101
|
Fernández-García M, Rey-Stolle F, Boccard J, Reddy VP, García A, Cumming BM, Steyn AJC, Rudaz S, Barbas C. Comprehensive Examination of the Mouse Lung Metabolome Following Mycobacterium tuberculosis Infection Using a Multiplatform Mass Spectrometry Approach. J Proteome Res 2020; 19:2053-2070. [PMID: 32285670 PMCID: PMC7199213 DOI: 10.1021/acs.jproteome.9b00868] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Indexed: 02/08/2023]
Abstract
The mechanisms whereby Mycobacterium tuberculosis (Mtb) rewires the host metabolism in vivo are surprisingly unexplored. Here, we used three high-resolution mass spectrometry platforms to track altered lung metabolic changes associated with Mtb infection of mice. The multiplatform data sets were merged using consensus orthogonal partial least squares-discriminant analysis (cOPLS-DA), an algorithm that allows for the joint interpretation of the results from a single multivariate analysis. We show that Mtb infection triggers a temporal and progressive catabolic state to satisfy the continuously changing energy demand to control infection. This causes dysregulation of metabolic and oxido-reductive pathways culminating in Mtb-associated wasting. Notably, high abundances of trimethylamine-N-oxide (TMAO), produced by the host from the bacterial metabolite trimethylamine upon infection, suggest that Mtb could exploit TMAO as an electron acceptor under anaerobic conditions. Overall, these new pathway alterations advance our understanding of the link between Mtb pathogenesis and metabolic dysregulation and could serve as a foundation for new therapeutic intervention strategies. Mass spectrometry data has been deposited in the Metabolomics Workbench repository (data-set identifier: ST001328).
Collapse
Affiliation(s)
- Miguel Fernández-García
- Centro
de Metabolómica y Bioanálisis (CEMBIO), Facultad de
Farmacia, Universidad San Pablo-CEU, CEU
Universities, Urbanización Montepríncipe, Boadilla del Monte 28660, Spain
| | - Fernanda Rey-Stolle
- Centro
de Metabolómica y Bioanálisis (CEMBIO), Facultad de
Farmacia, Universidad San Pablo-CEU, CEU
Universities, Urbanización Montepríncipe, Boadilla del Monte 28660, Spain
| | - Julien Boccard
- School
of Pharmaceutical Sciences, University of
Lausanne and University of Geneva, Geneva 1211, Switzerland
| | - Vineel P. Reddy
- Department
of Microbiology, University of Alabama at
Birmingham, Birmingham, Alabama 35294, United States
| | - Antonia García
- Centro
de Metabolómica y Bioanálisis (CEMBIO), Facultad de
Farmacia, Universidad San Pablo-CEU, CEU
Universities, Urbanización Montepríncipe, Boadilla del Monte 28660, Spain
| | | | - Adrie J. C. Steyn
- Department
of Microbiology, University of Alabama at
Birmingham, Birmingham, Alabama 35294, United States
- Africa
Health Research Institute, Durban 4001, South Africa
- UAB
Centers for AIDS Research and Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294, United States
| | - Serge Rudaz
- School
of Pharmaceutical Sciences, University of
Lausanne and University of Geneva, Geneva 1211, Switzerland
| | - Coral Barbas
- Centro
de Metabolómica y Bioanálisis (CEMBIO), Facultad de
Farmacia, Universidad San Pablo-CEU, CEU
Universities, Urbanización Montepríncipe, Boadilla del Monte 28660, Spain
| |
Collapse
|
102
|
Nitrogen Metabolism in Cancer and Immunity. Trends Cell Biol 2020; 30:408-424. [PMID: 32302552 DOI: 10.1016/j.tcb.2020.02.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/16/2022]
Abstract
As one of the fundamental requirements for cell growth and proliferation, nitrogen acquisition and utilization must be tightly regulated. Nitrogen can be generated from amino acids (AAs) and utilized for biosynthetic processes through transamination and deamination reactions. Importantly, limitations of nitrogen availability in cells can disrupt the synthesis of proteins, nucleic acids, and other important nitrogen-containing compounds. Rewiring cellular metabolism to support anabolic processes is a feature common to both cancer and proliferating immune cells. In this review, we discuss how nitrogen is utilized in biosynthetic pathways and highlight different metabolic and oncogenic programs that alter the flow of nitrogen to sustain biomass production and growth, an important emerging feature of cancer and immune cell proliferation.
Collapse
|
103
|
Metabolomics analysis of human acute graft-versus-host disease reveals changes in host and microbiota-derived metabolites. Nat Commun 2019; 10:5695. [PMID: 31836702 PMCID: PMC6910937 DOI: 10.1038/s41467-019-13498-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 11/13/2019] [Indexed: 02/08/2023] Open
Abstract
Despite improvement in clinical management, allogeneic hematopoietic stem cell transplantation (HSCT) is still hampered by high morbidity and mortality rates, mainly due to graft versus host disease (GvHD). Recently, it has been demonstrated that the allogeneic immune response might be influenced by external factors such as tissues microenvironment or host microbiota. Here we used high throughput metabolomics to analyze two cohorts of genotypically HLA-identical related recipient and donor pairs. Metabolomic profiles markedly differ between recipients and donors. At the onset of acute GvHD, in addition to host-derived metabolites, we identify significant variation in microbiota-derived metabolites, especially in aryl hydrocarbon receptor (AhR) ligands, bile acids and plasmalogens. Altogether, our findings support that the allogeneic immune response during acute GvHD might be influenced by bile acids and by the decreased production of AhR ligands by microbiota that could limit indoleamine 2,3-dioxygenase induction and influence allogeneic T cell reactivity. Graft versus host disease (GvHD) still hinders allogeneic hematopoietic stem cell transplantation. Here, the authors use metabolomics to analyze two cohorts of paired transplant recipients and donors, identifying significant differences in both host- and microbiota-derived metabolites.
Collapse
|
104
|
Sanderson SM, Gao X, Dai Z, Locasale JW. Methionine metabolism in health and cancer: a nexus of diet and precision medicine. Nat Rev Cancer 2019; 19:625-637. [PMID: 31515518 DOI: 10.1038/s41568-019-0187-8] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2019] [Indexed: 01/11/2023]
Abstract
Methionine uptake and metabolism is involved in a host of cellular functions including methylation reactions, redox maintenance, polyamine synthesis and coupling to folate metabolism, thus coordinating nucleotide and redox status. Each of these functions has been shown in many contexts to be relevant for cancer pathogenesis. Intriguingly, the levels of methionine obtained from the diet can have a large effect on cellular methionine metabolism. This establishes a link between nutrition and tumour cell metabolism that may allow for tumour-specific metabolic vulnerabilities that can be influenced by diet. Recently, a number of studies have begun to investigate the molecular and cellular mechanisms that underlie the interaction between nutrition, methionine metabolism and effects on health and cancer.
