovative intrapleural strategy based on FOLactis, exhibiting remarkable efficacy and favorable biosafety profiles. These findings suggest prospective clinical translation of engineered probiotics for managing MPE through direct administration into the pleural cavity.
Collapse
Affiliation(s)
- Yue Fan
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China; The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Aoxing Chen
- The Clinical Cancer Institute of Nanjing University, Nanjing, China; Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China
| | - Junmeng Zhu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Rui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China; The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yi Mei
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lin Li
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, 210008, China; Department of Pathology, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xiaoxuan Sha
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xiaonan Wang
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China; The Comprehensive Cancer Centre, China Pharmaceutical University Nanjing Drum Tower Hospital, 321 Zhongshan Road, Nanjing, 210008, China
| | - Wei Ren
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Lifeng Wang
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China.
| | - Baorui Liu
- The Comprehensive Cancer Centre, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China; The Clinical Cancer Institute of Nanjing University, Nanjing, China.
| |
Collapse
|
3
|
Xu JJ, Lucero MY, Herndon NL, Lee MC, Chan J. Comparison of a Minimally Invasive Transthoracic Approach and a Surgical Method For Intrapleural Injection of Tumor Cells in Mice. Comp Med 2023; 73:120-126. [PMID: 36922006 PMCID: PMC10162381 DOI: 10.30802/aalas-cm-22-000044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/26/2022] [Accepted: 12/09/2022] [Indexed: 03/17/2023]
Abstract
Intrapleural injections can be used in mice to deliver therapeutic and diagnostic agents and to model human disease processes (for example, pleural fluid accumulation, malignant pleural disease, and lung cancers). In the context of establishing cancer models, minimally invasive methods of intrapleural injection are desirable because inflammation at the injection site can have a major impact on tumor growth and progression. Common approaches for intrapleural injection include surgical exposure of the thoracic wall or the diaphragm prior to injection; however, these invasive procedures require tissue dissection that triggers an undesirable inflammatory response and increases the risk of pneumothorax. While nonsurgical procedures can minimize this concern, 'blind' injections may lead to off-target inoculation. In this study, we hypothesized that a minimally invasive transthoracic approach (MI-TT) would produce a tumor distribution and burden similar to that of a surgical transabdominal approach (SX-TA). Prior to performing the procedures on live mice, surgeons were trained using cadavers and terminal procedures. Then a total of 14 nude mice (female, 4 to 6 wk old) were injected with 50 μL (5 million) A549-Luc2 human cancer cells either using the MI-TT (n = 8) or SX-TA (n = 6) approach under carprofen analgesia and isoflurane anesthesia. Our results indicate that with training, a minimally invasive transthoracic approach for intrapleural injection provides more consistent tumor placement and a greater tumor burden than does the surgical method. However, additional studies are necessary to confirm anatomic placement and characterize tumor profiles.
Collapse
Affiliation(s)
- Jiajie Jessica Xu
- Division of Animal Resources, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois;,
| | - Melissa Y Lucero
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Nicole L Herndon
- Division of Animal Resources, University of Illinois at Urbana-Champaign, Urbana, Illinois; Department of Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Michael C Lee
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jefferson Chan
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
4
|
Xiang Z, Deng X, He W, Yang Q, Ni L, Dehghan Shasaltaneh M, Maghsoudloo M, Yang G, Wu J, Imani S, Wen Q. Treatment of malignant pleural effusion in non-small cell lung cancer with VEGF-directed therapy. Ann Med 2022; 54:1357-1371. [PMID: 35543207 PMCID: PMC9103356 DOI: 10.1080/07853890.2022.2071977] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) is a critical regulator of malignant pleural effusion (MPE) in non-small-cell lung cancer (NSCLC). Bevacizumab (BEV) and apatinib (APA) are novel VEGF blockers that inhibit lung cancer cell proliferation and the development of pleural effusion. METHODS In this study, we established Lewis lung cancer (LLC) xenograft mouse models to compare the therapeutic effect of APA and BEV in combination with cisplatin (CDDP) against MPE. The anti-tumour and anti-angiogenic effects of this combination therapy were evaluated by 18F-FDG PET/CT imaging, TUNEL assay and Immunohistochemistry. RESULTS The triple drug combination significantly prolonged the overall survival of the tumour-bearing mice by reducing MPE and glucose metabolism and was more effective in lowering VEGF/soluble VEGFR-2 levels in the serum and pleural exudates compared to either of the monotherapies. Furthermore, CDDP + APA + BEV promoted in vivo apoptosis and decreased microvessel density. CONCLUSIONS Mechanistically, LLC-induced MPE was inhibited by targeting the VEGF-MEK/ERK pathways. Further studies are needed to establish the synergistic therapeutic effect of these drugs in NSCLC patients with MPE.KEY MESSAGESCombined treatment of MPE with apatinib, bevacizumab and cisplatin can prolong the survival time of mice, reduce the content of MPE, decrease the SUVmax of thoracic tumour tissue, down-regulate the content of VEGF and sVEGFR-2 in serum and pleural fluid, and promote the apoptosis of tumour cells. Angiogenesis and MPE formation can be inhibited by down-regulation of HIF-1α, VEGF, VEGFR-2, MEK1 and MMP-2 molecular signalling pathway proteins.
