1
|
Lee J, Park J, Song KM, Lee YG, Choi HK. Actinidia arguta Extract Containing Myo-Inositol Suppresses TNF- α-Induced VCAM-1 Expression and Monocyte Adhesion to Endothelial Cells via Inhibition of the PTEN/Akt/GSK-3 β and NF- κB Signaling Pathways. J Med Food 2024; 27:419-427. [PMID: 38656897 DOI: 10.1089/jmf.2023.k.0326] [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] [Indexed: 04/26/2024] Open
Abstract
The primary inflammatory process in atherosclerosis, a major contributor to cardiovascular disease, begins with monocyte adhering to vascular endothelial cells. Actinidia arguta (kiwiberry) is an edible fruit that contains various bioactive components. While A. arguta extract (AAE) has been recognized for its anti-inflammatory characteristics, its specific inhibitory effect on early atherogenic events has not been clarified. We used tumor necrosis factor-α (TNF-α)-stimulated human umbilical vein endothelial cells (HUVECs) for an in vitro model. AAE effectively hindered the attachment of THP-1 monocytes and reduced the expression of vascular cell adhesion molecule-1 (VCAM-1) in HUVECs. Transcriptome analysis revealed that AAE treatment upregulated phosphatase and tensin homolog (PTEN), subsequently inhibiting phosphorylation of AKT and glycogen synthase kinase 3β (GSK3β) in HUVECs. AAE further hindered phosphorylation of AKT downstream of the nuclear factor kappa B (NF-κB) signaling pathway, leading to suppression of target gene expression. Oral administration of AAE suppressed TNF-α-stimulated VCAM-1 expression, monocyte-derived macrophage infiltration, and proinflammatory cytokine expression in C57BL/6 mouse aortas. Myo-inositol, identified as the major compound in AAE, played a key role in suppressing THP-1 monocyte adhesion in HUVECs. These findings suggest that AAE could serve as a nutraceutical for preventing atherosclerosis by inhibiting its initial pathogenesis.
Collapse
Affiliation(s)
- Jangho Lee
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Joon Park
- Divisions of Strategic Food Technology Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Kyung-Mo Song
- Divisions of Strategic Food Technology Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Yu Geon Lee
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Hyo-Kyoung Choi
- Divisions of Functional Food Research, Korea Food Research Institute, Wanju-gun, Republic of Korea
| |
Collapse
|
2
|
Czarnecka-Chrebelska KH, Kordiak J, Brzeziańska-Lasota E, Pastuszak-Lewandoska D. Respiratory Tract Oncobiome in Lung Carcinogenesis: Where Are We Now? Cancers (Basel) 2023; 15:4935. [PMID: 37894302 PMCID: PMC10605430 DOI: 10.3390/cancers15204935] [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: 09/08/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The importance of microbiota in developing and treating diseases, including lung cancer (LC), is becoming increasingly recognized. Studies have shown differences in microorganism populations in the upper and lower respiratory tracts of patients with lung cancer compared to healthy individuals, indicating a link between dysbiosis and lung cancer. However, it is not only important to identify "which bacteria are present" but also to understand "how" they affect lung carcinogenesis. The interactions between the host and lung microbiota are complex, and our knowledge of this relationship is limited. This review presents research findings on the bacterial lung microbiota and discusses the mechanisms by which lung-dwelling microorganisms may directly or indirectly contribute to the development of lung cancer. These mechanisms include influences on the host immune system regulation and the local immune microenvironment, the regulation of oncogenic signaling pathways in epithelial cells (causing cell cycle disorders, mutagenesis, and DNA damage), and lastly, the MAMPs-mediated path involving the effects of bacteriocins, TLRs signaling induction, and TNF release. A better understanding of lung microbiota's role in lung tumor pathology could lead to identifying new diagnostic and therapeutic biomarkers and developing personalized therapeutic management for lung cancer patients.
Collapse
Affiliation(s)
| | - Jacek Kordiak
- Department of Thoracic, General and Oncological Surgery, Medical University of Lodz, 90-151 Lodz, Poland
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Dorota Pastuszak-Lewandoska
- Department of Microbiology and Laboratory Medical Immunology, Medical University of Lodz, Pomorska 251, 90-151 Lodz, Poland;
| |
Collapse
|
3
|
Miller YE, Ghosh M, Merrick DT, Kubala B, Szabo E, Bengtson L, Kocherginsky M, Helenowski IB, Benante K, Schering T, Kim J, Kim H, Ha D, Bergan RC, Khan SA, Keith RL. Phase Ib trial of inhaled iloprost for the prevention of lung cancer with predictive and response biomarker assessment. Front Oncol 2023; 13:1204726. [PMID: 37711198 PMCID: PMC10499515 DOI: 10.3389/fonc.2023.1204726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/26/2023] [Indexed: 09/16/2023] Open
Abstract
Introduction Iloprost, a prostacyclin analog, has lung cancerpreventive activity in preclinical models and improved dysplasia in former smokers in a phase IIb trial. Oral iloprost is currently unavailable. We performed a phase Ib trial of inhaled iloprost in former smokers to assess tolerance and compliance. Methods Participants self-administered nebulized iloprost (5ug) or placebo four (QID) or two (BID) times daily. As QID dose was well tolerated and due to expiration of the placebo, the BID dosing and placebo were eliminated early on in the trial. Bronchoscopy with biopsyat six standard sites was performed at treatment initiation and two months post-iloprost, with exploratory histological analysis. Bulk RNA sequencing, single cell RNA sequencing and an in vitro assay of epithelial progenitor cell iloprost response were performed on a subset of biopsies in an exploratory investigation of response mechanisms and predictive biomarkers. Results and discussion Thirty-four of a planned 48 participants were recruited to the trial.Inhaled iloprost was well tolerated with no adverse events > grade 2. Compliance was 67% in the QID group. The trial was not powered to detect histologic response and none was found. Bulk RNA sequencing of biopsies pre/post iloprost suggest that iloprost is immunomodulatory and downregulates cell proliferation pathways. Single cell RNA sequencing showed an increase in CD8-positive T cells with upregulation of genes in interferon γ signaling. In vitro iloprost response by epithelial progenitor cells correlated with histologic response with kappa coefficient of 0.81 (95% CI 0.47, 1.0). Inhaled iloprost was well tolerated with suboptimal compliance. Molecular analysis suggested that iloprosthas immunomodulatory and antiproliferative effects.The progenitor cell iloprost response assay may be a promising avenue to develop predictive biomarkers. Clinical trial registration https://clinicaltrials.gov/study/NCT02237183, identifier NCT02237183.
