1
|
Tsay JCJ, Darawshy F, Wang C, Kwok B, Wong KK, Wu BG, Sulaiman I, Zhou H, Isaacs B, Kugler MC, Sanchez E, Bain A, Li Y, Schluger R, Lukovnikova A, Collazo D, Kyeremateng Y, Pillai R, Chang M, Li Q, Vanguri RS, Becker AS, Moore WH, Thurston G, Gordon T, Moreira AL, Goparaju CM, Sterman DH, Tsirigos A, Li H, Segal LN, Pass HI. Lung Microbial and Host Genomic Signatures as Predictors of Prognosis in Early-Stage Adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2024; 33:1433-1444. [PMID: 39225784 PMCID: PMC11530314 DOI: 10.1158/1055-9965.epi-24-0661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/15/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Risk of early-stage lung adenocarcinoma recurrence after surgical resection is significant, and the postrecurrence median survival is approximately 2 years. Currently, there are no commercially available biomarkers that predict recurrence. In this study, we investigated whether microbial and host genomic signatures in the lung can predict recurrence. METHODS In 91 patients with early-stage (stage IA/IB) lung adenocarcinoma with extensive follow-up, we used 16s rRNA gene sequencing and host RNA sequencing to map the microbial and host transcriptomic landscape in tumor and adjacent unaffected lung samples. RESULTS Of 91 subjects, 23 had tumor recurrence over 5-year period. In tumor samples, lung adenocarcinoma recurrence was associated with enrichment in Dialister and Prevotella, whereas in unaffected lung samples, recurrence was associated with enrichment in Sphingomonas and Alloiococcus. The strengths of the associations between microbial and host genomic signatures with lung adenocarcinoma recurrence were greater in adjacent unaffected lung samples than in the primary tumor. Among microbial-host features in the unaffected lung samples associated with recurrence, enrichment in Stenotrophomonas geniculata and Chryseobacterium was positively correlated with upregulation of IL2, IL3, IL17, EGFR, and HIF1 signaling pathways among the host transcriptome. In tumor samples, enrichment in Veillonellaceae (Dialister), Ruminococcaceae, Haemophilus influenzae, and Neisseria was positively correlated with upregulation of IL1, IL6, IL17, IFN, and tryptophan metabolism pathways. CONCLUSIONS Overall, modeling suggested that a combined microbial/transcriptome approach using unaffected lung samples had the best biomarker performance (AUC = 0.83). IMPACT This study suggests that lung adenocarcinoma recurrence is associated with distinct pathophysiologic mechanisms of microbial-host interactions in the unaffected lung rather than those present in the resected tumor.
Collapse
Affiliation(s)
- Jun-Chieh J. Tsay
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Division of Pulmonary and Critical Care Medicine, VA New York Harbor Healthcare System, New York, NY
| | - Fares Darawshy
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- The Institute of Pulmonary Medicine, Hadassah Medical Center, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Chan Wang
- Department of Population Health, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Benjamin Kwok
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Kendrew K. Wong
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Benjamin G. Wu
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Division of Pulmonary and Critical Care Medicine, VA New York Harbor Healthcare System, New York, NY
| | - Imran Sulaiman
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Respiratory Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Respiratory Medicine, Beaumont Hospital, Dublin, Ireland
| | - Hua Zhou
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine New York, USA
| | - Bradley Isaacs
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Matthias C. Kugler
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Elizabeth Sanchez
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Alexander Bain
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Yonghua Li
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Rosemary Schluger
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Alena Lukovnikova
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Destiny Collazo
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Yaa Kyeremateng
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Ray Pillai
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Miao Chang
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Qingsheng Li
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Rami S. Vanguri
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Anton S. Becker
- Department of Radiology, NYU Grossman School of Medicine New York, USA
| | - William H. Moore
- Department of Radiology, NYU Grossman School of Medicine New York, USA
| | - George Thurston
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Population Health, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Terry Gordon
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Division of Environmental Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Andre L. Moreira