Collapse
Affiliation(s)
- Zhangqiang Xiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Phase 1 Clinical Trial Center, Deyang People's Hospital, Deyang, China
| | - Xiangyu Deng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenfeng He
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qian Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Laichao Ni
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | | | - Mazaher Maghsoudloo
- Laboratory of Systems Biology and Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.,Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Gang Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, Anyue Hospital of Traditional Chinese Medicine, Second Ziyang Hospital of Traditional Chinese Medicine, Ziyang, China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China. The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Saber Imani
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Ekeke CN, Russell KL, Murthy P, Guo ZS, Soloff AC, Weber D, Pan W, Lotze MT, Dhupar R. Intrapleural interleukin-2-expressing oncolytic virotherapy enhances acute antitumor effects and T-cell receptor diversity in malignant pleural disease. J Thorac Cardiovasc Surg 2022; 163:e313-e328. [PMID: 33485667 PMCID: PMC9594625 DOI: 10.1016/j.jtcvs.2020.11.160] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The mainstay of treatment for patients with malignant pleural disease is fluid drainage and systemic therapy. A tumor-specific oncolytic virus or T-cell-activating interleukin-2 immunotherapy may provide an opportunity for local control. We previously developed a vaccinia virus-expressing interleukin-2, an oncolytic virus that mediated tumor regression in preclinical peritoneal tumor models with expansion of tumor-infiltrating lymphocytes. We evaluated the antitumor efficacy and immune modulatory effects of vaccinia virus-expressing interleukin-2 in malignant pleural disease. METHODS A murine model of malignant pleural disease was established with percutaneous intrapleural deposition of the Lewis lung carcinoma cell line and monitored with bioluminescent imaging. After intrapleural or systemic administration of vaccinia viruses (vaccinia virus yellow fluorescent protein control, vaccinia virus-expressing interleukin-2), systemic anti-programmed cell death-1 antibody, or combination therapy (vaccinia virus-expressing interleukin-2 and anti-programmed cell death-1), tumor mass, immune cell infiltration, T-cell receptor diversity, and survival were assessed. RESULTS Intrapleural vaccinia virus resulted in significant tumor regression compared with phosphate-buffered saline control (P < .05). Inclusion of the interleukin-2 transgene further increased intratumoral CD8+ T cells (P < .01) and programmed cell death-1 expression on CD8+ tumor-infiltrating lymphocytes (P < .001). Intrapleural vaccinia virus-expressing interleukin-2 was superior to systemic vaccinia virus-expressing interleukin-2, with reduced tumor burden (P < .0001) and improved survival (P < .05). Intrapleural vaccinia virus-expressing interleukin-2 alone or combined treatment with systemic anti-programmed cell death-1 reduced tumor burden (P < .01), improved survival (P < .01), and increased intratumoral αβ T-cell receptor diversity (P < .05) compared with systemic anti-programmed cell death-1 monotherapy. CONCLUSIONS Intrapleural vaccinia virus-expressing interleukin-2 reduced tumor burden and enhanced survival in a murine malignant pleural disease model. Increased CD8+ tumor-infiltrating lymphocytes and αβ T-cell receptor diversity are associated with enhanced response. Clinical trials will enable assessment of intrapleural vaccinia virus-expressing interleukin-2 therapy in patients with malignant pleural disease.
