1
|
Cho Y, Lim J, Kim YH, Min J. ReV as a Novel S. cerevisiae-Derived Drug Carrier to Enhance Anticancer Therapy through Daunorubicin Delivery. Appl Biochem Biotechnol 2025; 197:2301-2311. [PMID: 39738861 DOI: 10.1007/s12010-024-05177-x] [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] [Accepted: 12/24/2024] [Indexed: 01/02/2025]
Abstract
This study explores the potential of vacuoles derived from Saccharomyces cerevisiae (S. cerevisiae) as a novel form of drug carrier, specifically focusing on their application in enhancing the delivery of the chemotherapeutic agent Daunorubicin (DNR). We isolated and reassembled these vacuoles, referred to as Reassembled Vacuoles (ReV), aiming to overcome the challenges of drug degradation caused by hydrolytic enzymes within traditional vacuoles. ReV encapsulating DNR were tested against HL-60 cells, a model for acute myeloid leukemia, to evaluate their therapeutic impact. Through various analyses, including Nanoparticle tracking analysis (NTA) and Field-emission electron scanning microscope (FE-SEM), we characterized the properties of ReV. Our findings revealed that ReV exhibited superior stability, drug release rate, and cytotoxic efficacy compared to normal vacuoles (NorV). Notably, ReV demonstrated a higher apoptosis rate in HL-60 cells, efficient and complete release of DNR within 24 h, and reduced cytotoxic side effects. These results suggest that ReV could represent a new and effective drug delivery system in anticancer therapy, paving the way for more targeted and safer cancer treatment modalities.
Collapse
Affiliation(s)
- Yunyoung Cho
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea
| | - Jiwoo Lim
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University 1, Chungdae-Ro, Seowon-Gu, Cheongju, 28644, South Korea.
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-Daero, Deokjin-Gu Jeonju, Jeonbuk, 54896, South Korea.
| |
Collapse
|
2
|
Xue D, Xu M, Madden MD, Lian X, Older EA, Pulliam C, Hui Y, Shang Z, Gupta G, Raja MK, Wang Y, Sardi A, Long Y, Chen H, Fan D, Bugni TS, Testerman TL, Wu Q, Li J. Discovery of a Chimeric Polyketide Family as Cancer Immunogenic Chemotherapeutic Leads. J Am Chem Soc 2025; 147:265-277. [PMID: 39731542 DOI: 10.1021/jacs.4c09582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Discovery of cancer immunogenic chemotherapeutics represents an emerging, highly promising direction for cancer treatment that uses a chemical drug to achieve the efficacy of both chemotherapy and immunotherapy. Herein, we report a high-throughput screening platform and the subsequent discovery of a new class of cancer immunogenic chemotherapeutic leads. Our platform integrates informatics-based activity metabolomics for the rapid identification of microbial natural products with both novel structures and potent activities. Additionally, we demonstrate the use of microcrystal electron diffraction (MicroED) for direct structure elucidation of lead compounds from partially purified mixtures. Using this strategy to screen geographically and phylogenetically diverse microbial metabolites against pseudomyxoma peritonei, a rare and severe cancer, we discovered a new class of leads, aspercyclicins. The aspercyclicins feature an unprecedented tightly packed polycyclic polyketide scaffold that comprises continuous fused, bridged, and spiro rings. The biogenesis of aspercyclicins involves two distinct biosynthetic pathways, leading to formation of chimeric compounds that cannot be predicted by bottom-up approaches mining natural product biosynthetic genes. With comparable potency to some clinically used anticancer drugs, aspercyclicins are active against multiple cancer cell types by inducing immunogenic cell death (ICD), including the release of damage-associated molecular patterns and subsequent phagocytosis of cancer cells. The broad-spectrum ICD-inducing activity of aspercyclicins, combined with their low toxicity to normal cells, represents a new class of potential cancer immunogenic chemotherapeutics and, particularly, the first drug lead for pseudomyxoma peritonei treatment.
Collapse
Affiliation(s)
- Dan Xue
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Mingming Xu
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Michael D Madden
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Xiaoying Lian
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Ethan A Older
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Conor Pulliam
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Yvonne Hui
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Zhuo Shang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Gourab Gupta
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Manikanda K Raja
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Armando Sardi
- Department of Surgical Oncology, The Institute for Cancer Care at Mercy, Mercy Medical Center, Baltimore, Maryland 21202, United States
| | - Yaoling Long
- Department of Biological and Physical Sciences, South Carolina State University, Orangeburg, South Carolina 29117, United States
| | - Hexin Chen
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29208, United States
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Lachman Institute for Pharmaceutical Development, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Traci L Testerman
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, South Carolina 29209, United States
| | - Qihao Wu
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jie Li
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina 29208, United States
| |
Collapse
|
3
|
Yu B, Zeng W, Zhou Y, Li N, Liang Z. Characterization and Bioactive Metabolite Profiling of Streptomyces sp. Y009: A Mangrove-Derived Actinomycetia with Anticancer and Antioxidant Potential. Microorganisms 2024; 12:2300. [PMID: 39597689 PMCID: PMC11596135 DOI: 10.3390/microorganisms12112300] [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: 09/26/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
Microorganisms from poorly explored environments are promising sources for the development of novel drugs. In our continuous efforts to screen for mangrove actinomycetes that produce metabolites with potential pharmaceutical applications, Streptomyces sp. Y009 was isolated from mangrove sediments in Guangxi, China. The phenotypic, physiological, biochemical, and phylogenetic characteristics of this strain were investigated. Analysis of phylogenetic and 16S rRNA gene sequences showed that it had the highest sequence similarity to Streptomyces thermolilacinus NBRC 14274 (98.95%). Further, the Y009 extract exhibited antioxidant activity, as indicated by DPPH and superoxide dismutase assays. The extract showed broad-spectrum and potent anticancer potential against six human cancer cell lines, with IC50 values ranging from 5.61 to 72.15 μg/mL. Furthermore, the selectivity index (SI) demonstrated that the Y009 extract exhibited less toxicity toward normal cell lines in comparison to the lung cancer cell line (A549) and hepatoma cell line (HepG2). GC-MS analysis revealed that the extract contained some biologically important secondary metabolites, mainly cyclic dipeptides and esters, which might be responsible for the antioxidant and anticancer properties. 3-Isobutylhexahydropyrrolo[1,2-a]pyrazine-1,4-dione (28.32%) was the major chemical compound available in the extract. The effect on cancer cells was then confirmed using nuclear staining and in silico docking. This study suggests that further exploration of the bioactive compounds of the newly isolated strain may be a promising approach for the development of novel chemopreventive drugs.
Collapse
Affiliation(s)
| | | | | | - Nan Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Microorganism and Enzyme Research Center of Engineering Technology, College of Life Science and Technology, Guangxi University, 100 Daxue Road, Nanning 530004, China; (B.Y.)
| | - Zhiqun Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Microorganism and Enzyme Research Center of Engineering Technology, College of Life Science and Technology, Guangxi University, 100 Daxue Road, Nanning 530004, China; (B.Y.)
| |
Collapse
|
4
|
Xue D, Xu M, Madden MD, Lian X, Older EA, Pulliam C, Hui Y, Shang Z, Gupta G, Raja MK, Wang Y, Sardi A, Long Y, Chen H, Fan D, Bugni TS, Testerman TL, Wu Q, Li J. Discovery of A Chimeric Polyketide Family as Cancer Immunogenic Chemotherapeutic Leads. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622009. [PMID: 39574732 PMCID: PMC11580922 DOI: 10.1101/2024.11.05.622009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Discovery of cancer immunogenic chemotherapeutics represents an emerging, highly promising direction for cancer treatment that uses a chemical drug to achieve the efficacy of both chemotherapy and immunotherapy. Herein we report a high-throughput screening platform and the subsequent discovery of a new class of cancer immunogenic chemotherapeutic leads. Our platform integrates informatics-based activity metabolomics for rapid identification of microbial natural products with both novel structures and potent activities. Additionally, we demonstrate the use of microcrystal electron diffraction (MicroED) for direct structure elucidation of the lead compounds from partially purified mixtures. Using this strategy to screen geographically and phylogenetically diverse microbial metabolites against pseudomyxoma peritonei, a rare and severe cancer, we discovered a new class of leads, aspercyclicins. The aspercyclicins feature an unprecedented tightly packed polycyclic polyketide scaffold that comprises continuous fused, bridged, and spiro rings. The biogenesis of aspercyclicins involves two distinct biosynthetic pathways, leading to formation of chimeric compounds that cannot be predicted by bottom-up approaches mining natural products biosynthetic genes. With comparable potency to some clinically used anticancer drugs, aspercyclicins are active against multiple cancer cell types by inducing immunogenic cell death (ICD), including the release of damage-associated molecular patterns and subsequent phagocytosis of cancer cells. The broad-spectrum ICD-inducing activity of aspercyclicins, combined with their low toxicity to normal cells, represents a new class of potential cancer immunogenic chemotherapeutics and particularly the first drug lead for pseudomyxoma peritonei treatment.
Collapse
|
5
|
Hong B, Du B, Chen R, Zheng C, Ni R, Liu M, Yang J. Comparison of immune checkpoint inhibitors related to pulmonary adverse events: a retrospective analysis of clinical studies and network meta-analysis. BMC Med 2024; 22:75. [PMID: 38373990 PMCID: PMC10875746 DOI: 10.1186/s12916-024-03285-3] [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: 07/05/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed tumor treatment. However, the risk of pulmonary adverse events (PAEs) associated with ICI combination therapy is still unclear. We aimed to provide a PAE overview and risk ordering of ICIs used in tumor treatment. METHODS We searched the databases of PubMed, PsycINFO, Embase, Cochrane Library, CINAHL, Web of Science, Scopus, and clinical trial websites during January 2011-April 2023 to identify phase II and III randomized clinical trials (RCTs) and single-arm clinical trials wherein at least one treatment arm received ICIs (e.g., ICI monotherapy, a combination of two ICIs, or ICIs in combination with conventional cancer therapy). We reported the results of PAEs. Additionally, we compared risks of PAEs between different drug classes using a Bayesian network meta-analysis. RESULTS Among 143 RCTs and 24 single-arm trials, the incidence of all-grade and grade 3-4 PAEs were highest with programmed death L1 (PD-L1) plus cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and plus chemotherapy and anti-PD1 plus anti-CTLA4, the lowest with targeted therapy drug plus chemotherapy and anti-PD1 plus anti-PDL1. Anti-PD1 plus anti-CTLA4 and plus chemotherapy was the intervention with the highest risk for all-grade and 3-4 grade PAEs, and the intervention with the lowest risk was chemotherapy and anti-PD1 plus anti-PDL1. In terms of all-grade PAEs, chemotherapy was safer than ICI monotherapy. Except for the anti-PD1 plus anti-PDL1 regimen, no significant difference in the risk of grade 3-4 PAEs was detected between dual-ICIs and single-ICIs. Furthermore, the risk of PAEs associated with nivolumab, pembrolizumab, and atezolizumab may be dose dependent. CONCLUSIONS In the single-drug regimen, anti-PD1 caused the greatest incidence of PAEs. The risk of PAEs was higher with all single-ICIs than with chemotherapy. However, no significant difference in the risk of PAEs was detected between single-ICIs. In the combined regimen, anti-PD1 plus anti-CTLA4 and plus chemotherapy showed the greatest risk of PAEs, but there were no significant differences in risk between dual-ICIs and single-ICIs.