- Department of Pathology, NYU Grossman School of Medicine New York, USA
| | - Chandra M. Goparaju
- Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, USA
| | - Daniel H. Sterman
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY
| | - Aristotelis Tsirigos
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine New York, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Huilin Li
- Department of Population Health, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
| | - Leopoldo N. Segal
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Department of Medicine, NYU Grossman School of Medicine, NYU Langone Health, New York, NY
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY
| | - Harvey I. Pass
- Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
2
|
Lim ESY, Ong Y, Chou Y, Then CK. Interconnected influences of tumour and host microbiota on treatment response and side effects in nasopharyngeal cancer. Crit Rev Oncol Hematol 2024; 202:104468. [PMID: 39103130 DOI: 10.1016/j.critrevonc.2024.104468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024] Open
Abstract
This study elucidates the intricate relationship between nasopharyngeal carcinoma (NPC), a significant malignancy predominant in Asia with notable global incidence and mortality rates, and the host microbiota, including those of tumour, nasal, nasopharyngeal, oral, oropharyngeal, and gut communities. It underscores how the composition and diversity of microbiota are altered in NPC, delving into their implications for disease pathogenesis, treatment response, and the side effects of therapies. A consistent reduction in alpha diversity across oral, nasal, and gut microbiomes in NPC patients compared to healthy individuals signals a distinct microbial signature indicative of the diseased state. The study also shows unique microbial changes tied to different NPC stages, indicating a dynamic interplay between disease progression and microbiota composition. Patients with specific microbial profiles exhibit varied responses to chemotherapy and immunotherapy, underscoring the potential for treatment personalisation based on microbiota analysis. Furthermore, the side effects of NPC treatments, such as oral mucositis, are intensified by shifts in microbial communities, suggesting a direct link between microbiota composition and treatment tolerance. This nexus offers opportunities for interventions aimed at modulating the microbiota to alleviate side effects, improve quality of life, and potentially enhance treatment efficacy. Highlighting the dual potential of microbiota as both a therapeutic target and a biomarker for NPC, this review emphasises its significance in influencing treatment outcomes and side effects, heralding a new era in NPC management through personalised treatment strategies and innovative approaches.
Collapse
Affiliation(s)
- Eugene Sheng Yao Lim
- Jeffery Cheah School of Medicine and Health Sciences, Monash University, Malaysia
| | - Yenyi Ong
- Jeffery Cheah School of Medicine and Health Sciences, Monash University, Malaysia
| | - Yang Chou
- Department of Otolaryngology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Chee Kin Then
- Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
3
|
Liao J, Dhilipkannah P, Jiang F. Improving CT scan for lung cancer diagnosis with an integromic signature. J Biol Methods 2024; 11:e99010023. [PMID: 39544186 PMCID: PMC11557295 DOI: 10.14440/jbm.2024.0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/14/2024] [Indexed: 11/17/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality globally, making early detection crucial for reducing death rates. Low-dose computed tomography (LDCT) screening helps detect lung cancer early but often identifies indeterminate pulmonary nodules (PNs), leading to potential overtreatment. This study aimed to develop a diagnostic test that accurately differentiates malignant from benign PNs detected on LDCT scans by analyzing non-coding RNAs, DNA methylation, and bacterial DNA in patient samples. Using droplet digital polymerase chain reaction, we analyzed samples from a training set of 150 patients with malignant PNs and 250 smokers with benign PNs. Individual biomarkers in plasma and sputum showed moderate effectiveness, with sensitivities ranging from 62% to 77% and specificities from 54% to 87%. We developed an integromic signature by combining two plasma biomarkers and one sputum biomarker, along with additional clinical data, which demonstrated a sensitivity of 90% and specificity of 95%. The signature's diagnostic performance was further validated in a cohort consisting of 30 patients with malignant PNs and 50 smokers with benign PNs. The integromic signature showed high sensitivity and specificity in distinguishing malignant from benign PNs identified through LDCT. This tool has the potential to significantly lower both mortality and health-care costs associated with the overtreatment of benign nodules, offering a promising approach to improving lung cancer screening protocols.
