1
|
Verhaegen M, Bergot T, Liebana E, Stancanelli G, Streissl F, Mingeot-Leclercq MP, Mahillon J, Bragard C. On the use of antibiotics to control plant pathogenic bacteria: a genetic and genomic perspective. Front Microbiol 2023; 14:1221478. [PMID: 37440885 PMCID: PMC10333595 DOI: 10.3389/fmicb.2023.1221478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Despite growing attention, antibiotics (such as streptomycin, oxytetracycline or kasugamycin) are still used worldwide for the control of major bacterial plant diseases. This raises concerns on their potential, yet unknown impact on antibiotic and multidrug resistances and the spread of their genetic determinants among bacterial pathogens. Antibiotic resistance genes (ARGs) have been identified in plant pathogenic bacteria (PPB), with streptomycin resistance genes being the most commonly reported. Therefore, the contribution of mobile genetic elements (MGEs) to their spread among PPB, as well as their ability to transfer to other bacteria, need to be further explored. The only well-documented example of ARGs vector in PPB, Tn5393 and its highly similar variants (carrying streptomycin resistance genes), is concerning because of its presence outside PPB, in Salmonella enterica and Klebsiella pneumoniae, two major human pathogens. Although its structure among PPB is still relatively simple, in human- and animal-associated bacteria, Tn5393 has evolved into complex associations with other MGEs and ARGs. This review sheds light on ARGs and MGEs associated with PPB, but also investigates the potential role of antibiotic use in resistance selection in plant-associated bacteria.
Collapse
Affiliation(s)
- Marie Verhaegen
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, Catholic University of Louvain (UCLouvain), Louvain-la-Neuve, Belgium
| | - Thomas Bergot
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, Catholic University of Louvain (UCLouvain), Louvain-la-Neuve, Belgium
| | | | | | | | - Marie-Paule Mingeot-Leclercq
- Cellular and Molecular Pharmacology Unit, Louvain Drug Research Institute, UCLouvain, Woluwe-Saint-Lambert, Belgium
| | - Jacques Mahillon
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, Catholic University of Louvain (UCLouvain), Louvain-la-Neuve, Belgium
| | - Claude Bragard
- Plant Health Laboratory, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, Belgium
| |
Collapse
|
2
|
Yao H, Liu J, Zhang C, Shao Y, Li X, Feng M, Wang X, Gan W, Zhou Y, Huang Y. Clinical study of apatinib plus temozolomide for the treatment of recurrent high-grade gliomas. J Clin Neurosci 2021; 90:82-88. [PMID: 34275586 DOI: 10.1016/j.jocn.2021.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/30/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Recurrent high-grade glioma, a malignant tumor of the brain or spinal cord associated with poor prognosis with a median survival of <6 months. Recurrent high-grade glioma does not have standard treatment even if some strategies have some effect in recurrent gliomas. Apatinib, as a tyrosine kinase inhibitor shown to be effective in treating the lung and gastric cancer. The present study investigated the efficacy and safety of apatinib in combination with dose-dense regimens of temozolomide for treating recurrent glioma. PATIENTS AND METHODS Eighteen patients with recurrent high-grade glioma were enrolled and treated with apatinib (500 mg/day) and TMZ (50 mg/m2/day). Patients who achieved partial response or stable disease continued treatment. Administration of drug was terminated for patients with progressive disease, who could not tolerate toxicity, and who required discontinuation due to other medical conditions. RESULTS From the 18 cases, only 17 were included in the evaluation of the curative effect of the drug and in that four showed partial responses, ten had stable disease, remaining three exhibited progressive disease. The disease control rate was 82.3% (14/17). Progression-free and overall survival was found to be 4 months and 9.1 months, respectively. Three patients became transiently capable of self-care (Karnofsky performance status >70). Cognition and quality of life improved after treatment and from the safety perspective, three most common adverse reactions included epilepsy (24.1%), hypertension (20.7%), and fatigue (17.2%). CONCLUSION Apatinib and TMZ may represent an alternative treatment option for patients with recurrent high-gradeglioma, especially those with a low Karnofsky performance status. However, studies using a larger sample size are required to confirm these findings.