Collapse
Affiliation(s)
- Sydney M Sanderson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Xia Gao
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Ziwei Dai
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
105
|
Guo T, Li B, Gu C, Chen X, Han M, Liu X, Xu C. PGC-1α inhibits polyamine metabolism in Cyclin E1-driven ovarian cancer. Cancer Med 2019; 8:7754-7761. [PMID: 31657115 PMCID: PMC6912055 DOI: 10.1002/cam4.2637] [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: 04/14/2019] [Revised: 10/03/2019] [Accepted: 10/10/2019] [Indexed: 12/16/2022] Open
Abstract
Aim Cyclin E1‐driven ovarian cancer (OvCa) is characterized with metabolic shift. In this study, we aim to pinpoint the metabolic pathway altered and assess its therapeutic potential. Methods In silico reproduction of TCGA ovarian cancer dataset and functional annotation using GSEA was performed. Candidate metabolic pathway was validated using in vitro and in vivo assays. Results From TCGA database, we found that polyamine metabolism was significantly enriched in Cyclin E1‐driven OvCa. Expressions of SMS, SRM, and ODC1 were positively correlated with that of CCNE1, respectively. ODC1 and SMS expressions were significantly correlated with decreased immune infiltrates. PGC‐1α silencing significantly decreased invasion and migration in both OvCa cell lines. Both spermidine and spermine levels were significantly increased when PGC‐1α was silenced. Targeting SRM significantly decreased spermine level in OVCAR3 cells, which was rescued when PGC‐1α was silenced. Silencing of PGC‐1α resulted in increased SRM in both OvCa cells. Dinaciclib significantly decreased invasion and migration of OVCAR3 cells. Expressions of PD‐L1 and PD‐L2 were predominantly in tumor‐infiltrating lymphocytes. Dinaciclib showed no notable effect of PD‐1 yet substantially induced the increased levels of PD‐L1 and PD‐L2. Conclusion Cyclin E1‐driven OvCa is characterized with activated polyamine synthesis, which is associated with decreased cancer immunity. Targeting polyamine and CDK2 may therefore sensitize this genotype to immune checkpoint blockade.
Collapse
Affiliation(s)
- Ting Guo
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| | - Bin Li
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| | - Chao Gu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| | - Xiuying Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| | - Mengxin Han
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| | - Xiaocheng Liu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| | - Congjian Xu
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, PR China
| |
Collapse
|
106
|
Zhang C, Aldrees M, Arif M, Li X, Mardinoglu A, Aziz MA. Elucidating the Reprograming of Colorectal Cancer Metabolism Using Genome-Scale Metabolic Modeling. Front Oncol 2019; 9:681. [PMID: 31417867 PMCID: PMC6682621 DOI: 10.3389/fonc.2019.00681] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/10/2019] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer is the third most incidental cancer worldwide, and the response rate of current treatment for colorectal cancer is very low. Genome-scale metabolic models (GEMs) are systems biology platforms, and they had been used to assist researchers in understanding the metabolic alterations in different types of cancer. Here, we reconstructed a generic colorectal cancer GEM by merging 374 personalized GEMs from the Human Pathology Atlas and used it as a platform for systematic investigation of the difference between tumor and normal samples. The reconstructed model revealed the metabolic reprogramming in glutathione as well as the arginine and proline metabolism in response to tumor occurrence. In addition, six genes including ODC1, SMS, SRM, RRM2, SMOX, and SAT1 associated with arginine and proline metabolism were found to be key players in this metabolic alteration. We also investigated these genes in independent colorectal cancer patients and cell lines and found that many of these genes showed elevated level in colorectal cancer and exhibited adverse effect in patients. Therefore, these genes could be promising therapeutic targets for treatment of a specific colon cancer patient group.
Collapse
Affiliation(s)
- Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Mohammed Aldrees
- Department of Medical Genomics, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud Bin Abdul Aziz University for Health Sciences, Riyadh, Saudi Arabia
- Ministry of the National Guard- Health Affairs, Riyadh, Saudi Arabia
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Centre for Host–Microbiome Interactions, Dental Institute, King's College London, London, United Kingdom
| | - Mohammad Azhar Aziz
- King Saud Bin Abdul Aziz University for Health Sciences, Riyadh, Saudi Arabia
- Ministry of the National Guard- Health Affairs, Riyadh, Saudi Arabia
- Colorectal Cancer Research Program, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
107
|
Manley K, Bravo-Nuevo A, Minton AR, Sedano S, Marcy A, Reichman M, Tobia A, Artlett CM, Gilmour SK, Laury-Kleintop LD, Prendergast GC. Preclinical study of the long-range safety and anti-inflammatory effects of high-dose oral meglumine. J Cell Biochem 2019; 120:12051-12062. [PMID: 30809852 DOI: 10.1002/jcb.28492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/14/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
Meglumine is a methylamino derivative of sorbitol that is an approved drug excipient. Recent preclinical studies suggest that administration of high-dose oral meglumine can exert beneficial medicinal effects to treat diabetes, obesity, and fatty liver disease (NAFLD/nonalcoholic steatohepatitis [NASH]). Here we address gaps in knowledge about the pharmacology and toxicology of this substance administered at high concentrations to explore its medicinal potential. We observed that high-dose meglumine limited secretion of proinflammatory cytokines and cell adhesion molecules from activated human THP-1 or murine RAW264.7 monocytes. Preclinical pharmacokinetic analysis in Swiss mice confirmed that meglumine was orally available. Informed by this data, oral doses of 18 to 75 mM meglumine were administered ad libitum in the drinking water of Sprague-Dawley rats and two cohorts of C57BL/6 mice housed in different vivariums. In a 32-week study, urinary isoprostane levels trended lower in subjects consistent with the possibility of anti-inflammatory effects. In full lifespan studies, there was no detrimental effect on longevity. Heart function evaluated in C57BL/6 mice using an established noninvasive cardiac imaging system showed no detrimental effects on ejection fraction, fractional shortening, left ventricle function or volume, and cardiac output in mice up to 15-month old, with a potential positive trend in heart function noted in elderly mice consistent with earlier reported benefits on muscle stamina. Finally, in a transgenic model of inflammation-associated skin carcinogenesis, the incidence, number, and growth of skin tumors trended lower in subjects receiving meglumine. Overall, the evidence obtained illustrating the long-range safety of high-dose oral meglumine support the rationale for its evaluation as a low-cost modality to limit diabetes, hypertriglyceridemia, and NAFLD/NASH.
Collapse
Affiliation(s)
- Kaylend Manley
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | - Allyson R Minton
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Summer Sedano
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Alice Marcy
- Dynamis Pharmaceuticals Inc, Jenkintown, Pennsylvania
| | - Melvin Reichman
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Annette Tobia
- Dynamis Pharmaceuticals Inc, Jenkintown, Pennsylvania
| | - Carol M Artlett
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Susan K Gilmour
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | | |
Collapse
|
108
|
Travers M, Brown SM, Dunworth M, Holbert CE, Wiehagen KR, Bachman KE, Foley JR, Stone ML, Baylin SB, Casero RA, Zahnow CA. DFMO and 5-Azacytidine Increase M1 Macrophages in the Tumor Microenvironment of Murine Ovarian Cancer. Cancer Res 2019; 79:3445-3454. [PMID: 31088836 DOI: 10.1158/0008-5472.can-18-4018] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/25/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Although ovarian cancer has a low incidence rate, it remains the most deadly gynecologic malignancy. Previous work has demonstrated that the DNMTi 5-Azacytidine (5AZA-C) activates type I interferon signaling to increase IFNγ+ T cells and natural killer (NK) cells and reduce the percentage of macrophages in the tumor microenvironment. To improve the efficacy of epigenetic therapy, we hypothesized that the addition of α-difluoromethylornithine (DFMO), an ornithine decarboxylase inhibitor, may further decrease immunosuppressive cell populations improving outcome. We tested this hypothesis in an immunocompetent mouse model for ovarian cancer and found that in vivo, 5AZA-C and DFMO, either alone or in combination, significantly increased survival, decreased tumor burden, and caused recruitment of activated (IFNγ+) CD4+ T cells, CD8+ T cells, and NK cells. The combination therapy had a striking increase in survival when compared with single-agent treatment, despite a smaller difference in recruited lymphocytes. Instead, combination therapy led to a significant decrease in immunosuppressive cells such as M2 polarized macrophages and an increase in tumor-killing M1 macrophages. In this model, depletion of macrophages with a CSF1R-blocking antibody reduced the efficacy of 5AZA-C + DFMO treatment and resulted in fewer M1 macrophages in the tumor microenvironment. These observations suggest our novel combination therapy modifies macrophage polarization in the tumor microenvironment, recruiting M1 macrophages and prolonging survival. SIGNIFICANCE: Combined epigenetic and polyamine-reducing therapy stimulates M1 macrophage polarization in the tumor microenvironment of an ovarian cancer mouse model, resulting in decreased tumor burden and prolonged survival.
