1
|
Co-Expression of Adaptor Protein FAM159B with Different Markers for Neuroendocrine Cells: An Immunocytochemical and Immunohistochemical Study. Int J Mol Sci 2022; 23:ijms232113503. [DOI: 10.3390/ijms232113503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/23/2022] [Accepted: 11/03/2022] [Indexed: 11/06/2022] Open
Abstract
Little is known about the adaptor protein FAM159B. Recently, FAM159B was shown to be particularly expressed in neuroendocrine cells and tissues, such as pancreatic islets and neuroendocrine cells of the bronchopulmonary and gastrointestinal tracts, as well as in different types of neuroendocrine tumours. To gain insights into possible interactions of FAM159B with other proteins and/or receptors, we analysed the co-expression of FAM159B and various neuroendocrine-specific markers in the cancer cell lines BON-1, PC-3, NCI-h82, OH-1, and A431 and also in human pancreatic tissues and pancreatic neuroendocrine tumours. The markers included prominent markers of neuroendocrine differentiation, such as chromogranin A (CgA), neuron-specific enolase (NSE), synaptophysin (SYP), insulinoma-associated protein 1 (INSM1), neural cell adhesion molecule 1 (NCAM1), serotonin (5-HT), somatostatin-14/28 (SST), and several receptors that are typically expressed by neuroendocrine cells, such as dopamine receptor 2 (D2R), somatostatin receptor (SSTR) 1, 2, 3, 4 and 5, and regulator of G-protein signalling 9 (RGS9). FAM159B was expressed evenly throughout the cytosol in all five cancer cell lines. Immunocytochemical and immunohistochemical analyses revealed co-expression of FAM159B with SYP, INSM1, RGS9, D2R, SSTR2, SSTR3, SSTR4, and SSTR5 and strong overlapping co-localisation with NSE. Double-labelling and co-immunoprecipitation Western blot analyses confirmed a direct association between FAM159B and NSE. These results suggest the involvement of FAM159B in several intracellular signalling pathways and a direct or indirect influence on diverse membrane proteins and receptors.
Collapse
|
2
|
Chen J, Song Y, Li M, Zhang Y, Lin T, Sun J, Wang D, Liu Y, Guo J, Yu W. Comprehensive analysis of ceRNA networks reveals prognostic lncRNAs related to immune infiltration in colorectal cancer. BMC Cancer 2021; 21:255. [PMID: 33750326 PMCID: PMC7941714 DOI: 10.1186/s12885-021-07995-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 02/28/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Competing endogenous RNA (ceRNA) represents a class of RNAs (e.g., long noncoding RNAs [lncRNAs]) with microRNA (miRNA) binding sites, which can competitively bind miRNA and inhibit its regulation of target genes. Increasing evidence has underscored the involvement of dysregulated ceRNA networks in the occurrence and progression of colorectal cancer (CRC). The purpose of this study was to construct a ceRNA network related to the prognosis of CRC and further explore the potential mechanisms that affect this prognosis. METHODS RNA-Seq and miRNA-Seq data from The Cancer Genome Atlas (TCGA) were used to identify differentially expressed lncRNAs (DElncRNAs), microRNAs (DEmiRNAs), and mRNAs (DEmRNAs), and a prognosis-related ceRNA network was constructed based on DElncRNA survival analysis. Subsequently, pathway enrichment, Pearson correlation, and Gene Set Enrichment Analysis (GSEA) were performed to determine the function of the genes in the ceRNA network. Gene Expression Profiling Interactive Analysis (GEPIA) and immunohistochemistry (IHC) were also used to validate differential gene expression. Finally, the correlation between lncRNA and immune cell infiltration in the tumor microenvironment was evaluated based on the CIBERSORT algorithm. RESULTS A prognostic ceRNA network was constructed with eleven key survival-related DElncRNAs (MIR4435-2HG, NKILA, AFAP1-AS1, ELFN1-AS1, AC005520.2, AC245884.8, AL354836.1, AL355987.4, AL591845.1, LINC02038, and AC104823.1), 54 DEmiRNAs, and 308 DEmRNAs. The MIR4435-2HG- and ELFN1-AS1-associated ceRNA subnetworks affected and regulated the expression of the COL5A2, LOX, OSBPL3, PLAU, VCAN, SRM, and E2F1 target genes and were found to be related to prognosis and tumor-infiltrating immune cell types. CONCLUSIONS MIR4435-2HG and ELFN1-AS1 are associated with prognosis and tumor-infiltrating immune cell types and could represent potential prognostic biomarkers or therapeutic targets in colorectal carcinoma.
Collapse
Affiliation(s)
- Jingyi Chen
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, 100044 China
| | - Yuxuan Song
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, 300052 China
| | - Mei Li
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
| | - Yu Zhang
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, 100044 China
| | - Tingru Lin
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, 100044 China
| | - Jie Sun
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
| | - Di Wang
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, 100044 China
| | - Jingzhu Guo
- Department of Paediatrics, Peking University People’s Hospital, Beijing, 100044 China
| | - Weidong Yu
- Department of Central Laboratory & Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing, 100044 China
| |
Collapse
|
3
|
Chalishazar MD, Wait SJ, Huang F, Ireland AS, Mukhopadhyay A, Lee Y, Schuman SS, Guthrie MR, Berrett KC, Vahrenkamp JM, Hu Z, Kudla M, Modzelewska K, Wang G, Ingolia NT, Gertz J, Lum DH, Cosulich SC, Bomalaski JS, DeBerardinis RJ, Oliver TG. MYC-Driven Small-Cell Lung Cancer is Metabolically Distinct and Vulnerable to Arginine Depletion. Clin Cancer Res 2019; 25:5107-5121. [PMID: 31164374 DOI: 10.1158/1078-0432.ccr-18-4140] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/30/2019] [Accepted: 05/30/2019] [Indexed: 01/12/2023]
Abstract
PURPOSE Small-cell lung cancer (SCLC) has been treated clinically as a homogeneous disease, but recent discoveries suggest that SCLC is heterogeneous. Whether metabolic differences exist among SCLC subtypes is largely unexplored. In this study, we aimed to determine whether metabolic vulnerabilities exist between SCLC subtypes that can be therapeutically exploited. EXPERIMENTAL DESIGN We performed steady state metabolomics on tumors isolated from distinct genetically engineered mouse models (GEMM) representing the MYC- and MYCL-driven subtypes of SCLC. Using genetic and pharmacologic approaches, we validated our findings in chemo-naïve and -resistant human SCLC cell lines, multiple GEMMs, four human cell line xenografts, and four newly derived PDX models. RESULTS We discover that SCLC subtypes driven by different MYC family members have distinct metabolic profiles. MYC-driven SCLC preferentially depends on arginine-regulated pathways including polyamine biosynthesis and mTOR pathway activation. Chemo-resistant SCLC cells exhibit increased MYC expression and similar metabolic liabilities as chemo-naïve MYC-driven cells. Arginine depletion with pegylated arginine deiminase (ADI-PEG 20) dramatically suppresses tumor growth and promotes survival of mice specifically with MYC-driven tumors, including in GEMMs, human cell line xenografts, and a patient-derived xenograft from a relapsed patient. Finally, ADI-PEG 20 is significantly more effective than the standard-of-care chemotherapy. CONCLUSIONS These data identify metabolic heterogeneity within SCLC and suggest arginine deprivation as a subtype-specific therapeutic vulnerability for MYC-driven SCLC.
Collapse
Affiliation(s)
- Milind D Chalishazar
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Sarah J Wait
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Fang Huang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas.,Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Abbie S Ireland
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Anandaroop Mukhopadhyay
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Younjee Lee
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Sophia S Schuman
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Matthew R Guthrie
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Kristofer C Berrett
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Jeffery M Vahrenkamp
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Zeping Hu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas.,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Marek Kudla
- Department of Molecular and Cell Biology, Center for RNA Systems Biology, University of California, Berkeley, California
| | - Katarzyna Modzelewska
- Preclinical Research Resource, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Guoying Wang
- Preclinical Research Resource, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Nicholas T Ingolia
- Department of Molecular and Cell Biology, Center for RNA Systems Biology, University of California, Berkeley, California
| | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - David H Lum
- Preclinical Research Resource, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah
| | - Sabina C Cosulich
- Bioscience Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | | | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Pediatrics and Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Trudy G Oliver
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, Utah.
| |
Collapse
|
4
|
Battaglia V, Shields CD, Murray-Stewart T, Casero RA. Polyamine catabolism in carcinogenesis: potential targets for chemotherapy and chemoprevention. Amino Acids 2014; 46:511-9. [PMID: 23771789 PMCID: PMC3795954 DOI: 10.1007/s00726-013-1529-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 05/30/2013] [Indexed: 01/01/2023]
Abstract
Polyamines, including spermine, spermidine, and the precursor diamine, putrescine, are naturally occurring polycationic alkylamines that are required for eukaryotic cell growth, differentiation, and survival. This absolute requirement for polyamines and the need to maintain intracellular levels within specific ranges require a highly regulated metabolic pathway primed for rapid changes in response to cellular growth signals, environmental changes, and stress. Although the polyamine metabolic pathway is strictly regulated in normal cells, dysregulation of polyamine metabolism is a frequent event in cancer. Recent studies suggest that the polyamine catabolic pathway may be involved in the etiology of some epithelial cancers. The catabolism of spermine to spermidine utilizes either the one-step enzymatic reaction of spermine oxidase (SMO) or the two-step process of spermidine/spermine N (1)-acetyltransferase (SSAT) coupled with the peroxisomal enzyme N (1)-acetylpolyamine oxidase. Both catabolic pathways produce hydrogen peroxide and a reactive aldehyde that are capable of damaging DNA and other critical cellular components. The catabolic pathway also depletes the intracellular concentrations of spermidine and spermine, which are free radical scavengers. Consequently, the polyamine catabolic pathway in general and specifically SMO and SSAT provide exciting new targets for chemoprevention and/or chemotherapy.