Collapse
Affiliation(s)
- Hui Yao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Jiangang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Chi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Yunxiang Shao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Xuetao Li
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Ming Feng
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Ximing Wang
- Department of Radiologly, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Wenjuan Gan
- Department of Pathology, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China
| | - Youxin Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China.
| | - Yulun Huang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, No. 188 Shizi Street, Suzhou 215007, Jiangsu, China.
| |
Collapse
|
3
|
Premkumar DR, Jane EP, Thambireddy S, Sutera PA, Cavaleri JM, Pollack IF. Mitochondrial dysfunction RAD51, and Ku80 proteolysis promote apoptotic effects of Dinaciclib in Bcl-xL silenced cells. Mol Carcinog 2017; 57:469-482. [PMID: 29240261 DOI: 10.1002/mc.22771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 10/17/2017] [Accepted: 12/07/2017] [Indexed: 01/08/2023]
Abstract
In the present study, we investigated the effect of CDK inhibitors (ribociclib, palbociclib, seliciclib, AZD5438, and dinaciclib) on malignant human glioma cells for cell viability, apoptosis, oxidative stress, and mitochondrial function using various assays. None of the CDK inhibitors induced cell death at a clinically relevant concentration. However, low nanomolar concentrations of dinaciclib showed higher cytotoxic activity against Bcl-xL silenced cells in a time- and concentration-dependent manner. This effect was not seen with other CDK inhibitors. The apoptosis-inducing capability of dinaciclib in Bcl-xL silenced cells was evidenced by cell shrinkage, mitochondrial dysfunction, DNA damage, and increased phosphatidylserine externalization. Dinaciclib was found to disrupt mitochondrial membrane potential, resulting in the release of cytochrome c, AIF, and smac/DIABLO into the cytoplasm. This was accompanied by the downregulation of cyclin-D1, D3, and total Rb. Dinaciclib caused cell cycle arrest in a time- and concentration-dependent manner and with accumulation of cells in the sub-G1 phase. Our results also revealed that dinaciclib, but not ribociclib or palbociclib or seliciclib or AZD5438 induced intrinsic apoptosis via upregulation of the levels of pro-apoptotic proteins (Bax and Bak), resulting in the activation of caspases and cleavage of PARP. We also found an additional mechanism for the dinaciclib-induced augmentation of apoptosis due to abrogation RAD51-cyclin D1 interaction, specifically proteolysis of the DNA repair proteins RAD51 and Ku80. Our results suggest that successfully interfering with Bcl-xL function may restore sensitivity to dinaciclib and could hold the promise for an effective combination therapeutic strategy.
Collapse
Affiliation(s)
- Daniel R Premkumar
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania
| | - Esther P Jane
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Swetha Thambireddy
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Philip A Sutera
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jonathon M Cavaleri
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ian F Pollack
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,University of Pittsburgh Cancer Institute Brain Tumor Center, Pittsburgh, Pennsylvania
| |
Collapse
|
4
|
Plaisier CL, O'Brien S, Bernard B, Reynolds S, Simon Z, Toledo CM, Ding Y, Reiss DJ, Paddison PJ, Baliga NS. Causal Mechanistic Regulatory Network for Glioblastoma Deciphered Using Systems Genetics Network Analysis. Cell Syst 2016; 3:172-186. [PMID: 27426982 DOI: 10.1016/j.cels.2016.06.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/20/2016] [Accepted: 06/09/2016] [Indexed: 12/30/2022]
Abstract
We developed the transcription factor (TF)-target gene database and the Systems Genetics Network Analysis (SYGNAL) pipeline to decipher transcriptional regulatory networks from multi-omic and clinical patient data, and we applied these tools to 422 patients with glioblastoma multiforme (GBM). The resulting gbmSYGNAL network predicted 112 somatically mutated genes or pathways that act through 74 TFs and 37 microRNAs (miRNAs) (67 not previously associated with GBM) to dysregulate 237 distinct co-regulated gene modules associated with patient survival or oncogenic processes. The regulatory predictions were associated to cancer phenotypes using CRISPR-Cas9 and small RNA perturbation studies and also demonstrated GBM specificity. Two pairwise combinations (ETV6-NFKB1 and romidepsin-miR-486-3p) predicted by the gbmSYGNAL network had synergistic anti-proliferative effects. Finally, the network revealed that mutations in NF1 and PIK3CA modulate IRF1-mediated regulation of MHC class I antigen processing and presentation genes to increase tumor lymphocyte infiltration and worsen prognosis. Importantly, SYGNAL is widely applicable for integrating genomic and transcriptomic measurements from other human cohorts.