Collapse
Affiliation(s)
- Baohui Hong
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Pharmacy, The Second Hospital of Sanming City, Sanming, China
- The School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bin Du
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rong Chen
- Department of Anesthesiology, The Second Hospital of Sanming City, Sanming, China
| | - Caiyun Zheng
- Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
- The School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Ruping Ni
- Department of Pharmacy, The First Hospital of Nanping City, Nanping, China
- The School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China.
- The School of Pharmacy, Fujian Medical University, Fuzhou, China.
| | - Jing Yang
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China.
- The School of Pharmacy, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
6
|
Li LZ, Lin SH, Wu LX, Chen T, Zhuang JN, Lai HQ, Liang ZP, Zhu WS, Li WX, Tang L, Zhang HT, Fang CT. Major pathological remissions in a patient with stage IIIA nonsmall cell lung cancer after neoadjuvant tislelizumab combined with chemotherapy: a case report and literature review. Anticancer Drugs 2023; 34:962-966. [PMID: 36602434 DOI: 10.1097/cad.0000000000001497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Currently, there are few reports of patients with locally advanced lung cancer achieving a clinical complete response by medical treatment. Preoperative neoadjuvant immunotherapy combined with chemotherapy is an option for patients with unresectable, locally advanced nonsmall cell lung cancer (NSCLC) which is of great potential, and may change traditional treatment paradigms. There are relatively few large-scale, high-quality randomized-controlled trials yet, and limitations such as short postoperative follow-up period and immature disease-free survival and overall survival data still persist. Thus, evidence-based medical evidence is urgently needed. It is worthy to explore the further treatment of patients who achieved complete response after initial treatment, though lacking of evidence by now. CASE PRESENTATION We report a stage IIIA lung squamous cell carcinoma case who achieved a major pathologic remission after neoadjuvant treatment with tislelizumab and chemotherapy. CONCLUSION Our case study contributes to the existing evidence on the feasibility, efficacy and safety of neoadjuvant immunotherapy combined with chemotherapy in locally advanced unresectable NSCLC.
Collapse
Affiliation(s)
- Lu-Zhen Li
- Department of Oncology, Zhongshan Affiliated Hospital, Guangzhou University of Chinese Medicine, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Khalil SR, Zheng C, Abou-Zeid SM, Farag MR, Elsabbagh HS, Siddique MS, Azzam MM, Cerbo AD, Elkhadrawey BA. Modulatory effect of thymol on the immune response and susceptibility to Aeromonas hydrophila infection in Nile tilapia fish exposed to zinc oxide nanoparticles. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 259:106523. [PMID: 37058790 DOI: 10.1016/j.aquatox.2023.106523] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 05/15/2023]
Abstract
Zinc oxide nanoparticles (ZnO-NPs) have many exciting properties that make their use in a continuous increase in various biomedical, industrial, and agricultural applications. This is associated with accumulation in the aquatic ecosystems and fish exposure with consequent deleterious effects. To determine the potential of thymol to counteract the immunotoxic effects of ZnO-NPs, Oreochromis niloticus was exposed to ZnO-NPs (⅕ LC50 =1.14 mg/L, for 28 days) with or without feeding a thymol-incorporated diet (1 or 2 g/kg diet). Our data demonstrated a reduction of aquaria water quality, leukopenia, and lymphopenia with a decrease in serum total protein, albumin, and globulin levels in exposed fish. At the same time, the stress indices (cortisol and glucose) were elevated in response to ZnO-NPs exposure. The exposed fish also revealed a decline in serum immunoglobulins, nitric oxide, and the activities of lysozyme and myeloperoxidase, in addition to reduced resistance to the Aeromonas hydrophila challenge. The RT-PCR analysis showed downregulation of antioxidant (SOD) superoxide dismutase and (CAT) catalase gene expression in the liver tissue with overexpression of the immune-related genes (TNF-α and IL-1β). Importantly, we found that thymol markedly protected against ZnO-NPs-induced immunotoxicity in fish co-supplemented with thymol (1 or 2 g/kg diet) in a dose-dependent manner. Our data confirm the immunoprotective and antibacterial effects of thymol in ZnO-NPs exposed fish, supporting the potential utility of thymol as a possible immunostimulant agent.
Collapse
Affiliation(s)
- Samah R Khalil
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Chuntian Zheng
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Key Laboratory of Animal Nutrition and Feed Science (South China) of Ministry of Agriculture, State Key Laboratory of Livestock and Poultry Breeding, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangdong Key Laboratory of Animal Breeding and Nutrition, 510640 Guangzhou, China.
| | - Shimaa M Abou-Zeid
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, University of Sadat City, 32897, Egypt
| | - Mayada R Farag
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Hesham S Elsabbagh
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, University of Sadat City, 32897, Egypt
| | - Mouhamed S Siddique
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, University of Sadat City, 32897, Egypt
| | - Mahmoud M Azzam
- Department of Animal Production College of Food & Agriculture Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, Matelica, Italy.
| | - Basma A Elkhadrawey
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, University of Sadat City, 32897, Egypt
| |
Collapse
|
8
|
Meng L, Wang Z, Hou Z, Wang H, Zhang X, Zhang X, He X, Zhang X, Qin B, Li J, Zhang Z, Xue X, Wei Y. Study of epirubicin sustained-release chemoablation in tumor suppression and tumor microenvironment remodeling. Front Immunol 2022; 13:1064047. [PMID: 36605217 PMCID: PMC9807901 DOI: 10.3389/fimmu.2022.1064047] [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: 10/07/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction Although intratumoral chemoablation can obtain an impressive therapeutic effect, there is still incomplete ablation and tumor recurrence in some patients. This could be due to the short retention time of the drug in the tumor, the limited distribution of intratumoral drugs, and, beyond that, the immunotolerance caused by the tumor microenvironment (TME). There is still an urgent need to find an optimal drug sustained-release carrier and figure out the impact of regional injection to TME. Methods In this study, we supposed to use polyethylene glycol (PEG) hydrogel as a drug carrier to improve the retention time of the drug to extend the exposure of tumor cells and investigate the feasibility of combination local Epirubicin injection with anti-PD-L1. Results The results revealed obvious tumor suppression based on the tumor volume and the inhibition time of tumor growth in the A549 lung cancer mouse model after local injection. Furthermore, the enhanced antitumor effects of the combination of systematic anti- programmed death ligand 1 (PD-L1) therapy with local chemoablation (EPI-GEL/PD-L1) for abscopal tumor reduction in the 4T1 breast model were also observed. Flow cytometry analysis of the tumor and blood samples showed significant variations in the proportions of PD-L1+ and CD3+CD8+PD-1+ cells before and after anti-PD-L1 therapy. On day 4 after local injection of the EPI gel, the expression of PD-L1 in abscopal tumors was upregulated, while the expression of PD-L1 in bilateral tumors in mice was significantly reduced after anti-PD-L1 treatment. The proportion of CD3+CD8+PD-1+ cells in the tumor and circulating blood in the EPI-GEL/PD-L1 group was decreased compared with that in the EPI-GEL (single injection of epirubicin) group. Discussion The combination of local injection of the chemoablation agent with anti-PD-L1 monoclonal antibody (mAb) therapy may strengthen the antitumor activity, and the use of PEG hydrogel as the drug carrier can extend the retention time of the chemoablation agent around the tumor, maintaining a long-term tumor-killing activity.
Collapse
Affiliation(s)
- Liangliang Meng
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Zhenjun Wang
- Department of Radiology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhonghui Hou
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Hufei Wang
- National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Xiao Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xiaobo Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xiaofeng He
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xin Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Boyu Qin
- Department of Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jing Li
- Department of Radiology, Characteristic Medical Center, Chinese People’s Armed Police Force, Tianjin, China
| | - Zhongliang Zhang
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Xiaodong Xue
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China
| | - Yingtian Wei
- Department of Radiology, Chinese People's Armed Police (PAP) Hospital of Beijing, Beijing, China,*Correspondence: Yingtian Wei,
| |
Collapse
|
9
|
Saetang J, Tedasen A, Sangkhathat S, Sangkaew N, Dokduang S, Prompat N, Taraporn S, Graidist P. The attenuation effect of low piperine Piper nigrum extract on doxorubicin-induced toxicity of blood chemical and immunological properties in mammary tumour rats. PHARMACEUTICAL BIOLOGY 2022; 60:96-107. [PMID: 34962450 PMCID: PMC8735876 DOI: 10.1080/13880209.2021.2018470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/26/2021] [Accepted: 12/10/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Many natural extracts have been shown to minimize the toxicity of doxorubicin (Dox). Low piperine Piper nigrum L. (Piperaceae) extract (PFPE) is a natural extract containing many types of antioxidants that may reduce Dox toxicities. OBJECTIVE To evaluate the effect of PFPE in attenuating the side effects of Dox. MATERIALS AND METHODS Tumour-bearing Sprague Dawley rats were divided into five groups including normal, vehicle, 100 mg/kg BW of PFPE plus 2 mg/kg BW of Dox (P100 + Dox), 100 mg/kg BW of PFPE plus 2 mg/kg BW of Dox (P200 + Dox) and Dox. Rats were treated with Dox and/or PFPE three times/week for 4 weeks. Tumour burden, blood parameters, weight of internal organs and immunological data were investigated. RESULTS The addition of 200 mg/kg PFPE significantly restored the levels of AST from 174.60 ± 45.67 U/L in the Dox group near to normal levels at 109.80 ± 4.99 U/L. The combination of PFPE and Dox also decreased the levels of CXCL7, TIMP-1, sICAM-1 and l-selectin about 1.4-1.6-fold compared to Dox group. Feeding rats with 200 mg/kg BW of PFPE combination with Dox slightly increased Th1 from 161.67 ± 14.28 cells in Dox group to 200.75 ± 5.8 cells meanwhile suppressed Treg from 3088 ± 78 cells in Dox to 2561 ± 71 cells. DISCUSSION AND CONCLUSIONS This study showed that PFPE ameliorated Dox toxicity in many aspects indicating the role of antioxidant and other substances in the extract on toxicity attenuation. This suggested the using of PFPE may be valuable for Dox treated patients.