Collapse
Affiliation(s)
- Jipei Liao
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St. Baltimore, MD, United States of America
| | - Pushpawallie Dhilipkannah
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St. Baltimore, MD, United States of America
| | - Feng Jiang
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St. Baltimore, MD, United States of America
| |
Collapse
|
4
|
Wang L, Mei X, Liu X, Guo L, Yang B, Chen R. The Interleukin-33/ST2 Axis Enhances Lung-Resident CD14+ Monocyte Function in Patients with Non-Small Cell Lung Cancer. Immunol Invest 2023; 52:67-82. [PMID: 36218388 DOI: 10.1080/08820139.2022.2130075] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interleukin-33 (IL-33) binds to its cognate receptor suppression of tumorigenicity 2 (ST2), leading to critical modulatory roles in immune responses during inflammation and cancers. The aim of this study was to investigate the role of IL-33/ST2 signaling in monocyte function in non-small cell lung cancer (NSCLC). Sixty-two NSCLC patients and nineteen controls were enrolled. IL-33 levels and ST2 expression were measured in peripheral blood and bronchoalveolar lavage fluid (BALF) by ELISA and flow cytometry. HLA-DR expression by CD14+ monocytes, granzyme B and proinflammatory cytokine secretion were also investigated in lipopolysaccharide-stimulated cells. CD14+ monocytes purified from BALF in the tumor site were stimulated with IL-33 in vitro, and co-cultured with a lung cancer cell line A549 cells. The cytotoxicity of monocytes with IL-33 stimulation was then assessed. IL-33 levels were lower in the peripheral blood and tumor microenvironment of NSCLC patients. There was no significant difference in peripheral ST2 expression between NSCLC patients and controls. Soluble ST2 levels were increased but membrane-bound ST2 expression in CD14+ monocytes was decreased in tumor microenvironment of NSCLC patients. There were no remarkable differences in either HLA-DR expression or proinflammatory cytokine secretion by circulating CD14+ monocytes between NSCLC patients and controls. CD14+ monocytes in the tumor microenvironment revealed a dysfunctional phenotype, which presented as lower HLA-DR expression and reduced granzyme B and proinflammatory cytokines. A higher concentration of IL-33 stimulation promoted tumor-resident CD14+ monocyte-induced target cell death. The present study indicates that IL-33/ST2 signaling pathway might enhance the activity of tumor-resident CD14+ monocytes in NSCLC.
Collapse
Affiliation(s)
- Lv Wang
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Xingke Mei
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Xiaogang Liu
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Lu Guo
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Bo Yang
- Department of Thoracic Surgery, Xi'an Daxing Hospital, Xi'an, Shaanxi, China
| | - Ren'an Chen
- The Second Department of Internal Medicine, Shaanxi Cancer Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
5
|
Peters BA, Pass HI, Burk RD, Xue X, Goparaju C, Sollecito CC, Grassi E, Segal LN, Tsay JCJ, Hayes RB, Ahn J. The lung microbiome, peripheral gene expression, and recurrence-free survival after resection of stage II non-small cell lung cancer. Genome Med 2022; 14:121. [PMID: 36303210 PMCID: PMC9609265 DOI: 10.1186/s13073-022-01126-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Cancer recurrence after tumor resection in early-stage non-small cell lung cancer (NSCLC) is common, yet difficult to predict. The lung microbiota and systemic immunity may be important modulators of risk for lung cancer recurrence, yet biomarkers from the lung microbiome and peripheral immune environment are understudied. Such markers may hold promise for prediction as well as improved etiologic understanding of lung cancer recurrence. METHODS In tumor and distant normal lung samples from 46 stage II NSCLC patients with curative resection (39 tumor samples, 41 normal lung samples), we conducted 16S rRNA gene sequencing. We also measured peripheral blood immune gene expression with nanoString®. We examined associations of lung microbiota and peripheral gene expression with recurrence-free survival (RFS) and disease-free survival (DFS) using 500 × 10-fold cross-validated elastic-net penalized Cox regression, and examined predictive accuracy using time-dependent receiver operating characteristic (ROC) curves. RESULTS Over a median of 4.8 years of follow-up (range 0.2-12.2 years), 43% of patients experienced a recurrence, and 50% died. In normal lung tissue, a higher abundance of classes Bacteroidia and Clostridia, and orders Bacteroidales and Clostridiales, were associated with worse RFS, while a higher abundance of classes Alphaproteobacteria and Betaproteobacteria, and orders Burkholderiales and Neisseriales, were associated with better RFS. In tumor tissue, a higher abundance of orders Actinomycetales and Pseudomonadales were associated with worse DFS. Among these taxa, normal lung Clostridiales and Bacteroidales were also related to worse survival in a previous small pilot study and an additional independent validation cohort. In peripheral blood, higher expression of genes TAP1, TAPBP, CSF2RB, and IFITM2 were associated with better DFS. Analysis of ROC curves revealed that lung microbiome and peripheral gene expression biomarkers provided significant additional recurrence risk discrimination over standard demographic and clinical covariates, with microbiome biomarkers contributing more to short-term (1-year) prediction and gene biomarkers contributing to longer-term (2-5-year) prediction. CONCLUSIONS We identified compelling biomarkers in under-explored data types, the lung microbiome, and peripheral blood gene expression, which may improve risk prediction of recurrence in early-stage NSCLC patients. These findings will require validation in a larger cohort.