Collapse
Affiliation(s)
| | - Sofie O'Brien
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA
| | - Brady Bernard
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA
| | - Sheila Reynolds
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA
| | - Zac Simon
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA
| | - Chad M Toledo
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Yu Ding
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - David J Reiss
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Nitin S Baliga
- Institute for Systems Biology, 401 Terry Avenue North, Seattle, WA 98109-5234, USA.
| |
Collapse
|
5
|
Jane EP, Premkumar DR, Cavaleri JM, Sutera PA, Rajasekar T, Pollack IF. Dinaciclib, a Cyclin-Dependent Kinase Inhibitor Promotes Proteasomal Degradation of Mcl-1 and Enhances ABT-737-Mediated Cell Death in Malignant Human Glioma Cell Lines. J Pharmacol Exp Ther 2015; 356:354-65. [PMID: 26585571 DOI: 10.1124/jpet.115.230052] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/17/2014] [Indexed: 02/04/2023] Open
Abstract
The prognosis for malignant glioma, the most common brain tumor, is still poor, underscoring the need to develop novel treatment strategies. Because glioma cells commonly exhibit genomic alterations involving genes that regulate cell-cycle control, there is a strong rationale for examining the potential efficacy of strategies to counteract this process. In this study, we examined the antiproliferative effects of the cyclin-dependent kinase inhibitor dinaciclib in malignant human glioma cell lines, with intact, deleted, or mutated p53 or phosphatase and tensin homolog on chromosome 10; intact or deleted or p14ARF or wild-type or amplified epidermal growth factor receptor. Dinaciclib inhibited cell proliferation and induced cell-cycle arrest at the G2/M checkpoint, independent of p53 mutational status. In a standard 72-hour 3-[4,5-dimethylthiazol- 2yl]-5-[3-carboxymethoxyphenyl]-2-[4-sulfophenyl]-2H, tetrazolium (MTS) assay, at clinically relevant concentrations, dose-dependent antiproliferative effects were observed, but cell death was not induced. Moreover, the combination of conventional chemotherapeutic agents and various growth-signaling inhibitors with dinaciclib did not yield synergistic cytotoxicity. In contrast, combination of the Bcl-2/Bcl-xL inhibitors ABT-263 (4-[4-[[2-(4-chlorophenyl)-5,5-dimethylcyclohexen-1-yl]methyl]piperazin-1-yl]-N-[4-[[(2R)-4-morpholin-4-yl-1-phenylsulfanylbutan-2-yl]amino]-3-(trifluoromethylsulfonyl)phenyl]sulfonylbenzamide) or ABT-737 (4-[4-[[2-(4-chlorophenyl)phenyl]methyl]piperazin-1-yl]-N-[4-[[(2R)-4-(dimethylamino)-1-phenylsulfanylbutan-2-yl]amino]-3-nitrophenyl]sulfonylbenzamide) with dinaciclib potentiated the apoptotic response induced by each single drug. The synergistic killing by ABT-737 with dinaciclib led to cell death accompanied by the hallmarks of apoptosis, including an early loss of the mitochondrial transmembrane potential; the release of cytochrome c, smac/DIABLO, and apoptosis-inducing factor; phosphatidylserine exposure on the plasma membrane surface and activation of caspases and poly ADP-ribose polymerase. Mechanistic studies revealed that dinaciclib promoted proteasomal degradation of Mcl-1. These observations may have important clinical implications for the design of experimental treatment protocols for malignant human glioma.