Collapse
Affiliation(s)
- Valentina Battaglia
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Christina DeStefano Shields
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Program in Molecular and Translational Toxicology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21231, USA
| | - Tracy Murray-Stewart
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert A. Casero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
5
|
Marton LJ, Feuerstein BG. Polyamine-DNA Interactions: Possible Site of New Cancer Chemotherapeutic Intervention. Pharm Res 2013; 3:311-7. [PMID: 24271829 DOI: 10.1023/a:1016375705487] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The effects of polyamines on the structure of nucleic acids in cell-free systems and in cell culture systems are reviewed. Evidence suggests that polyamine depletion inhibits cell growth and may cause conformational changes in DNA. These effects may be exploited to cause changes in the action of drugs and may be used to advantage in combination treatment protocols. A discussion of theoretical models of the interactions, physicochemical evidence for conformational changes, and the effects of anticancer drugs in cells depleted of polyamines is presented.
Collapse
Affiliation(s)
- L J Marton
- Department of Laboratory Medicine, School of Medicine, University of California, San Francisco, California, 94143
| | | |
Collapse
|
6
|
Polyamines and cancer: implications for chemotherapy and chemoprevention. Expert Rev Mol Med 2013; 15:e3. [PMID: 23432971 DOI: 10.1017/erm.2013.3] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Polyamines are small organic cations that are essential for normal cell growth and development in eukaryotes. Under normal physiological conditions, intracellular polyamine concentrations are tightly regulated through a dynamic network of biosynthetic and catabolic enzymes, and a poorly characterised transport system. This precise regulation ensures that the intracellular concentration of polyamines is maintained within strictly controlled limits. It has frequently been observed that the metabolism of, and the requirement for, polyamines in tumours is frequently dysregulated. Elevated levels of polyamines have been associated with breast, colon, lung, prostate and skin cancers, and altered levels of rate-limiting enzymes in both biosynthesis and catabolism have been observed. Based on these observations and the absolute requirement for polyamines in tumour growth, the polyamine pathway is a rational target for chemoprevention and chemotherapeutics. Here we describe the recent advances made in the polyamine field and focus on the roles of polyamines and polyamine metabolism in neoplasia through a discussion of the current animal models for the polyamine pathway, chemotherapeutic strategies that target the polyamine pathway, chemotherapeutic clinical trials for polyamine pathway-specific drugs and ongoing clinical trials targeting polyamine biosynthesis.
Collapse
|
7
|
Polyamine transport as a target for treatment of Pneumocystis pneumonia. Antimicrob Agents Chemother 2009; 53:5259-64. [PMID: 19805570 DOI: 10.1128/aac.00662-09] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Polyamine levels are greatly increased in alveolar macrophages (AMs) during Pneumocystis pneumonia (PCP), leading to increased production of H(2)O(2), which causes AMs to undergo apoptosis. One of the mechanisms by which polyamine levels in AMs are elevated is enhanced uptake of exogenous polyamines. In this study, the possibility of targeting polyamine uptake as a treatment for PCP was examined. Four anthracene- and one benzene-polyamine conjugates that are potential polyamine transport inhibitors, including N1-anthracen-9-ylmethyl-butane-1,4-diamine; N-(4-aminobutyl)-N-anthracen-9-ylmethylbutane-1,4-diamine; N-[4-(4-aminobutylamino)butyl]-N-anthracen-9-ylmethylbutane-1,4-diamine; N-(4-amino-butyl)-N'-(10-[[4-(4-amino-butylamino)butylamino]-methyl]anthracen-9-ylmethyl)butane-1,4-diamine (44-Ant-44); and benzene-polyamine conjugate N-(4-amino-butyl)-N'-(4-[[4-(4-amino-butylamino)butylamino]-methyl]benzyl)butane-1,4-diamine (44-Bn-44), were tested. Compounds 44-Ant-44 and 44-Bn-44 were found to have a very low toxicity to AMs in vitro and were evaluated for their therapeutic effect on PCP in vivo. Sprague-Dawley rats infected with P. carinii for 28 days were intranasally instilled with 50 microl of a 1 mM solution of 44-Bn-44 or 44-Ant-44 every 2 days. Twenty-one days after initiation of the treatment, three to five rats from each group were sacrificed and examined for lung pathology, organism burden, and apoptosis of AMs. Both 44-Bn-44 and 44-Ant-44 reduced organism burdens; however, only 44-Ant-44 decreased the severity of the infection with reduced lung inflammation, increased clearance of exudates, increased air space, and decreased apoptosis of AMs. 44-Ant-44 also significantly prolonged the survival of treated animals. These results suggest that polyamine uptake is a potential target for treatment of PCP.
Collapse
|
8
|
Casero RA, Marton LJ. Targeting polyamine metabolism and function in cancer and other hyperproliferative diseases. Nat Rev Drug Discov 2007; 6:373-90. [PMID: 17464296 DOI: 10.1038/nrd2243] [Citation(s) in RCA: 576] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The polyamines spermidine and spermine and their diamine precursor putrescine are naturally occurring, polycationic alkylamines that are essential for eukaryotic cell growth. The requirement for and the metabolism of polyamines are frequently dysregulated in cancer and other hyperproliferative diseases, thus making polyamine function and metabolism attractive targets for therapeutic intervention. Recent advances in our understanding of polyamine function, metabolic regulation, and differences between normal cells and tumour cells with respect to polyamine biology, have reinforced the interest in this target-rich pathway for drug development.
Collapse
Affiliation(s)
- Robert A Casero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|
9
|
Allgayer H, Roisch U, Zehnter E, Ziegenhagen DJ, Dienes HP, Kruis W. Colonic ornithine decarboxylase in inflammatory bowel disease: ileorectal activity gradient, guanosine triphosphate stimulation, and association with epithelial regeneration but not the degree of inflammation and clinical features. Dig Dis Sci 2007; 52:25-30. [PMID: 17171446 DOI: 10.1007/s10620-006-9515-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Accepted: 07/05/2006] [Indexed: 12/09/2022]
Abstract
The role of colonic mucosal ornithine decarboxylase (ODC) in inflammatory bowel disease (IBD) remains controversial. This study assessed mucosal ODC activity in IBD patients segment by segment with regard to patient characteristics, disease activity/duration, medication, degree of mucosal inflammation, and presence/absence of epithelial regeneration and guanosine triphosphate (GTP) stimulation. Mucosal ODC activity was determined in biopsy specimens from the terminal ileum, cecum/ascending, transverse, and descending colon, and the sigmoid/rectum of 35 patients with IBD (18 with Crohn's disease, 17 with ulcerative colitis) and 29 controls, using the amount of 14CO2 liberated from (carboxyl-14C)ornithine hydrochloride. GTP-stimulatable activity was expressed as the ratio of ODC activity in the presence and absence of GTP (70 micromol/L). Mucosal inflammation was assessed endoscopically/microscopically with previously described criteria. Presence/absence of mucosal regeneration also was determined by predefined criteria. Mucosal ODC-activity did not significantly differ in IBD patients and controls. There was a 4.4-fold activity gradient from the ileum to the rectum. Mucosal ODC activity was significantly higher in areas with epithelial regeneration compared to those without regeneration, and was stimulated by GTP by a factor of 1.42 in Crohn's disease and 1.19 in ulcerative colitis patients compared to controls (p < 0.004). On the other hand, there was no significant association/relationship of mucosal ODC activity with disease activity/duration and the endoscopic/histologic degree of mucosal inflammation. The observation of unchanged mucosal ODC activity in patients with IBD and the absence of a significant relationship with clinical and endoscopic/histologic disease characteristics speaks against a major role of ODC in IBD as a major disease marker. The role of the ileorectal gradient, the enhanced activity in areas with epithelial regeneration, and the GTP-stimulatable form, however, need further investigation with regard to a possible involvement in carcinogenesis in IBD.