Collapse
Affiliation(s)
- Jirakrit Saetang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
- EZ-Mol-Design Laboratory, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Aman Tedasen
- Department of Medical Technology, School of Allied Health Sciences and Public Health, Walailak University, Thai Buri, Thailand
| | - Surasak Sangkhathat
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Natnaree Sangkaew
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Sirinapa Dokduang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Napat Prompat
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Siriporn Taraporn
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| | - Potchanapond Graidist
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Thailand
| |
Collapse
|
10
|
Cao L, Xu C, Wang MD, Qi WX, Cai G, Cai R, Wang SB, Ou D, Li M, Shen KW, Chen JY. Influence of Adjuvant Radiotherapy Timing on Survival Outcomes in High-Risk Patients Receiving Neoadjuvant Treatments. Front Oncol 2022; 12:905223. [PMID: 35912233 PMCID: PMC9334789 DOI: 10.3389/fonc.2022.905223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeTo determine the relationship between time to radiotherapy (TTR) and survival outcomes in breast cancer (BC) patients treated with neoadjuvant treatments (NATs).MethodsContinuous non-metastatic BC patients receiving NAT and adjuvant radiotherapy (RT) from 2009 to 2016 were retrospectively reviewed. A multivariable Cox model with restricted cubic splines (RCSs) was used to determine the panoramic relationship between TTR and survival outcomes. Multivariable analysis was used to control for confounding factors between the groups of TTR.ResultsA total of 315 patients were included. The RCS modeling demonstrated a non-linear relationship between TTR and survival outcomes. The lowest risk for distant metastasis-free survival (DMFS) and recurrence-free survival (RFS) was observed at the TTR of 12 weeks, and the lowest risk of BC-specific survival (BCSS) at 10 weeks. TTR was accordingly transformed into categorical variables as ≤10, 11–20, and >20 weeks. Multivariable analysis revealed that the TTR of ≤10 weeks was an independent prognostic factor for worse DMFS (HR = 2.294, 95% CI 1.079–4.881) and RFS (HR = 2.126, 95% CI 1.038–4.356) compared with the TTR of 10–20 weeks, while the is no difference in DMFS, RFS, and BCSS between TTR >20 weeks and TTR of 10–20 weeks.ConclusionThere exists a non-linear relationship between TTR after surgery and survival outcomes in patients treated with NAT. Early initiation of RT following surgery does not seem to be associated with a better therapeutic outcome. A relatively flexible recommendation of TTR could be adopted in clinical practice.
Collapse
Affiliation(s)
- Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cheng Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Meng-Di Wang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei-Xiang Qi
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Gang Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Rong Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shu-Bei Wang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dan Ou
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Li
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kun-Wei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jia-Yi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Jia-Yi Chen,
| |
Collapse
|
11
|
Yan YD, Cui JJ, Fu J, Su YJ, Chen XY, Gu ZC, Lin HW. A Network Comparison on Safety Profiling of Immune Checkpoint Inhibitors in Advanced Lung Cancer. Front Immunol 2021; 12:760737. [PMID: 34925331 PMCID: PMC8677695 DOI: 10.3389/fimmu.2021.760737] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/16/2021] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have become one of the standard treatment options for advanced lung cancer. However, adverse events (AEs), particularly immune–related AEs (irAEs), caused by these drugs have aroused public attention. The current network meta-analysis (NMA) aimed to compare the risk of AEs across different ICI–based regimens in patients with advanced lung cancer. Methods We systematically searched the PubMed, EMBASE, and Cochrane Library databases (from inception to 19 April 2021) for relevant randomized controlled trials (RCTs) that compared two or more treatments, with at least one ICI administered to patients with advanced lung cancer. The primary outcomes were treatment–related AEs and irAEs, including grade 1–5 and grade 3–5. The secondary outcomes were grade 1–5 and grade 3–5 irAEs in specific organs. Both pairwise and network meta-analyses were conducted for chemotherapy, ICI monotherapy, ICI monotherapy + chemotherapy, dual ICIs therapy, and dual ICIs + chemotherapy for all safety outcomes. Node–splitting analyses were performed to test inconsistencies in network. Sensitivity analyses were adopted by restricting phase III RCTs and studies that enrolled patients with non–small cell lung cancer. Results Overall, 38 RCTs involving 22,178 patients with advanced lung cancer were enrolled. Both pooled incidence and NMA indicated that treatments containing chemotherapy increased the risk of treatment–related AEs when compared with ICI-based regimens without chemotherapy. As for grade 1–5 irAEs, dual ICIs + chemotherapy was associated with the highest risk of irAEs (probability in ranking first: 50.5%), followed by dual-ICI therapy (probability in ranking second: 47.2%), ICI monotherapy (probability in ranking third: 80.0%), ICI monotherapy + chemotherapy (probability in ranking fourth: 98.0%), and finally chemotherapy (probability in ranking fifth: 100.0%). In grade 3–5 irAEs, subtle differences were observed; when ranked from least safe to safest, the trend was dual ICIs therapy (60.4%), dual ICIs + chemotherapy (42.5%), ICI monotherapy (76.3%), ICI monotherapy + chemotherapy (95.0%), and chemotherapy (100.0%). Furthermore, detailed comparisons between ICI–based options provided irAE profiles based on specific organ/system and severity. Conclusions In consideration of overall immune–related safety profiles, ICI monotherapy + chemotherapy might be a better choice among ICI–based treatments for advanced lung cancer. The safety profiles of ICI–based treatments are various by specific irAEs and their severity. Systematic Review Registration https://www.crd.york.ac.uk/prospero, identifier CRD42021268650
Collapse
Affiliation(s)
- Yi-Dan Yan
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiu-Jie Cui
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Fu
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying-Jie Su
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Yu Chen
- Department of Pharmacy, The People's Hospital of Guangxi Zhuang Autonomous Region (Guangxi Academy of Medical Sciences), Nanning, China
| | - Zhi-Chun Gu
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hou-Wen Lin
- Department of Pharmacy, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Farag MR, Moselhy AAA, El-Mleeh A, Aljuaydi SH, Ismail TA, Di Cerbo A, Crescenzo G, Abou-Zeid SM. Quercetin Alleviates the Immunotoxic Impact Mediated by Oxidative Stress and Inflammation Induced by Doxorubicin Exposure in Rats. Antioxidants (Basel) 2021; 10:antiox10121906. [PMID: 34943009 PMCID: PMC8750303 DOI: 10.3390/antiox10121906] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent against hematogenous and solid tumors with undesirable side effects including immunosuppression. Quercetin (QUR), a natural flavonoid abundant in fruits and vegetables, has a potent antioxidant activity. The aim of the current study was to assess the impact of QUR on DOX-induced hematological and immunological dysfunctions in a rodent model. Randomly grouped rats were treated as follows: control, QUR alone (50 mg/kg for 15 days per os), DOX alone (2.5 mg/kg I/P, three times a week, for two weeks), and co-treated rats with QUR for 15 days prior to and concomitantly with DOX (for two weeks), at the doses intended for groups two and three. DOX alone significantly disrupted the erythrogram and leukogram variables. Serum immunoglobulin (IgG, IgM, and IgE) levels and the activities of catalase (CAT) and superoxide dismutase (SOD) in spleen were declined. The DNA damage traits in spleen were elevated with an upregulation of the expression of the apoptotic markers (p53 and Caspase-3 genes) and the proinflammatory cytokines (IL-6 and TNF-α genes), while the expression of CAT gene was downregulated. These biochemical changes were accompanied by morphological changes in the spleen of DOX-treated rats. Co-treatment with QUR abated most of the DOX-mediated alterations in hematological variables, serum immunoglobulins, and spleen antioxidant status, pro-inflammatory and apoptotic responses, and histopathological alterations. In essence, these data suggest that QUR alleviated DOX-induced toxicities on the bone marrow, spleen, and antibody-producing cells. Supplementation of chemotherapy patients with QUR could circumvent the DOX-induced inflammation and immunotoxicity, and thus prevent chemotherapy failure.
Collapse
Affiliation(s)
- Mayada R. Farag
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.R.F.); (A.D.C.)
| | - Attia A. A. Moselhy
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt;
| | - Amany El-Mleeh
- Department of Pharmacology, Faculty of Veterinary Medicine, Menoufia University, Shebin Elkoum 32511, Egypt;
| | - Samira H. Aljuaydi
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Tamer Ahmed Ismail
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Alessandro Di Cerbo
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
- Correspondence: (M.R.F.); (A.D.C.)
| | - Giuseppe Crescenzo
- Department of Veterinary Medicine, University of Bari ‘Aldo Moro’, 70121 Bari, Italy;
| | - Shimaa M. Abou-Zeid
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City 6012201, Egypt;
| |
Collapse
|
13
|
Johnson BA, Lindgren BR, Blaes AH, Parsons HM, LaRocca CJ, Farah R, Hui JYC. The New Normal? Patient Satisfaction and Usability of Telemedicine in Breast Cancer Care. Ann Surg Oncol 2021; 28:5668-5676. [PMID: 34275045 PMCID: PMC8286165 DOI: 10.1245/s10434-021-10448-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Telemedicine was adopted to minimize exposure risks for patients and staff during the coronavirus disease 2019 pandemic. This study measured patient satisfaction and telemedicine usability in breast cancer care. METHODS Adult breast cancer patients who had a telemedicine visit at a single academic institution (with surgical, radiation, or medical oncology) from 15 June 2020 to 4 September 2020 were surveyed anonymously. Patient and cancer characteristics were collected, and patient satisfaction and telemedicine usability were assessed using a modified Telehealth Usability Questionnaire with a 7-point Likert scale. Associations of satisfaction and usability with patient characteristics were analyzed using Wilcoxon rank-sum and Kruskal-Wallis tests. RESULTS Of 203 patients who agreed to be contacted, 78 responded, yielding a response rate of 38%. The median age of the respondents was 63 years (range 25-83 years). The majority lived in an urban area (61%), were white (92%), and saw a medical oncologist (62%). The median patient satisfaction score was 5.5 (interquartile range [IQR] 4.25-6.25). The median telemedicine usability score was 5.6 (IQR 4.4-6.2). A strong positive correlation was seen between satisfaction and usability, with a Spearman correlation coefficient (ρ) of 0.80 (p < 0.001). Satisfaction and usability scores did not vary significantly according to patient age, race, location of residence, insurance status, previous visit commute time, oncology specialty seen, prior telemedicine visits, or whether patients were actively receiving cancer treatment. CONCLUSIONS Breast cancer patients were satisfied with telemedicine and found it usable. Patient satisfaction and telemedicine usability should not limit the use of telemedicine in future post-pandemic breast cancer care.
Collapse
Affiliation(s)
- Bryan A Johnson
- University of Minnesota Medical School, Minneapolis, MN, USA
| | - Bruce R Lindgren
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Anne H Blaes
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Helen M Parsons
- Division of Health Policy and Management, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Christopher J LaRocca
- Division of Surgical Oncology, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Ronda Farah
- Department of Dermatology, University of Minnesota, Minneapolis, MN, USA
| | - Jane Yuet Ching Hui
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Division of Surgical Oncology, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
14
|
Lowder LL, Powell M, Miller SE, Kishton RJ, Kelly CB, Cribb CB, Mastro-Kishton K, Chelvanambi M, Do PT, Govindapur RR, Wardell SE, McDonnell DP, Bartolotti LJ, Akkaraju GR, Frampton AR, Varadarajan S. Mechanistic Investigation of Site-specific DNA Methylating Agents Targeting Breast Cancer Cells. J Med Chem 2021; 64:12651-12669. [PMID: 34415160 DOI: 10.1021/acs.jmedchem.1c00615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We previously described the development of a DNA-alkylating compound that showed selective toxicity in breast cancer cells. This compound contained an estrogen receptor α (ERα)-binding ligand and a DNA-binding/methylating component that could selectively methylate the N3-position of adenines at adenine-thymine rich regions of DNA. Herein, we describe mechanistic investigations that demonstrate that this class of compounds facilitate the translocation of the ERα-compound complex to the nucleus and induce the expression of ERα target genes. We confirm that the compounds show selective toxicity in ERα-expressing cells, induce ERα localization in the nucleus, and verify the essential role of ERα in modulating the toxicity. Minor alterations in the compound structure significantly affects the DNA binding ability, which correlates to the DNA-methylating ability. These studies demonstrate the utility of DNA-alkylating compounds to accomplish targeted inhibition of the growth of specific cancer cells; an approach that may overcome shortcomings of currently used chemotherapy agents.