Collapse
Affiliation(s)
- York E. Miller
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, CO, United States
- Pulmonary and Critical Care Section, RMR VAMC Rocky Mountain Regional Veteran Administration Medical Center, Aurora, CO, United States
| | - Moumita Ghosh
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, CO, United States
| | - Daniel T. Merrick
- Department of Pathology, University of Colorado, Aurora, CO, United States
| | - Brandi Kubala
- Cancer Center Clinical Trial Core, University of Colorado, Aurora, CO, United States
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Lisa Bengtson
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Masha Kocherginsky
- Department of Preventative Medicine, Northwestern University, Evanston, IL, United States
| | - Irene B. Helenowski
- Department of Preventative Medicine, Northwestern University, Evanston, IL, United States
| | - Kelly Benante
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, United States
| | - Tia Schering
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Evanston, IL, United States
| | - Jihye Kim
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States
| | - Hyunmin Kim
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Duc Ha
- Pulmonary and Critical Care Section, RMR VAMC Rocky Mountain Regional Veteran Administration Medical Center, Aurora, CO, United States
| | - Raymond C. Bergan
- Fred and Pamela Buffett Cancer Center, Division of Oncology & Hematology, Genitourinary Oncology, University of Nebraska, Evanston, IL, United States
| | - Seema A. Khan
- Department of Surgery, Northwestern University, Omaha, NE, United States
| | - Robert L. Keith
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Aurora, CO, United States
- Pulmonary and Critical Care Section, RMR VAMC Rocky Mountain Regional Veteran Administration Medical Center, Aurora, CO, United States
| |
Collapse
|
4
|
Li JM, Chang WH, Li L, Yang DC, Hsu SW, Kenyon NJ, Chen CH. Inositol possesses antifibrotic activity and mitigates pulmonary fibrosis. Respir Res 2023; 24:132. [PMID: 37194070 PMCID: PMC10189934 DOI: 10.1186/s12931-023-02421-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/13/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Myo-inositol (or inositol) and its derivatives not only function as important metabolites for multiple cellular processes but also act as co-factors and second messengers in signaling pathways. Although inositol supplementation has been widely studied in various clinical trials, little is known about its effect on idiopathic pulmonary fibrosis (IPF). Recent studies have demonstrated that IPF lung fibroblasts display arginine dependency due to loss of argininosuccinate synthase 1 (ASS1). However, the metabolic mechanisms underlying ASS1 deficiency and its functional consequence in fibrogenic processes are yet to be elucidated. METHODS Metabolites extracted from primary lung fibroblasts with different ASS1 status were subjected to untargeted metabolomics analysis. An association of ASS1 deficiency with inositol and its signaling in lung fibroblasts was assessed using molecular biology assays. The therapeutic potential of inositol supplementation in fibroblast phenotypes and lung fibrosis was evaluated in cell-based studies and a bleomycin animal model, respectively. RESULTS Our metabolomics studies showed that ASS1-deficient lung fibroblasts derived from IPF patients had significantly altered inositol phosphate metabolism. We observed that decreased inositol-4-monophosphate abundance and increased inositol abundance were associated with ASS1 expression in fibroblasts. Furthermore, genetic knockdown of ASS1 expression in primary normal lung fibroblasts led to the activation of inositol-mediated signalosomes, including EGFR and PKC signaling. Treatment with inositol significantly downregulated ASS1 deficiency-mediated signaling pathways and reduced cell invasiveness in IPF lung fibroblasts. Notably, inositol supplementation also mitigated bleomycin-induced fibrotic lesions and collagen deposition in mice. CONCLUSION These findings taken together demonstrate a novel function of inositol in fibrometabolism and pulmonary fibrosis. Our study provides new evidence for the antifibrotic activity of this metabolite and suggests that inositol supplementation may be a promising therapeutic strategy for IPF.
Collapse
Affiliation(s)
- Ji-Min Li
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Wen-Hsin Chang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Linhui Li
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - David C Yang
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Ssu-Wei Hsu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA
| | - Ching-Hsien Chen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, CA, USA.
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA, 95616, USA.
| |
Collapse
|
5
|
Guarnieri G, Iervolino M, Cavallone S, Unfer V, Vianello A. The "Asthma-Polycystic Ovary Overlap Syndrome" and the Therapeutic Role of Myo-Inositol. Int J Mol Sci 2023; 24:ijms24086959. [PMID: 37108123 PMCID: PMC10138395 DOI: 10.3390/ijms24086959] [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: 02/21/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Asthma is a heterogeneous inflammatory disease characterized by abnormalities in immune response. Due to the inherent complexity of the disease and the presence of comorbidities, asthma control is often difficult to obtain. In asthmatic patients, an increased prevalence of irregular menstrual cycles, infertility, obesity, and insulin resistance has been reported. Given that these conditions are also common in patients with polycystic ovary syndrome (PCOS), we propose the definition of "asthma-PCOS overlap syndrome" to indicate a medical condition which shares characteristics of both diseases. The aim of this review is to analyze the links between asthma and PCOS and evaluate the therapeutic role of myo-inositol, a natural compound currently utilized in patients with PCOS, in the management of asthma patients.
Collapse
Affiliation(s)
- Gabriella Guarnieri
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | | | | | - Vittorio Unfer
- Systems Biology Group Laboratory, 00163 Rome, Italy
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), 00161 Rome, Italy
| | - Andrea Vianello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| |
Collapse
|
6
|
Pujol A, Sanchis P, Grases F, Masmiquel L. Phytate Intake, Health and Disease: "Let Thy Food Be Thy Medicine and Medicine Be Thy Food". Antioxidants (Basel) 2023; 12:antiox12010146. [PMID: 36671007 PMCID: PMC9855079 DOI: 10.3390/antiox12010146] [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: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Phytate (myo-inositol hexakisphosphate or InsP6) is the main phosphorus reservoir that is present in almost all wholegrains, legumes, and oilseeds. It is a major component of the Mediterranean and Dietary Approaches to Stop Hypertension (DASH) diets. Phytate is recognized as a nutraceutical and is classified by the Food and Drug Administration (FDA) as Generally Recognized As Safe (GRAS). Phytate has been shown to be effective in treating or preventing certain diseases. Phytate has been shown to inhibit calcium salt crystallization and, therefore, to reduce vascular calcifications, calcium renal calculi and soft tissue calcifications. Moreover, the adsorption of phytate to the crystal faces can inhibit hydroxyapatite dissolution and bone resorption, thereby playing a role in the treatment/prevention of bone mass loss. Phytate has a potent antioxidation and anti-inflammatory action. It is capable of inhibiting lipid peroxidation through iron chelation, reducing iron-related free radical generation. As this has the effect of mitigating neuronal damage and loss, phytate shows promise in the treatment/prevention of neurodegenerative disease. It is reported that phytate improves lipid and carbohydrate metabolism, increases adiponectin, decreases leptin and reduces protein glycation, which is linked with macrovascular and microvascular diabetes complications. In this review, we summarize the benefits of phytate intake as seen in in vitro, animal model, epidemiological and clinical trials, and we also identify questions to answer in the future.