Collapse
Affiliation(s)
- Brandilyn A Peters
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, #1315AB, The Bronx, New York, NY, 10461, USA.
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
- NYU Perlmutter Cancer Center, New York, NY, USA
| | - Robert D Burk
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, #1315AB, The Bronx, New York, NY, 10461, USA
- Department of Pediatrics, Albert Einstein College of Medicine, The Bronx, New York, NY, USA
- Department of Microbiology & Immunology, and Obstetrics & Gynecology & Women's Health, Albert Einstein College of Medicine, The Bronx, New York, NY, USA
| | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, #1315AB, The Bronx, New York, NY, 10461, USA
| | - Chandra Goparaju
- Department of Cardiothoracic Surgery, NYU Langone Health, New York, NY, USA
| | | | - Evan Grassi
- Department of Pediatrics, Albert Einstein College of Medicine, The Bronx, New York, NY, USA
| | | | | | - Richard B Hayes
- NYU Perlmutter Cancer Center, New York, NY, USA
- Department of Population Health, NYU Langone Health, New York, NY, USA
| | - Jiyoung Ahn
- NYU Perlmutter Cancer Center, New York, NY, USA
- Department of Population Health, NYU Langone Health, New York, NY, USA
| |
Collapse
|
6
|
Sun K, Wu L, Wang S, Deng W. Antitumor effects of Chinese herbal medicine compounds and their nano-formulations on regulating the immune system microenvironment. Front Oncol 2022; 12:949332. [PMID: 36212483 PMCID: PMC9540406 DOI: 10.3389/fonc.2022.949332] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
Traditional Chinese medicine (TCM), including herbal medicine, acupuncture and meditation, has a wide range of applications in China. In recent years, herbal compounding and active ingredients have been used to control tumor growth, reduce suffering, improve quality of life, and prolong the life span of cancer patients. To reduce side effects, herbal medicine can be used in conjunction with radiotherapy and chemotherapy or can be used as an adjuvant to strengthen the immune effect of anticancer vaccines. In particular, in the immunosuppressed tumor microenvironment, herbal medicine can have antitumor effects by stimulating the immune response. This paper reviews the advances in research on antitumor immunomodulation in Chinese herbal medicine, including the regulation of the innate immune system, which includes macrophages, MDSCs, and natural killer cells, and the adaptive immune system, which includes CD4+ T cells, CD8+ T cells, and regulatory T cells (Tregs), to influence tumor-associated inflammation. In addition, a combination of active ingredients of herbal medicine and modern nanotechnology alter the tumor immune microenvironment. In recent years, immunological antitumor therapy in TCM has been applied on a reasonably large scale both nationally and internationally, and there is potential for further clinical expansion. Investigation of immune modulation mechanisms in Chinese herbal medicine will provide novel perspectives of how herbal medicine controls tumor growth and metastasis, which will contribute to the evolution of tumor research.
Collapse
|
7
|
Yeoh WJ, Vu VP, Krebs P. IL-33 biology in cancer: An update and future perspectives. Cytokine 2022; 157:155961. [PMID: 35843125 DOI: 10.1016/j.cyto.2022.155961] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 family of cytokines that is constitutively expressed in the nucleus of epithelial, endothelial and fibroblast-like cells. Upon cell stress, damage or necrosis, IL-33 is released into the cytoplasm to exert its prime role as an alarmin by binding to its specific receptor moiety, ST2. IL-33 exhibits pleiotropic function in inflammatory diseases and particularly in cancer. IL-33 may play a dual role as both a pro-tumorigenic and anti-tumorigenic cytokine, dependent on tumor and cellular context, expression levels, bioactivity and the nature of the inflammatory environment. In this review, we discuss the differential contribution of IL-33 to malignant or inflammatory conditions, its multifaceted effects on the tumor microenvironment, while providing possible explanations for the discrepant findings described in the literature. Additionally, we examine the emerging and divergent functions of IL-33 in the nucleus, and aspects of IL-33 biology that are currently under-addressed.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Vivian P Vu
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
8
|
Delgado A, Guddati AK. Infections in Hospitalized Cancer Patients. World J Oncol 2022; 12:195-205. [PMID: 35059079 PMCID: PMC8734501 DOI: 10.14740/wjon1410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/01/2021] [Indexed: 11/11/2022] Open
Abstract
Cancer patients are at an increased risk of developing infections that are primarily treatment-driven but may also be malignancy-driven. While cancer treatments such as chemotherapy, radiotherapy, and surgery have been known to improve malignancy morbidity and mortality, they also have the potential to weaken immune defenses and induce periods of severe cytopenia. These adverse effects pave the way for opportunistic infections to complicate a hospitalized cancer patient's clinical course. Understanding the risk each patient inherently has for developing a bacterial, fungal, or viral infection is critical to choosing the correct prophylactic treatment in conjunction with their scheduled cancer therapy. This review discusses the most common types of infections found in hospitalized cancer patients as well as the current guidelines for prophylactic and antimicrobial treatment in cancer patients. In addition, it describes the interaction between antibiotics and cancer therapies for consideration when treating infection in a cancer patient.