Collapse
Affiliation(s)
- Esther P Jane
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh (E.P.J., D.R.P., I.F.P.), University of Pittsburgh, School of Medicine (E.P.J., D.R.P., J.M.C., P.A.S., T.R., I.F.P), and University of Pittsburgh Brain Tumor Center (D.R.P., I.F.P.), Pittsburgh, Pennsylvania
| | - Daniel R Premkumar
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh (E.P.J., D.R.P., I.F.P.), University of Pittsburgh, School of Medicine (E.P.J., D.R.P., J.M.C., P.A.S., T.R., I.F.P), and University of Pittsburgh Brain Tumor Center (D.R.P., I.F.P.), Pittsburgh, Pennsylvania
| | - Jonathon M Cavaleri
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh (E.P.J., D.R.P., I.F.P.), University of Pittsburgh, School of Medicine (E.P.J., D.R.P., J.M.C., P.A.S., T.R., I.F.P), and University of Pittsburgh Brain Tumor Center (D.R.P., I.F.P.), Pittsburgh, Pennsylvania
| | - Philip A Sutera
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh (E.P.J., D.R.P., I.F.P.), University of Pittsburgh, School of Medicine (E.P.J., D.R.P., J.M.C., P.A.S., T.R., I.F.P), and University of Pittsburgh Brain Tumor Center (D.R.P., I.F.P.), Pittsburgh, Pennsylvania
| | - Thatchana Rajasekar
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh (E.P.J., D.R.P., I.F.P.), University of Pittsburgh, School of Medicine (E.P.J., D.R.P., J.M.C., P.A.S., T.R., I.F.P), and University of Pittsburgh Brain Tumor Center (D.R.P., I.F.P.), Pittsburgh, Pennsylvania
| | - Ian F Pollack
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh (E.P.J., D.R.P., I.F.P.), University of Pittsburgh, School of Medicine (E.P.J., D.R.P., J.M.C., P.A.S., T.R., I.F.P), and University of Pittsburgh Brain Tumor Center (D.R.P., I.F.P.), Pittsburgh, Pennsylvania
| |
Collapse
|
6
|
de Araujo CB, Russo LC, Castro LM, Forti FL, do Monte ER, Rioli V, Gozzo FC, Colquhoun A, Ferro ES. A novel intracellular peptide derived from g1/s cyclin d2 induces cell death. J Biol Chem 2014; 289:16711-26. [PMID: 24764300 DOI: 10.1074/jbc.m113.537118] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Intracellular peptides are constantly produced by the ubiquitin-proteasome system, and many are probably functional. Here, the peptide WELVVLGKL (pep5) from G1/S-specific cyclin D2 showed a 2-fold increase during the S phase of HeLa cell cycle. pep5 (25-100 μm) induced cell death in several tumor cells only when it was fused to a cell-penetrating peptide (pep5-cpp), suggesting its intracellular function. In vivo, pep5-cpp reduced the volume of the rat C6 glioblastoma by almost 50%. The tryptophan at the N terminus of pep5 is essential for its cell death activity, and N terminus acetylation reduced the potency of pep5-cpp. WELVVL is the minimal active sequence of pep5, whereas Leu-Ala substitutions totally abolished pep5 cell death activity. Findings from the initial characterization of the cell death/signaling mechanism of pep5 include caspase 3/7 and 9 activation, inhibition of Akt2 phosphorylation, activation of p38α and -γ, and inhibition of proteasome activity. Further pharmacological analyses suggest that pep5 can trigger cell death by distinctive pathways, which can be blocked by IM-54 or a combination of necrostatin-1 and q-VD-OPh. These data further support the biological and pharmacological potential of intracellular peptides.