Collapse
Affiliation(s)
- Meghan Travers
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Stephen M Brown
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Matthew Dunworth
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Cassandra E Holbert
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | | | | | - Jackson R Foley
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Meredith L Stone
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland.,Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen B Baylin
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Robert A Casero
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland.
| | - Cynthia A Zahnow
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland.
| |
Collapse
|
109
|
Abstract
Advances in our understanding of the metabolism and molecular functions of polyamines and their alterations in cancer have led to resurgence in the interest of targeting polyamine metabolism as an anticancer strategy. Increasing knowledge of the interplay between polyamine metabolism and other cancer-driving pathways, including the PTEN-PI3K-mTOR complex 1 (mTORC1), WNT signalling and RAS pathways, suggests potential combination therapies that will have considerable clinical promise. Additionally, an expanding number of promising clinical trials with agents targeting polyamines for both therapy and prevention are ongoing. New insights into molecular mechanisms linking dysregulated polyamine catabolism and carcinogenesis suggest additional strategies that can be used for cancer prevention in at-risk individuals. In addition, polyamine blocking therapy, a strategy that combines the inhibition of polyamine biosynthesis with the simultaneous blockade of polyamine transport, can be more effective than therapies based on polyamine depletion alone and may involve an antitumour immune response. These findings open up new avenues of research into exploiting aberrant polyamine metabolism for anticancer therapy.
Collapse
Affiliation(s)
- Robert A Casero
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | - Tracy Murray Stewart
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Anthony E Pegg
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
110
|
Orillion A, Damayanti NP, Shen L, Adelaiye-Ogala R, Affronti H, Elbanna M, Chintala S, Ciesielski M, Fontana L, Kao C, Elzey BD, Ratliff TL, Nelson DE, Smiraglia D, Abrams SI, Pili R. Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy. Clin Cancer Res 2018; 24:6383-6395. [PMID: 30190370 DOI: 10.1158/1078-0432.ccr-18-0980] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/03/2018] [Accepted: 08/31/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Diet and healthy weight are established means of reducing cancer incidence and mortality. However, the impact of diet modifications on the tumor microenvironment and antitumor immunity is not well defined. Immunosuppressive tumor-associated macrophages (TAMs) are associated with poor clinical outcomes and are potentially modifiable through dietary interventions. We tested the hypothesis that dietary protein restriction modifies macrophage function toward antitumor phenotypes. EXPERIMENTAL DESIGN Macrophage functional status under different tissue culture conditions and in vivo was assessed by Western blot, immunofluorescence, qRT-PCR, and cytokine array analyses. Tumor growth in the context of protein or amino acid (AA) restriction and immunotherapy, namely, a survivin peptide-based vaccine or a PD-1 inhibitor, was examined in animal models of prostate (RP-B6Myc) and renal (RENCA) cell carcinoma. All tests were two-sided. RESULTS Protein or AA-restricted macrophages exhibited enhanced tumoricidal, proinflammatory phenotypes, and in two syngeneic tumor models, protein or AA-restricted diets elicited reduced TAM infiltration, tumor growth, and increased response to immunotherapies. Further, we identified a distinct molecular mechanism by which AA-restriction reprograms macrophage function via a ROS/mTOR-centric cascade. CONCLUSIONS Dietary protein restriction alters TAM activity and enhances the tumoricidal capacity of this critical innate immune cell type, providing the rationale for clinical testing of this supportive tool in patients receiving cancer immunotherapies.
Collapse
Affiliation(s)
- Ashley Orillion
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Nur P Damayanti
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Li Shen
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Remi Adelaiye-Ogala
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.,Department of Cancer Pathology and Prevention, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Hayley Affronti
- Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - May Elbanna
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Sreenivasulu Chintala
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | - Michael Ciesielski
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Luigi Fontana
- Charles Perkins Centre and Central Clinical School, The University of Sydney, New South Wales, Australia
| | - Chinghai Kao
- Department of Urology, Indiana University, Indianapolis, Indiana
| | - Bennett D Elzey
- Department of Urology, Indiana University, Indianapolis, Indiana.,Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana
| | - Timothy L Ratliff
- Center for Cancer Research, Purdue University, West Lafayette, Indiana
| | - David E Nelson
- Department of Microbiology and Immunology, Indiana University, Indianapolis, Indiana
| | - Dominic Smiraglia
- Department of Cellular and Molecular Biology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York
| | - Scott I Abrams
- Department of Immunology, University at Buffalo, Roswell Park Cancer Institute, Buffalo, New York.
| | - Roberto Pili
- Genitourinary Malignancies Program, Simon Cancer Center, Indiana University, Indianapolis, Indiana.
| |
Collapse
|
111
|
Sun L, Suo C, Li ST, Zhang H, Gao P. Metabolic reprogramming for cancer cells and their microenvironment: Beyond the Warburg Effect. Biochim Biophys Acta Rev Cancer 2018; 1870:51-66. [PMID: 29959989 DOI: 10.1016/j.bbcan.2018.06.005] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023]
Abstract
While metabolic reprogramming of cancer cells has long been considered from the standpoint of how and why cancer cells preferentially utilize glucose via aerobic glycolysis, the so-called Warburg Effect, the progress in the following areas during the past several years has substantially advanced our understanding of the rewired metabolic network in cancer cells that is intertwined with oncogenic signaling. First, in addition to the major nutrient substrates glucose and glutamine, cancer cells have been discovered to utilize a variety of unconventional nutrient sources for survival. Second, the deregulated biomass synthesis is intertwined with cell cycle progression to coordinate the accelerated progression of cancer cells. Third, the reciprocal regulation of cancer cell's metabolic alterations and the microenvironment, involving extensive host immune cells and microbiota, have come into view as critical mechanisms to regulate cancer progression. These and other advances are shaping the current and future paradigm of cancer metabolism.
Collapse
Affiliation(s)
- Linchong Sun
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Caixia Suo
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shi-Ting Li
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Huafeng Zhang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| | - Ping Gao
- Laboratory of Cancer and Stem Cell metabolism, Guangzhou First Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China; CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
112
|
Kim CH. Immune regulation by microbiome metabolites. Immunology 2018; 154:220-229. [PMID: 29569377 PMCID: PMC5980225 DOI: 10.1111/imm.12930] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/18/2018] [Accepted: 03/06/2018] [Indexed: 02/06/2023] Open
Abstract
Commensal microbes and the host immune system have been co-evolved for mutual regulation. Microbes regulate the host immune system, in part, by producing metabolites. A mounting body of evidence indicates that diverse microbial metabolites profoundly regulate the immune system via host receptors and other target molecules. Immune cells express metabolite-specific receptors such as P2X7 , GPR41, GPR43, GPR109A, aryl hydrocarbon receptor precursor (AhR), pregnane X receptor (PXR), farnesoid X receptor (FXR), TGR5 and other molecular targets. Microbial metabolites and their receptors form an extensive array of signals to respond to changes in nutrition, health and immunological status. As a consequence, microbial metabolite signals contribute to nutrient harvest from diet, and regulate host metabolism and the immune system. Importantly, microbial metabolites bidirectionally function to promote both tolerance and immunity to effectively fight infection without developing inflammatory diseases. In pathogenic conditions, adverse effects of microbial metabolites have been observed as well. Key immune-regulatory functions of the metabolites, generated from carbohydrates, proteins and bile acids, are reviewed in this article.