Collapse
Affiliation(s)
- Amanda Delgado
- Division of Hematology/Oncology, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Achuta Kumar Guddati
- Division of Hematology/Oncology, Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
9
|
Salidroside Suppresses the Proliferation and Migration of Human Lung Cancer Cells through AMPK-Dependent NLRP3 Inflammasome Regulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6614574. [PMID: 34457117 PMCID: PMC8390167 DOI: 10.1155/2021/6614574] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 01/22/2023]
Abstract
Inflammatory reactions mediated by the NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) inflammasome contributes to non-small-cell lung cancer (NSCLC) progression, particularly in patients with bacterial infections. Salidroside (SAL) has recently been shown to suppress lipopolysaccharide- (LPS-) induced NSCLC proliferation and migration, but its mechanism of action remains unclear. It has been shown that SAL improves metabolic inflammation in diabetic rodents through AMP-activated protein kinase- (AMPK-) dependent inhibition of the NLRP3 inflammasome. However, whether the NLRP3 inflammasome is regulated by SAL in NSCLC cells and how its underlying mechanism(s) can be determined require clarification. In this study, human lung alveolar basal carcinoma epithelial (A549) cells were treated with LPS, and the effects of SAL on cell proliferation, migration, AMPK activity, reactive oxygen species (ROS) production, and NLRP3 inflammasome activation were investigated. We found that LPS induction increases the proliferation and migration of A549 cells which was suppressed by SAL. Moreover, SAL protected A549 cells against LPS-induced AMPK inhibition, ROS production, and NLRP3 inflammasome activation. Blocking AMPK using Compound C almost completely suppressed the beneficial effects of SAL. In summary, these results indicate that SAL suppresses the proliferation and migration of human lung cancer cells through AMPK-dependent NLRP3 inflammasome regulation.
Collapse
|
10
|
Mughal MJ, Kwok HF. Multidimensional role of bacteria in cancer: Mechanisms insight, diagnostic, preventive and therapeutic potential. Semin Cancer Biol 2021; 86:1026-1044. [PMID: 34119644 DOI: 10.1016/j.semcancer.2021.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
The active role of bacteria in oncogenesis has long been a topic of debate. Although, it was speculated to be a transmissible cause of cancer as early as the 16th-century, yet the idea about the direct involvement of bacteria in cancer development has only been explored in recent decades. More recently, several studies have uncovered the mechanisms behind the carcinogenic potential of bacteria which are inflammation, immune evasion, pro-carcinogenic metabolite production, DNA damage and genomic instability. On the other side, the recent development on the understanding of tumor microenvironment and technological advancements has turned this enemy into an ally. Studies using bacteria for cancer treatment and detection have shown noticeable effects. Therapeutic abilities of bioengineered live bacteria such as high specificity, selective cytotoxicity to cancer cells, responsiveness to external signals and control after ingestion have helped to overcome the challenges faced by conventional cancer therapies and highlighted the bacterial based therapy as an ideal approach for cancer treatment. In this review, we have made an effort to compile substantial evidence to support the multidimensional role of bacteria in cancer. We have discussed the multifaceted role of bacteria in cancer by highlighting the wide impact of bacteria on different cancer types, their mechanisms of actions in inducing carcinogenicity, followed by the diagnostic and therapeutic potential of bacteria in cancers. Moreover, we have also highlighted the existing gaps in the knowledge of the association between bacteria and cancer as well as the limitation and advantage of bacteria-based therapies in cancer. A better understanding of these multidimensional roles of bacteria in cancer can open up the new doorways to develop early detection strategies, prevent cancer, and develop therapeutic tactics to cure this devastating disease.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau.