Collapse
Affiliation(s)
| | - Lilian C Russo
- the Department of Biochemistry, Support Center for Research in Proteolysis and Cell Signaling (NAPPS), Institute of Chemistry, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | | | - Fábio L Forti
- the Department of Biochemistry, Support Center for Research in Proteolysis and Cell Signaling (NAPPS), Institute of Chemistry, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | | | - Vanessa Rioli
- the Special Laboratory of Applied Toxinology (LETA), Center of Toxins, Immune Response, and Cell Signaling (CETICS), Butantan Institute, 05503-000, São Paulo, SP, Brazil, and
| | - Fabio C Gozzo
- the Institute of Chemistry, State University of Campinas, 13083-862, Campinas, SP, Brazil
| | - Alison Colquhoun
- Cell Biology and Development, Support Center for Research in Proteolysis and Cell Signaling (NAPPS), Biomedical Science Institute, University of São Paulo, São Paulo, 05508-000, SP, Brazil
| | | |
Collapse
|
7
|
Abstract
Although advances in surgery, radiation therapy and stereotactic radiosurgery have significantly improved the treatment of meningiomas, there remains an important subset of patients who remain refractory to conventional therapy. Treatment with chemotherapeutic agents such as hydroxyurea and alpha-interferon has provided minimal benefit. In this review, the role of newly emerging novel therapies for meningiomas, with a focus on targeted molecular agents, will be discussed.
Collapse
Affiliation(s)
- Patrick Y Wen
- Center for Neuro-Oncology Dana-Farber/Brigham and Women's Cancer Center, and Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | |
Collapse
|
8
|
Penas-Prado M, Gilbert MR. Molecularly targeted therapies for malignant gliomas: advances and challenges. Expert Rev Anticancer Ther 2014; 7:641-61. [PMID: 17492929 DOI: 10.1586/14737140.7.5.641] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The identification of molecular markers associated with tumor but not with normal tissue has allowed the development of highly specific, targeted therapies for the treatment of cancer. Over the last several years, tremendous advances in our understanding of the genetic and molecular changes involved in the progression of malignant gliomas have triggered a large effort in the development of targeted therapies to treat these tumors. However, to date only a modest clinical benefit, limited to subsets of patients, has been demonstrated. Furthermore, despite a high degree of target selectivity, the use of targeted therapies often has systemic toxicity. The reasons behind this limited clinical success are complex and include the intricacy of the signaling pathways in gliomas and the heterogeneity of the disease process, compounded by existing limitations in assessing the efficacy of these novel agents when conventional end points and clinical trial designs are utilized. However, despite these difficulties targeted therapies remain a very attractive avenue of treatment for malignant gliomas. Three basic approaches are needed to overcome the hurdles associated with targeted therapies: first, further development of genetic profiling techniques will help to better determine the genetic changes and molecular pathways involved in gliomas and will potentially allow the design of individualized therapies based on the genetic and molecular signature of each tumor. Second, there is a need for the development of better combination strategies (complementary targeted agents or targeted agents with chemotherapy drugs) directed towards disease heterogeneity. Third, we need to optimize the design of preclinical and clinical trials to obtain the maximum amount of information in the shortest period of time.
Collapse
Affiliation(s)
- Marta Penas-Prado
- The UT MD Anderson Cancer Center, Department of Neuro-Oncology, Houston, 77030 TX, USA.
| | | |
Collapse
|
9
|
Munsch-Alatossava P, Gauchi JP, Chamlagain B, Alatossava T. Trends of Antibiotic Resistance in Mesophilic and Psychrotrophic Bacterial Populations during Cold Storage of Raw Milk. ISRN MICROBIOLOGY 2012; 2012:918208. [PMID: 23724333 PMCID: PMC3658802 DOI: 10.5402/2012/918208] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 12/25/2011] [Indexed: 11/23/2022]
Abstract
Psychrotrophic bacteria in raw milk are most well known for their spoilage potential and cause significant economic losses in the dairy industry. Despite their ability to produce several exoenzyme types at low temperatures, psychrotrophs that dominate the microflora at the time of spoilage are generally considered benign bacteria. It was recently reported that raw milk-spoiling Gram-negative-psychrotrophs frequently carried antibiotic resistance (AR) features. The present study evaluated AR to four antibiotics (ABs) (gentamicin, ceftazidime, levofloxacin, and trimethoprim-sulfamethoxazole) in mesophilic and psychrotrophic bacterial populations recovered from 18 raw milk samples, after four days storage at 4°C or 6°C. Robust analysis of variance and non parametric statistics (e.g., REGW and NPS) revealed that AR prevalence among psychrotrophs, for milk samples stored at 4°C, often equalled the initial levels and equalled or increased during the cold storage at 6°C, depending on the AB. The study performed at 4°C with an intermediate sampling point at day 2 suggested that (1) different psychrotrophic communities with varying AR levels dominate over time and (2) that AR (determined from relative amounts) was most prevalent, transiently, after 2-day storage in psychrotrophic or mesophilic populations, most importantly at a stage where total counts were below or around 10(5) CFU/mL, at levels at which the milk is acceptable for industrial dairy industrial processes.