Collapse
Affiliation(s)
- Antelm Pujol
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07198 Palma de Mallorca, Spain
- Correspondence: (A.P.); (L.M.)
| | - Pilar Sanchis
- Laboratory of Renal Lithiasis Research, Deptartment of Chemistry, University of Balearic Islands, Health Research Institute of Balearic Islands, (IdISBa), 07122 Palma de Mallorca, Spain
| | - Felix Grases
- Laboratory of Renal Lithiasis Research, Deptartment of Chemistry, University of Balearic Islands, Health Research Institute of Balearic Islands, (IdISBa), 07122 Palma de Mallorca, Spain
| | - Luis Masmiquel
- Vascular and Metabolic Diseases Research Group, Endocrinology Department, Son Llàtzer University Hospital, Health Research Institute of the Balearic Islands (IdISBa), 07198 Palma de Mallorca, Spain
- Correspondence: (A.P.); (L.M.)
| |
Collapse
|
7
|
Keith RL, Miller YE, Ghosh M, Franklin WA, Nakachi I, Merrick DT. Lung cancer: Premalignant biology and medical prevention. Semin Oncol 2022; 49:254-260. [PMID: 35305831 DOI: 10.1053/j.seminoncol.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 11/11/2022]
Abstract
Lung cancer (both adenocarcinoma and squamous cell) progress through a series of pre-malignant histologic changes before the development of invasive disease. Each of these carcinogenic cascades is defined by genetic and epigenetic alterations in pulmonary epithelial cells. Additionally, alterations in the immune response, progenitor cell function, mutational burden, and microenvironmental mediated survival of mutated clones contribute to the risk of pre-malignant lesions progressing to cancer. Medical preventions studies have been completed and current and future trials are informed by the improved understanding of pre-malignancy. This will lead to precision chemoprevention trials based on lesional biology and histologic characteristics.
Collapse
Affiliation(s)
- R L Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO.
| | - Y E Miller
- Division of Pulmonary Sciences and Critical Care Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - M Ghosh
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - I Nakachi
- Department of Pulmonary Medicine, Keio University, Tokyo, Japan
| | - D T Merrick
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
8
|
Kassie F, Jian Bagherpoor A, Kovacs K, Seelig D. Combinatory lung tumor inhibition by myo-inositol and iloprost/rapamycin: association with immunomodulation. Carcinogenesis 2022; 43:547-556. [PMID: 35147705 DOI: 10.1093/carcin/bgac019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/06/2022] [Accepted: 02/09/2022] [Indexed: 11/12/2022] Open
Abstract
Although both preclinical and clinical studies have suggested that myo-inositol (MI) may be a safe and effective lung cancer chemopreventive agent, its efficacy is moderate. To test whether the chemopreventive agents iloprost (IL) or rapamycin enhance the lung tumor inhibitory effects of MI, A/J mice were treated with the tobacco smoke carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and, beginning one week after the end of NNK treatment, given MI, IL, rapamycin, MI+IL or MI+rapamycin for 17 weeks. Analyses of the number and size of tumors on the surface of the lung have indicated that MI, IL, rapamycin, MI+IL and MI+rapamycin reduced the multiplicity of NNK-induced lung tumors by 41%, 34%, 46%, 79% and 67%, respectively, and larger tumors (lung tumors with a diameter of 1-2 mm or > 2 mm) were absent in the MI+IL and MI+rapamycin groups. These results clearly indicated that MI+IL and MI+rapamycin are more effective than MI alone in inhibiting the formation and growth of lung tumors. Assessment of the immunomodulatory effects of the drugs showed that whereas MI+rapamycin and MI+IL increased the infiltration of lung tumors by CD4 + and CD8 + T cells, MI+rapamycin reduced the expression of the immune checkpoint protein programmed-death ligand-1 (PD-L1). Moreover, all treatments, except IL, increased apoptosis, whereas cell proliferation was markedly suppressed in all treated groups. In summary, these results suggest that IL and rapamycin could enhance the efficacy of MI in lung cancer chemoprevention trials.
Collapse
Affiliation(s)
- Fekadu Kassie
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | | | - Katalin Kovacs
- College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Davis Seelig
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.,College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
9
|
Geyer T, Rübenthaler J, Alunni-Fabbroni M, Schinner R, Weber S, Mayerle J, Schiffer E, Höckner S, Malfertheiner P, Ricke J. NMR-Based Lipid Metabolite Profiles to Predict Outcomes in Patients Undergoing Interventional Therapy for a Hepatocellular Carcinoma (HCC): A Substudy of the SORAMIC Trial. Cancers (Basel) 2021; 13:cancers13112787. [PMID: 34205110 PMCID: PMC8199928 DOI: 10.3390/cancers13112787] [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: 05/02/2021] [Revised: 05/25/2021] [Accepted: 05/31/2021] [Indexed: 11/18/2022] Open
Abstract
Simple Summary A hepatocellular carcinoma (HCC) is the most common cause of death in patients suffering from chronic liver diseases. In order to improve the prediction of outcomes in HCC patients, there is a need for new biomarkers. This pilot study aimed at identifying serum metabolites for the prediction of outcomes of HCC patients using nuclear magnetic resonance (NMR) spectroscopy. This analysis revealed that high serum concentrations of myo-inositol or dimethylamine were associated with an improved overall survival. In contrast, high concentrations of total cholesterol, LDL-cholesterol and LDL particles (LDL-P) were associated with a decreased overall survival. The identification of novel biomarkers using this NMR-based technology holds promise for opening new directions in the conduction of interventional trials in HCCs. Abstract Background: This exploratory study aimed to evaluate lipidomic and metabolomic profiles in patients with early and advanced HCCs and to investigate whether certain metabolic parameters may predict the overall survival in these patients. Methods: A total of 60 patients from the prospective, randomized-controlled, multicenter phase II SORAMIC trial were included in this substudy; among them were 30 patients with an early HCC who underwent radiofrequency ablation combined with sorafenib or a placebo and 30 patients with an advanced HCC who were treated with a selective internal radiation therapy (SIRT) plus sorafenib vs. sorafenib alone. The blood serum of these patients was analyzed using a standardized nuclear magnetic resonance (NMR) platform. All tested metabolites were correlated with the overall survival. Results: The overall survival (OS) was significantly higher in patients with an early HCC (median OS: 34.0 months) compared with patients with an advanced HCC (median OS: 12.0 months) (p < 0.0001). Patients with high serum concentrations of myo-inositol (MI) had a higher overall survival compared with patients with low concentrations (21.6 vs. 13.8 months) with a Pearson correlation coefficient of 0.331 (p = 0.011). Patients with high serum concentrations of dimethylamine had a higher overall survival compared with patients with low concentrations (25.1 vs. 19.7 months) with a Pearson correlation coefficient of 0.279 (p = 0.034). High concentrations of total cholesterol, LDL-cholesterol and LDL particles (LDL-P) were associated with a decreased overall survival. Conclusions: NMR-based lipidomic and metabolomic profiling has the potential to identify individual metabolite biomarkers that predict the outcome of patients with an HCC exposed to non-invasive therapeutic management.
Collapse
Affiliation(s)
- Thomas Geyer
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.R.); (M.A.-F.); (R.S.); (P.M.); (J.R.)
- Correspondence: ; Tel.: +49-89-4400-73620
| | - Johannes Rübenthaler
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.R.); (M.A.-F.); (R.S.); (P.M.); (J.R.)
| | - Marianna Alunni-Fabbroni
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.R.); (M.A.-F.); (R.S.); (P.M.); (J.R.)
| | - Regina Schinner
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.R.); (M.A.-F.); (R.S.); (P.M.); (J.R.)
| | - Sabine Weber
- Department of Medicine II, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (S.W.); (J.M.)
| | - Julia Mayerle
- Department of Medicine II, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (S.W.); (J.M.)
| | - Eric Schiffer
- Numares AG, Am BioPark 9, 93053 Regensburg, Germany; (E.S.); (S.H.)
| | | | - Peter Malfertheiner
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.R.); (M.A.-F.); (R.S.); (P.M.); (J.R.)