Collapse
Affiliation(s)
- Leah L Lowder
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Matthew Powell
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Sean E Miller
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Rigel J Kishton
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Charles B Kelly
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Connor B Cribb
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Kelly Mastro-Kishton
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Manoj Chelvanambi
- Department of Biology, Texas Christian University, Fort Worth, Texas 76129, United States
| | - Phat T Do
- Department of Biology, Texas Christian University, Fort Worth, Texas 76129, United States
| | - Rajeshwar Reddy Govindapur
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Libero J Bartolotti
- Department of Chemistry, East Carolina University, Greenville, North Carolina 27858, United States
| | - Giridhar R Akkaraju
- Department of Biology, Texas Christian University, Fort Worth, Texas 76129, United States
| | - Arthur R Frampton
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| | - Sridhar Varadarajan
- Department of Chemistry and Biochemistry, University of North Carolina Wilmington, Wilmington, North Carolina 28403, United States
| |
Collapse
|
15
|
The high dose of vitamin D supplementation combined with yoga training improve the leukocytes cell survival-related gene expression in breast cancer survivors. Nutr Metab (Lond) 2021; 18:80. [PMID: 34454533 PMCID: PMC8403369 DOI: 10.1186/s12986-021-00607-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study aimed to examine the effect of yoga training combined with vitamin D supplementation on the expression of survival-related genes in leukocytes and psycho-physical status in breast cancer survivors. METHODS Thirty breast cancer survivor women (age, 48 ± 8 yrs) were randomly assigned into three groups: high dose (4000 IU) of vitamin D supplementation (HD) (n = 10); yoga training with a high dose of vitamin D (Y + HD); (n = 10); yoga training with a low dose (2000 IU) of vitamin D (Y + LD) (n = 10). Participants performed the Hatha yoga style twice a week. Blood samples and a battery of psychological and physical tests were taken before and after the completion of interventions. Expression of p53, NF-κB, Bcl2, and Bax genes was measured in leukocytes. RESULTS Body fat percentage (ηp2 = 0.36), shoulder flexibility (ηp2 = 0.38), Rockport walk tests (ηp2 = 0.49), and anxiety (ηp2 = 0.52) were significantly improved in both the Y + HD and Y + LD groups compared to the HD group (p < 0.05). P53 was significantly over-expressed in the Y + HD group while Bcl2 upregulated in both the Y + HD and Y + LD groups. NF-κB and Bax expression downregulated in all groups but were not statistically significant. CONCLUSION yoga training combined with low and high doses of VD improved physical fitness and psychological measures while only in combination with a high dose of VD positively modified the leukocytes cell survival-related gene expression.
Collapse
|
16
|
Zhu H, Liu X. Advances of Tumorigenesis, Diagnosis at Early Stage, and Cellular Immunotherapy in Gastrointestinal Malignancies. Front Oncol 2021; 11:666340. [PMID: 34434889 PMCID: PMC8381364 DOI: 10.3389/fonc.2021.666340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023] Open
Abstract
Globally, in 2018, 4.8 million new patients have a diagnosis of gastrointestinal (GI) cancers, while 3.4 million people died of such disorders. GI malignancies are tightly relevant to 26% of the world-wide cancer incidence and occupies 35% of all cancer-associated deaths. In this article, we principally investigated molecular and cellular mechanisms of tumorigenesis in five major GI cancers occurring at esophagus, stomach, liver, pancreas, and colorectal region that illustrate high morbidity in Eastern and Western countries. Moreover, through this investigation, we not only emphasize importance of the tumor microenvironment in development and treatment of malignant tumors but also identify significance of M2PK, miRNAs, ctDNAs, circRNAs, and CTCs in early detection of GI cancers, as well as systematically evaluate contribution of personalized precision medicine including cellular immunotherapy, new antigen and vaccine therapy, and oncolytic virotherapy in treatment of GI cancers.
Collapse
Affiliation(s)
- Haipeng Zhu
- Precision and Personalized Cancer Treatment Center, Division of Cancer Diagnosis & Therapy, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China.,Stem Cell and Biotherapy Technology Research Center, Xinxiang Medical College, Xinxiang, China
| | - Xiaojun Liu
- Division of Cellular & Biomedical Science, Ciming Boao International Hospital, Boao Lecheng International Medical Tourism Pilot Zone, Qionghai, China
| |
Collapse
|
17
|
Suwanpakdee D, Taweesith W, Traivaree C, Rujkijyanont P. HBV Seroprotection and Anamnestic Response to Booster Vaccination in Pediatric Cancer Survivors. Glob Pediatr Health 2021; 8:2333794X211033452. [PMID: 34350309 PMCID: PMC8287357 DOI: 10.1177/2333794x211033452] [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: 05/17/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022] Open
Abstract
Hepatitis B is a major global health concern and can be prevented in the era of
vaccination. Impaired immunological memory to primary immunization is a common
chemotherapy-related complication among cancer survivors. The study aimed to
determine protective immunity against hepatitis B virus (HBV) and anamnestic
response to booster vaccination. In all, 107 pediatric cancer survivors
previously immunized with primary hepatitis B vaccination were enrolled. A
hepatitis B booster dose was administered to those with suboptimal
seroprotection (anti-HBs < 10 mIU/mL) and 2 additional doses were
subsequently administered at 1 and 6 months to those whose anti-HBs remained
low. Clinical and serologic parameters were analyzed. Sero-protective rate
against HBV (anti-HBs ≥ 10 mIU/mL) among survivors was 20.6% with geometric mean
titer (GMT) of 95.7 ± 265.6 mIU/mL. Anamnestic response was 61% after a booster
vaccine among those with suboptimal seroprotection and 100% after 2 additional
booster doses among those whose anti-HBs remained low. GMTs among those
survivors after the First and third booster vaccines were 320.0 ± 412.4 mIU/mL
and 826.5 ± 343.8 mIU/mL, respectively. Age at diagnosis was a significant
independent risk factor for adequate seroprotection (adjusted OR = 0.84, 95%CI:
0.71-0.99) with a P-value of .034. No associated risk factors
to predict optimal anamnestic response to booster vaccination were identified.
Loss of immunological memory to primary hepatitis B immunization is an
inevitable complication among most pediatric cancer survivors; therefore,
assessing adequate seroprotection is essentially required. For those with
limited accessibility to serologic tests, completion of full 3-booster-dose
series is alternative and highly recommended.
Collapse
Affiliation(s)
| | | | - Chanchai Traivaree
- Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Piya Rujkijyanont
- Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| |
Collapse
|
18
|
The Combinatorial Effect of Cisplatin and Moxibustion on Tumor Growth Inhibition with Special Reference to Modulation of the Immune Microenvironment in Lewis Lung Cancer Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2020:3170803. [PMID: 33456484 PMCID: PMC7785363 DOI: 10.1155/2020/3170803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/08/2020] [Accepted: 11/27/2020] [Indexed: 12/15/2022]
Abstract
Objective As a first-line treatment for non-small cell lung cancer (NSCLC), the efficacy of chemotherapy is still unsatisfactory. Moxibustion has been shown to improve the side effects of radiotherapy and chemotherapy and regulate immune function. This study aimed to explore the antitumor effects and potential mechanisms of combinatorial cisplatin and moxibustion treatment for NSCLC by targeting the tumor microenvironment. Methods Lewis lung cancer (LLC)-bearing mice were induced and treated with cisplatin or/and moxibustion at ST36 (Zusanli), and the growth, weight, and area of the tumor were evaluated. The numbers of various T cell subsets and myeloid cells in the tumor were assessed by flow cytometry, and the gene expression of related markers and cytokines was detected with real-time quantitative polymerase chain reaction (RT-qPCR). In addition, the tumor vascular structure was investigated using CD31 and α-smooth muscle actin (α-SMA) immunofluorescence staining. The expression of the vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α) was detected by immunohistochemical staining. Results Both cisplatin and moxibustion inhibited LLC tumor growth and reduced both the tumor area and weight, with the combinatorial therapy showing superior outcomes. Moxibustion upregulated the infiltration of CD4+ T cells and Th1 cells in the tumor, and the combinatorial therapy increased the proportion of CD8+ cytotoxic T cells (CTLs), CD4+T cells, Th1, Th9 cells, and M1 macrophages, as well as the expression of Cd69, Ifng, and Cd86 mRNA. The combinatorial therapy improved vascular normalization by increasing both the microvessel density (MVD) and pericyte coverage (α-SMA area density) and inhibiting the expression of the VEGF. Conclusions Combinatorial cisplatin and moxibustion treatment inhibited the LLC tumor growth by mechanisms related to the improvement of the tumor immune microenvironment and vascular normalization, providing an effective combinatorial therapy beneficial for patients with NSCLC.
Collapse
|
19
|
Chen X, Zhang Z, Hou X, Zhang Y, Zhou T, Liu J, Lin Z, Fang W, Yang Y, Ma Y, Huang Y, Zhao H, Zhang L. Immune-related pneumonitis associated with immune checkpoint inhibitors in lung cancer: a network meta-analysis. J Immunother Cancer 2020; 8:jitc-2020-001170. [PMID: 32863271 PMCID: PMC7462235 DOI: 10.1136/jitc-2020-001170] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2020] [Indexed: 12/30/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have dramatically revolutionized lung cancer treatment, providing unprecedented clinical benefits. However, immune-related pneumonitis (IRP) caused by ICIs has aroused widespread concern due to its high rate of discontinuation and mortality. This network meta-analysis (NMA) aims to compare the risks of IRP among different regimens for advanced lung cancer. Methods Phase II and III randomized clinical trials (RCTs) were searched from electronic databases. The rates of grade 1–5 IRP and grade 3–5 IRP were systematically extracted. An NMA was conducted among chemotherapy, ICIs monotherapy, dual ICIs combination, and ICIs+chemotherapy. Subgroup analysis was also compared based on specific types of ICIs. Results Twenty-five RCTs involving 17,310 patients were eligible for inclusion. Compared with chemotherapy, ICI-based regimens were associated with an increased risk of grade 1–5 IRP and grade 3–5 IRP. Compared with ICIs+chemotherapy, ICIs monotherapy (grade 1–5: OR 2.14, 95% credible interval 1.12 to 4.80; grade 3–5: 3.03, 1.491 to 6.69) and dual ICIs combination (grade 1–5: 3.86, 1.69 to 9.89; grade 3–5: 5.12, 2.01 to 13.68) were associated with a higher risk of grade 1–5 IRP and grade 3–5 IRP. No significant difference was found between dual ICIs combination and ICIs monotherapy in grade 1–5 IRP (1.85, 0.91 to 3.37) or in grade 3–5 IRP (1.65, 0.81 to 3.37). Besides, compared with programmed cell death protein 1 (PD-1) inhibitors (2.56, 1.12 to 6.60), a lower risk of grade 1–5 IRP was observed in programmed cell death ligand 1 (PD-L1) inhibitors. Conclusion Compared with chemotherapy, using ICIs is associated with an increased risk of IRP. ICIs+chemotherapy is associated with a lower risk of IRP compared with dual ICIs combination and ICIs monotherapy. PD-1 inhibitors are associated with a higher risk of 1–5 grade IRP compared with PD-L1 inhibitors.