Collapse
Affiliation(s)
- Patricia Munsch-Alatossava
- Division of Food Technology, Department of Food and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
| | - Jean-Pierre Gauchi
- Unité de Mathématiques et Informatique Appliquées (UR 341), Centre de Jouy en Josas, Institut National de la Recherche Agronomique, Domaine de Vilvert, 78352 Jouy en Josas, France
| | - Bhawani Chamlagain
- Division of Food Technology, Department of Food and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
| | - Tapani Alatossava
- Division of Food Technology, Department of Food and Environmental Sciences, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
10
|
Abstract
Gliobastoma multiform (GBM) is the most common and aggressive brain tumor, which is characterized by its infiltrative nature. Current standard therapy for GBMs consists of surgery followed by radiotherapy combined with the alkylating agent temozolomide (TMZ). Recent clinical trials have demonstrated that this chemo-irradiation approach results in a significant increase in survival compared to radiotherapy alone. Nevertheless, due to tumor recurrence, the median survival time is still limited to approximately 15 months. Recently, several studies have focused on aberrant signal transduction in GBM, resistance mechanisms of GBM to TMZ and to radiotherapy. Attention has been focused on molecular targets including phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway, protein kinase C (pKC) pathway, Ras/mitogen-activated protein kinase pathway (MAPK), Wnt pathway and intrinsic or extrinsic apoptosis pathways. In addition, research has been directed to radiotherapy and radiosensitizing agents, and cancer gene therapy as well. This article will address several resistance mechanisms of GBM to chemotherapy and radiotherapy and the recent preclinical and clinical studies on targeted therapy.
Collapse
Affiliation(s)
- N H Rekers
- Department of Medical Oncology, VU University Medical Center, The Netherlands
| | | | | |
Collapse
|
11
|
Affiliation(s)
- Marta Penas-Prado
- Department of Neuro-oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
12
|
General and neurological complications of targeted therapy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 105:937-45. [PMID: 22230543 DOI: 10.1016/b978-0-444-53502-3.00033-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
13
|
Sathornsumetee S, Rich JN. Molecularly targeted therapy in neuro-oncology. HANDBOOK OF CLINICAL NEUROLOGY 2012; 104:255-78. [PMID: 22230448 DOI: 10.1016/b978-0-444-52138-5.00018-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
14
|
Affiliation(s)
- Andrew S Chi
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
15
|
Alexandru O, Dragutescu L, Tataranu L, Ciubotaru V, Sevastre A, Georgescu AM, Purcaru O, Danoiu S, Bäcklund LM, Dricu A. Helianthin induces antiproliferative effect on human glioblastoma cells in vitro. J Neurooncol 2010; 102:9-18. [PMID: 20635119 DOI: 10.1007/s11060-010-0285-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Accepted: 06/21/2010] [Indexed: 02/07/2023]
Abstract
A major focus of brain cancer research today is to translate understanding of glioma biology into advances in treatment, by exploring the potential of target therapy. Here we investigated the ability of three compounds belonging to the chemical class of azo dyes (methyl red, methyl yellow, and helianthin) to inhibit glioblastoma (GB) cell growth in vitro. Our results showed that helianthin induced cytotoxicity in two GB cell cultures, cell lines 18 and 38, whereas methyl red and methyl yellow were not cytotoxic. The effect of helianthin on EGFR, IGF-1R, and their common intracellular signaling via PI3-K and ERK1/2 was also analyzed. Treatment with helianthin down-regulated EGFR and IGF-1R activity in both cell lines. Helianthin treatment blocked ERK1/2 phosphorylation without affecting PI3K activity in cell line 18 and reduced both PI3K and ERK1/2 in GB 38 cell line. The cell death was accompanied by degradation of PARP without affecting BCL2 expression in both GB cell cultures. Because of the genetic heterogeneity of malignant gliomas, we tested the effect of helianthin on other two primary GB lines (11 and 15) and two early-passage GB cultures (BT1GB and BT2GB), to assess the general nature of the anti-tumor effect of the drug in GB cells. We found that helianthin treatment induced cell death in all the GB cell cultures analyzed. To our knowledge, this is the first report indicating that helianthin can reduce GB cell growth.