- Department of Medicine II, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (S.W.); (J.M.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany; (J.R.); (M.A.-F.); (R.S.); (P.M.); (J.R.)
| |
Collapse
|
10
|
Wiśniewski K, Jozwik M, Wojtkiewicz J. Cancer Prevention by Natural Products Introduced into the Diet-Selected Cyclitols. Int J Mol Sci 2020; 21:E8988. [PMID: 33256104 PMCID: PMC7729485 DOI: 10.3390/ijms21238988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 12/23/2022] Open
Abstract
Cancer is now the second leading cause of death worldwide. It is estimated that every year, approximately 9.6 million people die of oncologic diseases. The most common origins of malignancy are the lungs, breasts, and colorectum. Even though in recent years, many new drugs and therapeutic options have been introduced, there are still no safe, effective chemopreventive agents. Cyclitols seem poised to improve this situation. There is a body of evidence that suggests that their supplementation can decrease the incidence of colorectal cancer, lower the risk of metastasis occurrence, lower the proliferation index, induce apoptosis in malignant cells, enhance natural killer (NK) cell activity, protect cells from free radical damage, and induce positive molecular changes, as well as reduce the side effects of anticancer treatments such as chemotherapy or surgery. Cyclitol supplementation appears to be both safe and well-tolerated. This review focuses on presenting, in a comprehensive way, the currently available knowledge regarding the use of cyclitols in the treatment of different malignancies, particularly in lung, breast, colorectal, and prostate cancers.
Collapse
Affiliation(s)
- Karol Wiśniewski
- Department Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland;
| | - Marcin Jozwik
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum University of Warmia and Mazury, 10-561 Olsztyn, Poland;
| | - Joanna Wojtkiewicz
- Department Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland;
| |
Collapse
|
11
|
Laganà AS, Unfer V, Garzon S, Bizzarri M. Role of inositol to improve surfactant functions and reduce IL-6 levels: A potential adjuvant strategy for SARS-CoV-2 pneumonia? Med Hypotheses 2020; 144:110262. [PMID: 33254564 PMCID: PMC7480225 DOI: 10.1016/j.mehy.2020.110262] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 08/30/2020] [Accepted: 09/05/2020] [Indexed: 01/18/2023]
Abstract
To date, the spread of SARS-CoV-2 infection is increasing worldwide and represents a primary healthcare emergency. Although the infection can be asymptomatic, several cases develop severe pneumonia and acute respiratory distress syndrome (ARDS) characterized by high levels of pro-inflammatory cytokines, primarily interleukin (IL)-6. Based on available data, the severity of ARDS and serum levels of IL-6 are key determinants for the prognosis. In this scenario, available in vitro and in vivo data suggested that myo-inositol is able to increase the synthesis and function of the surfactant phosphatidylinositol, acting on the phosphoinositide 3-kinase (PI3K)-regulated signaling, with amelioration of both immune system and oxygenation at the bronchoalveolar level. In addition, myo-inositol has been found able to decrease the levels of IL-6 in several experimental settings, due to an effect on the inositol-requiring enzyme 1 (IRE1)-X-box-binding protein 1 (XBP1) and on the signal transducer and activator of transcription 3 (STAT3) pathways. In this scenario, treatment with myo-inositol may be able to reduce IL-6 dependent inflammatory response and improve oxygenation in patients with severe ARDS by SARS-CoV-2. In addition, the action of myo-inositol on IRE1 endonuclease activity may also inhibit the replication of SARS-CoV-2, as was reported for the respiratory syncytial virus. Since the available data are extremely limited, if this potential therapeutic approach will be considered valid in the clinical practice, the necessary future investigations should aim to identify the best dose, administration route (oral, intravenous and/or aerosol nebulization), and cluster(s) of patients which may get beneficial effects from this treatment.
Collapse
Affiliation(s)
- Antonio Simone Laganà
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy; The Experts Group on Inositol in Basic and Clinical Research (EGOI), Italy(1).
| | - Vittorio Unfer
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), Italy(1); Systems Biology Group Lab, "La Sapienza" University, Rome, Italy
| | - Simone Garzon
- Department of Obstetrics and Gynecology, "Filippo Del Ponte" Hospital, University of Insubria, Varese, Italy
| | - Mariano Bizzarri
- The Experts Group on Inositol in Basic and Clinical Research (EGOI), Italy(1); Systems Biology Group Lab, "La Sapienza" University, Rome, Italy; Department of Experimental Medicine, "La Sapienza" University, Rome, Italy
| |
Collapse
|
12
|
Recent Advances and the Potential for Clinical Use of Autofluorescence Detection of Extra-Ophthalmic Tissues. Molecules 2020; 25:molecules25092095. [PMID: 32365790 PMCID: PMC7248908 DOI: 10.3390/molecules25092095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
Abstract
The autofluorescence (AF) characteristics of endogenous fluorophores allow the label-free assessment and visualization of cells and tissues of the human body. While AF imaging (AFI) is well-established in ophthalmology, its clinical applications are steadily expanding to other disciplines. This review summarizes clinical advances of AF techniques published during the past decade. A systematic search of the MEDLINE database and Cochrane Library databases was performed to identify clinical AF studies in extra-ophthalmic tissues. In total, 1097 articles were identified, of which 113 from internal medicine, surgery, oral medicine, and dermatology were reviewed. While comparable technological standards exist in diabetology and cardiology, in all other disciplines, comparability between studies is limited due to the number of differing AF techniques and non-standardized imaging and data analysis. Clear evidence was found for skin AF as a surrogate for blood glucose homeostasis or cardiovascular risk grading. In thyroid surgery, foremost, less experienced surgeons may benefit from the AF-guided intraoperative separation of parathyroid from thyroid tissue. There is a growing interest in AF techniques in clinical disciplines, and promising advances have been made during the past decade. However, further research and development are mandatory to overcome the existing limitations and to maximize the clinical benefits.