| |
Collapse
|
11
|
Giannakou LE, Giannopoulos AS, Hatzoglou C, Gourgoulianis KI, Rouka E, Zarogiannis SG. Investigation and Functional Enrichment Analysis of the Human Host Interaction Network with Common Gram-Negative Respiratory Pathogens Predicts Possible Association with Lung Adenocarcinoma. PATHOPHYSIOLOGY 2021; 28:20-33. [PMID: 35366267 PMCID: PMC8830454 DOI: 10.3390/pathophysiology28010003] [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: 11/09/2020] [Revised: 12/26/2020] [Accepted: 12/27/2020] [Indexed: 11/16/2022] Open
Abstract
Haemophilus influenzae (Hi), Moraxella catarrhalis (MorCa) and Pseudomonas aeruginosa (Psa) are three of the most common gram-negative bacteria responsible for human respiratory diseases. In this study, we aimed to identify, using the functional enrichment analysis (FEA), the human gene interaction network with the aforementioned bacteria in order to elucidate the full spectrum of induced pathogenicity. The Human Pathogen Interaction Database (HPIDB 3.0) was used to identify the human proteins that interact with the three pathogens. FEA was performed via the ToppFun tool of the ToppGene Suite and the GeneCodis database so as to identify enriched gene ontologies (GO) of biological processes (BP), cellular components (CC) and diseases. In total, 11 human proteins were found to interact with the bacterial pathogens. FEA of BP GOs revealed associations with mitochondrial membrane permeability relative to apoptotic pathways. FEA of CC GOs revealed associations with focal adhesion, cell junctions and exosomes. The most significantly enriched annotations in diseases and pathways were lung adenocarcinoma and cell cycle, respectively. Our results suggest that the Hi, MorCa and Psa pathogens could be related to the pathogenesis and/or progression of lung adenocarcinoma via the targeting of the epithelial cellular junctions and the subsequent deregulation of the cell adhesion and apoptotic pathways. These hypotheses should be experimentally validated.
Collapse
Affiliation(s)
- Lydia-Eirini Giannakou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (L.-E.G.); (A.-S.G.); (C.H.); (S.G.Z.)
| | - Athanasios-Stefanos Giannopoulos
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (L.-E.G.); (A.-S.G.); (C.H.); (S.G.Z.)
| | - Chrissi Hatzoglou
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (L.-E.G.); (A.-S.G.); (C.H.); (S.G.Z.)
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece;
| | - Konstantinos I. Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece;
| | - Erasmia Rouka
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (L.-E.G.); (A.-S.G.); (C.H.); (S.G.Z.)
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece;
- Correspondence:
| | - Sotirios G. Zarogiannis
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece; (L.-E.G.); (A.-S.G.); (C.H.); (S.G.Z.)
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, 41500 Larissa, Greece;
| |
Collapse
|
12
|
Kashani B, Zandi Z, Pourbagheri-Sigaroodi A, Bashash D, Ghaffari SH. The role of toll-like receptor 4 (TLR4) in cancer progression: A possible therapeutic target? J Cell Physiol 2020; 236:4121-4137. [PMID: 33230811 DOI: 10.1002/jcp.30166] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/13/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
The toll-like receptor (TLR) family consists of vital receptors responsible for pattern recognition in innate immunity, making them the core proteins involved in pathogen detection and eliciting immune responses. The most studied member of this family, TLR4, has been the center of attention regarding its contributory role in many inflammatory diseases including sepsis shock and asthma. Notably, mounting pieces of evidence have proved that this receptor is aberrantly expressed on the tumor cells and the tumor microenvironment in a wide range of cancer types and it is highly associated with the initiation of tumorigenesis as well as tumor progression and drug resistance. Cancer therapy using TLR4 inhibitors has recently drawn scientists' attention, and the promising results of such studies may pave the way for more investigation in the foreseeable future. This review will introduce the key proteins of the TLR4 pathway and how they interact with major growth factors in the tumor microenvironment. Moreover, we will discuss the many aspects of tumor progression affected by the activation of this receptor and provide an overview of the recent therapeutic approaches using various TLR4 antagonists.