Collapse
Affiliation(s)
- Hubert Allgayer
- Department of Gastroenterology and Metabolism, Rehaklinik Ob der Tauber, Bad Mergentheim, Academic Teaching Hospital, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
10
|
Abstract
Neuroendocrine (NE) cancers occur in multiple anatomic locations and range in prognosis from indolent to aggressive. In addition, adenocarcinomas can express gene products associated with NE cells, referred to as NE differentiation (NED), which correlates with poor prognosis and aggressive disease. Several metabolites and peptides produced by NE cells have been discovered that engage in cellular signaling and have autocrine and paracrine effects on cancer cell proliferation. This review focuses on the current knowledge of small molecule metabolism in NE cancers involving the synthesis of biogenic amine, polyamine, and amino acid neurotransmitters. Systems biology-directed approaches to NE cancer metabolism using gene expression profiling, liquid chromatography/mass spectrometry (LC/MS) and nuclear magnetic resonance (NMR) are also discussed. Furthermore, knowledge of metabolic and signaling pathways in NE cancers has led to the successful implementation of therapeutic regimens in cell culture and animal models of NE carcinogenesis.
Collapse
Affiliation(s)
- Joseph E Ippolito
- Center for Genome Sciences, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
11
|
Bisceglia JÁ, García MB, Massa R, Magri ML, Zani M, Gutkind GO, Orelli LR. Synthesis, characterization and biological activity of bis(3-Aryl-1-hexahydropyrimidinyl)methanes. Novel heterocyclic polyamine derivatives. J Heterocycl Chem 2004. [DOI: 10.1002/jhet.5570410113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
12
|
Orelli LR, Blanco MM, García MB, Hedrera ME, Perillo IA. A NEW SYNTHETIC APPROACH TON,N′-DISUBSTITUTED 1,n-ALKANEDIAMINES. SYNTHETIC COMMUN 2001. [DOI: 10.1081/scc-100103257] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
13
|
Casero RA, Mank AR, Saab NH, Wu R, Dyer WJ, Woster PM. Growth and biochemical effects of unsymmetrically substituted polyamine analogues in human lung tumor cells 1. Cancer Chemother Pharmacol 1995; 36:69-74. [PMID: 7720179 DOI: 10.1007/bf00685735] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Three unsymmetrically substituted polyamine analogues demonstrate significant and selective antitumor effects. Each of the analogues N1-ethyl-N11-propargyl-4,8-diazaundecane (PENSpm), N1-ethyl-N11-(cyclobutyl)methyl-4,8-diazaundecane (CBENSpm), and N1-ethyl-N11-(cyclopropyl)methyl-4,8-diazaundecane (CPENSpm) is cytotoxic to a representative non-small-cell lung carcinoma line, NCI H157, while being only growth-inhibitory to a representative small-cell-lung carcinoma line, NCI H82. Cytotoxicity is accompanied by a significant increase in expression of the polyamine catabolic enzyme spermidine/spermine N1-acetyltransferase (SSAT) at the levels of activity and steady-state mRNA. These new analogues are significant both for their cell-type-specific activity and as synthetic prototypes for the addition of SSAT-activated functional groups.
Collapse
Affiliation(s)
- R A Casero
- Oncology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | |
Collapse
|
14
|
Fredlund JO, Johansson M, Baldetorp B, Oredsson SM. Abnormal DNA synthesis in polyamine deficient cells revealed by bromodeoxyuridine-flow cytometry technique. Cell Prolif 1994; 27:243-56. [PMID: 10465009 DOI: 10.1111/j.1365-2184.1994.tb01422.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Chinese hamster ovary cells were seeded in the absence or presence of the polyamine synthesis inhibitor 2-difluoromethylornithine (DFMO). At 1-4 days after seeding, the cells were labelled for 15-120 min with the thymidine analogue bromodeoxyuridine (BrdUrd) and they were then fixed directly after the labelling period. In addition, cells were labelled for 30 min and they were then allowed to progress in BrdUrd-free medium during a defined post-labelling time before fixation. An indirect immunofluorescence technique, using the monoclonal BrdUrd antibody and the intercalating stoichiometric DNA stain, propidium iodide, was applied to enable quantification of cellular BrdUrd and DNA contents, respectively, by flow cytometry (FCM). By comparing the mean DNA content of BrdUrd-labelled cells to the mean DNA contents of G1 and G2 cells, a relative measure of the position of the BrdUrd-labelled cells was obtained (relative movement). Relative movement data, obtained from control and DFMO-treated cells fixed directly after BrdUrd labelling, indicated that DFMO-treated cells entered S phase at a normal rate, while their progression through S phase was impaired. DNA histograms of BrdUrd-labelled control cells fixed directly after labelling showed that most cells were found in early and late S phase, while DNA histograms of BrdUrd-labelled DFMO-treated cells showed that most cells were in early S phase, indicating a delayed progression through S phase. Analysis of relative movement of cells that were allowed to progress in BrdUrd-free medium after labelling showed that DFMO treatment resulted in a significant lengthening of the DNA synthesis time. Labelling index was significantly higher in DFMO-treated, growth-inhibited cells than in early plateau phase control cells indicating an S phase accumulation in the former cells.
Collapse
Affiliation(s)
- J O Fredlund
- Department of Animal Physiology, University of Lund, Sweden
| | | | | | | |
Collapse
|
15
|
Hessels J, Kingma AW, Muskiet FA, Sarhan S, Seiler N. Growth inhibition of two solid tumors in mice, caused by polyamine depletion, is not attended by alterations in cell-cycle phase distribution. Int J Cancer 1991; 48:697-703. [PMID: 2071231 DOI: 10.1002/ijc.2910480512] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We studied the effect of polyamine depletion on growth and cell cycle characteristics of subcutaneously grown Lewis lung carcinoma (LLC) and fibrosarcoma (FIO 26) in mice. Polyamine depletion was achieved by inhibition of ornithine decarboxylase using 2-(difluoromethyl)ornithine, limitation of exogenous polyamines by administration of a polyamine-poor diet and decontamination of the gastrointestinal tract, and inhibition of endogenous polyamine reutilization by N,N'-bis-(2,3-butadienyl)putrescine (MDL 72527). Determination of S-phase cells was performed in tumor-cell suspensions by flow cytometry and in tumor tissue sections by microscopy, following in vivo labelling with 5-bromo-2'-deoxyuridine (BUdR). DNA synthesis rate was estimated from the incorporation of in vivo-injected [3H]-thymidine (3H-TdR). Both solid tumors almost completely stopped growing after access to polyamines was blocked. Growth inhibition was, however, not attended by changes in cell-cycle-phase distribution. Paradoxically, we measured increased in vivo 3H-TdR incorporation rates and unaltered BUdR-linked staining intensity in treated tumors. Injection of putrescine into treated LLC-bearing mice resulted in an increase in intracellular putrescine and spermidine concentrations, a slight increase in the number of S-phase cells and a marked drop in DNA synthesis rate within the following 9 hr.
Collapse
Affiliation(s)
- J Hessels
- Central Laboratory for Clinical Chemistry, University Hospital Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
16
|
Casero RA, Celano P, Ervin SJ, Applegren NB, Wiest L, Pegg AE. Isolation and characterization of a cDNA clone that codes for human spermidine/spermine N1-acetyltransferase. J Biol Chem 1991. [DOI: 10.1016/s0021-9258(17)35245-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
17
|
Neelam SS, Bernabei A, Freedland C, Thompson R, Corbett TH, Luk GD. Combination of flavone acetic acid (FAA) with adriamycin, cis-platinum and difluoromethylornithine (DFMO) in vitro against human colon cancer cells. Invest New Drugs 1990; 8:263-8. [PMID: 2272766 DOI: 10.1007/bf00171835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Unresectable solid tumors in the metastatic stage are quite resistant to current chemotherapy and radiation therapy regimens. Flavone acetic acid (FAA) is a novel antitumor agent which appears to work through a different mechanism than the conventional chemotherapeutic agents. In preclinical studies it has shown effectiveness against a variety of transplantable murine and human tumors and appears to be solid tumor selective. It also has non-overlapping toxicities as compared to conventional agents. We therefore investigated FAA in vitro against human colon cancer cells and explored whether its effectiveness could be enhanced in combination with other agents such as adriamycin (ADR), cis-platinum (CP) and difluoromethylornithine (DFMO)--an inhibitor of polyamine biosynthesis. Addition of FAA for 24 hours in liquid media produced dose dependent growth inhibition. Using soft agar colony assay, growth was inhibited by 58% by 3mM FAA and only 1.4% by 0.375mM FAA. The combination of FAA and cis-platinum produced synergism at the lower doses tested. The combination of FAA and adriamycin produced antagonism at all doses tested and the combination of FAA with DFMO did not produce results significantly different from DFMO alone. We conclude that enhancement of FAA activity can be achieved in combination with conventional antitumor agents, but may be drug and dose specific.