Collapse
|
13
|
Lippman SM, Abate-Shen C, Colbert Maresso KL, Colditz GA, Dannenberg AJ, Davidson NE, Disis ML, DuBois RN, Szabo E, Giuliano AR, Hait WN, Lee JJ, Kensler TW, Kramer BS, Limburg P, Maitra A, Martinez ME, Rebbeck TR, Schmitz KH, Vilar E, Hawk ET. AACR White Paper: Shaping the Future of Cancer Prevention - A Roadmap for Advancing Science and Public Health. Cancer Prev Res (Phila) 2019; 11:735-778. [PMID: 30530635 DOI: 10.1158/1940-6207.capr-18-0421] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 11/02/2018] [Indexed: 12/09/2022]
Abstract
The recent pace, extent, and impact of paradigm-changing cancer prevention science has been remarkable. The American Association for Cancer Research (AACR) convened a 3-day summit, aligned with five research priorities: (i) Precancer Atlas (PCA). (ii) Cancer interception. (iii) Obesity-cancer linkage, a global epidemic of chronic low-grade inflammation. (iv) Implementation science. (v) Cancer disparities. Aligned with these priorities, AACR co-led the Lancet Commission to formally endorse and accelerate the NCI Cancer Moonshot program, facilitating new global collaborative efforts in cancer control. The expanding scope of creative impact is perhaps most startling-from NCI-funded built environments to AACR Team Science Awarded studies of Asian cancer genomes informing global primary prevention policies; cell-free epigenetic marks identifying incipient neoplastic site; practice-changing genomic subclasses in myeloproliferative neoplasia (including germline variant tightly linked to JAK2 V617F haplotype); universal germline genetic testing for pancreatic cancer; and repurposing drugs targeting immune- and stem-cell signals (e.g., IL-1β, PD-1, RANK-L) to cancer interception. Microbiota-driven IL-17 can induce stemness and transformation in pancreatic precursors (identifying another repurposing opportunity). Notable progress also includes hosting an obesity special conference (connecting epidemiologic and molecular perspectives to inform cancer research and prevention strategies), co-leading concerted national implementation efforts in HPV vaccination, and charting the future elimination of cancer disparities by integrating new science tools, discoveries and perspectives into community-engaged research, including targeted counter attacks on e-cigarette ad exploitation of children, Hispanics and Blacks. Following this summit, two unprecedented funding initiatives were catalyzed to drive cancer prevention research: the NCI Cancer Moonshot (e.g., PCA and disparities); and the AACR-Stand Up To Cancer bold "Cancer Interception" initiative.
Collapse
Affiliation(s)
| | - Cory Abate-Shen
- Departments of Urology, Medicine, Systems Biology, and Pathology & Cell Biology, Institute of Cancer Genetics, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Karen L Colbert Maresso
- Division of Cancer Prevention & Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Graham A Colditz
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | | | - Nancy E Davidson
- Fred Hutchinson Cancer Center and University of Washington, Seattle, Washington
| | - Mary L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, Seattle, Washington
| | - Raymond N DuBois
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, NIH, Bethesda, Maryland
| | - Anna R Giuliano
- Center for Infection Research in Cancer, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - William N Hait
- Janssen Research and Development LLC., Raritan, New Jersey
| | - J Jack Lee
- Department of Biostatistics, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Thomas W Kensler
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Paul Limburg
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anirban Maitra
- Sheikh Ahmed Pancreatic Cancer Research Center, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Maria Elena Martinez
- Department of Family Medicine and Public Health, UC San Diego, LaJolla, California
| | - Timothy R Rebbeck
- Cancer Epidemiology & Cancer Risk and Disparity, Dana-Farber Cancer Institute, Boston, MA
| | | | - Eduardo Vilar
- Departments of Clinical Cancer Prevention and GI Medical Oncology, UT MD Anderson Cancer Center, Houston, TX
| | - Ernest T Hawk
- Division of Cancer Prevention & Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
14
|
Zhao Y, Johnson WE. Exploring Host-Microbe Interactions in Lung Cancer. Am J Respir Crit Care Med 2019; 198:1116-1118. [PMID: 30063838 DOI: 10.1164/rccm.201807-1225ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Yue Zhao
- 1 Division of Computational Biomedicine Boston University School of Medicine Boston, Massachusetts
| | - W Evan Johnson
- 1 Division of Computational Biomedicine Boston University School of Medicine Boston, Massachusetts
| |
Collapse
|
15
|
Keith RL, Blatchford PJ, Merrick DT, Bunn PA, Bagwell B, Dwyer-Nield LD, Jackson MK, Geraci MW, Miller YE. A Randomized Phase II Trial of Pioglitazone for Lung Cancer Chemoprevention in High-Risk Current and Former Smokers. Cancer Prev Res (Phila) 2019; 12:721-730. [PMID: 31308004 DOI: 10.1158/1940-6207.capr-19-0006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/01/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023]
Abstract
Lung cancer chemoprevention, especially in high-risk former smokers, has great potential to reduce lung cancer incidence and mortality. Thiazolidinediones prevent lung cancer in preclinical studies, and diabetics receiving thiazolidinediones have lower lung cancer rates which led to our double-blind, randomized, phase II placebo-controlled trial of oral pioglitazone in high-risk current or former smokers with sputum cytologic atypia or known endobronchial dysplasia. Bronchoscopy was performed at study entry and after completing 6 months of treatment. Biopsies were histologically scored, and primary endpoint analysis tested worst biopsy scores (Max) between groups; Dysplasia index (DI) and average score (Avg) changes were secondary endpoints. Biopsies also received an inflammation score. The trial accrued 92 subjects (47 pioglitazone, 45 placebo), and 76 completed both bronchoscopies (39 pioglitazone, 37 placebo). Baseline dysplasia was significantly worse for current smokers, and 64% of subjects had mild or greater dysplasia at study entry. Subjects receiving pioglitazone did not exhibit improvement in bronchial dysplasia. Former smokers treated with pioglitazone exhibited a slight improvement in Max, while current smokers exhibited slight worsening. While statistically significant changes in Avg and DI were not observed in the treatment group, former smokers exhibited a slight decrease in both Avg and DI. Negligible Avg and DI changes occurred in current smokers. A trend toward decreased Ki-67 labeling index occurred in former smokers with baseline dysplasia receiving pioglitazone. While pioglitazone did not improve endobronchial histology in this high-risk cohort, specific lesions showed histologic improvement, and further study is needed to better characterize responsive dysplasia.
Collapse
Affiliation(s)
- Robert L Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Eastern Colorado VA Healthcare System, Rocky Mountain Regional VA Medical Center, Aurora, Colorado. .,Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Patrick J Blatchford
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel T Merrick
- Division of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paul A Bunn
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brandi Bagwell
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lori D Dwyer-Nield
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mary K Jackson
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mark W Geraci
- Department of Medicine, IU School of Medicine, Indianapolis, Indiana
| | - York E Miller
- Division of Pulmonary Sciences and Critical Care Medicine, Eastern Colorado VA Healthcare System, Rocky Mountain Regional VA Medical Center, Aurora, Colorado.,Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
16
|
Premalignant lesions of squamous cell carcinoma of the lung: The molecular make-up and factors affecting their progression. Lung Cancer 2019; 135:21-28. [PMID: 31446997 DOI: 10.1016/j.lungcan.2019.07.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/23/2019] [Accepted: 07/01/2019] [Indexed: 01/06/2023]
Abstract
Squamous cell carcinoma (SCC), one of the most common forms of lung cancer, shows accelerated progression and aggressive growth and usually is observed at advanced stages. SCC originates from morphological changes in the bronchial epithelium that occur during chronic inflammation: basal cell hyperplasia, squamous metaplasia, and dysplasia I-III. However, the process is not inevitable; it can be stopped at any stage, remain in the stable state indefinitely and either progress or regress. The reasons and mechanisms of different scenarios of the evolution of premalignant lesions in the respiratory epithelium are not fully understood. In this review, we summarized the literature data (including our own data) regarding genetic, epigenetic, transcriptomic and proteomic profiles of the premalignant lesions and highlighted factors (environmental causes, inflammation, and gene polymorphism) that may govern their progression or regression. In conclusion, we reviewed strategies for lung cancer prevention and proposed new models and research directions for studying premalignant lesions and developing new tools to predict the risk of their malignant transformation.