Collapse
Affiliation(s)
- Chang H. Kim
- Department of Pathology and Mary H. Weiser Food Allergy CenterUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
113
|
Myc, Oncogenic Protein Translation, and the Role of Polyamines. Med Sci (Basel) 2018; 6:medsci6020041. [PMID: 29799508 PMCID: PMC6024823 DOI: 10.3390/medsci6020041] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/19/2018] [Accepted: 05/22/2018] [Indexed: 01/21/2023] Open
Abstract
Deregulated protein synthesis is a common feature of cancer cells, with many oncogenic signaling pathways directly augmenting protein translation to support the biomass needs of proliferating tissues. MYC’s ability to drive oncogenesis is a consequence of its essential role as a governor linking cell cycle entry with the requisite increase in protein synthetic capacity, among other biomass needs. To date, direct pharmacologic inhibition of MYC has proven difficult, but targeting oncogenic signaling modules downstream of MYC, such as the protein synthetic machinery, may provide a viable therapeutic strategy. Polyamines are essential cations found in nearly all living organisms that have both direct and indirect roles in the control of protein synthesis. Polyamine metabolism is coordinately regulated by MYC to increase polyamines in proliferative tissues, and this is further augmented in the many cancer cells harboring hyperactivated MYC. In this review, we discuss MYC-driven regulation of polyamines and protein synthetic capacity as a key function of its oncogenic output, and how this dependency may be perturbed through direct pharmacologic targeting of components of the protein synthetic machinery, such as the polyamines themselves, the eukaryotic translation initiation factor 4F (eIF4F) complex, and the eukaryotic translation initiation factor 5A (eIF5A).
Collapse
|
114
|
Cho HE, Kang MH. pH gradient-liquid chromatography tandem mass spectrometric assay for determination of underivatized polyamines in cancer cells. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1085:21-29. [DOI: 10.1016/j.jchromb.2018.03.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/18/2018] [Accepted: 03/26/2018] [Indexed: 10/17/2022]
|
115
|
Abstract
The immune system is remarkably responsive to a myriad of invading microorganisms and provides continuous surveillance against tissue damage and developing tumor cells. To achieve these diverse functions, multiple soluble and cellular components must react in an orchestrated cascade of events to control the specificity, magnitude and persistence of the immune response. Numerous catabolic and anabolic processes are involved in this process, and prominent roles for l-arginine and l-glutamine catabolism have been described, as these amino acids serve as precursors of nitric oxide, creatine, agmatine, tricarboxylic acid cycle intermediates, nucleotides and other amino acids, as well as for ornithine, which is used to synthesize putrescine and the polyamines spermidine and spermine. Polyamines have several purported roles and high levels of polyamines are manifest in tumor cells as well in autoreactive B- and T-cells in autoimmune diseases. In the tumor microenvironment, l-arginine catabolism by both tumor cells and suppressive myeloid cells is known to dampen cytotoxic T-cell functions suggesting there might be links between polyamines and T-cell suppression. Here, we review studies suggesting roles of polyamines in normal immune cell function and highlight their connections to autoimmunity and anti-tumor immune cell function.
Collapse
Affiliation(s)
- Rebecca S Hesterberg
- University of South Florida Cancer Biology Graduate Program, University of South Florida, 4202 East Fowler Ave, Tampa, FL 33620, USA.
- Department Immunology, PharmD, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, 23033 SRB, Tampa, FL 33612, USA.
| | - John L Cleveland
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | - Pearlie K Epling-Burnette
- Department Immunology, PharmD, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, 23033 SRB, Tampa, FL 33612, USA.
| |
Collapse
|
116
|
Reigada C, Phanstiel O, Miranda MR, Pereira CA. Targeting polyamine transport in Trypanosoma cruzi. Eur J Med Chem 2018; 147:1-6. [DOI: 10.1016/j.ejmech.2018.01.083] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 01/09/2018] [Accepted: 01/26/2018] [Indexed: 02/05/2023]
|
117
|
Alpha-Difluoromethylornithine, an Irreversible Inhibitor of Polyamine Biosynthesis, as a Therapeutic Strategy against Hyperproliferative and Infectious Diseases. Med Sci (Basel) 2018; 6:medsci6010012. [PMID: 29419804 PMCID: PMC5872169 DOI: 10.3390/medsci6010012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/02/2018] [Accepted: 02/05/2018] [Indexed: 12/18/2022] Open
Abstract
The fluorinated ornithine analog α-difluoromethylornithine (DFMO, eflornithine, ornidyl) is an irreversible suicide inhibitor of ornithine decarboxylase (ODC), the first and rate-limiting enzyme of polyamine biosynthesis. The ubiquitous and essential polyamines have many functions, but are primarily important for rapidly proliferating cells. Thus, ODC is potentially a drug target for any disease state where rapid growth is a key process leading to pathology. The compound was originally discovered as an anticancer drug, but its effectiveness was disappointing. However, DFMO was successfully developed to treat African sleeping sickness and is currently one of few clinically used drugs to combat this neglected tropical disease. The other Food and Drug Administration (FDA) approved application for DFMO is as an active ingredient in the hair removal cream Vaniqa. In recent years, renewed interest in DFMO for hyperproliferative diseases has led to increased research and promising preclinical and clinical trials. This review explores the use of DFMO for the treatment of African sleeping sickness and hirsutism, as well as its potential as a chemopreventive and chemotherapeutic agent against colorectal cancer and neuroblastoma.
Collapse
|
118
|
Gitto SB, Pandey V, Oyer JL, Copik AJ, Hogan FC, Phanstiel O, Altomare DA. Difluoromethylornithine Combined with a Polyamine Transport Inhibitor Is Effective against Gemcitabine Resistant Pancreatic Cancer. Mol Pharm 2018; 15:369-376. [PMID: 29299930 DOI: 10.1021/acs.molpharmaceut.7b00718] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly chemo-resistant and has an extremely poor patient prognosis, with a survival rate at five years of <8%. There remains an urgent need for innovative treatments. Targeting polyamine biosynthesis through inhibition of ornithine decarboxylase with difluoromethylornithine (DFMO) has had mixed clinical success due to tumor escape via an undefined transport system, which imports exogenous polyamines and sustains intracellular polyamine pools. Here, we tested DFMO in combination with a polyamine transport inhibitor (PTI), Trimer44NMe, against Gemcitabine-resistant PDAC cells. DFMO alone and with Trimer44NMe significantly reduced PDAC cell viability by inducing apoptosis or diminishing proliferation. DFMO alone and with Trimer44NMe also inhibited in vivo orthotopic PDAC growth and resulted in decreased c-Myc expression, a readout of polyamine pathway dysfunction. Moreover, dual inhibition significantly prolonged survival of tumor-bearing mice. Collectively, these studies demonstrate that targeting polyamine biosynthesis and import pathways in PDAC can lead to increased survival in pancreatic cancer.
Collapse
Affiliation(s)
- Sarah B Gitto
- Burnett School of Biomedical Sciences, University of Central Florida , 6900 Lake Nona Blvd., Orlando, Florida 32827, United States
| | - Veethika Pandey
- Burnett School of Biomedical Sciences, University of Central Florida , 6900 Lake Nona Blvd., Orlando, Florida 32827, United States
| | - Jeremiah L Oyer
- Burnett School of Biomedical Sciences, University of Central Florida , 6900 Lake Nona Blvd., Orlando, Florida 32827, United States
| | - Alicja J Copik
- Burnett School of Biomedical Sciences, University of Central Florida , 6900 Lake Nona Blvd., Orlando, Florida 32827, United States
| | - Frederick C Hogan
- Burnett School of Biomedical Sciences, University of Central Florida , 6900 Lake Nona Blvd., Orlando, Florida 32827, United States
| | - Otto Phanstiel
- Department of Medical Education, University of Central Florida , 12722 Research Parkway, Orlando, Florida 32826, United States
| | - Deborah A Altomare
- Burnett School of Biomedical Sciences, University of Central Florida , 6900 Lake Nona Blvd., Orlando, Florida 32827, United States
| |
Collapse
|
119
|
Inflammatory Reprogramming with IDO1 Inhibitors: Turning Immunologically Unresponsive 'Cold' Tumors 'Hot'. Trends Cancer 2017; 4:38-58. [PMID: 29413421 DOI: 10.1016/j.trecan.2017.11.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 11/07/2017] [Accepted: 11/14/2017] [Indexed: 01/24/2023]
Abstract
We discuss how small-molecule inhibitors of the tryptophan (Trp) catabolic enzyme indoleamine 2,3-dioxygenase (IDO) represent a vanguard of new immunometabolic adjuvants to safely enhance the efficacy of cancer immunotherapy, radiotherapy, or 'immunogenic' chemotherapy by leveraging responses to tumor neoantigens. IDO inhibitors re-program inflammatory processes to help clear tumors by blunting tumor neovascularization and restoring immunosurveillance. Studies of regulatory and effector pathways illuminate IDO as an inflammatory modifier. Recent work suggests that coordinate targeting of the Trp catabolic enzymes tryptophan 2,3-dioxygenase (TDO) and IDO2 may also safely broaden efficacy. Understanding IDO inhibitors as adjuvants to turn immunologically 'cold' tumors 'hot' can seed new concepts in how to improve the efficacy of cancer therapy while limiting collateral damage.