Collapse
Affiliation(s)
- S S Neelam
- Department of Medicine, Wayne State University School of Medicine, Detroit, MI
| | | | | | | | | | | |
Collapse
|
18
|
Ajani JA, Ota DM, Grossie VB, Abbruzzese JL, Faintuch JS, Patt YZ, Jackson DE, Levin B, Nishioka K. Evaluation of continuous-infusion alpha-difluoromethylornithine therapy for colorectal carcinoma. Cancer Chemother Pharmacol 1990; 26:223-6. [PMID: 2113439 DOI: 10.1007/bf02897204] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A total of 32 evaluable patients with measurable advanced colorectal carcinoma were treated with continuous-infusion alpha-difluoromethylornithine (DFMO) at a median daily dose of 8 g/m2 (range, 6-14 g/m2). DFMO was infused over 24 h daily for 28 days, followed by a rest period of 7 days. Of the 32 patients, 14 had received no prior chemotherapy. A total of 65 courses was given, with the median being 2 (range, 1-9 courses). None of the patients achieved a partial or complete response; however, 3 patients achieved a minor response and 14 had stable disease. The frequent toxic effects of DFMO included thrombocytopenia (which was dose-limiting), malaise, nausea, vomiting, reversible hearing loss, and diarrhea. Our data suggest that continuous-infusion DFMO therapy is feasible and results in only mild gastrointestinal toxicity. Although DFMO proved to be ineffective as a single agent in this trial, it could probably best be used in combination with cytotoxic agents known to enhance its antitumor activity in a preclinical setting.
Collapse
Affiliation(s)
- J A Ajani
- Department of Medical Oncology, University of Texas M. D. Anderson Cancer Center, Houston 77030
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Simon MS, Eckenrode J, Natale RB. Phase II trial of methylglyoxal bis-guanylhydrazone (MGBG) in refractory small cell lung cancer. Invest New Drugs 1990; 8 Suppl 1:S79-81. [PMID: 2166008 DOI: 10.1007/bf00171989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Methylglyoxal bis-guanylhydrazone (MGBG), a potent inhibitor of polyamine synthesis, has demonstrated single agent activity against a number of tumor types including malignant lymphomas and head and neck, esophageal and non-small cell lung cancers. The growth of small cell lung cancer (SCLC) cell lines can be arrested by polyamine inhibition. Therefore a phase II trial was conducted in twenty-four patients with refractory SCLC. MGBG was administered by intravenous infusion at a dose of 500 mg/m2 per week for four cycles and then every two weeks thereafter. The dose was escalated by 100 mg/m2 every two weeks in the absence of toxicity greater than or equal to grade 2. One patient achieved a partial response of objectively measurable lung disease and supraclavicular adenopathy. Three patients had stable disease. Dose limiting toxicity consisted primarily of mild to moderate nausea, vomiting, stomatitis and/or diarrhea. Myelosuppression was uncommon and rarely dose limiting. We conclude that MGBG in the dose and schedule used does not have significant activity as a single agent in previously treated small cell lung cancer.
Collapse
Affiliation(s)
- M S Simon
- Department of Internal Medicine, University of Michigan, Ann Arbor
| | | | | |
Collapse
|
20
|
Zirvi KA, Dasmahapatra KS, Atabek U, Lyons MA. alpha-Difluoromethylornithine inhibits liver metastasis produced by intrasplenic injection of human tumor cells into nude mice. Clin Exp Metastasis 1989; 7:591-8. [PMID: 2505959 DOI: 10.1007/bf01753670] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The purpose of these studies was to examine metastatic potentials of a human colon tumor xenograft (T6) and three different human tumor cell lines (LS174T, HT29 and A549) using the intrasplenic-nude mouse model system (ISMS model system). A further objective was to study the activity of alpha-difluoromethyl-ornithine (DFMO) against primary and metastatic growth of the xenograft and the three cell lines. DFMO is an irreversible inhibitor of ornithine decarboxylase, a rate-limiting step in polyamine biosynthesis. Tumor burdens in the liver of nude mice were observed 6 weeks after the intrasplenic injection with LS174T and 12-14 weeks after intrasplenic injections with T6, HT29 and A549. Most of the mice developed primary tumor growth in the spleens. DFMO showed significant activity against liver metastases but had little or no activity against primary tumor growth in the spleens of the ISMS model and against s.c. growth of the xenograft. The studies demonstrated that the ISMS model system is an excellent system for studying metastatic behavior of human tumors and for studying the antimetastatic activity of experimental drugs.
Collapse
Affiliation(s)
- K A Zirvi
- Surgical Service, VA Medical Center, East Orange, NJ 07019
| | | | | | | |
Collapse
|
21
|
Horn Y, Spigel L, Marton LJ. Urinary polyamine levels in cancer patients treated with D,L-alpha-difluoromethylornithine, an inhibitor of polyamine biosynthesis. J Surg Oncol 1989; 41:177-82. [PMID: 2501593 DOI: 10.1002/jso.2930410309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The polyamine biosynthesis inhibitor D,L-alpha-Difluoromethylornithine hydrochloride monohydrate (DFMO) has cytostatic and cytotoxic effects against various human tumor cell lines in vitro. We measured levels of the polyamines putrescine and spermidine in the urine of cancer patients undergoing "conventional" chemotherapy in a two-arm randomized phase I-II study with and without additional DFMO administered orally at a dose of 1.7 g/sq.m. t.i.d. The study group included 38 patients with carcinoma of the breast, stomach, prostate, or female genital organs or metastatic carcinoma of unknown origin. A control group of 32 patients with similar malignancies received "conventional" chemotherapy without DFMO. Polyamine levels were determined periodically in the urine of all patients. In DFMO-treated patients, a significant decrease in putrescine and spermidine levels was observed after 3 weeks of DFMO therapy (the first time point evaluated) that usually persisted throughout the course of treatment. Significant differences in polyamine levels between DFMO-treated and control patients were observed for patients in remission. Less significant differences were noted, however, for patients with static or progressive disease between DFMO-treated and control groups. DFMO activity appears to be reflected by a long-term decrease in urinary polyamine levels.
Collapse
Affiliation(s)
- Y Horn
- Department of Oncology, Assaf Harofeh Medical Center, Sackler School of Medicine, Zerifin, Israel
| | | | | |
Collapse
|
22
|
Lipton A, Harvey HA, Glenn J, Weidner WA, Strauss M, Miller SE, Taylor JB, White-Hershey D, Barlow JL. A phase I study of hepatic arterial infusion using difluoromethylornithine. Cancer 1989; 63:433-7. [PMID: 2492203 DOI: 10.1002/1097-0142(19890201)63:3<433::aid-cncr2820630306>3.0.co;2-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Ten patients with metastatic disease to the liver were treated with difluoromethylornithine (DFMO) administered by continuous hepatic arterial infusion. Two of nine evaluable patients had an objective partial response. Stable disease was recorded in three patients. Ototoxicity was encountered in all patients who received a daily dose of DFMO equal to or greater than 1.0 g/m2.
Collapse
Affiliation(s)
- A Lipton
- Division of Oncology, Milton S. Hershey Medical Center, Pennsylvania State University, Hershey 17033
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Mabry M, Nakagawa T, Nelkin BD, McDowell E, Gesell M, Eggleston JC, Casero RA, Baylin SB. v-Ha-ras oncogene insertion: a model for tumor progression of human small cell lung cancer. Proc Natl Acad Sci U S A 1988; 85:6523-7. [PMID: 2842776 PMCID: PMC282005 DOI: 10.1073/pnas.85.17.6523] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Small cell lung cancer (SCLC) manifests a range of phenotypes in culture that may be important in understanding its relationship to non-SCLCs and to tumor progression events in patients. Most SCLC-derived cell lines, termed "classic" SCLC lines, have properties similar to SCLC tumors in patients, including high expression of neuroendocrine markers and low c-myc oncogene expression. A significant number of SCLC lines characterized as "biochemical or morphologic variant" SCLC lines have decreased levels of endocrine differentiation markers associated with increased proliferative indices, amplification of the c-myc oncogene, and growth patterns and biochemical markers more typical of non-SCLCs. To delineate further the relationships between these phenotypes and the molecular events involved, we have inserted the v-Ha-ras gene in SCLC cell lines with (biochemical variant) and without (classic) an amplified c-myc gene. These two SCLC subtypes had markedly different phenotypic responses to similar levels of expression of v-Ha-ras RNA. No biochemical or morphologic changes were observed in classic SCLC cells. In contrast, in biochemical variant SCLC cells, v-Ha-ras expression induced features typical of large cell undifferentiated lung carcinoma, including adherent monolayer growth patterns, increased cloning efficiency, increased levels of non-SCLC cell markers, ultrastructural characteristics and an acquired resistance to polyamine depletion typical of large cell carcinoma, but not SCLC, in vitro. Expression of v-Ha-ras in biochemical variant SCLC cells directly demonstrates that important transitions can occur between phenotypes of human lung cancer cells and that these may play a critical role in tumor progression events in patients. The findings provide a model system to study molecular events involved in tumor progression steps within a series of related tumor types.