Collapse
Affiliation(s)
- Chigozirim N Ekeke
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Kira L Russell
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Pranav Murthy
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Zong Sheng Guo
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Adam C Soloff
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Daniel Weber
- iRepertoire, Inc, Hudson Alpha Institute for Biotechnology, Huntsville, Ala
| | - Wenjing Pan
- iRepertoire, Inc, Hudson Alpha Institute for Biotechnology, Huntsville, Ala
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh, Pittsburgh, Pa; Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pa
| | - Rajeev Dhupar
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pa; Surgical Services Division, VAPHS, Pittsburgh, Pa.
| |
Collapse
|
6
|
Sabbion RO, Terra RM, Teixeira LR, Acencio MMP, Augusto MC, Costa PB, Fernandes PMP. Influence of the progression of pleural neoplasia on the outcome of pleurodesis in mice. Oncotarget 2020; 11:2002-2009. [PMID: 32523654 PMCID: PMC7260117 DOI: 10.18632/oncotarget.27610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/19/2020] [Indexed: 11/25/2022] Open
Abstract
Purpose: Experimental study aimed at evaluating whether pleural neoplastic disease is associated with the degree of pleural fibrosis over time caused by talc pleurodesis. The study describes changes in levels of inflammatory mediators and determines whether the course of time involved in progression of neoplastic pleural disease is the factor that influences safety of talc pleurodesis usage in mice. Materials and Methods: Animals were randomized into two groups: Cancer group (CG) that received intrapleural injection of Lewis cells or Saline group (SG) that received saline injection. After, the animals were subdivided into Early (pleurodesis 3 days after pleural injection) and Late (pleurodesis 7 days after pleural injection) groups. Half of the animals in each group were euthanized 24 hours after pleurodesis (to obtain the inflammatory data); the remaining animals were killed after 8 days (to obtain the scores of pleural fibrosis). Results: CGs had lower fibrosis scores than SGs comparing early phases to late phases. Inflammation scores were lower in CGs, particularly in Late group. In SGs the inflammation was intense in 100% of the animals. In Late CG group pleural adhesions had the lowest scores; we found intense fibrosis only in SGs. VEGF and LDH levels had increased in animals with cancer, particularly in Late group. Systemic distribution of talc occurred only in Late CG. Conclusions: The time for pleural neoplasia to evolve is inversely proportional to the degree of pleural fibrosis. Earlier pleurodesis yielded the best results related to fibrosis, with less systemic inflammation and is safer in mice.
Collapse
Affiliation(s)
- Rodrigo Olivio Sabbion
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Ricardo Mingarini Terra
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Lisete Ribeiro Teixeira
- Laboratorio de Pleura-Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Milena Marques Pagliarelli Acencio
- Laboratorio de Pleura-Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Marcia Cristina Augusto
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Priscila Berenice Costa
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Paulo Manuel Pego Fernandes
- Division of Thoracic Surgery, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
7
|
Jin Z, Shen C, Zhang H, Qi R, Guo Q, Liu R, Hua B, Shi Z. Chinese medicine Xiaoshui decoction inhibits malignant pleural effusion in mice and mediates tumor-associated macrophage polarization by activating autophagy. JOURNAL OF ETHNOPHARMACOLOGY 2020; 249:112412. [PMID: 31751649 DOI: 10.1016/j.jep.2019.112412] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/23/2019] [Accepted: 11/17/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoshui decoction (XSD) is a traditional Chinese medicine compound prescription that has been shown to reinforce the spleen and remove the fluid retention, while being widely used in the treatment of malignant pleural effusion (MPE). We previously reported that XSD alleviates symptoms and improves the quality of life in patients with MPE; however, the mechanism employed by XSD on MPE has not yet been elucidated. AIM OF THE STUDY To investigate the role and mechanism of XSD in inhibiting the development of MPE, and in regulating macrophage polarization in vitro and in vivo. MATERIALS AND METHODS A murine MPE model was used to study the effect of XSD on MPE. Mice with MPE were randomly allocated to a control group and XSD-low-dose (1.144 g/mL), XSD-middle-dose (2.288 g/mL), XSD-high-dose (4.576 g/mL), or cisplatin groups. RAW264.7 cells were induced to form tumor-associated macrophages (TAMs) as well as M1 and M2 macrophages using different conditioned media in vitro. RESULTS XSD effectively inhibited MPE formation, reduced pleural permeability and angiogenesis, and prolonged mice survival. Particularly, XSD treatment induced the polarization of TAMs to the M1 phenotype in MPE. Moreover, in-vitro XSD remarkably promoted the expansion of M1 macrophages and reduced M2 macrophages by enhancing autophagy. CONCLUSIONS XSD inhibits MPE development and regulates macrophage polarization by activating autophagy, indicating that XSD may serve as a novel option for integrative MPE therapies.