Collapse
|
120
|
Phanstiel O. An overview of polyamine metabolism in pancreatic ductal adenocarcinoma. Int J Cancer 2017; 142:1968-1976. [PMID: 29134652 DOI: 10.1002/ijc.31155] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/19/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest major cancers, with a five year survival rate of less than 8%. With current therapies only giving rise to modest life extension, new approaches are desperately needed. Even though targeting polyamine metabolism is a proven anticancer strategy, there are no reports, which thoroughly survey the literature describing the role of polyamine biosynthesis and transport in PDAC. This review seeks to fill this void by describing what is currently known about polyamine metabolism in PDAC and identifies new targets and opportunities to treat this disease. Due to the pleiotropic effects that polyamines play in cells, this review covers diverse areas ranging from polyamine metabolism (biosynthesis, catabolism and transport), as well as the potential role of polyamines in desmoplasia, autophagy and immune privilege. Understanding these diverse roles provides the opportunity to design new therapies to treat this deadly cancer via polyamine depletion.
Collapse
Affiliation(s)
- Otto Phanstiel
- Department of Medical Education, College of Medicine, University of Central Florida, Orlando, FL
| |
Collapse
|
121
|
Investigation of Polyamine Metabolism and Homeostasis in Pancreatic Cancers. Med Sci (Basel) 2017; 5:medsci5040032. [PMID: 29215586 PMCID: PMC5753661 DOI: 10.3390/medsci5040032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancers are currently the fourth leading cause of cancer-related death and new therapies are desperately needed. The most common pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC). This report describes the development of therapies, which effectively deplete PDAC cells of their required polyamine growth factors. Of all human tissues, the pancreas has the highest level of the native polyamine spermidine. To sustain their high growth rates, PDACs have altered polyamine metabolism, which is reflected in their high intracellular polyamine levels and their upregulated import of exogenous polyamines. To understand how these cancers respond to interventions that target their specific polyamine pools, L3.6pl human pancreatic cancer cells were challenged with specific inhibitors of polyamine biosynthesis. We found that pancreatic cell lines have excess polyamine pools, which they rebalance to address deficiencies induced by inhibitors of specific steps in polyamine biosynthesis (e.g., ornithine decarboxylase (ODC), spermidine synthase (SRM), and spermine synthase (SMS)). We also discovered that combination therapies targeting ODC, SMS, and polyamine import were the most effective in reducing intracellular polyamine pools and reducing PDAC cell growth. A combination therapy containing difluoromethylornithine (DFMO, an ODC inhibitor) and a polyamine transport inhibitor (PTI) were shown to significantly deplete intracellular polyamine pools. The additional presence of an SMS inhibitor as low as 100 nM was sufficient to further potentiate the DFMO + PTI treatment.
Collapse
|
122
|
Wang M, Phanstiel O, von Kalm L. Evaluation of Polyamine Transport Inhibitors in a Drosophila Epithelial Model Suggests the Existence of Multiple Transport Systems. ACTA ACUST UNITED AC 2017; 5:medsci5040027. [PMID: 29135915 PMCID: PMC5753656 DOI: 10.3390/medsci5040027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 12/19/2022]
Abstract
Increased polyamine biosynthesis activity and an active polyamine transport system are characteristics of many cancer cell lines and polyamine depletion has been shown to be a viable anticancer strategy. Polyamine levels can be depleted by difluoromethylornithine (DFMO), an inhibitor of the key polyamine biosynthesis enzyme ornithine decarboxylase (ODC). However, malignant cells frequently circumvent DFMO therapy by up-regulating polyamine import. Therefore, there is a need to develop compounds that inhibit polyamine transport. Collectively, DFMO and a polyamine transport inhibitor (PTI) provide the basis for a combination therapy leading to effective intracellular polyamine depletion. We have previously shown that the pattern of uptake of a series of polyamine analogues in a Drosophila model epithelium shares many characteristics with mammalian cells, indicating a high degree of similarity between the mammalian and Drosophila polyamine transport systems. In this report, we focused on the utility of the Drosophila epithelial model to identify and characterize polyamine transport inhibitors. We show that a previously identified inhibitor of transport in mammalian cells has a similar activity profile in Drosophila. The Drosophila model was also used to evaluate two additional transport inhibitors. We further demonstrate that a cocktail of polyamine transport inhibitors is more effective than individual inhibitors, suggesting the existence of multiple transport systems in Drosophila. Our findings reinforce the similarity between the Drosophila and mammalian transport systems and the value of the Drosophila model to provide inexpensive early screening of molecules targeting the transport system.
Collapse
Affiliation(s)
- Minpei Wang
- Department of Biology, University of Central Florida, Orlando, FL 32816, USA.
| | - Otto Phanstiel
- Department of Medical Education, College of Medicine, University of Central Florida, Orlando, FL 32827, USA.
| | - Laurence von Kalm
- Department of Biology, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
123
|
Alexander ET, Minton A, Peters MC, Phanstiel O, Gilmour SK. A novel polyamine blockade therapy activates an anti-tumor immune response. Oncotarget 2017; 8:84140-84152. [PMID: 29137411 PMCID: PMC5663583 DOI: 10.18632/oncotarget.20493] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 07/23/2017] [Indexed: 01/22/2023] Open
Abstract
Most tumors maintain elevated levels of polyamines to support their growth and survival. This study explores the anti-tumor effect of polyamine starvation via both inhibiting polyamine biosynthesis and blocking the upregulated import of polyamines into the tumor. We demonstrate that polyamine blockade therapy (PBT) co-treatment with both DFMO and a novel polyamine transport inhibitor, Trimer PTI, significantly inhibits tumor growth more than treatment with DFMO or the Trimer PTI alone. The anti-tumor effect of PBT was lost in mice where CD4+ and CD8+ T cells were antibody depleted, implying that PBT stimulates an anti-tumor immune effect that is T-cell dependent. The PBT anti-tumor effect was accompanied by an increase in granzyme B+, IFN-γ+ CD8+ T-cells and a decrease in immunosuppressive tumor infiltrating cells including Gr-1+CD11b+ myeloid derived suppressor cells (MDSCs), CD4+CD25+ Tregs, and CD206+F4/80+ M2 macrophages. Stimulation with tumor-specific peptides elicited elevated antigen-specific IFN-γ secretion in splenocytes from PBT-treated mice, indicating that PBT treatment stimulates the activation of T-cells in a tumor-specific manner. These data show that combined treatment with both DFMO and the Trimer PTI not only deprives polyamine-addicted tumor cells of polyamines, but also relieves polyamine-mediated immunosuppression in the tumor microenvironment, thus allowing the activation of tumoricidal T-cells.