Collapse
|
17
|
Beane JE, Mazzilli SA, Campbell JD, Duclos G, Krysan K, Moy C, Perdomo C, Schaffer M, Liu G, Zhang S, Liu H, Vick J, Dhillon SS, Platero SJ, Dubinett SM, Stevenson C, Reid ME, Lenburg ME, Spira AE. Molecular subtyping reveals immune alterations associated with progression of bronchial premalignant lesions. Nat Commun 2019; 10:1856. [PMID: 31015447 PMCID: PMC6478943 DOI: 10.1038/s41467-019-09834-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/28/2019] [Indexed: 12/13/2022] Open
Abstract
Bronchial premalignant lesions (PMLs) are precursors of lung squamous cell carcinoma, but have variable outcome, and we lack tools to identify and treat PMLs at risk for progression to cancer. Here we report the identification of four molecular subtypes of PMLs with distinct differences in epithelial and immune processes based on RNA-Seq profiling of endobronchial biopsies from high-risk smokers. The Proliferative subtype is enriched with bronchial dysplasia and exhibits up-regulation of metabolic and cell cycle pathways. A Proliferative subtype-associated gene signature identifies subjects with Proliferative PMLs from normal-appearing uninvolved large airway brushings with high specificity. In progressive/persistent Proliferative lesions expression of interferon signaling and antigen processing/presentation pathways decrease and immunofluorescence indicates a depletion of innate and adaptive immune cells compared with regressive lesions. Molecular biomarkers measured in PMLs or the uninvolved airway can enhance histopathological grading and suggest immunoprevention strategies for intercepting the progression of PMLs to lung cancer.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biopsy
- Bronchi/diagnostic imaging
- Bronchi/immunology
- Bronchi/pathology
- Bronchoscopy
- Carcinoma, Bronchogenic/genetics
- Carcinoma, Bronchogenic/immunology
- Carcinoma, Bronchogenic/pathology
- Carcinoma, Bronchogenic/prevention & control
- Cohort Studies
- Datasets as Topic
- Disease Progression
- Early Detection of Cancer/methods
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/immunology
- Gene Regulatory Networks/genetics
- Gene Regulatory Networks/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Cellular/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Mass Screening/methods
- Middle Aged
- Precancerous Conditions/diagnostic imaging
- Precancerous Conditions/genetics
- Precancerous Conditions/immunology
- Precancerous Conditions/pathology
- RNA, Messenger/genetics
- Respiratory Mucosa/cytology
- Respiratory Mucosa/diagnostic imaging
- Respiratory Mucosa/immunology
- Respiratory Mucosa/pathology
- Sequence Analysis, RNA
- T-Lymphocytes/immunology
- Tomography, X-Ray Computed
- Up-Regulation
Collapse
Affiliation(s)
| | | | | | - Grant Duclos
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Kostyantyn Krysan
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | | | | | - Gang Liu
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sherry Zhang
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hanqiao Liu
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Jessica Vick
- Boston University School of Medicine, Boston, MA, 02118, USA
| | | | | | - Steven M Dubinett
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | - Mary E Reid
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Marc E Lenburg
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Avrum E Spira
- Boston University School of Medicine, Boston, MA, 02118, USA
- Johnson and Johnson Innovation, Cambridge, MA, 02142, USA
| |
Collapse
|
18
|
Piranlioglu R, Korkaya H, Hassan KA. Dietary myo-inositol chemoprevents lung carcinogenesis via boosting immune system in Kras mouse model. J Thorac Dis 2019; 11:632-635. [PMID: 31019745 DOI: 10.21037/jtd.2019.02.37] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Raziye Piranlioglu
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Hasan Korkaya
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| | - Khaled A Hassan
- Rogel Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Xie YD, Zhao CQ, Wang JP, Cheng CS, Zhou JY, Zhang Y, Xia H, Wei L. Alcohol consumption analysis among patients with liver disease in China. Chin Med J (Engl) 2019; 132:420-430. [PMID: 30707167 PMCID: PMC6595713 DOI: 10.1097/cm9.0000000000000067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Alcohol consumption has been observed to be a contributing factor in liver damage. However, very few studies have tried to decipher the correlation between patients with liver disease and alcohol consumption. Therefore, this study was planned to determine the prevalence of alcohol consumption among patients with liver disease, and to evaluate the risk factors, liver diseases, and chronic medical conditions associated with alcohol drinking. METHODS A cross-sectional study was conducted among patients with liver disease in 30 provinces, autonomous regions, and municipalities across China. All participants answered the questionnaire, which led to the calculation of Alcohol Use Disorders Inventory Test (AUDIT) score for each patient. Based on this score, low-risk drinkers, hazardous drinkers, and harmful drinkers were defined as having AUDIT score of <8, between 8 and 15, and ≥16, respectively. RESULTS A total of 1489 participants completed the questionnaire. Based on this information, 900 (60.44%) participants were classified as alcohol drinkers. Among these, 8.66% were ex-drinkers, 22.10% were low-risk drinkers, 17.13% were hazardous drinkers, and 12.56% were harmful drinkers. Further investigation of the association between alcohol consumption and other baseline characteristics of patients with liver disease revealed that usually men <40 years old, participants having higher family annual income, having college degree or higher education, living alone, having higher body mass index (BMI), current smokers, and ex-smokers had significant association with higher risk of alcohol consumption. In addition, among the 18.07% of the participants with cirrhosis, it was observed that risk of cirrhosis increased with higher alcohol consumption. Furthermore, harmful drinkers showed greater odds of hypertension and heart diseases, while hazardous drinkers and harmful drinkers, both had greater odds of hyperlipidemia. CONCLUSIONS Overall our analyses indicated that among the patients with liver disease in China, there was high rate of alcohol consumption and dependence. Alcohol consumption usually associated with men <40 years old, higher family income, education level, living alone, high BMI, and smoking. Increased alcohol consumption not only increased the risk of cirrhosis, but also enhanced the risk of hypertension, heart diseases, and hyperlipidemia.