Collapse
Affiliation(s)
- Zhichao Jin
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimennei, Beijing, 100700, China; Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Chunfeng Shen
- Changzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, Jiangsu, 213003, China.
| | - Haidong Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Runzhi Qi
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| | - Qiujun Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| | - Rui Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| | - Zhan Shi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| |
Collapse
|
8
|
Beck TN, Deneka AY, Chai L, Kanach C, Johal P, Alvarez NJ, Boumber Y, Golemis EA, Laub GW. An improved method of delivering a sclerosing agent for the treatment of malignant pleural effusion. BMC Cancer 2019; 19:614. [PMID: 31234819 PMCID: PMC6589887 DOI: 10.1186/s12885-019-5777-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022] Open
Abstract
Background Malignant pleural effusion (MPE) is a devastating sequela associated with cancer. Talc pleurodesis is a common treatment strategy for MPE but has been estimated to be unsuccessful in up to 20–50% of patients. Clinical failure of talc pleurodesis is thought to be due to poor dispersion. This monograph reports the development of a foam delivery system designed to more effectively coat the pleural cavity. Methods C57BL/6 mice were injected with Lewis lung carcinoma (LL/2) cells intrapleurally to induce MPE. The mice then received either normal saline (NS) control, foam control (F), talc slurry (TS, 2 mg/g) or talc foam (TF, 2 mg/g). Airspace volume was evaluated by CT, lungs/pleura were collected, and percent fibrosis was determined. Results The TF group had significantly better survival than the TS group (21 vs 13.5 days, p < 0.0001). The average effusion volume was less in the talc groups compared to the control group (140 vs 628 μL, p < 0.001). TF induced significant lung fibrosis (p < 0.01), similar to TS. On CT, TF significantly (p < 0.05) reduced loss of right lung volume (by 30–40%) compared to the control group. This was not seen with TS (p > 0.05). Conclusions This report describes using a novel talc foam delivery system for the treatment of MPE. In the LL/2 model, mice treated with the TF had better survival outcomes and less reduction of lung volume than mice treated with the standard of care TS. These data provide support for translational efforts to move talc foam from animal models into clinical trials.
Collapse
Affiliation(s)
- Tim N Beck
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA. .,Digestive Disease & Surgery Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| | - Alexander Y Deneka
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.,Department of Biochemistry, Kazan Federal University, Kazan, Russia
| | - Louis Chai
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Hahnemann University Hospital, 230 North Broad Street, Philadelphia, PA, 19102, USA
| | - Colin Kanach
- Department of Pathology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Priya Johal
- Department of Pathology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Nicolas J Alvarez
- Department of Chemical and Biological Engineering, Drexel University, Philadelphia, PA, 19129, USA
| | - Yanis Boumber
- Department of Biochemistry, Kazan Federal University, Kazan, Russia.,Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Glenn W Laub
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Hahnemann University Hospital, 230 North Broad Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
9
|
Intrapleural targeted therapies (anti-VEGF and anti-EGFR) in the model of malignant pleural effusion. Oncotarget 2017; 8:105093-105102. [PMID: 29285236 PMCID: PMC5739623 DOI: 10.18632/oncotarget.21362] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/28/2017] [Indexed: 01/09/2023] Open
Abstract
Rationale Malignant pleural effusion has few options of treatment and drugs administrated by different routes can lead to a less permissive microenvironment for the development of malignant pleural disease. Objectives To analyze therapies administered intrapleurally in malignant pleural disease and to study EGFR and KRAS mutations in adenocarcinoma. Methods Mice received LLC cells and were treated intrapleurally with anti-VEGF, anti-EGFR, anti-VEGF+anti-EGFR or saline. Animal survival, weight and mobility, volume, biochemistry and immunology of fluid, gene expression, KRAS and EGFR mutation were evaluated. Results All animals developed malignant effusion and presented progressive weight loss without difference between groups; however, groups treated with anti-EGFR were more active. No difference in mortality was observed. Temporal increase of volume and inflammatory markers was observed mainly in the untreated group. Gene expression in tumors was overexpressed in VEGF, EGFR and KRAS compared with normal tissue. Mutation in exon 2 of the KRAS gene was observed. Conclusions Intrapleural Anti-VEGF and/or anti-EGFR reduced volume and inflammatory mediators in pleural fluid. Anti-EGFR and anti-VEGF+anti-EGFR decreased morbidity although without impact on survival. LLC tumors presented KRAS mutation, this could have influenced the action of these therapies.
Collapse
|