Collapse
Affiliation(s)
- Eric T Alexander
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Allyson Minton
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Molly C Peters
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Otto Phanstiel
- University of Central Florida, Biomolecular Research Annex, Orlando, FL 32826-3227, USA
| | - Susan K Gilmour
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| |
Collapse
|
124
|
The influence of the commensal microbiota on distal tumor-promoting inflammation. Semin Immunol 2017; 32:62-73. [PMID: 28687194 DOI: 10.1016/j.smim.2017.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023]
Abstract
Commensal microbes inhabit barrier surfaces, providing a first line of defense against invading pathogens, aiding in metabolic function of the host, and playing a vital role in immune development and function. Several recent studies have demonstrated that commensal microbes influence systemic immune function and homeostasis. For patients with extramucosal cancers, or cancers occurring distal to barrier surfaces, the role of commensal microbes in influencing tumor progression is beginning to be appreciated. Extrinsic factors such as chronic inflammation, antibiotics, and chemotherapy dysregulate commensal homeostasis and drive tumor-promoting systemic inflammation through a variety of mechanisms, including disruption of barrier function and bacterial translocation, release of soluble inflammatory mediators, and systemic changes in metabolic output. Conversely, it has also been demonstrated that certain immune therapies, immunogenic chemotherapies, and checkpoint inhibitors rely on the commensal microbiota to facilitate anti-tumor immune responses. Thus, it is evident that the mechanisms associated with commensal microbe facilitation of both pro- and anti-tumor immune responses are context dependent and rely upon a variety of factors present within the tumor microenvironment and systemic periphery. The goal of this review is to highlight the various contexts during which commensal microbes orchestrate systemic immune function with a focus on describing possible scenarios where the loss of microbial homeostasis enhances tumor progression.
Collapse
|
125
|
Targeting polyamine metabolism for cancer therapy and prevention. Biochem J 2017; 473:2937-53. [PMID: 27679855 DOI: 10.1042/bcj20160383] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
Abstract
The chemically simple, biologically complex eukaryotic polyamines, spermidine and spermine, are positively charged alkylamines involved in many crucial cellular processes. Along with their diamine precursor putrescine, their normally high intracellular concentrations require fine attenuation by multiple regulatory mechanisms to keep these essential molecules within strict physiologic ranges. Since the metabolism of and requirement for polyamines are frequently dysregulated in neoplastic disease, the metabolic pathway and functions of polyamines provide rational drug targets; however, these targets have been difficult to exploit for chemotherapy. It is the goal of this article to review the latest findings in the field that demonstrate the potential utility of targeting the metabolism and function of polyamines as strategies for both chemotherapy and, possibly more importantly, chemoprevention.
Collapse
|
126
|
Abstract
The microbiota - the collection of microorganisms that live within and on all mammals - provides crucial signals for the development and function of the immune system. Increased availability of technologies that profile microbial communities is facilitating the entry of many immunologists into the evolving field of host-microbiota studies. The microbial communities, their metabolites and components are not only necessary for immune homeostasis, they also influence the susceptibility of the host to many immune-mediated diseases and disorders. In this Review, we discuss technological and computational approaches for investigating the microbiome, as well as recent advances in our understanding of host immunity and microbial mutualism with a focus on specific microbial metabolites, bacterial components and the immune system.
Collapse
|
127
|
Trypanocidal Effect of Isotretinoin through the Inhibition of Polyamine and Amino Acid Transporters in Trypanosoma cruzi. PLoS Negl Trop Dis 2017; 11:e0005472. [PMID: 28306713 PMCID: PMC5371382 DOI: 10.1371/journal.pntd.0005472] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 03/29/2017] [Accepted: 03/09/2017] [Indexed: 12/04/2022] Open
Abstract
Polyamines are essential compounds to all living organisms and in the specific case of Trypanosoma cruzi, the causative agent of Chagas disease, they are exclusively obtained through transport processes since this parasite is auxotrophic for polyamines. Previous works reported that retinol acetate inhibits Leishmania growth and decreases its intracellular polyamine concentration. The present work describes a combined strategy of drug repositioning by virtual screening followed by in vitro assays to find drugs able to inhibit TcPAT12, the only polyamine transporter described in T. cruzi. After a screening of 3000 FDA-approved drugs, 7 retinoids with medical use were retrieved and used for molecular docking assays with TcPAT12. From the docked molecules, isotretinoin, a well-known drug used for acne treatment, showed the best interaction score with TcPAT12 and was selected for further in vitro studies. Isotretinoin inhibited the polyamine transport, as well as other amino acid transporters from the same protein family (TcAAAP), with calculated IC50 values in the range of 4.6–10.3 μM. It also showed a strong inhibition of trypomastigote burst from infected cells, with calculated IC50 of 130 nM (SI = 920) being significantly less effective on the epimastigote stage (IC50 = 30.6 μM). The effect of isotretinoin on the parasites plasma membrane permeability and on mammalian cell viability was tested, and no change was observed. Autophagosomes and apoptotic bodies were detected as part of the mechanisms of isotretinoin-induced death indicating that the inhibition of transporters by isotretinoin causes nutrient starvation that triggers autophagic and apoptotic processes. In conclusion, isotretinoin is a promising trypanocidal drug since it is a multi-target inhibitor of essential metabolites transporters, in addition to being an FDA-approved drug largely used in humans, which could reduce significantly the requirements for its possible application in the treatment of Chagas disease. Polyamines are polycationic compounds essential for the regulation of cell growth and differentiation. In contrast with other protozoa, Trypanosoma cruzi, the etiological agent of Chagas disease, is auxotrophic for polyamines; therefore the intracellular availability of these molecules depends exclusively on transport processes. It was previously demonstrated that the lack of polyamines in T. cruzi leads to its death, making the polyamine transporter an excellent therapeutic target for Chagas disease. In this work, the polyamine permease TcPAT12 was selected as a target for drug screening using 3000 FDA-approved compounds and computational simulation techniques. Using two combined virtual screening methods, isotretinoin, a well-known and safe drug used for acne treatment, bound to substrate recognition residues of TcPAT12 and was chosen for further in vitro studies. Isotretinoin inhibited not only the polyamine transport but also all tested amino acid transporters from the same protein family as TcPAT12. Interestingly, isotretinoin showed a high trypanocidal effect on trypomastigotes, with an IC50 in the nanomolar range. Autophagy and apoptosis were proposed as mechanisms of parasites death induced by isotretinoin. These results suggest that isotretinoin is a promising trypanocidal drug, being a multi-target inhibitor of essential metabolites transporters.
Collapse
|
128
|
Alexander ET, Minton AR, Peters MC, van Ryn J, Gilmour SK. Thrombin inhibition and cisplatin block tumor progression in ovarian cancer by alleviating the immunosuppressive microenvironment. Oncotarget 2016; 7:85291-85305. [PMID: 27852034 PMCID: PMC5356737 DOI: 10.18632/oncotarget.13300] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
Cancer is often associated with an increased risk of thrombotic complications which can be aggravated by treatment with chemotherapeutics such as cisplatin. Multiple lines of evidence suggest that thrombin activity promotes tumor growth and metastasis. We examined the effect of co-treatment with dabigatran etexilate, a direct thrombin inhibitor, and cisplatin using the murine ID8 ovarian cancer model. Mice receiving co-treatment with both dabigatran etexilate and low dose cisplatin had significantly smaller tumors, developed less ascites and had lower levels of circulating activated platelets and tissue factor (TF) positive microparticles than those treated with dabigatran etexilate or cisplatin alone. Co-treatment with dabigatran etexilate and cisplatin significantly decreased the number of Gr1+/CD11b+ myeloid derived suppresser cells and CD11b+/CD11c+ dendritic cells in the ascites of ID8 tumor-bearing mice. Co-treatment also significantly reduced levels of pro-tumorigenic cytokines including TGF-β, VEGF, IL-6, IL-10, and MCP-1 in the ascites while increasing IFN-γ production by CD8+ effector T cells in the tumor ascites. These results demonstrate that co-treatment with dabigatran etexilate significantly augments the anti-tumor activity of cisplatin in ovarian tumor progression by alleviating the immunosuppressive microenvironment, suggesting that thrombin may be a potential therapeutic target for treatment of ovarian cancer.