Collapse
Affiliation(s)
- Xinru Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhonghan Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Xue Hou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yaxiong Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ting Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jiaqing Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Zhihuan Lin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yuxiang Ma
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Tan LTH, Chan CK, Chan KG, Pusparajah P, Khan TM, Ser HL, Lee LH, Goh BH. Streptomyces sp. MUM256: A Source for Apoptosis Inducing and Cell Cycle-Arresting Bioactive Compounds against Colon Cancer Cells. Cancers (Basel) 2019; 11:E1742. [PMID: 31698795 PMCID: PMC6896111 DOI: 10.3390/cancers11111742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 10/25/2019] [Accepted: 10/25/2019] [Indexed: 12/23/2022] Open
Abstract
New and effective anticancer compounds are much needed as the incidence of cancer continues to rise. Microorganisms from a variety of environments are promising sources of new drugs; Streptomyces sp. MUM256, which was isolated from mangrove soil in Malaysia as part of our ongoing efforts to study mangrove resources, was shown to produce bioactive metabolites with chemopreventive potential. This present study is a continuation of our previous efforts and aimed to investigate the underlying mechanisms of the ethyl acetate fraction of MUM256 crude extract (MUM256 EA) in inhibiting the proliferation of HCT116 cells. Our data showed that MUM256 EA reduced proliferation of HCT116 cells via induction of cell-cycle arrest. Molecular studies revealed that MUM256 EA regulated the expression level of several important cell-cycle regulatory proteins. The results also demonstrated that MUM256 EA induced apoptosis in HCT116 cells mediated through the intrinsic pathway. Gas chromatography-mass spectrometry (GC-MS) analysis detected several chemical compounds present in MUM256 EA, including cyclic dipeptides which previous literature has reported to demonstrate various pharmacological properties. The cyclic dipeptides were further shown to inhibit HCT116 cells while exerting little to no toxicity on normal colon cells in this study. Taken together, the findings of this project highlight the important role of exploring the mangrove microorganisms as a bioresource which hold tremendous promise for the development of chemopreventive drugs against colorectal cancer.
Collapse
Affiliation(s)
- Loh Teng-Hern Tan
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; or (L.T.-H.T.); (H.-L.S.)
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Chim-Kei Chan
- de Duve Institute, Université catholique de Louvain, Avenue Hippocrate 74, 1200 Brussels, Belgium;
| | - Kok-Gan Chan
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Priyia Pusparajah
- Medical Health and Translational Research Group, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia;
| | - Tahir Mehmood Khan
- Institute of Pharmaceutical Science, University of Veterinary and Animal Science Lahore, Punjab 54000, Pakistan;
| | - Hooi-Leng Ser
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; or (L.T.-H.T.); (H.-L.S.)
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia; or (L.T.-H.T.); (H.-L.S.)
- Institute of Pharmaceutical Science, University of Veterinary and Animal Science Lahore, Punjab 54000, Pakistan;
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bey-Hing Goh
- Institute of Pharmaceutical Science, University of Veterinary and Animal Science Lahore, Punjab 54000, Pakistan;
- Health and Well-Being Cluster, Global Asia in the 21st Century (GA21) Platform, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
21
|
Kim J, Manspeaker MP, Thomas SN. Augmenting the synergies of chemotherapy and immunotherapy through drug delivery. Acta Biomater 2019; 88:1-14. [PMID: 30769136 DOI: 10.1016/j.actbio.2019.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/25/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
Abstract
Despite the recent approvals of multiple cancer immunotherapies, low tumor immunogenicity and immunosuppressive tumor microenvironments prevent a large portion of patients from responding to these treatment modalities. Given the immunomodulatory and adjuvant effects of conventional chemotherapy as well as its widespread clinical use, the use of chemotherapy in combination with immunotherapy (so-called chemoimmunotherapy) is an attractive approach to potentiate the effects of immunotherapy in more patient populations. However, due to the limited extent of tumor accumulation, poorly controlled interactions with the immune system, and effects on systemic healthy tissues by chemotherapeutic drugs, the incorporation of anti-cancer agents into biomaterial-based structures, such as nanocarriers, is highly attractive to improve the safety and efficacy of chemoimmunotherapy. Herein, we review the recent progress in drug delivery systems (DDSs) for potentiating the immunomodulatory effects of chemotherapeutics in chemoimmunotherapy, which represent among the most promising next generation strategies for cancer treatment in the immunotherapy era. STATEMENT OF SIGNIFICANCE: Given the benefits of cancer immunotherapy in inducing durable, albeit low rates, of patient response, interest in the immunomodulatory and adjuvant effects of conventional chemotherapy has been re-invigorated. This review article discusses the recent progress towards understanding the synergies between these two treatment types, how they can be used in combination (so-called chemoimmunotherapy), and the potential for drug delivery systems to optimize their effects in translational settings.
Collapse
|
22
|
Jeong J, Kim YJ, Lee DY, Moon BG, Sohn KY, Yoon SY, Kim JW. 1-Palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) attenuates gemcitabine-induced neutrophil extravasation. Cell Biosci 2019; 9:4. [PMID: 30622698 PMCID: PMC6317242 DOI: 10.1186/s13578-018-0266-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/24/2018] [Indexed: 11/10/2022] Open
Abstract
Cancer patients treated with chemotherapy often experience a rapid decline of blood neutrophils, a dose-limiting side effect called chemotherapy-induced neutropenia. This complication brings about dose reductions or cessation of chemotherapy during treatment of cancer patients because a rapid decline of neutrophil counts increases susceptibility to infection. Here, we found that 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) attenuates gemcitabine-induced neutrophil extravasation via the inhibition of neutrophil-attracting chemokine production in macrophages using in vivo and in vitro approaches. A single intraperitoneal administration of gemcitabine induced the migration of circulating neutrophils into the peritoneal cavity in normal mice, and PLAG effectively decreased neutrophil migration by inhibiting the expression of adhesion molecules, L-selectin and LFA-1. Inhibition of CXCR2 by its antagonist, reparixin, abrogated gemcitabine-induced neutrophil migration, indicating that chemokines produced by gemcitabine mainly support neutrophil activation. In vitro experiments demonstrated that PLAG inhibited NADPH oxidase 2 (NOX2)-mediated reactive oxygen species production induced by gemcitabine, which is the upstream of MIP-2 and/or CXCL8. Importantly, PLAG down-regulated gemcitabine-induced membrane translocation of the cytosolic NOX subunit, Rac1, and phosphorylation of p47phox. The activation of upstream signaling molecules of p47phox phosphorylation, phospholipase C β3 and protein kinase C, were effectively regulated by PLAG. We also demonstrated that 1-palmitoyl-2-linoleic-3-hydroxyl-rac-glycerol (PLH), the natural form of diacylglycerol, has no effects on gemcitabine-induced CXCL8 production and dHL-60 migration, suggesting that an acetyl group at the third position of the glycerol backbone may have a key role in the regulation of neutrophil activation. Altogether, this study suggests the potential of PLAG as a therapeutic strategy to modulate chemotherapy-induced neutrophil activation for cancer patients undergoing chemotherapeutic treatment.
Collapse
Affiliation(s)
- Jinseon Jeong
- 1Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-333 Republic of Korea.,2Department of Functional Genomics, University of Science & Technology, Daejeon, Republic of Korea.,Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Yong-Jae Kim
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Do Young Lee
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Byoung-Gon Moon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Ki-Young Sohn
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Sun Young Yoon
- Division of Global New Drug Development, ENZYCHEM Lifesciences, Jecheon, 27159 Republic of Korea
| | - Jae Wha Kim
- 1Cell Factory Research Center, Division of Systems Biology and Bioengineering, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 305-333 Republic of Korea.,2Department of Functional Genomics, University of Science & Technology, Daejeon, Republic of Korea
| |
Collapse
|
23
|
Xiao Z, Wang CQ, Feng JH, Zhou MH, Wang YZ, Li NN, Sun YP, Liu SY, Yao XS, Li CW, Ma B, Ding J, Chen L. Effectiveness and safety of chemotherapy with cytokine-induced killer cells in non-small cell lung cancer: A systematic review and meta-analysis of 32 randomized controlled trials. Cytotherapy 2018; 21:125-147. [PMID: 30554868 DOI: 10.1016/j.jcyt.2018.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND AIMS Cytokine-induced killer (CIK) cells are the most commonly used cellular immunotherapy for multiple tumors. To further confirm whether chemotherapy with CIK cells improves clinical effectiveness and to reveal its optimal use in non-small cell lung cancer (NSCLC), we systematically reevaluated all relevant studies. METHODS We collected all studies about chemotherapy with CIK cells for NSCLC from the Medline, Embase, Web of Science, China National Knowledge Infrastructure Database (CNKI), Chinese Scientific Journals Full-Text Database (VIP), Wanfang Data, China Biological Medicine Database (CBM), Cochrane Central Register of Controlled Trials (CENTRAL), Chinese clinical trial registry (Chi-CTR), World Health Organization (WHO) International Clinical Trials Registry Platform (ICTRP) and U.S. clinical trials. We evaluated their quality according to the Cochrane evaluation handbook of randomized controlled trials (RCTs) (version 5.1.0), extracted the data using a standard data extraction form, synthesized the data using meta-analysis and finally rated the evidence quality using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. RESULTS Thirty-two RCTs with 2250 patients were included, and most trials had unclear risk of bias. The merged risk ratios values and their 95% confidence intervals of meta-analysis for objective response rate, disease control rate, 1- and 2-year overall survival rates, 1- and 2-year progression-free survival rates were as following: 1.45 (1.31-1.61), 1.26 (1.16-.37), 1.42 (1.23-1.63), 2.06 (1.36-3.12), 1.93 (1.38-2.69) and 3.30 (1.13-9.67). Compared with chemotherapy alone, all differences were statistically significant. CIK cells could increase the CD3+ T cells, CD3+ CD4+ T cells, NK cells and the ratio of CD4+/CD8+ T cells. The chemotherapy with CIK cells had a lower risk of hematotoxicity, gastrointestinal toxicity, liver injury and a higher fever than that of chemotherapy alone. The evidence quality was "moderate" to "very low." CONCLUSIONS The available moderate evidences indicate that chemotherapy with CIK cells, especially autologous CIK cells, can significantly improve the tumor responses, 1- and 2-year overall and progression-free survival rates in patients with advanced NSCLC. This treatment does have a high risk of fever. The optimal use may be treatment with one or two cycles and in combination with vinorelbine and cisplatin, paclitaxel and cisplatin, or docetaxel and cisplatin.