Collapse
Affiliation(s)
- M Mabry
- Oncology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Tsai DC, Luk GD. The growth-inhibitory effect of 4-hydroperoxycyclophosphamide against human non-small cell lung carcinoma is enhanced by low-dose difluoromethylornithine. Cancer Chemother Pharmacol 1988; 22:36-40. [PMID: 2840221 DOI: 10.1007/bf00254178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Surgically unresectable human non-small cell lung carcinoma (NSCC) is highly resistant to present chemotherapy and radiation therapy regimens. Cyclophosphamide, a potent alkylating agent, has shown some efficacy, especially in combination chemotherapy. Difluoromethylornithine (DFMO), a specific and irreversible inhibitor of ornithine decarboxylase (ODC) which produces minimal toxicity in animals and humans, has shown antiproliferative effect against human SCC in culture but a much smaller effect (cytostatic) against NSCC. We therefore investigated 4-hydroperoxycyclophosphamide (4HC) and DFMO alone and in combination against a human NSCC line (NCI-H157). Cells were treated with DFMO at graded concentrations of 0 to 800 microM from day 0 to day 7. On day 3, cells were exposed for 1 h to 4HC at graded concentrations of 0 to 80 microM, washed, and refed with media containing DFMO at initial concentrations. On day 7, cells were counted by hemacytometer. Cells treated with DFMO or 4HC alone exhibited dose-dependent growth inhibition. Growth inhibition by 4HC was enhanced through combination with DFMO. On day 7, 50 microM (5 x 10(-5) M) DFMO effected a 37% inhibition, 8 microM 4HC 47% inhibition, and the combination of 50 microM DFMO and 8 microM 4HC yielded an elevated 71% inhibition. The growth inhibitory effect and potentiating effect of DFMO were reversible upon addition of putrescine (PU) to the culture medium. The combination of DFMO and 4HC, two agents with different toxicity spectra, may represent an effective chemotherapeutic regimen for the treatment of lung cancer.
Collapse
Affiliation(s)
- D C Tsai
- Oncology Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | |
Collapse
|
25
|
Manni A, Badger B, Luk G, Wright C, Caplan R, Rockette H, Bartholomew M, Ahmed SR. Role of polyamines in the growth of hormone-responsive experimental breast cancer in vivo. Breast Cancer Res Treat 1988; 11:231-40. [PMID: 3139111 DOI: 10.1007/bf01807281] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We have provided evidence for a critical role of polyamines in the growth of the hormone-responsive N-nitrosomethyl-urea (NMU)-induced rat mammary tumor in vitro. The present experiments were designed to test whether polyamines are involved in the growth of this experimental tumor in vivo. To test this hypothesis, groups of rats bearing NMU-induced mammary cancers were randomly allocated to receive no treatment or escalating doses of the polyamine biosynthesis inhibitor alpha-difluoromethyl-ornithine (DFMO) (0.5%, 1%, 2%, 3% in drinking water). DFMO inhibited tumor growth in a dose-dependent fashion and consistently reduced tumor putrescine level. To evaluate the time dependency of this effect, additional groups of rats received either no treatment or 2% DFMO for 3, 7, 14, and 21 days. At all times DFMO suppressed tumor putrescine level as well as spermidine to spermine ratio. Finally, exogenous administration of putrescine (200 mg/kg/i.p./day x 21 days) given concomitantly with DFMO restored tumor growth, partially repleted tumor putrescine level, and raised the spermidine to spermine ratio to control levels. Putrescine, given alone, had no significant effect on either tumor polyamine levels or tumor growth. Except for modest weight loss, no major toxicity was encountered. These results indicate that polyamines play an important role in the growth of the NMU rat mammary tumor in vivo. The interaction between polyamines and hormones in supporting NMU mammary tumor growth in vivo remains to be elucidated.
Collapse
Affiliation(s)
- A Manni
- Department of Medicine, Milton S. Hershey Medical Center, Pennsylvania State University, Hershey 17033
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Maddox AM, Freireich EJ, Keating MJ, Frasier-Scott KF, Haddox MK. Alterations in human circulating and bone marrow mononuclear cell polyamine levels in hematologic malignancies as a consequence of difluoromethylornithine administration. Invest New Drugs 1988; 6:125-34. [PMID: 3139580 DOI: 10.1007/bf00195371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effect of administering increasing intravenous doses of difluoromethylornithine on human tumor cell polyamine levels was determined in patients with hematologic malignancies. Difluoromethylornithine from 5.5. to 64 gm/m2 per day was administered to nine patients with refractory acute leukemia or multiple myeloma. Putrescine, spermidine, and spermine levels were determined on a daily basis in the circulating mononuclear cells and on a weekly basis in the mononuclear cells of the bone marrow. Tumor cell putrescine levels declined in 5 patients, spermidine levels declined in 4 patients, and spermine levels declined in 3 patients. Alterations in the polyamine levels of the bone marrow mononuclear cells paralleled those occurring in the peripheral blood mononuclear cells in the patients with leukemia. Seven to ten days of DFMO treatment were required for mononuclear cell polyamine levels to decrease. The higher drug doses were not significantly more effective than the lower doses in bringing about a decline in tumor cell polyamine levels, either with respect to treatment time required for onset of response or with respect to the ultimate extent of response.
Collapse
Affiliation(s)
- A M Maddox
- Department of Hematology/Oncology, University of Texas Medical School, Houston
| | | | | | | | | |
Collapse
|
27
|
Bahnson RR, Ratliff TL. Inhibition of mouse bladder tumor proliferation by alpha difluoromethylornithine and interferon in vitro and in vivo. J Urol 1988; 139:1367-71. [PMID: 3131549 DOI: 10.1016/s0022-5347(17)42921-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Previous studies have reported that alpha difluoromethylornithine (DFMO), an enzyme-activated, irreversible inhibitor of ornithine decarboxylase (ODC), has anti-tumor activity in several tumor systems. Recently, investigations have revealed that combinations of DFMO and Interferon (IFN) are synergistic in inhibiting tumor cell growth. We tested the effects of DFMO and IFN alpha, beta alone and in combination on the growth of mouse bladder tumor (MBT-2) cells both in vitro and in vivo. MBT-2 cells were incubated for 72 hours in 96 well microtiter plates with DFMO, IFN alpha, beta and combinations of both agents and percentage inhibition was calculated. In vivo studies utilized the intravesical implantation method as well as subcutaneous implantation. DFMO was administered as a 1% solution in the drinking water. IFN alpha, beta was given bi-weekly by intravesical administration. DFMO effectively inhibited MBT-2 growth in vitro. The ID50 was 0.08 mM and peak inhibitory activity was reached at concentrations of 0.16 mM and remained constant with concentrations of up to 10 mM. IFN alpha, beta also inhibited the in vitro proliferation of MBT-2 with maximum inhibition (46%) at 2,000 U/ml. Combinations of DFMO and IFN alpha, beta showed increased anti-proliferative activity. The degree of enhancement varied with synergism, additivity, or sub-additivity at varying drug concentrations. In vivo, DFMO significantly retarded the growth of tumors implanted subcutaneously (p less than .05) and significantly delayed the outgrowth of tumors implanted intravesically (p less than .01). IFN alpha, beta alone was ineffective in vivo and produced no additive effect in vivo when used in combination with DFMO. Results of our investigation show that DFMO inhibits proliferation of MBT-2 cells in vitro and exhibits a similar effect in vivo against subcutaneous and intravesical tumor implants. IFN alpha, beta alone demonstrated anti-proliferative activity in vitro but did not affect MBT-2 growth in vivo. Although the combination of DFMO and IFN alpha, beta exhibited enhanced activity in vitro, no enhancement was observed with combination therapy in vivo.
Collapse
Affiliation(s)
- R R Bahnson
- Division of Urology, Washington University School of Medicine, St. Louis, MO 63110
| | | |
Collapse
|
28
|
Sunkara PS, Zwolshen JH, Prakash NJ, Bowlin TL. Mechanism of antitumor activity of norspermidine, a structural homologue of spermidine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1988; 250:707-16. [PMID: 3255245 DOI: 10.1007/978-1-4684-5637-0_62] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- P S Sunkara
- Merrell Dow Research Institute, Cincinnati, Ohio 45215
| | | | | | | |
Collapse
|
29
|
Porter CW, Bergeron RJ. Regulation of polyamine biosynthetic activity by spermidine and spermine analogs--a novel antiproliferative strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1988; 250:677-90. [PMID: 3076344 DOI: 10.1007/978-1-4684-5637-0_60] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Interference with polyamine biosynthesis by analog-mediated regulatory mechanisms represents a viable alternative to the use of specific enzyme inhibitors as an antiproliferative strategy. The approach is unique among antimetabolite approaches and is made possible by unusual characteristics inherent to the polyamines and their biosynthetic pathway. Current antitumor data obtained with these analogs provides indication of their potential usefulness as antitumor agents but, at the same time, demonstrates the need for improvement. This latter might be attained by the rational design of analogs which (a) bind more tightly at enzyme regulatory sites, (b) which are less able to substitute for natural polyamines in growth related functions and (c) which are eliminated less rapidly from tumor-bearing animals. At the same time, the continued preclinical development of available analogs might proceed most productively by targeting large cell lung carcinoma and melanoma and by examining the generality of the relationship between oncogene expression and the accompanying sensitivity to regulatory analogs.