Collapse
Affiliation(s)
| | | | - Molly C. Peters
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Joanne van Ryn
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany
| | - Susan K. Gilmour
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| |
Collapse
|
129
|
Musolino C, Allegra A, Pioggia G, Gangemi S. Immature myeloid-derived suppressor cells: A bridge between inflammation and cancer (Review). Oncol Rep 2016; 37:671-683. [PMID: 27922687 DOI: 10.3892/or.2016.5291] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/29/2015] [Indexed: 11/06/2022] Open
Abstract
Chronic inflammation is considered to be one of the hallmarks of tumor initiation and progression. Changes occurring in the microenvironment of progressing tumors resemble the process of chronic inflammation, which begins with ischemia followed by interstitial and cellular edema, appearance of immune cells, growth of blood vessels and tissue repair, and development of inflammatory infiltrates. Moreover, long‑term production and accumulation of inflammatory factors lead to local and systemic immunosuppression associated with cancer progression. Of the several mechanisms described to explain this anergy, the accumulation of myeloid cells in the tumor, spleen, and peripheral blood of cancer patients has gained considerable interest. A population of suppressive CD11b+Gr-1+ cells has in fact been designated as myeloid-derived suppressor cells (MDSCs). MDSCs are a unique category of the myeloid lineage, and they induce the prevention of the development of cytotoxic T lymphocytes (CTLs) in vitro, and the induction of antigen-specific CD8+ T-cell tolerance in vivo. Therapeutic approaches directed toward the manipulation of the MDSC population and their function may improve chemoimmune-enhancing therapy for advanced malignancies.
Collapse
Affiliation(s)
- Caterina Musolino
- Division of Hematology, Department of General Surgery, Pathological Anatomy and Oncology, University of Messina, Messina, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of General Surgery, Pathological Anatomy and Oncology, University of Messina, Messina, Italy
| | - Govanni Pioggia
- Institute of Clinical Physiology, IFN CNR, Messina Unit, Messina, Italy
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital 'G. Martino', Messina, Italy
| |
Collapse
|
130
|
Mahmoud AA, Farouk A, Goneim A, Hafez MFA, Saleem TH. Ornithine decarboxylase gene expression and activity in lung cancer. GENE REPORTS 2016. [DOI: 10.1016/j.genrep.2016.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
131
|
Evageliou NF, Haber M, Vu A, Laetsch TW, Murray J, Gamble LD, Cheng NC, Liu K, Reese M, Corrigan KA, Ziegler DS, Webber H, Hayes CS, Pawel B, Marshall GM, Zhao H, Gilmour SK, Norris MD, Hogarty MD. Polyamine Antagonist Therapies Inhibit Neuroblastoma Initiation and Progression. Clin Cancer Res 2016; 22:4391-404. [PMID: 27012811 DOI: 10.1158/1078-0432.ccr-15-2539] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/15/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Deregulated MYC drives oncogenesis in many tissues yet direct pharmacologic inhibition has proven difficult. MYC coordinately regulates polyamine homeostasis as these essential cations support MYC functions, and drugs that antagonize polyamine sufficiency have synthetic-lethal interactions with MYC Neuroblastoma is a lethal tumor in which the MYC homologue MYCN, and ODC1, the rate-limiting enzyme in polyamine synthesis, are frequently deregulated so we tested optimized polyamine depletion regimens for activity against neuroblastoma. EXPERIMENTAL DESIGN We used complementary transgenic and xenograft-bearing neuroblastoma models to assess polyamine antagonists. We investigated difluoromethylornithine (DFMO; an inhibitor of Odc, the rate-limiting enzyme in polyamine synthesis), SAM486 (an inhibitor of Amd1, the second rate-limiting enzyme), and celecoxib (an inducer of Sat1 and polyamine catabolism) in both the preemptive setting and in the treatment of established tumors. In vitro assays were performed to identify mechanisms of activity. RESULTS An optimized polyamine antagonist regimen using DFMO and SAM486 to inhibit both rate-limiting enzymes in polyamine synthesis potently blocked neuroblastoma initiation in transgenic mice, underscoring the requirement for polyamines in MYC-driven oncogenesis. Furthermore, the combination of DFMO with celecoxib was found to be highly active, alone, and combined with numerous chemotherapy regimens, in regressing established tumors in both models, including tumors harboring highest risk genetic lesions such as MYCN amplification, ALK mutation, and TP53 mutation with multidrug resistance. CONCLUSIONS Given the broad preclinical activity demonstrated by polyamine antagonist regimens across diverse in vivo models, clinical investigation of such approaches in neuroblastoma and potentially other MYC-driven tumors is warranted. Clin Cancer Res; 22(17); 4391-404. ©2016 AACR.
Collapse
Affiliation(s)
- Nicholas F Evageliou
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. Center for Childhood Cancer Research, University of New South Wales, Sydney, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia, Sydney, Australia
| | - Annette Vu
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Jayne Murray
- Children's Cancer Institute Australia, Sydney, Australia
| | - Laura D Gamble
- Children's Cancer Institute Australia, Sydney, Australia
| | | | - Kangning Liu
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Megan Reese
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kelly A Corrigan
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David S Ziegler
- Children's Cancer Institute Australia, Sydney, Australia. Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia. School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Kensington, Sydney, Australia
| | - Hannah Webber
- Children's Cancer Institute Australia, Sydney, Australia
| | - Candice S Hayes
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Bruce Pawel
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Glenn M Marshall
- Children's Cancer Institute Australia, Sydney, Australia. Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Huaqing Zhao
- Department of Biostatistics, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Susan K Gilmour
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Murray D Norris
- Children's Cancer Institute Australia, Sydney, Australia. Center for Childhood Cancer Research, University of New South Wales, Sydney, Australia
| | - Michael D Hogarty
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania. Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
132
|
Verlinden BK, Louw A, Birkholtz LM. Resisting resistance: is there a solution for malaria? Expert Opin Drug Discov 2016; 11:395-406. [PMID: 26926843 DOI: 10.1517/17460441.2016.1154037] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Currently, widely used antimalarial drugs have a limited clinical lifespan due to parasite resistance development. With resistance continuously rising, antimalarial drug discovery requires strategies to decrease the time of delivering a new antimalarial drug while simultaneously increasing the drug's therapeutic lifespan. Lessons learnt from various chemotherapeutic resistance studies in the fields of antibiotic and cancer research offer potentially useful strategies that can be applied to antimalarial drug discovery. AREAS COVERED In this review the authors discuss current strategies to circumvent resistance in malaria and alternatives that could be employed. EXPERT OPINION Scientists have been 'beating back' the malaria parasite with novel drugs for the past 49 years but the constant rise in antimalarial drug resistance is forcing the drug discovery community to explore alternative strategies. Avant-garde anti-resistance strategies from alternative fields may assist our endeavors to manage, control and prevent antimalarial drug resistance to progress beyond beating the resistant parasite back, to stopping it dead in its tracks.