Collapse
Affiliation(s)
- Yan-Di Xie
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing 100044, China
| | - Chang-Qing Zhao
- Department of Liver Cirrhosis, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Jiu-Ping Wang
- Department of Infectious Disease, Xijing Hospital, the First Affiliated Hospital of the Air Force Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chang-Sheng Cheng
- Department of Hepatology, Guidong People's Hospital of Guangxi Zhuang Autonomous Region, Wuzhou, Guangxi 543001, China
| | - Jun-Ying Zhou
- Department of Infectious Disease, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, China
| | - Ying Zhang
- Sixth Department of Liver Disease, Dalian Sixth People's Hospital, Dalian Medical University, Dalian, Liaoning 116001, China
| | - Hong Xia
- Department of Infectious Disease, the Dazu District People's Hospital of Chongqing, Chongqing 402360, China
| | - Lai Wei
- Peking University People's Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing 100044, China
| |
Collapse
|
20
|
Tsay JCJ, Wu BG, Badri MH, Clemente JC, Shen N, Meyn P, Li Y, Yie TA, Lhakhang T, Olsen E, Murthy V, Michaud G, Sulaiman I, Tsirigos A, Heguy A, Pass H, Weiden MD, Rom WN, Sterman DH, Bonneau R, Blaser MJ, Segal LN. Airway Microbiota Is Associated with Upregulation of the PI3K Pathway in Lung Cancer. Am J Respir Crit Care Med 2018; 198:1188-1198. [PMID: 29864375 PMCID: PMC6221574 DOI: 10.1164/rccm.201710-2118oc] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
Abstract
RATIONALE In lung cancer, upregulation of the PI3K (phosphoinositide 3-kinase) pathway is an early event that contributes to cell proliferation, survival, and tissue invasion. Upregulation of this pathway was recently described as associated with enrichment of the lower airways with bacteria identified as oral commensals. OBJECTIVES We hypothesize that host-microbe interactions in the lower airways of subjects with lung cancer affect known cancer pathways. METHODS Airway brushings were collected prospectively from subjects with lung nodules at time of diagnostic bronchoscopy, including 39 subjects with final lung cancer diagnoses and 36 subjects with noncancer diagnoses. In addition, samples from 10 healthy control subjects were included. 16S ribosomal RNA gene amplicon sequencing and paired transcriptome sequencing were performed on all airway samples. In addition, an in vitro model with airway epithelial cells exposed to bacteria/bacterial products was performed. MEASUREMENTS AND MAIN RESULTS The composition of the lower airway transcriptome in the patients with cancer was significantly different from the control subjects, which included up-regulation of ERK (extracellular signal-regulated kinase) and PI3K signaling pathways. The lower airways of patients with lung cancer were enriched for oral taxa (Streptococcus and Veillonella), which was associated with up-regulation of the ERK and PI3K signaling pathways. In vitro exposure of airway epithelial cells to Veillonella, Prevotella, and Streptococcus led to upregulation of these same signaling pathways. CONCLUSIONS The data presented here show that several transcriptomic signatures previously identified as relevant to lung cancer pathogenesis are associated with enrichment of the lower airway microbiota with oral commensals.
Collapse
Affiliation(s)
| | | | - Michelle H. Badri
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, New York
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nan Shen
- Department of Genetics and Genomic Sciences and Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter Meyn
- New York University Genomic Technology Center, New York, New York; and
| | - Yonghua Li
- Division of Pulmonary and Critical Care Medicine
| | - Ting-An Yie
- Division of Pulmonary and Critical Care Medicine
| | - Tenzin Lhakhang
- New York University Genomic Technology Center, New York, New York; and
| | - Evan Olsen
- Division of Pulmonary and Critical Care Medicine
| | - Vivek Murthy
- Division of Pulmonary and Critical Care Medicine
| | | | | | | | - Adriana Heguy
- New York University Genomic Technology Center, New York, New York; and
| | | | | | | | | | - Richard Bonneau
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, New York
- New York University Center for Data Science, New York, New York
| | - Martin J. Blaser
- Department of Medicine, New York University School of Medicine, New York, New York
| | | |
Collapse
|
21
|
Affiliation(s)
- Jae-Il Roh
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Sauter ER. Breast Cancer Prevention: Current Approaches and Future Directions. Eur J Breast Health 2018; 14:64-71. [PMID: 29774312 DOI: 10.5152/ejbh.2018.3978] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/25/2018] [Indexed: 11/22/2022]
Abstract
The topic of breast cancer prevention is very broad. All aspects of the topic, therefore, cannot be adequately covered in a single review. The objective of this review is to discuss strategies in current use to prevent breast cancer, as well as potential approaches that could be used in the future. This review does not discuss early detection strategies for breast cancer, including breast cancer screening. The breast is the most common site among women worldwide of noncutaneous cancer. Many clinical and genetic factors have been found to increase a woman's risk of developing the disease. Current strategies to decrease a woman's risk of developing breast cancer include primary prevention, such as avoiding tobacco, exogenous hormone use and excess exposure to ionizing radiation, maintaining a normal weight, exercise, breastfeeding, eating a healthy diet and minimizing alcohol intake. Chemoprevention medications are available for those at high risk, though they are underutilized in eligible women. Mastectomy and/or bilateral oophorectomy are reasonable strategies for women who have deleterious mutations in genes that dramatically increase the risk of developing cancer in either breast. There are a variety of strategies in development for the prevention of breast cancer. Personalized approaches to prevent breast cancer that are being developed focus on advances in precision medicine, knowledge of the immune system and the tumor microenvironment and their role in cancer development. Advances in our understanding of how breast cancer develops are allowing investigators to specifically target populations who are most likely to benefit. Additionally, prevention clinical trials are starting to evaluate multi-agent cancer prevention approaches, with the hope of improved efficacy over single agents. Finally, there is a push to increase the use of chemopreventive agents with proven efficacy, such as tamoxifen and raloxifene, in the prevention of breast cancer.
Collapse
Affiliation(s)
- Edward R Sauter
- University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
23
|
Unver N, Delgado O, Zeleke K, Cumpian A, Tang X, Caetano MS, Wang H, Katayama H, Yu H, Szabo E, Wistuba II, Moghaddam SJ, Hanash SM, Ostrin EJ. Reduced IL-6 levels and tumor-associated phospho-STAT3 are associated with reduced tumor development in a mouse model of lung cancer chemoprevention with myo-inositol. Int J Cancer 2017; 142:1405-1417. [PMID: 29134640 DOI: 10.1002/ijc.31152] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/12/2017] [Accepted: 10/26/2017] [Indexed: 01/30/2023]
Abstract
Several promising chemopreventive agents have for lung cancer emerged in preclinical models and in retrospective trials. These agents have been shown to modulate pathways altered in carcinogenesis and reduce markers of carcinogenesis in animal and cell culture models. Cancer-prone transgenic mice with oncogenic Kras expressed in the airway epithelium (CcspCre/+ ; KrasLSL-G12D/+ ) were raised on diets compounded with myo-inositol. These animals form lung premalignant lesions in a stereotypical fashion over the ten weeks following weaning. Mice raised on myo-inositol containing diets showed potent reduction in the number, size, and stage of lesions as compared to those raised on control diets. myo-inositol has previously been reported to inhibit phosphoinositide 3-kinase (PI3K) signaling. However, in mice raised on myo-inositol, total PI3K signaling was largely unaffected. Proteomic and cytokine analyses revealed large reduction in IL-6 related pathways, including STAT3 phosphorylation. This effect was not due to direct inhibition of IL-6 production and autocrine signaling within the tumor cell, but rather through alteration in macrophage recruitment and in phenotype switching, with an increase in antitumoral M1 macrophages.
Collapse
Affiliation(s)
- Nese Unver
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Oliver Delgado
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Kirubel Zeleke
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Amber Cumpian
- Department of Pulmonary Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Ximing Tang
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Mauricio S Caetano
- Department of Pulmonary Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Hong Wang
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Hua Yu
- Department of Immuno-Oncology, City of Hope, Duarte, CA, 91010
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, 20892
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| | - Edwin J Ostrin
- Department of Pulmonary Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030.,Department of General Internal Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030
| |
Collapse
|
24
|
Beane J, Campbell JD, Lel J, Vick J, Spira A. Genomic approaches to accelerate cancer interception. Lancet Oncol 2017; 18:e494-e502. [PMID: 28759388 DOI: 10.1016/s1470-2045(17)30373-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 12/13/2022]
Abstract
Although major advances have been reported in the last decade in the treatment of late-stage cancer with targeted and immune-based therapies, there is a crucial unmet need to develop new approaches to improve the prevention and early detection of cancer. Advances in genomics and computational biology offer unprecedented opportunities to understand the earliest molecular events associated with carcinogenesis, enabling novel strategies to intercept the development of invasive cancers. This Series paper will highlight emerging big data genomic approaches with the potential to accelerate advances in cancer prevention, screening, and early detection across various tumour types, and the challenges inherent in the development of these tools for clinical use. Through coordinated multicentre consortia, these genomic approaches are likely to transform the landscape of cancer interception in the coming years.