Collapse
Affiliation(s)
- C W Porter
- Grace Cancer Drug Center, Roswell Park Memorial Institute, Buffalo, New York 14263
| | | |
Collapse
|
30
|
Kubota S, Ohsawa N, Takaku F. Effects of DL-alpha-difluoromethylornithine on the growth and metastasis of B16 melanoma in vivo. Int J Cancer 1987; 39:244-7. [PMID: 3100460 DOI: 10.1002/ijc.2910390220] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effects of DL-alpha-difluoromethylornithine (DFMO), a specific irreversible inhibitor of ornithine decarboxylase, on the growth of experimental mouse B16-F10 melanoma cells were investigated. DFMO (3%) in drinking water was administered to B16-F10 melanoma-bearing mice. At 24 days, B16-F10 melanomas in DFMO-fed mice weighed 75% less than those in control mice (p less than 0.001). DFMO reduced putrescine and spermidine levels in B16-F10 melanoma by 98% and 84%, respectively, and prolonged the mean survival time from 25.9 +/- 1.2 to 35.7 +/- 2.2 days (p less than 0.001). The effects of DFMO on experimental metastasis were also investigated. DFMO treatment resulted in a significant decrease in pulmonary metastasis induced by i.v. injection of B16-F10 melanoma cells.
Collapse
|
31
|
Griffin CA, Slavik M, Chien SC, Hermann J, Thompson G, Blanc O, Luk GD, Baylin SB, Abeloff MD. Phase I trial and pharmacokinetic study of intravenous and oral alpha-difluoromethylornithine. Invest New Drugs 1987; 5:177-86. [PMID: 3115911 DOI: 10.1007/bf00203544] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Eflornithine-HCl (alpha-difluoromethylornithine or DFMO), an irreversible inhibitor of ornithine decarboxylase, blocks polyamine synthesis and has demonstrated antitumor activity in cell culture and animal tumor models. This phase I study was designed to determine and compare toxicity and the maximally tolerated dose of a 4-day course of DFMO given to patients in oral, continuous intravenous infusion or pulse intravenous infusion forms. Twenty-four patients were entered into this study: 8 received intravenous pulse drug, 10 intravenous continuous infusion of drug, and 6 oral DFMO. The most frequent toxicity was nausea and vomiting which occurred in 9 courses of oral drug. Only two patients receiving intravenous DFMO had nausea and vomiting. Clinically significant thrombocytopenia and audiometric abnormalities were not encountered in contrast to previous experience with 28-day courses of oral DFMO. The maximally tolerated dose of a four-day course of oral DFMO was 3.75 gm/M2 every 6 hours. The maximally tolerated dose of intravenous pulse and continuous infusion DFMO was not attained. Pharmacokinetic studies demonstrated that the intravenous schedules achieved higher plasma levels of DFMO than those previously obtained with chronic oral dosing.
Collapse
Affiliation(s)
- C A Griffin
- Johns Hopkins Oncology Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kaur K, Emmett K, McCann PP, Sjoerdsma A, Ullman B. Effects of DL-alpha-difluoromethylornithine on Leishmania donovani promastigotes. THE JOURNAL OF PROTOZOOLOGY 1986; 33:518-21. [PMID: 3098971 DOI: 10.1111/j.1550-7408.1986.tb05654.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Alpha-difluoromethylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, has been demonstrated to be an effective agent against a variety of parasitic protozoa but not against Leishmania spp. In this report, we show that Leishmania donovani promastigotes in continuous culture are sensitive to the growth inhibitory and cytotoxic effects of DFMO. Incubation of the promastigotes with DFMO obliterates intracellular putrescine pools and depletes spermidine concentrations, which correlates with the onset of growth inhibition. The effects of DFMO on the growth and the intracellular polyamine pools can be reversed completely by the addition of 10 microM putrescine to the culture medium. These results suggest that the treatment of leishmaniasis may be amenable to chemotherapeutic manipulation by DFMO.
Collapse
|
33
|
Courdi A, Milano G, Bouclier M, Lalanne CM. Radiosensitization of human tumor cells by alpha-difluoromethylornithine. Int J Cancer 1986; 38:103-7. [PMID: 3087889 DOI: 10.1002/ijc.2910380117] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effect of polyamine depletion on the radiosensitivity of a human tumor cell line was investigated. CAL 18 A cells, derived from a breast carcinoma, were incubated with alpha-difluoromethylornithine (DFMO)--a specific and irreversible inhibitor of ornithine decarboxylase (ODC)--at a 1 mM or 10-mM concentration for either 1 hr or 24 hr and irradiated thereafter. Survival curves of exponentially growing cells revealed a moderate but significant enhancement of radiosensitivity as compared to untreated irradiated cells. Maximum radiosensitization was observed at a concentration of 10 mM after 1 hr incubation. Plateau-phase cells were used to study the effect of polyamine inhibition on repair of radiation-induced potentially lethal damage (PLD). DFMO enhanced the radiation response and significantly inhibited PLD repair in these cells. Measurement of ODC indicated that this enzyme was markedly inactivated upon brief incubation of CAL 18 A cells with DFMO, reflecting a depletion of polyamine synthesis. These results extend findings that have demonstrated enhancement of drug-induced cytotoxicity, and raise the possibility of clinical use of this substance for potentiation of radiation response.
Collapse
|
34
|
Watanabe T, Sherman M, Shafman T, Iwata T, Kufe D. Effects of ornithine decarboxylase inhibition on c-myc expression during murine erythroleukemia cell proliferation and differentiation. J Cell Physiol 1986; 127:480-4. [PMID: 3458709 DOI: 10.1002/jcp.1041270319] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The polyamines putrescine, spermidine, and spermine have been implicated in the regulation of cellular proliferation and differentiation. We have previously demonstrated that spermidine is required for proliferation of murine Friend erythroleukemia (MEL) cells. We have also shown that spermidine is required at a transcriptional level for induction of MEL globin synthesis. Since studies monitoring c-myc expression have suggested that this gene also plays a role in both growth and differentiation, we have monitored the effects of inhibiting ornithine decarboxylase (ODCase) activity and polyamine synthesis on levels of c-myc transcripts. The results demonstrate that the level of c-myc RNA is independent of ODCase inhibition and depletion of intracellular spermidine. More importantly, arrest of MEL proliferation is not associated with detectable changes in c-myc expression, while under these conditions there is a decline in ODCase transcripts. During induction of MEL differentiation with dimethyl sulfoxide (DMSO) and hexamethylene bisacetamide (HMBA), c-myc and ODCase undergo similar changes in patterns of expression. However, although spermidine is required for appearance of the differentiated MEL phenotype, depletion of this polyamine by ODCase inhibition had no detectable effect on the biphasic changes in c-myc RNA observed during MEL differentiation. Thus, these biphasic changes in c-myc expression are not sufficient for induction of the mature phenotype. Finally, these results would indicate that the regulation of c-myc expression during both proliferation and differentiation is independent of ODCase activity and inhibition of proliferation by spermidine depletion.
Collapse
|
35
|
McGovern KA, Clark RS, Pegg AE. Effect of 1,3,6-triaminohexane and 1,4,7-triaminoheptane on growth and polyamine metabolism in SV-3T3 cells treated with 2-difluoromethylornithine. J Cell Physiol 1986; 127:311-6. [PMID: 3009500 DOI: 10.1002/jcp.1041270219] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The possibility that one or both of the synthetic triamines, 1,3,6-triaminohexane and 1,4,7-triaminoheptane, could substitute for the naturally occurring polyamines in the growth of SV-3T3 cells was investigated. It was found that these triamines did lead to a restoration of growth in cells in which spermidine content had been depleted by exposure to the ornithine decarboxylase inhibitor 2-difluoromethylornithine. This resumption of a normal growth rate occurred prior to the reduction in the content of cellular decarboxylated S-adenosylmethionine, suggesting that this nucleoside (which increases in concentration several hundred-fold in cells treated with 2-difluoromethylornithine) does not cause the reduction of cell growth. However, unlike the increase in cell growth brought about by spermidine, which continued indefinitely, the increase produced by 1,3,6-triaminohexane or 1,4,7-triaminoheptane was transient. Cell growth in the presence of 2-difluoromethylornithine and these triamines stopped after about three or four population doublings. This corresponded to the time at which the intracellular spermine content of the cells was reduced to values less than 20% of normal. It is suggested that the increased growth rate of spermidine-depleted cells in response to these triamines is due to their uptake into the cell and ability to displace spermine from intracellular sites, thus making spermine available to fulfill the polyamine function(s) essential for growth. These results indicate that the naturally occurring polyamines spermidine or spermine are essential for continued cell growth and cannot be replaced by analogues containing only primary amino groups.