Collapse
Affiliation(s)
- Zheng Xiao
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of major infectious diseases), Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China.
| | - Cheng-Qiong Wang
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of major infectious diseases), Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ji-Hong Feng
- Department of Oncology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Ming-Hua Zhou
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yu-Zhi Wang
- Department of Immunology, Southwest Medical University, Luzhou, Sichuan, China
| | - Na-Na Li
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of major infectious diseases), Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yong-Ping Sun
- Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Shi-Yu Liu
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xin-Sheng Yao
- Department of Immunology, Zunyi Medical University, Zunyi,Guizhou, China
| | - Cheng-Wen Li
- Department of Immunology, Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Ma
- Evidence-Based Medicine Center of Lanzhou University (School of Basic Medical Sciences), Lanzhou University, Lanzhou 730000, Gansu, China
| | - Jie Ding
- Outpatient Department of Psychological Counseling Clinic (Center for Evidence-Based and Translational Medicine of major infectious diseases), Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Ling Chen
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of major infectious diseases), Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
24
|
Xiao Z, Wang CQ, Zhou MH, Li NN, Sun YP, Wang YZ, Liu SY, Yu HS, Li CW, Zeng XT, Chen L, Yao XS, Feng JH. The Antitumor Immunity and Tumor Responses of Chemotherapy with or without DC-CIK for Non-Small-Cell Lung Cancer in China: A Meta-Analysis of 28 Randomized Controlled Trials. J Immunol Res 2018; 2018:9081938. [PMID: 30648123 PMCID: PMC6311820 DOI: 10.1155/2018/9081938] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/04/2018] [Accepted: 10/03/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE DC-CIK therapy included DC-CIK cells and Ag-DC-CIK cells. To further confirm whether DC-CIK reconstructs the antitumor immunity and improves the tumor responses and reveals its optimal usage and combination with chemotherapy, we systematically reevaluated all the related studies. MATERIALS AND METHODS All studies about DC-CIK plus chemotherapy for NSCLC were collected from the published and ongoing database as CBM, CNKI, VIP, Wanfang, ISI, Embase, MEDLINE, CENTRAL, WHO-ICTRP, Chi-CTR, and US clinical trials (established on June 2017). We evaluated their methodological bias risk according to the Cochrane evaluation handbook of RCTs (5.1.0), extracted data following the predesigned data extraction form, and synthesized the data using meta-analysis. RESULTS We included 28 RCTs (phase IV) with 2242 patients, but most trials had unclear bias risk. The SMD and 95% CI of meta-analysis for CD3+ T cells, CD3+ CD4+ T cells, CD3+ CD8+ T cells, CD4+/CD8+ T cell ratio, CIK cells, NK cells, and Treg cells were as follows: 1.85 (1.39 to 2.31), 0.87 (0.65 to 1.10), 1.04 (0.58 to 1.50), 0.75 (0.27 to 1.22), 3.87 (2.48 to 5.25), 1.51 (0.99 to 2.03), and -2.31(-3.84 to -0.79). The RR and 95% CI of meta-analysis for ORR and DCR were as follows: 1.38 (1.24 to 1.54) and 1.27 (1.20 to 1.34). All differences were statistically significant between DC-CIK plus chemotherapy and chemotherapy alone. Subgroup analysis showed that only DC-CIK cells could increase the CD3+T cells, CD3+ CD4+T cells, CD3+ CD8+T cells, and CD4+/CD8+ T cell ratio. In treatment with one cycle or two cycles and combination with NP or GP, DC-CIK could increase the CD4+/CD8+ T cell ratio. All results had good stability. CONCLUSIONS DC-CIK therapy can simultaneously improve the antitumor immunity and tumor responses. DC-CIK therapy, especially DC-CIK cells, can improve antitumor immunity through increasing the T lymphocyte subsets, CIK cell, and NK cells in peripheral blood. The one cycle to two cycles may be optimal cycle, and the NP or GP may be optimal combination.
Collapse
Affiliation(s)
- Zheng Xiao
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-Based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
- Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of Major Infectious Diseases), Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Cheng-qiong Wang
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-Based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
- Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of Major Infectious Diseases), Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Ming-hua Zhou
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-Based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Na-na Li
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-Based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
- Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of Major Infectious Diseases), Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Yong-ping Sun
- Teaching and Research Group of Evidence-based Medicine, Zhuhai Campus of Zunyi Medical College, Zhuhai, 519000 Guangdong, China
| | - Yu-zhi Wang
- Department of immunology, Southwest Medical University, Luzhou, 646000 Sichuan, China
| | - Shi-yu Liu
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-Based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Hong-song Yu
- Department of Immunology, Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Cheng-wen Li
- Department of immunology, Southwest Medical University, Luzhou, 646000 Sichuan, China
| | - Xian-tao Zeng
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071 Hubei, China
| | - Ling Chen
- Evidence-Based Medicine Center, MOE Virtual Research Center of Evidence-Based Medicine at Zunyi Medical College, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
- Department of Respiratory Medicine (Center for Evidence-Based and Translational Medicine of Major Infectious Diseases), Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Xin-sheng Yao
- Department of Immunology, Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical College, Zunyi, 563000 Guizhou, China
| | - Ji-hong Feng
- Department of Oncology, Affiliated Hospital of Zunyi Medical College, Zunyi, 563000 Guizhou, China
| |
Collapse
|
25
|
Zhang J, Du Z, Bi J, Wu Z, Lv Y, Zhang X, Wu R. The impact of previous abdominal surgery on clinical characteristics and prognosis of pyogenic liver abscess: A 10-year retrospective study of 392 patients. Medicine (Baltimore) 2018; 97:e12290. [PMID: 30278501 PMCID: PMC6181538 DOI: 10.1097/md.0000000000012290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/17/2018] [Indexed: 01/24/2023] Open
Abstract
Many pyogenic liver abscess (PLA) patients underwent abdominal surgery before. However, little is known about the impact of previous abdominal surgery on the clinical characteristics and prognosis of PLA.The clinical data of 392 adult PLA patients who received treatment at our hospital from January 1, 2007 to December 31, 2016 were collected. The demographic data, cause, comorbidities, surgery history, clinical features, laboratory results, imaging findings, microbiological characteristics, choices of treatment, and clinical outcomes were analyzed.In all, 177 PLA patients (45.2%) underwent abdominal surgery before. The median time for the occurrence of PLA after the most recent abdominal surgery was 2.0 (interquartile range 0.25, 6.0) years. PLA patients with a previous abdominal surgery history were more likely to have underlying diseases and presented with more abnormal laboratory values. Klebsiella pneumonia and Escherichia coli were the most common pathogens. Previous abdominal surgery appeared to increase the incidence of E coli. More PLA patients without a previous abdominal surgery history required surgical drainage. However, there were no differences in PLA-related complications, days required for temperature normalization, and length of hospital stay between the 2 groups.Because a large number of PLA patients had a history of abdominal surgery, and proper screening should be performed for patients with any suspicion of a liver abscess after abdominal surgery. Despite the differences in the coexisting conditions, clinical and microbiological characteristics between PLA patients with and without a previous abdominal surgery history, the overall short-term outcomes were comparable.
Collapse
Affiliation(s)
- Jia Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
- Institute of Advanced Surgical Technology and Engineering
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Zhaoqing Du
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
- Institute of Advanced Surgical Technology and Engineering
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Jianbin Bi
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
- Institute of Advanced Surgical Technology and Engineering
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
- Institute of Advanced Surgical Technology and Engineering
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Xufeng Zhang
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
- Institute of Advanced Surgical Technology and Engineering
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering
- Institute of Advanced Surgical Technology and Engineering
| |
Collapse
|
26
|
Jia X, Liang Y, Zhang C, Wang K, Tu Y, Chen M, Li P, Wan JB, He C. Polysaccharide PRM3 from Rhynchosia minima root enhances immune function through TLR4-NF-κB pathway. Biochim Biophys Acta Gen Subj 2018; 1862:1751-1759. [DOI: 10.1016/j.bbagen.2018.05.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 04/29/2018] [Accepted: 05/11/2018] [Indexed: 12/20/2022]
|
27
|
Effects of combined and resistance training on the inflammatory profile in breast cancer survivors: A systematic review. Complement Ther Med 2018; 36:73-81. [DOI: 10.1016/j.ctim.2017.11.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 12/18/2022] Open
|
28
|
Theunissen PMJ, van den Branden A, Van Der Sluijs-Gelling A, De Haas V, Beishuizen A, van Dongen JJM, Van Der Velden VHJ. Understanding the reconstitution of the B-cell compartment in bone marrow and blood after treatment for B-cell precursor acute lymphoblastic leukaemia. Br J Haematol 2017; 178:267-278. [PMID: 28542787 DOI: 10.1111/bjh.14685] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/16/2017] [Indexed: 01/08/2023]
Abstract
A better understanding of the reconstitution of the B-cell compartment during and after treatment in B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) will help to assess the immunological status and needs of post-treatment BCP-ALL patients. Using 8-colour flow cytometry and proliferation-assays, we studied the composition and proliferation of both the B-cell precursor (BCP) population in the bone marrow (BM) and mature B-cell population in peripheral blood (PB) during and after BCP-ALL therapy. We found a normal BCP differentiation pattern and a delayed formation of classical CD38dim -naive mature B-cells, natural effector B-cells and memory B-cells in patients after chemotherapy. This B-cell differentiation/maturation pattern was strikingly similar to that during initial B-cell development in healthy infants. Tissue-resident plasma cells appeared to be partly protected from chemotherapy. Also, we found that the fast recovery of naive mature B-cell numbers after chemotherapy was the result of increased de novo BCP generation, rather than enhanced B-cell proliferation in BM or PB. These results indicate that post-treatment BCP-ALL patients will eventually re-establish a B-cell compartment with a composition and B-cell receptor repertoire similar to that in healthy children. Additionally, the formation of a new memory B-cell compartment suggests that revaccination might be beneficial after BCP-ALL therapy.
Collapse
Affiliation(s)
- Prisca M J Theunissen
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Anouk van den Branden
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | | | - Auke Beishuizen
- Department of Paediatric Haematology and Oncology, Sophia Children's Hospital/Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Jacques J M van Dongen
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|
29
|
Zhang Z, Yu X, Wang Z, Wu P, Huang J. Anthracyclines potentiate anti-tumor immunity: A new opportunity for chemoimmunotherapy. Cancer Lett 2015; 369:331-5. [PMID: 26454214 DOI: 10.1016/j.canlet.2015.10.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023]
Abstract
Anthracyclines are a class of drugs, including doxorubicin, epirubicin and idarubicin, used in cancer chemotherapy which are derived from Streptomyces bacterium Streptomyces peucetius var. caesius. Traditionally, substantial pieces of evidence have demonstrated that anthracyclines could harness the host immune system to prevent cancer progression. But nowadays, researches also implied that anthracyclines could sensitize tumor cells to immune cell driven cytotoxicity, like dendritic cells and CD8+ T cell. The ability of anthracyclines in tumor immune cycle, including trigger direct tumor cell death, enhance immune effector cell activation and eliminate immunosuppressive myeloid-derived suppressor cells (MDSCs), explained its capacity to relieve tumor induced immunosuppression and restore anticancer immune responses. And current pre-clinical and clinical trials implied that combination therapies using anthracyclines with immunotherapy have further enhanced the clinical benefit. Here, we discuss how the increased understanding of the immune-driven effects of anthracyclines prompts the design of relevant cancer chemoimmunotherapy strategies.