Collapse
Affiliation(s)
- Bianca K Verlinden
- a Department of Biochemistry, Centre for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences , University of Pretoria , Pretoria , South Africa
| | - Abraham Louw
- a Department of Biochemistry, Centre for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences , University of Pretoria , Pretoria , South Africa
| | - Lyn-Marié Birkholtz
- a Department of Biochemistry, Centre for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences , University of Pretoria , Pretoria , South Africa
| |
Collapse
|
133
|
Ye C, Geng Z, Dominguez D, Chen S, Fan J, Qin L, Long A, Zhang Y, Kuzel TM, Zhang B. Targeting Ornithine Decarboxylase by α-Difluoromethylornithine Inhibits Tumor Growth by Impairing Myeloid-Derived Suppressor Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:915-923. [PMID: 26663722 PMCID: PMC4707077 DOI: 10.4049/jimmunol.1500729] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 11/04/2015] [Indexed: 01/04/2023]
Abstract
α-Difluoromethylornithine (DFMO) is currently used in chemopreventive regimens primarily for its conventional direct anticarcinogenesic activity. However, little is known about the effect of ornithine decarboxylase (ODC) inhibition by DFMO on antitumor immune responses. We showed in this study that pharmacologic blockade of ODC by DFMO inhibited tumor growth in intact immunocompetent mice, but abrogated in the immunodeficient Rag1(-/-) mice, suggesting that antitumor effect of DFMO is dependent on the induction of adaptive antitumor T cell immune responses. Depletion of CD8(+) T cells impeded the tumor-inhibiting advantage of DFMO. Moreover, DFMO treatment enhanced antitumor CD8(+) T cell infiltration and IFN-γ production and augmented the efficacy of adoptive T cell therapy. Importantly, DFMO impaired Gr1(+)CD11b(+) myeloid-derived suppressor cells (MDSCs) suppressive activity through at least two mechanisms, including reducing arginase expression and activity and inhibiting the CD39/CD73-mediated pathway. MDSCs were one primary cellular target of DFMO as indicated by both adoptive transfer and MDSC-depletion analyses. Our findings establish a new role of ODC inhibition by DFMO as a viable and effective immunological adjunct in effective cancer treatment, thereby adding to the growing list of chemoimmunotherapeutic applications of these agents.
Collapse
Affiliation(s)
- Cong Ye
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; and
| | - Zhe Geng
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611; Hubei Maternity and Child Health Hospital, Wuhan 430070, China
| | - Donye Dominguez
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Siqi Chen
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Jie Fan
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Lei Qin
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Alan Long
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Timothy M Kuzel
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Bin Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611;
| |
Collapse
|
134
|
Nowotarski SL, Feith DJ, Shantz LM. Skin Carcinogenesis Studies Using Mouse Models with Altered Polyamines. CANCER GROWTH AND METASTASIS 2015; 8:17-27. [PMID: 26380554 PMCID: PMC4558889 DOI: 10.4137/cgm.s21219] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/30/2015] [Accepted: 07/01/2015] [Indexed: 12/16/2022]
Abstract
Nonmelanoma skin cancer (NMSC) is a major health concern worldwide. With increasing numbers in high-risk groups such as organ transplant recipients and patients taking photosensitizing medications, the incidence of NMSC continues to rise. Mouse models of NMSC allow us to better understand the molecular signaling cascades involved in skin tumor development in order to identify novel therapeutic strategies. Here we review the models designed to determine the role of the polyamines in NMSC development and maintenance. Elevated polyamines are absolutely required for tumor growth, and dysregulation of their biosynthetic and catabolic enzymes has been observed in NMSC. Studies using mice with genetic alterations in epidermal polyamines suggest that they play key roles in tumor promotion and epithelial cell survival pathways, and recent clinical trials indicate that pharmacological inhibitors of polyamine metabolism show promise in individuals at high risk for NMSC.
Collapse
Affiliation(s)
- Shannon L Nowotarski
- Department of Biochemistry, The Pennsylvania State University Berks College, Reading, PA, USA
| | - David J Feith
- University of Virginia Cancer Center and Department of Medicine, Hematology and Oncology, University of Virginia, Charlottesville, VA, USA
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
135
|
Bassiri H, Benavides A, Haber M, Gilmour SK, Norris MD, Hogarty MD. Translational development of difluoromethylornithine (DFMO) for the treatment of neuroblastoma. Transl Pediatr 2015; 4:226-38. [PMID: 26835380 PMCID: PMC4729051 DOI: 10.3978/j.issn.2224-4336.2015.04.06] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/08/2015] [Indexed: 01/01/2023] Open
Abstract
Neuroblastoma is a childhood tumor in which MYC oncogenes are commonly activated to drive tumor progression. Survival for children with high-risk neuroblastoma remains poor despite treatment that incorporates high-dose chemotherapy, stem cell support, surgery, radiation therapy and immunotherapy. More effective and less toxic treatments are sought and one approach under clinical development involves re-purposing the anti-protozoan drug difluoromethylornithine (DFMO; Eflornithine) as a neuroblastoma therapeutic. DFMO is an irreversible inhibitor of ornithine decarboxylase (Odc), a MYC target gene, bona fide oncogene, and the rate-limiting enzyme in polyamine synthesis. DFMO is approved for the treatment of Trypanosoma brucei gambiense encephalitis ("African sleeping sickness") since polyamines are essential for the proliferation of these protozoa. However, polyamines are also critical for mammalian cell proliferation and the finding that MYC coordinately regulates all aspects of polyamine metabolism suggests polyamines may be required to support cancer promotion by MYC. Pre-emptive blockade of polyamine synthesis is sufficient to block tumor initiation in an otherwise fully penetrant transgenic mouse model of neuroblastoma driven by MYCN, underscoring the necessity of polyamines in this process. Moreover, polyamine depletion regimens exert potent anti-tumor activity in pre-clinical models of established neuroblastoma as well, in combination with numerous chemotherapeutic agents and even in tumors with unfavorable genetic features such as MYCN, ALK or TP53 mutation. This has led to the testing of DFMO in clinical trials for children with neuroblastoma. Current trial designs include testing lower dose DFMO alone (2,000 mg/m(2)/day) starting at the completion of standard therapy, or higher doses combined with chemotherapy (up to 9,000 mg/m(2)/day) for patients with relapsed disease that has progressed. In this review we will discuss important considerations for the future design of DFMO-based clinical trials for neuroblastoma, focusing on the need to better define the principal mechanisms of anti-tumor activity for polyamine depletion regimens. Putative DFMO activities that are both cancer cell intrinsic (targeting the principal oncogenic driver, MYC) and cancer cell extrinsic (altering the tumor microenvironment to support anti-tumor immunity) will be discussed. Understanding the mechanisms of DFMO activity are critical in determining how it might be best leveraged in upcoming clinical trials. This mechanistic approach also provides a platform by which iterative pre-clinical testing using translational tumor models may complement our clinical approaches.
Collapse
|
136
|
Liao C, Wang Y, Tan X, Sun L, Liu S. Discovery of novel inhibitors of human S-adenosylmethionine decarboxylase based on in silico high-throughput screening and a non-radioactive enzymatic assay. Sci Rep 2015; 5:10754. [PMID: 26030749 PMCID: PMC5377238 DOI: 10.1038/srep10754] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 04/27/2015] [Indexed: 12/12/2022] Open
Abstract
Natural polyamines are small polycationic molecules essential for cell growth and development, and elevated level of polyamines is positively correlated with various cancers. As a rate-limiting enzyme of the polyamine biosynthetic pathway, S-adenosylmethionine decarboxylase (AdoMetDC) has been an attractive drug target. In this report, we present the discovery of novel human AdoMetDC (hAdoMetDC) inhibitors by coupling computational and experimental tools. We constructed a reasonable computational structure model of hAdoMetDC that is compatible with general protocols for high-throughput drug screening, and used this model in in silico screening of hAdoMetDC inhibitors against a large compound library using a battery of computational tools. We also established and validated a simple, economic, and non-radioactive enzymatic assay, which can be adapted for experimental high-throughput screening of hAdoMetDC inhibitors. Finally, we obtained an hAdoMetDC inhibitor lead with a novel scaffold. This study provides both new tools and a new lead for the developing of novel hAdoMetDC inhibitors.
Collapse
Affiliation(s)
- Chenzeng Liao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yanlin Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Xiao Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Lidan Sun
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| | - Sen Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
- College of Medical Science, China Three Gorges University, Yichang 443002, China
| |
Collapse
|