Collapse
|
36
|
Kingsnorth AN. The chemotherapeutic potential of polyamine antimetabolites. Ann R Coll Surg Engl 1986; 68:76-81. [PMID: 3082276 PMCID: PMC2497796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The polyamines, putrescine, spermidine and spermine are small cationic molecules essential for DNA synthesis and cell replication. Because the cytotoxicity of most anti-cancer drugs can be attributed to inhibitory effects on DNA synthesis and cell replication it led to speculation that inhibition of polyamine synthesis could be a useful tool in the control of neoplastic growth. In 1978 alpha-difluoromethylornithine (DFMO), a powerful inhibitor of ornithine decarboxylase, the rate limiting enzyme in polyamine synthesis, was synthesized by Metcalf. Since then numerous investigators have tested the potential of DFMO and other polyamine antimetabolites as chemotherapeutic agents in experimental animals and cell cultures. The accumulated knowledge is now being evaluated in the treatment of human proliferative disorders and cancer.
Collapse
|
37
|
Gupta PK, Myers JD, Baylin SB, Mulshine JL, Cuttitta F, Gazdar AF. Improved antigen detection in ethanol-fixed cytologic specimens. A modified avidin-biotin-peroxidase complex (ABC) method. Diagn Cytopathol 1985; 1:133-6. [PMID: 3013531 DOI: 10.1002/dc.2840010209] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A new amplification technique is described for the detection of small amounts of antigen in ethanol-fixed cytologic specimens and formalin-fixed tissues. By adding a third antibody layer to the already sensitive avidin-biotin immunoperoxidase system, we have demonstrated a considerable increase in both the quantitative and qualitative aspects of this immunostaining method. In evaluating this technique, we employed a previously defined small cell lung carcinoma cell surface antigen system and monoclonal antibodies.
Collapse
|
38
|
Bowlin TL, Rosenberger AL, Sunkara PS. The effect of combination treatment with alpha-difluoromethylornithine and Corynebacterium parvum on B16 melanoma growth and tumoricidal effector cell generation in vivo. Cancer Immunol Immunother 1985; 20:214-8. [PMID: 3933819 PMCID: PMC11038897 DOI: 10.1007/bf00205579] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/1985] [Accepted: 05/13/1985] [Indexed: 01/08/2023]
Abstract
The objective of the present investigation was to establish whether a known lymphoreticular-stimulating agent Corynebacterium parvum would augment the established antitumor activity of alpha-difluoromethylornithine in vivo. Furthermore, since C. parvum is known to boost cell mediated cytotoxicity, the effect of DFMO (DL-alpha-difluoromethylornithine X HCl X H2O) treatment was evaluated on macrophage and natural killer (NK) cell tumoricidal activity. DFMO administered alone, 1% or 2% in drinking water, inhibited 49.4% or 88.0% of B16 melanoma growth in vivo, respectively. Administration of C. parvum alone, three doses of 300 micrograms each, inhibited tumor growth 57.4%. When administered together, DFMO and C. parvum treatment resulted in 89.8% (1% DFMO) or 97.4% (2% DFMO) inhibition of melanoma growth depending upon the dose of DFMO. C. parvum-treated animals had increased levels of macrophage-mediated tumoricidal activity directed against B16 melanoma cells in vitro, however, NK cell activity was reduced. DFMO treatment alone had no effect on macrophage or NK cell tumoricidal activity. In animals receiving both C. parvum and DFMO treatments macrophage-mediated tumoricidal activity was augmented. These results demonstrate that C. parvum can augment the antitumor activity of DFMO in vivo, possibly through macrophage activation. Furthermore, in contrast to many other cancer chemotherapeutic drugs, DFMO is apparently not immunosuppressive regarding tumoricidal effector cells.
Collapse
|
39
|
Russell DH. Ornithine decarboxylase: a key regulatory enzyme in normal and neoplastic growth. Drug Metab Rev 1985; 16:1-88. [PMID: 3905315 DOI: 10.3109/03602538508991430] [Citation(s) in RCA: 179] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
40
|
Maddox AM, Keating MJ, McCredie KE, Estey E, Freireich EJ. Phase I evaluation of intravenous difluoromethylornithine--a polyamine inhibitor. Invest New Drugs 1985; 3:287-92. [PMID: 3934106 DOI: 10.1007/bf00179433] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Difluoromethylornithine (DFMO), a non-competitive inhibitor of ornithine decarboxylase (ODC), the rate limiting enzyme of the polyamine synthetic pathway was evaluated in a Phase I trial. Intravenous DFMO was given to twenty patients with refractory leukemia by continuous infusion in doses from 5.5 to 64 g/m2. Toxicity clearly attributable to the drug was not severe and other than nausea and vomiting did not increase with dose. The previously reported ototoxicity which occurred with the oral form appeared to be less frequent. Loss of hearing which improved when the drug was stopped was seen in four patients, three of whom were simultaneously receiving aminoglycosides. Anorexia occurred in some patients at all doses. Vomiting, necessitating dosage reduction, was a significant problem at the highest dose administered. No patient achieved a remission but there was stabilization or decrease in circulating blast cells in several patients. This growth inhibition did not appear to be dosage related.
Collapse
|
41
|
|
42
|
Pettengill OS, Wurster-Hill DH, Cate CC, Sorenson GD. Morphological growth characteristics and hormone secretion of small cell carcinoma of the lung in vitro. Recent Results Cancer Res 1985; 99:130-42. [PMID: 2999913 DOI: 10.1007/978-3-642-82533-0_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
43
|
Oredsson SM, Linden M, Heby O. Increased rate of tumor cell death caused by polyamine synthesis inhibitors. VIRCHOWS ARCHIV. B, CELL PATHOLOGY INCLUDING MOLECULAR PATHOLOGY 1984; 47:131-8. [PMID: 6151293 DOI: 10.1007/bf02890195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
This investigation was designed to determine whether cell death plays a role in the antiproliferative action exerted by polyamine synthesis inhibitors. To estimate the rate of tumor cell death, we measured the loss of 125I from mice harboring Ehrlich ascites tumor cells in which DNA was labeled with 5-125I-iodo-2'-deoxyuridine. DL-alpha-difluoromethylornithine (0.85 mumoles/g body weight/6 h), and enzyme-activated irreversible inhibitor of ornithine decarboxylase, and methylglyoxal-bis(guanylhydrazone) (45 nmoles/g body weight/6 h), an inhibitor of S-adenosylmethionine decarboxylase, were both found to increase the rate of 125I excretion. Our data suggest that these polyamine synthesis inhibitors provoke an increase in the rate of tumor cell death beyond that normally occurring during growth, methylglyoxal-bis(guanylhydrazone) being considerably more potent than DL-alpha-difluoromethylornithine. These in vivo data were corroborated by a study where the host-mediated responses did not have to be considered. Thus, Ehrlich ascites tumor cells were adapted for suspension growth in culture and treated with methylglyoxal-bis(guanylhydrazone) or DL-alpha-difluoromethylornithine. The growth kinetics and the colony forming efficiency of the drug-treated cells clearly show that polyamine synthesis inhibitors not only slow the growth rate but also cause an increase in tumor cell death.