Collapse
Affiliation(s)
- Zhigang Zhang
- Department of Gynecology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | | | - Zhen Wang
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Pin Wu
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jian Huang
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education, Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; Department of Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
30
|
Iyer JG, Parvathaneni U, Gooley T, Miller NJ, Markowitz E, Blom A, Lewis CW, Doumani RF, Parvathaneni K, Anderson A, Bestick A, Liao J, Kane G, Bhatia S, Paulson K, Nghiem P. Single-fraction radiation therapy in patients with metastatic Merkel cell carcinoma. Cancer Med 2015; 4:1161-70. [PMID: 25908228 PMCID: PMC4559027 DOI: 10.1002/cam4.458] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/27/2015] [Accepted: 03/08/2015] [Indexed: 12/20/2022] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive, polyomavirus-associated cancer with limited therapeutic options for metastatic disease. Cytotoxic chemotherapy is associated with high response rates, but responses are seldom durable and toxicity is considerable. Here, we report our experience with palliative single-fraction radiotherapy (SFRT) in patients with metastatic MCC. We conducted retrospective analyses of safety and efficacy outcomes in patients that received SFRT (8 Gy) to MCC metastases between 2010 and 2013. Twenty-six patients were treated with SFRT to 93 MCC tumors located in diverse sites that included skin, lymph nodes, and visceral organs. Objective responses were observed in 94% of the measurable irradiated tumors (86/92). Complete responses were observed in 45% of tumors (including bulky tumors up to 16 cm). “In field” lesion control was durable with no progression in 77% (69/89) of treated tumors during median follow-up of 277 days among 16 living patients. Clinically significant toxicity was seen in only two patients who had transient side effects. An exploratory analysis suggested a higher rate of in-field progression in patients with an immunosuppressive comorbidity or prior recent chemotherapy versus those without (30% and 9%, respectively; P = 0.03). Use of SFRT in palliating MCC patients was associated with an excellent in field control rate and durable responses at treated sites, and with minimal toxicity. SFRT may represent a convenient and appealing alternative to systemic chemotherapy for palliation, for which most patients with oligometastatic MCC are eligible. SFRT may also synergize with emerging systemic immune stimulants by lowering tumor burden and enhancing presentation of viral/tumor antigens.
Collapse
Affiliation(s)
- Jayasri G Iyer
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | | | - Ted Gooley
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Natalie J Miller
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Elan Markowitz
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Astrid Blom
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Christopher W Lewis
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Ryan F Doumani
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Kaushik Parvathaneni
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Austin Anderson
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Amy Bestick
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Jay Liao
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Gabrielle Kane
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Shailender Bhatia
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,Seattle Cancer Care Alliance, Seattle, Washington.,Department of Medicine/Oncology, University of Washington, Seattle, Washington
| | - Kelly Paulson
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington
| | - Paul Nghiem
- Department of Medicine/Dermatology, University of Washington, Seattle, Washington.,Fred Hutchinson Cancer Research Center, Seattle, Washington.,Seattle Cancer Care Alliance, Seattle, Washington
| |
Collapse
|
31
|
Samdani T, Pieracci FM, Eachempati SR, Benarroch-Gampel J, Weiss A, Pietanza MC, Barie PS, Nash GM. Colonic diverticulitis in chemotherapy patients: should operative indications change? A retrospective cohort study. Int J Surg 2014; 12:1489-94. [PMID: 25448673 DOI: 10.1016/j.ijsu.2014.10.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 10/17/2014] [Accepted: 10/22/2014] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Management of the immunosuppressed patient with diverticular disease remains controversial. We report the largest series of colon cancer patients undergoing chemotherapy and hospitalized for acute diverticulitis, to determine whether recent treatment with systemic chemotherapy is associated with increased risk for/increased severity of recurrent diverticulitis. METHODS Retrospective cohort study of adult patients hospitalized for an initial episode of acute colonic diverticulitis at Memorial Sloan Kettering Cancer Center, 1988-2004. Outcomes in patients receiving systemic chemotherapy within one month of admission for diverticulitis ("Chemo") were compared to outcomes of patients not receiving chemotherapy within the past month ("No-chemo"). RESULTS A total 131 patients met inclusion criteria. Chemo patients did not differ significantly from No-chemo group in terms of severity of acute diverticulitis at index admission (13.2% vs. 4.4%, respectively, p = 0.12), resumption of chemotherapy (median 2 months), failure of non-operative management (13.2% vs 4.4%, respectively, p = 0.12), frequency of recurrence (20.5% vs 18.5%), hospital length of stay (p = 0.08), and likelihood of interval resection (24.0% vs. 16.2%, respectively, p = 0.39). Chemo patients recurred with more severe disease, were more likely to undergo emergent surgery (75.0% vs. 23.5%, respectively, p = 0.03), and were more likely to be diverted (100.0% vs. 25.0%, respectively, p = 0.03). Chemo patients were significantly more likely to incur a postoperative complication (100% vs 9.1% p < 0.01) following interval resection. Overall mortality was significantly higher in the Chemo vs. No-chemo group. Median survival in Chemo patients was 3.4 years; in No-chemo patients, median survival was not reached at 10 years. CONCLUSION Our data do not support routine elective surgery for acute diverticulitis in patients receiving chemotherapy. Non-operative management in the acute or interval setting appears preferable whenever possible.
Collapse
Affiliation(s)
- Tushar Samdani
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Fredric M Pieracci
- Departments of Surgery and Public Health, Weill Cornell Medical College, 445 East 69th Street, New York, NY 10065, USA
| | - Soumitra R Eachempati
- Departments of Surgery and Public Health, Weill Cornell Medical College, 445 East 69th Street, New York, NY 10065, USA
| | - Jaime Benarroch-Gampel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Alex Weiss
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - M Cathy Pietanza
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Philip S Barie
- Departments of Surgery and Public Health, Weill Cornell Medical College, 445 East 69th Street, New York, NY 10065, USA
| | - Garrett M Nash
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
32
|
Joshi VB, Geary SM, Gross BP, Wongrakpanich A, Norian LA, Salem AK. Tumor lysate-loaded biodegradable microparticles as cancer vaccines. Expert Rev Vaccines 2014; 13:9-15. [PMID: 24219096 DOI: 10.1586/14760584.2014.851606] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer vaccines that use tumor lysate (TL) as a source of tumor-associated antigens (TAAs) have significant potential for generating therapeutic anti-tumor immune responses. Vaccines encompassing TL bypass the limitations of single antigen vaccines by simultaneously stimulating immunity against multiple TAAs, thereby broadening the repertoire of TAA-specific T-cell clones available for activation. Administration of TL in particulate form, such as when encapsulated in biodegradable microparticles, increases its immunostimulatory capacity and produces more robust immune responses than when TL is given in soluble form. These effects can be further enhanced by co-administering TL with adjuvants. A number of recent studies using polymeric microparticle delivery of TL, with or without adjuvants, have produced promising results in preclinical studies. In this review, we will discuss current experimental approaches involving TL being pursued in the oncoimmunology field, and comment on strategies such as combining specific chemotherapeutic agents with TL microparticle delivery that may eventually lead to improved survival outcomes for cancer patients.
Collapse
Affiliation(s)
- Vijaya B Joshi
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
33
|
Merzoug S, Toumi ML, Tahraoui A. Quercetin mitigates Adriamycin-induced anxiety- and depression-like behaviors, immune dysfunction, and brain oxidative stress in rats. Naunyn Schmiedebergs Arch Pharmacol 2014; 387:921-33. [DOI: 10.1007/s00210-014-1008-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 06/10/2014] [Indexed: 12/11/2022]
|
34
|
Adriamycin-related anxiety-like behavior, brain oxidative stress and myelotoxicity in male Wistar rats. Pharmacol Biochem Behav 2011; 99:639-47. [DOI: 10.1016/j.pbb.2011.06.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 06/08/2011] [Accepted: 06/10/2011] [Indexed: 11/20/2022]
|
35
|
Faber J, van Limpt K, Kegler D, Luiking Y, Garssen J, van Helvoort A, Vos AP, Knol J. Bacterial translocation is reduced by a specific nutritional combination in mice with chemotherapy-induced neutropenia. J Nutr 2011; 141:1292-8. [PMID: 21562235 DOI: 10.3945/jn.110.136986] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Immune function is compromised in many cancer patients, leading to an increased risk of (infectious) complications. Chemotherapy-induced neutropenia is a common cause of treatment-induced immune suppression. In the present study, the effect of a specific nutritional combination (SNC) on bacterial translocation was studied in a model of chemotherapy-induced neutropenia in C3H/HeN mice colonized with Pseudomonas aeruginosa PAO-1. Dietary intervention started after stable colonization with P. aeruginosa to compare the SNC containing high protein, l-leucine, fish oil, and specific oligosaccharides to an isoenergetic control diet. After 3 wk, the mice were treated with cyclophosphamide to induce neutropenia. This rendered the mice susceptible to Pseudomonas translocation, which was quantified 5 d later. Intervention with the SNC resulted in a reduced incidence and intensity of bacterial translocation to the liver (P < 0.05) and a similar trend in the lungs (P ≤ 0.057). In addition, the SNC reduced the fecal pH (P < 0.05) and decreased P. aeruginosa counts in fecal samples (P < 0.05). Moreover, plasma concentrations of proinflammatory cytokines were correlated with the reduced bacterial translocation to the liver (ρ > 0.78; P < 0.001). In conclusion, dietary intervention with the SNC significantly reduced the incidence and severity of P. aeruginosa translocation in a mouse model of chemotherapy-induced immune suppression. Several mechanisms might have played a role, including the modulation of the intestinal microbiota, an improved gut barrier function, immune function, and a reduced inflammatory state. These results suggest an opportunity to develop new applications in cancer patients, with the aim to reduce infectious and other complications.
Collapse
Affiliation(s)
- Joyce Faber
- Nutricia Advanced Medical Nutrition, Danone Research, Centre for Specialised Nutrition, Wageningen, The Netherlands 6704 PH.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Monkman K, Mahony J, Lazo-langner A, Chin-yee BH, Minuk LA. The pandemic H1N1 influenza vaccine results in low rates of seroconversion for patients with hematological malignancies. Leuk Lymphoma 2011; 52:1736-41. [DOI: 10.3109/10428194.2011.584003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Mattarollo SR, Loi S, Duret H, Ma Y, Zitvogel L, Smyth MJ. Pivotal Role of Innate and Adaptive Immunity in Anthracycline Chemotherapy of Established Tumors. Cancer Res 2011; 71:4809-20. [DOI: 10.1158/0008-5472.can-11-0753] [Citation(s) in RCA: 236] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
van Tilburg CM, van der Velden VH, Sanders EA, Wolfs TF, Gaiser JF, de Haas V, Pieters R, Bloem AC, Bierings MB. Reduced versus intensive chemotherapy for childhood acute lymphoblastic leukemia: Impact on lymphocyte compartment composition. Leuk Res 2011; 35:484-91. [DOI: 10.1016/j.leukres.2010.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 10/10/2010] [Accepted: 10/11/2010] [Indexed: 01/18/2023]
|
39
|
|
40
|
Abstract
BACKGROUND Although immunosuppression from cancer adjuvant therapy has been documented, how these suppressed immune responses recover to baseline values after completion of cancer adjuvant therapy has not been studied systematically. OBJECTIVES The objective of this study was to examine the probability of immune recovery after cancer adjuvant therapy and the potential impact of cancer adjuvant therapy type and cancer stage on immune recovery in patients with newly diagnosed breast cancer. METHODS In a repeated-measures design, immune responses were measured four times in 80 patients with early-stage breast cancer: before and at 2, 6, and 12 months from the beginning of cancer adjuvant therapy. Natural killer cell activity, lymphokine-activated killer cell activity, lymphocyte proliferation, CD subsets (CD4, CD8, and CD56), and cytokines (interferon-gamma, interleukin [IL]-2, IL-4, IL-6, and IL-1alpha) were selected for their relevance to breast cancer. Immune recovery was defined by the level of immune response reaching to and above baseline levels. Data were analyzed using a multivariate generalized linear mixed-model approach. RESULTS Delayed immune recovery to pretreatment baseline levels continued to the 12-month time point in all parameters. The percentages of immune recovery ranged from 6% to 76% of the patients, varying among immune parameters. Overall, immune recovery was poorer for interferon-gamma, IL-2, IL-4, lymphocyte proliferation, and natural killer cell activity than was for CD subsets and IL-6. The type of cancer adjuvant therapy, not cancer stage, showed selective influence on immune recovery. Chemotherapy or chemotherapy and radiotherapy combination significantly delayed IL-2 recovery, whereas radiotherapy significantly delayed IL-4 recovery. DISCUSSION Immune recovery after breast cancer adjuvant therapy is delayed significantly for an extended time period in numerous immune parameters. The type of cancer adjuvant therapy has selective influence on immune recovery. Future investigations are warranted to elucidate the time course of immune recovery, clinical significance of poor immune recovery, and factors influencing immune recovery to develop potential interventions.