Collapse
Affiliation(s)
- Jennifer Beane
- Department of Medicine and BU-BMC Cancer Center, Boston University, Boston, MA, USA
| | - Joshua D Campbell
- Department of Medicine and BU-BMC Cancer Center, Boston University, Boston, MA, USA
| | - Julian Lel
- Department of Medicine and BU-BMC Cancer Center, Boston University, Boston, MA, USA
| | - Jessica Vick
- Department of Medicine and BU-BMC Cancer Center, Boston University, Boston, MA, USA
| | - Avrum Spira
- Department of Medicine and BU-BMC Cancer Center, Boston University, Boston, MA, USA.
| |
Collapse
|
25
|
Vachani A, Sequist LV, Spira A. AJRCCM: 100-Year Anniversary. The Shifting Landscape for Lung Cancer: Past, Present, and Future. Am J Respir Crit Care Med 2017; 195:1150-1160. [PMID: 28459327 PMCID: PMC5439022 DOI: 10.1164/rccm.201702-0433ci] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 03/16/2017] [Indexed: 12/13/2022] Open
Abstract
The past century has witnessed a transformative shift in lung cancer from a rare reportable disease to the leading cause of cancer death among men and women worldwide. This historic shift reflects the increase in tobacco consumption worldwide, spurring public health efforts over the past several decades directed at tobacco cessation and control. Although most lung cancers are still diagnosed at a late stage, there have been significant advances in screening high-risk smokers, diagnostic modalities, and chemopreventive approaches. Improvements in surgery and radiation are advancing our ability to manage early-stage disease, particularly among patients considered unfit for traditional open resection. Arguably, the most dramatic progress has occurred on the therapeutic side, with the development of targeted and immune-based therapy over the past decade. This article reviews the major shifts in the lung cancer landscape over the past 100 years. Although many ongoing clinical challenges remain, this review will also highlight emerging molecular and imaging-based approaches that represent opportunities to transform the prevention, early detection, and treatment of lung cancer in the years ahead.
Collapse
Affiliation(s)
- Anil Vachani
- Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lecia V. Sequist
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Avrum Spira
- Section of Computational Biomedicine, Department of Medicine, Boston University Medical Center, Boston, Massachusetts
| |
Collapse
|
26
|
Spira A, Yurgelun MB, Alexandrov L, Rao A, Bejar R, Polyak K, Giannakis M, Shilatifard A, Finn OJ, Dhodapkar M, Kay NE, Braggio E, Vilar E, Mazzilli SA, Rebbeck TR, Garber JE, Velculescu VE, Disis ML, Wallace DC, Lippman SM. Precancer Atlas to Drive Precision Prevention Trials. Cancer Res 2017; 77:1510-1541. [PMID: 28373404 PMCID: PMC6681830 DOI: 10.1158/0008-5472.can-16-2346] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023]
Abstract
Cancer development is a complex process driven by inherited and acquired molecular and cellular alterations. Prevention is the holy grail of cancer elimination, but making this a reality will take a fundamental rethinking and deep understanding of premalignant biology. In this Perspective, we propose a national concerted effort to create a Precancer Atlas (PCA), integrating multi-omics and immunity - basic tenets of the neoplastic process. The biology of neoplasia caused by germline mutations has led to paradigm-changing precision prevention efforts, including: tumor testing for mismatch repair (MMR) deficiency in Lynch syndrome establishing a new paradigm, combinatorial chemoprevention efficacy in familial adenomatous polyposis (FAP), signal of benefit from imaging-based early detection research in high-germline risk for pancreatic neoplasia, elucidating early ontogeny in BRCA1-mutation carriers leading to an international breast cancer prevention trial, and insights into the intricate germline-somatic-immunity interaction landscape. Emerging genetic and pharmacologic (metformin) disruption of mitochondrial (mt) respiration increased autophagy to prevent cancer in a Li-Fraumeni mouse model (biology reproduced in clinical pilot) and revealed profound influences of subtle changes in mt DNA background variation on obesity, aging, and cancer risk. The elaborate communication between the immune system and neoplasia includes an increasingly complex cellular microenvironment and dynamic interactions between host genetics, environmental factors, and microbes in shaping the immune response. Cancer vaccines are in early murine and clinical precancer studies, building on the recent successes of immunotherapy and HPV vaccine immune prevention. Molecular monitoring in Barrett's esophagus to avoid overdiagnosis/treatment highlights an important PCA theme. Next generation sequencing (NGS) discovered age-related clonal hematopoiesis of indeterminate potential (CHIP). Ultra-deep NGS reports over the past year have redefined the premalignant landscape remarkably identifying tiny clones in the blood of up to 95% of women in their 50s, suggesting that potentially premalignant clones are ubiquitous. Similar data from eyelid skin and peritoneal and uterine lavage fluid provide unprecedented opportunities to dissect the earliest phases of stem/progenitor clonal (and microenvironment) evolution/diversity with new single-cell and liquid biopsy technologies. Cancer mutational signatures reflect exogenous or endogenous processes imprinted over time in precursors. Accelerating the prevention of cancer will require a large-scale, longitudinal effort, leveraging diverse disciplines (from genetics, biochemistry, and immunology to mathematics, computational biology, and engineering), initiatives, technologies, and models in developing an integrated multi-omics and immunity PCA - an immense national resource to interrogate, target, and intercept events that drive oncogenesis. Cancer Res; 77(7); 1510-41. ©2017 AACR.
Collapse
Affiliation(s)
- Avrum Spira
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew B Yurgelun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ludmil Alexandrov
- Theoretical Division, Center for Nonlinear Studies, Los Alamos National Laboratory, Los Alamos, New Mexico
| | - Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Rafael Bejar
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Madhav Dhodapkar
- Department of Hematology and Immunology, Yale Cancer Center, New Haven, Connecticut
| | - Neil E Kay
- Department of Hematology, Mayo Clinic Hospital, Rochester, Minnesota
| | - Esteban Braggio
- Department of Hematology, Mayo Clinic Hospital, Phoenix, Arizona
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah A Mazzilli
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology and Bioinformatics, Boston University School of Medicine, Boston, Massachusetts
| | - Timothy R Rebbeck
- Division of Hematology and Oncology, Dana-Farber Cancer Institute and Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Victor E Velculescu
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
- Department of Pathology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Mary L Disis
- Department of Medicine, Center for Translational Medicine in Women's Health, University of Washington, Seattle, Washington
| | - Douglas C Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott M Lippman
- Department of Medicine, Moores Cancer Center, University of California San Diego, La Jolla, California.
| |
Collapse
|