Collapse
|
44
|
Pegg AE. The role of polyamine depletion and accumulation of decarboxylated S-adenosylmethionine in the inhibition of growth of SV-3T3 cells treated with alpha-difluoromethylornithine. Biochem J 1984; 224:29-38. [PMID: 6439194 PMCID: PMC1144394 DOI: 10.1042/bj2240029] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The effects of alpha-difluoromethylornithine, a specific inhibitor of ornithine decarboxylase, on cell growth rate, polyamine content and the content of decarboxylated S-adenosylmethionine in SV-3T3 transformed mouse fibroblasts were studied. DL-alpha-Difluoromethylornithine at 1 mM or higher concentrations decreased the growth rate by over 90% after 2 or more days of exposure, but the cells remained viable, although quiescent for at least 9 days. Addition of 10 microM-spermidine or -spermine or 50 microM-putrescine at any time throughout this period completely reversed the inhibition of growth. Treatment with alpha-difluoromethylornithine decreased putrescine and spermidine contents by more than 98% and that of spermine by 60%, but cells exposed to exogenous polyamines did not require complete replenishment of the polyamine pools to resume growth. In fact, a virtually normal growth rate was obtained in cells lacking putrescine, having 2% of normal spermidine content and 156% of normal spermine. These results suggest that the well-known increase in putrescine and spermidine in cells stimulated for growth is not essential for this to occur and that mammalian cells can utilize spermine as their only polyamine. A substantial reversal of the growth-inhibitory effect of alpha-difluoromethylornithine was produced by a number of polyamines not normally found in mammalian cells, including the spermidine analogues aminopropylcadaverine and sym-homospermidine, which were partially converted into their respective spermine analogues by addition of an aminopropyl group within the cell. The spermine analogue sym-norspermine was also effective, but the maximal growth rate produced by these unphysiological polyamines was only 60-70% of that produced by the normal polyamines. These results indicate that spermidine and spermine have the optimal length for activation of the cellular processes critically dependent on polyamines and should help in identifying these processes. Exposure to alpha-difluoromethylornithine leads to an enormous rise in the concentration of decarboxylated S-adenosylmethionine, which reached a peak at 530-fold after 3 days of exposure and steadily declined to 140-fold after 11 days. This increase was abolished by addition of exogenous polyamines, which rapidly decreased the activity of S-adenosylmethionine decarboxylase. The increase in decarboxylated S-adenosylmethionine is unlikely to be solely responsible for the decrease to the same extent by spermine, sym-norspermidine and sym-homospermidine, which produce 97%, 16% and 60% of the control growth rate, respectively.(ABSTRACT TRUNCATED AT 400 WORDS)
Collapse
|
45
|
Levin VA, Byrd D, Campbell J, Davis RL, Borcich JK. CNS toxicity and CSF pharmacokinetics of intraventricular DFMO and MGBG in beagle dogs. Cancer Chemother Pharmacol 1984; 13:200-5. [PMID: 6435895 DOI: 10.1007/bf00269029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We have developed a beagle dog model to study the pharmacology and toxicology of anticancer drugs administered through the 3rd or lateral ventricles. A Foltz-type reservoir was implanted SC and connected by tube into a cerebral ventricle. Drugs were administered directly into the reservoir; CSF sampling of drugs administered into the ventricle was achieved directly by tapping the reservoir or by percutaneous puncture of the cisterna magna. In the current study, we evaluated the CSF pharmacokinetics and CNS toxicity of two inhibitors of polyamine metabolism, alpha-difluoromethylornitine (DFMO) and methylglyoxal bisguanylhydrazone (MGBG). Both drugs were judged too toxic to justify intrathecal or intraventricular studies with these agents in patients.
Collapse
|
46
|
Abstract
The data reviewed here demonstrate that there are many similarities in growth and cellular characteristics for different types of tumor cells grown as multicell spheroids. Furthermore, where comparisons have been made many of the features of spheroids also occur in tumors in vivo. However, as for tumors, there are also many characteristics of individual types of spheroids which are relatively specific and cannot be generalized as properties of all spheroid model systems. The results also demonstrate the marked influence which cellular microenvironments regulated by a supply of oxygen and nutrients may have on the development of cellular heterogeneity. Furthermore, using spheroids it was shown that dynamic cellular and metabolic interactions exist in regulating the development of cellular subpopulations and microenvironments. Spheroids are more sensitive to alterations in culture environment than are monolayer or single-cell suspension cultures. Consequently, researchers who use this model system must characterize, optimize, and standardize the growth conditions for the spheroid cell type being investigated. This information then provides a base from which to undertake detailed studies, which are not possible in experimental tumors, of controlled manipulation of microenvironments in spheroids. The ranges of cellular microenvironments and cellular heterogeneity which exist at different stages of spheroid growth provide a model, at least in part, for coexisting size ranges of microregions in many solid tumors. Thus, spheroids provide a model, which at different stages of growth is readily manipulated and controlled experimentally, to facilitate studies of contributions of individual environmental factors, or concomitant changes in these, on cellular phenotypic expression. It is probable that the cellular changes which can be demonstrated to occur during spheroid growth, also occur in vivo. Modulation of cellular characteristics revealed by research with spheroids requires much more study to determine the mechanisms and effects on tumor cell behavior, as well as response to therapeutic agents and their relevance to tumors in vivo.
Collapse
|
47
|
McGarrity GJ, Vanaman V, Sarama J. Cytogenetic effects of mycoplasmal infection of cell cultures: a review. IN VITRO 1984; 20:1-18. [PMID: 6199287 DOI: 10.1007/bf02633326] [Citation(s) in RCA: 57] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
48
|
Warrell RP, Coonley CJ, Burchenal JH. Sequential inhibition of polyamine synthesis. A phase I trial of DFMO (alpha-difluoromethylornithine) and methyl-GAG [methylglyoxal-bis(guanylhydrazone)]. Cancer Chemother Pharmacol 1983; 11:134-6. [PMID: 6414731 DOI: 10.1007/bf00254263] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Both DFMO and methyl-GAG inhibit sequential enzymatic reactions in the pathway of polyamine biosynthesis. Since polyamines may be important factors in proliferation of cancer cells, we initiated a phase-I study of these agents in patients with advanced cancer. DFMO was given by mouth at a constant daily dose of 4 g/m2 starting on day 1 of the treatment protocol. The dose of methyl-GAG ranged from 200 to 700 mg/m2 administered IV every 2 weeks beginning on day 4. Twenty-two patients were entered into the protocol. Toxic reactions to this therapy were dose-related and included nausea, fatigue, diarrhea, and myelosuppression. One patient with colon cancer experienced a greater than 50% decrease in measurable disease but developed severe myelotoxicity. While DFMO was well tolerated, the combination of drugs appeared to cause substantially more hematologic and gastrointestinal toxicity than encountered during our recent experience with methyl-GAG used alone. We suggest that future studies of this drug combination carefully evaluate levels of polyamines and inhibition of enzymatic activity to minimize toxicity.
Collapse
|
49
|
Luk GD, Sharkis SJ, Abeloff MD, McCann PP, Sjoerdsma A, Baylin SB. Polyamine biosynthesis is required for the maintenance of peripheral blood cell elements in the rat. Proc Natl Acad Sci U S A 1983; 80:5090-3. [PMID: 6410400 PMCID: PMC384194 DOI: 10.1073/pnas.80.16.5090] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The specific ornithine decarboxylase inhibitor alpha-difluoromethylornithine, when given to adult rats in vivo for 5 wk, resulted in a decrease in peripheral blood cell elements in normal rats and a marked suppression of marrow recovery in rats with chemotherapy-induced marrow hypoplasia. In normal rats, alpha-difluoromethylornithine resulted in a reduction of the leukocyte count to 73% of control, erythrocyte count to 61% of control, and platelet count to 24% of control. The bleeding time was increased to twice normal and 67% of the animals had epistaxis and 42% had melena. In rats treated with the S phase-specific chemotherapeutic agent 1-beta-D-arabinofuranosylcytosine, the simultaneous administration of alpha-difluoromethylornithine prevented the recovery of the bone marrow. The peripheral blood cell counts remained low--leukocyte count was 10% of control, and erythrocyte and platelet counts were 6% of control. All the animals developed epistaxis and melena and there was a 72% mortality. The administration of putrescine (4 mmol/kg, intraperitoneally, daily), the specific polyamine product of ornithine decarboxylase, reversed these hematologic effects in both normal and recovering marrow and resulted in rapid clinical improvement. Thus, the maintenance of normal, adult rat hematologic parameters, as with the proliferation of neoplastic and transformed cells in culture, is critically dependent on continued polyamine biosynthesis.
Collapse
|
50
|
McConlogue LC, Marton LJ, Coffino P. Growth regulatory effects of cyclic AMP and polyamine depletion are dissociable in cultured mouse lymphoma cells. J Cell Biol 1983; 96:762-7. [PMID: 6300139 PMCID: PMC2112398 DOI: 10.1083/jcb.96.3.762] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Treatment of mouse lymphoma S49 cells with D,L-alpha-difluoromethylornithine (DFMO), an inhibitor of ornithine decarboxylase, depleted cellular polyamine levels and stopped cell growth. The cells were arrested predominantly in G1. Thus, polyamine depletion may lead to a regulatory growth arrest in S49 cells. We tested two hypotheses regarding the relationship of growth arrest mediated by polyamine limitation to that mediated by cyclic AMP (cAMP). The hypothesis that cAMP-induced arrest results from polyamine depletion is not tenable, because the arrest could not be reversed by addition of exogenous polyamines, and because cellular polyamine levels do not drop in dibuturyl cyclic AMP (Bt2cAMP)-arrested cells. The hypothesis that polyamine-mediated growth arrest is effected via modulation of cAMP levels or cAMP-dependent protein kinase activity was also shown to be incorrect, because a S49 variant deficient in cAMP-dependent protein kinase was arrested by DFMO. The activities of the polyamine-synthesizing enzymes ornithine decarboxylase (ODC) and S-adenosyl methionine decarboxylase (SAMD) are both reduced in Bt2cAMP-treated cells to about 10% of that in control populations, as shown previously. DFMO diminishes ODC activity and augments SAMD activity in both untreated and Bt2cAMP-treated cells, leading to polyamine depletion in both cases.
Collapse
|