Collapse
|
41
|
Shin JW, Lee MM, Son JY, Lee NH, Cho CK, Chung WK, Cho JH, Son CG. Myelophil, a mixture of Astragali Radix and Salviae Radix extract, moderates toxic side effects of fluorouracil in mice. World J Gastroenterol 2008; 14:2323-2328. [PMID: 18416457 PMCID: PMC2705085 DOI: 10.3748/wjg.14.2323] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Revised: 02/21/2008] [Indexed: 02/06/2023] Open
Abstract
AIM To evaluate the efficacy of Myelophil, an extract containing Astragali Radix and Salviae Radix, for reducing complications induced by 5-fluorouracil (5-FU) in a gastrointestinal cancer model. METHODS We injected 5-FU into mice and then administered Myelophil to examine the ability of the drug to treat the side effects of 5-FU in mice. Peripheral blood counts, histological examinations, and colony-forming assays of bone marrow were conducted, followed by swimming tests and assessment of survival times. RESULTS Myelophil restored red and white blood cells and platelets in blood, and recovered cell density in bone marrow to levels comparable to those observed within the control group. In addition, Myelophil significantly increased colony-forming unit granulocyte-macrophage (CFU-GM) and CFU-erythroid (CFU-E) compared to the control group. We confirmed that interleukin-3 gene expression was upregulated by Myelophil in spleen cells. Myelophil administration also doubled the survival rate of mice that were severely myelosuppressed as a result of 5-FU injection at a lethal dose of 70%. Finally, the swimming performance of mice significantly improved as a result of Myelophil treatment. CONCLUSION These results provide experimental evidence in support of clinical applications of Myelophil to minimize 5-FU-induced myelosuppression and improve general post-chemotherapy health.
Collapse
|
42
|
Zhou Q, Zhu XQ, Zhang J, Xu ZL, Lu P, Wu F. Changes in circulating immunosuppressive cytokine levels of cancer patients after high intensity focused ultrasound treatment. ULTRASOUND IN MEDICINE & BIOLOGY 2008; 34:81-7. [PMID: 17854983 DOI: 10.1016/j.ultrasmedbio.2007.07.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2007] [Revised: 06/26/2007] [Accepted: 07/23/2007] [Indexed: 05/17/2023]
Abstract
Immunosuppression in a patient with malignant tumor is a major obstacle in cancer treatment. In this study, we investigated changes in the circulating level of all measured immunosuppressive cytokines in patients with malignancy before and after high intensity focused ultrasound (HIFU) treatment. Fifteen patients with solid malignancy were enrolled in this study and an enzyme-linked immunoabsorbent assay (ELISA) method was used to measure serum level of vascular endothelial growth factor (VEGF), transforming growth factor-beta1 (TGF-beta1), transforming growth factor-beta2 (TGF-beta2), interleukin 6 (IL-6) and interleukin 10 (IL-10), respectively before and 1 wk after HIFU treatment. Among them, seven patients had distant metastasis and the remaining eight had no metastasis. All patients received one-session HIFU treatment for primary cancer, including complete ablation in eight patients without metastasis, and partial ablation in seven patients with metastases. The results showed that serum immunosuppressive cytokine levels decreased after HIFU treatment, and there were significant decreases of VEGF, TGF-beta1, and TGF-beta2 before and after HIFU treatment. Compared with the values in the metastatic patients, serum levels of immunosuppressive cytokines were significantly lower in the nonmetastatic patients after HIFU treatment. It is concluded that HIFU can decrease tumor-secreted immunosuppressive cytokine production in addition to its direct tumor destruction. This change may lessen tumor-induced immunosuppression and renew antitumor immunity after HIFU in cancer patients.
Collapse
Affiliation(s)
- Qiang Zhou
- Clinical Center for Tumor Therapy of the 2nd Affiliated Hospital and Institute of Ultrasonic Engineering in Medicine, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
43
|
Pal S, Bhattacharyya S, Choudhuri T, Datta GK, Das T, Sa G. Amelioration of immune cell number depletion and potentiation of depressed detoxification system of tumor-bearing mice by curcumin. ACTA ACUST UNITED AC 2005; 29:470-8. [PMID: 16188398 DOI: 10.1016/j.cdp.2005.05.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2005] [Indexed: 11/21/2022]
Abstract
BACKGROUND The true therapeutic benefit of the use of natural products, especially acceptable dietary components such as curcumin, which can spare the normal cells and boost host immunity, has opened new horizons in cancer prevention and treatment. METHODS In our model system we used Ehrlich's ascites carcinoma cells grown in peritoneal carity of Swiss albino mice and curcumin was fed every alternative day. RESULTS Here, we report that curcumin administration to tumor-bearing mice decreased tumor cell number significantly in a dose-dependent manner. Furthermore, tumor-induced depletion of immune cell number of the host, as was evidenced from the decrease in bone marrow progenitor as well as thymic and splenic mononuclear cell numbers, was reintrated by curcumin. In fact, curcumin inhibited tumor-induced apoptosis of both thymocytes and splenocytes thereby restoring immune cell numbers to normal level in treated Ehrlich's ascites carcinoma-bearing mice. Moreover, curcumin was not toxic to the host; rather in tumor-bearing mice it inhibited hematopoietic toxicity, acted as a hepatoprotective agent and activated depressed anti-oxidant and detoxification systems. CONCLUSION The ability of curcumin to regress tumor as well as to protect the host from tumor-induced immunosuppression and toxicity strongly supports the candidacy of curcumin as a potential agent for the dietary therapy of cancer.
Collapse
Affiliation(s)
- Suman Pal
- Bose Institute, P-1/12 CIT Scheme VII M, Kolkata 700 054, India
| | | | | | | | | | | |
Collapse
|
44
|
Caras I, Grigorescu A, Stavaru C, Radu DL, Mogos I, Szegli G, Salageanu A. Evidence for immune defects in breast and lung cancer patients. Cancer Immunol Immunother 2004; 53:1146-52. [PMID: 15185014 PMCID: PMC11034324 DOI: 10.1007/s00262-004-0556-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2004] [Accepted: 04/23/2004] [Indexed: 10/26/2022]
Abstract
Immunosuppression is often identified in cancer patients. The aim of this study was to evaluate several immune parameters for patients with breast and lung cancer. Immunophenotyping analysis showed that the cancer patients investigated had significantly lower absolute numbers of peripheral blood lymphocytes than controls. The immunosuppression was more evident for the breast cancer subgroup. The most severe immune defect noticed was the marked impairment of IFN-gamma secretion. A shift toward the Th2 phenotype as revealed by assessment of intracellular level of IFN-gamma and IL-4 was also noticed. The secretion of proinflammatory cytokines IL-1beta and TNF-alpha in whole blood cultures was not impaired. Although the proportion of activated cells was slightly lower than in the control group, our results showed that both peripheral T lymphocytes and NK cells of cancer patients could be induced to express early activation marker CD69 after ex vivo mitogen stimulation. In conclusion, our study revealed several immune defects in cancer patients. This suggests that an appropriate immunotherapeutical approach might be used to restore compromised immune functions with beneficial effects on both antitumor and general immunity.
Collapse
Affiliation(s)
- Iuliana Caras
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - A. Grigorescu
- Oncology Institute “Prof. Dr. Alex. Trestioreanu”, Bucharest, Romania
| | - Crina Stavaru
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - D. L. Radu
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - I. Mogos
- Oncology Institute “Prof. Dr. Alex. Trestioreanu”, Bucharest, Romania
| | - G. Szegli
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - Aurora Salageanu
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
- Immunomodulators Group, Department of Immunology, Cantacuzino Institute, 103 Spl. Independentei, PO 050096, Bucharest, Romania
| |
Collapse
|
45
|
Induction of comprehensible models for gene expression datasets by subgroup discovery methodology. J Biomed Inform 2004; 37:269-84. [DOI: 10.1016/j.jbi.2004.07.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2004] [Indexed: 11/23/2022]
|
46
|
Schlezinger JJ, Jensen BA, Mann KK, Ryu HY, Sherr DH. Peroxisome proliferator-activated receptor gamma-mediated NF-kappa B activation and apoptosis in pre-B cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:6831-41. [PMID: 12471115 DOI: 10.4049/jimmunol.169.12.6831] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of peroxisome proliferator-activated receptor gamma (PPARgamma) in adipocyte physiology has been exploited for the treatment of diabetes. The expression of PPARgamma in lymphoid organs and its modulation of macrophage inflammatory responses, T cell proliferation and cytokine production, and B cell proliferation also implicate it in immune regulation. Despite significant human exposure to PPARgamma agonists, little is known about the consequences of PPARgamma activation in the developing immune system. Here, well-characterized models of B lymphopoiesis were used to investigate the effects of PPARgamma ligands on nontransformed pro/pre-B (BU-11) and transformed immature B (WEHI-231) cell development. Treatment of BU-11, WEHI-231, or primary bone marrow B cells with PPARgamma agonists (ciglitazone and GW347845X) resulted in rapid apoptosis. A role for PPARgamma and its dimerization partner, retinoid X receptor (RXR)alpha, in death signaling was supported by 1) the expression of RXRalpha mRNA and cytosolic PPARgamma protein, 2) agonist-induced binding of PPARgamma to a PPRE, and 3) synergistic increases in apoptosis following cotreatment with PPARgamma agonists and 9-cis-retinoic acid, an RXRalpha agonist. PPARgamma agonists activated NF-kappaB (p50, Rel A, c-Rel) binding to the upstream kappaB regulatory element site of c-myc. Only doses of agonists that induced apoptosis stimulated NF-kappaB-DNA binding. Cotreatment with 9-cis-retinoic acid and PPARgamma agonists decreased the dose required to activate NF-kappaB. These data suggest that activation of PPARgamma-RXR initiates a potent apoptotic signaling cascade in B cells, potentially through NF-kappaB activation. These results have implications for the nominal role of the PPARgamma in B cell development and for the use of PPARgamma agonists as immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Jennifer J Schlezinger
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA.
| | | | | | | | | |
Collapse
|