Collapse
Affiliation(s)
- Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Jarosz J, Papiernik D, Wietrzyk J. IL-33 – positive or negative role in cancer progression? POSTEP HIG MED DOSW 2019. [DOI: 10.5604/01.3001.0013.5955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interleukin-33 (IL-33) is a IL-1 family member of cytokines which binds the ST2 (suppression of tumorigenicity 2) receptor. This cytokine has a dual function. It may act both as a traditional cytokine and as an intracellular nuclear factor. IL-33 plays a role in many diseases such as: allergy, inflammatory diseases, diabetes and heart diseases. The role of IL-33 in the development of cancer has been intensively studied in recent years and researchers observe both its pro- -and anti-cancer effects. IL-33 promotes the development of tumors by affecting expression of cytokines promoting proliferation, angiogenesis, migration, matrix remodeling, the inhibition of apoptosis and recruitment of individual cells of the immune system. Antitumor action of IL-33 is carried out by recruiting and activating CD8+T lymphocytes, natural killer (NK) cells and by promoting second type immune response by the type 2 innate lymphoid cells (ILC2). Despite numerous studies on the role of IL-33 in the development of cancer, we still do not fully understand the mechanisms by which IL-33 impacts the development and malignancy of various types of cancers. This review summarizes the dual role of IL-33 in the development of the most common cancers in the world to better understand its importance in the carcinogenesis.
Collapse
Affiliation(s)
- Joanna Jarosz
- Instytut Immunologii i Terapii Doświadczalnej im. L. Hirszfelda PAN we Wrocławiu
| | | | - Joanna Wietrzyk
- Instytut Immunologii i Terapii Doświadczalnej im. L. Hirszfelda PAN we Wrocławiu
| |
Collapse
|
14
|
Interleukin-33 Involvement in Nonsmall Cell Lung Carcinomas: An Update. Biomolecules 2019; 9:biom9050203. [PMID: 31130612 PMCID: PMC6572046 DOI: 10.3390/biom9050203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
Lung carcinogenesis is a multistep process involving genetic mutations and epigenetic changes, with the acquisition of a malignant phenotype characterized by apoptosis resistance, unregulated proliferation and differentiation, invasion, and metastatic abilities. However, neoplastic development and progression seem to be aided by non-neoplastic cells; the molecules they produced can either promote the immune response or, alternatively, support tumor pathogenesis. Consequently, the relative contribution of tumor-associated inflammatory pathways to cancer development has become crucial information. Interleukin-33 (IL-33) is an IL-1-like alarmin, and it is a ligand for the suppressor of tumorigenicity 2 (ST2) receptor. IL-33 functions as a dual role cytokine with the ability to induce T-helper-type 2 (Th2) immune cells and translocate into the nucleus, suppressing gene transcription. Although its function in immunity- and immune-related disorders is well known, its role in tumorigenesis is still debated. The IL-33/ST2 axis is emerging as a powerful modulator of the tumor microenvironment (TME) by recruiting immune cells, able to modify the TME, supporting malignant proliferation or improving antitumor immunity. In the present review, we discuss IL-33′s potential role in lung carcinogenesis and its possible application as a therapeutic target.
Collapse
|
15
|
The Role of IL-33/ST2 Pathway in Tumorigenesis. Int J Mol Sci 2018; 19:ijms19092676. [PMID: 30205617 PMCID: PMC6164146 DOI: 10.3390/ijms19092676] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 12/13/2022] Open
Abstract
Cancer is initiated by mutations in critical regulatory genes; however, its progression to malignancy is aided by non-neoplastic cells and molecules that create a permissive environment known as the tumor stroma or microenvironment (TME). Interleukin 33 (IL-33) is a dual function cytokine that also acts as a nuclear factor. IL-33 typically resides in the nucleus of the cells where it is expressed. However, upon tissue damage, necrosis, or injury, it is quickly released into extracellular space where it binds to its cognate receptor suppression of tumorigenicity 2 (ST2)L found on the membrane of target cells to potently activate a T Helper 2 (Th2) immune response, thus, it is classified as an alarmin. While its role in immunity and immune-related disorders has been extensively studied, its role in tumorigenesis is only beginning to be elucidated and has revealed opposing roles in tumor development. The IL-33/ST2 axis is emerging as a potent modulator of the TME. By recruiting a cohort of immune cells, it can remodel the TME to promote malignancy or impose tumor regression. Here, we review its multiple functions in various cancers to better understand its potential as a therapeutic target to block tumor progression or as adjuvant therapy to enhance the efficacy of anticancer immunotherapies.
Collapse
|