Collapse
Affiliation(s)
- Oana Alexandru
- University of Medicine and Pharmacy, Petru Rares Street, No 2, 200349 Craiova, Romania
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ribeiro G, Benadiba M, de Oliveira Silva D, Colquhoun A. The novel rutheniumâγ-linolenic complex [Ru2(aGLA)4Cl] inhibits C6 rat glioma cell proliferation and induces changes in mitochondrial membrane potential, increased reactive oxygen species generation and apoptosisin vitro. Cell Biochem Funct 2010; 28:15-23. [DOI: 10.1002/cbf.1626] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
17
|
Management of newly diagnosed glioblastoma: guidelines development, value and application. J Neurooncol 2009; 93:1-23. [DOI: 10.1007/s11060-009-9838-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 02/23/2009] [Indexed: 11/25/2022]
|
18
|
Goldhoff P, Warrington NM, Limbrick DD, Hope A, Woerner BM, Jackson E, Perry A, Piwnica-Worms D, Rubin JB. Targeted inhibition of cyclic AMP phosphodiesterase-4 promotes brain tumor regression. Clin Cancer Res 2009; 14:7717-25. [PMID: 19047098 DOI: 10.1158/1078-0432.ccr-08-0827] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
PURPOSE As favorable outcomes from malignant brain tumors remain limited by poor survival and treatment-related toxicity, novel approaches to cure are essential. Previously, we identified the cyclic AMP phosphodiesterase-4 (PDE4) inhibitor Rolipram as a potent antitumor agent. Here, we investigate the role of PDE4 in brain tumors and examine the utility of PDE4 as a therapeutic target. EXPERIMENTAL DESIGN Immunohistochemistry was used to evaluate the expression pattern of a subfamily of PDE4, PDE4A, in multiple brain tumor types. To evaluate the effect of PDE4A on growth, a brain-specific isoform, PDE4A1 was overexpressed in xenografts of Daoy medulloblastoma and U87 glioblastoma cells. To determine therapeutic potential of PDE4 inhibition, Rolipram, temozolomide, and radiation were tested alone and in combination on mice bearing intracranial U87 xenografts. RESULTS We found that PDE4A is expressed in medulloblastoma, glioblastoma, oligodendroglioma, ependymoma, and meningioma. Moreover, when PDE4A1 was overexpressed in Daoy medulloblastoma and U87 glioblastoma cells, in vivo doubling times were significantly shorter for PDE4A1-overexpressing xenografts compared with controls. In long-term survival and bioluminescence studies, Rolipram in combination with first-line therapy for malignant gliomas (temozolomide and conformal radiation therapy) enhanced the survival of mice bearing intracranial xenografts of U87 glioblastoma cells. Bioluminescence imaging indicated that whereas temozolomide and radiation therapy arrested intracranial tumor growth, the addition of Rolipram to this regimen resulted in tumor regression. CONCLUSIONS This study shows that PDE4 is widely expressed in brain tumors and promotes their growth and that inhibition with Rolipram overcomes tumor resistance and mediates tumor regression.
Collapse
Affiliation(s)
- Patricia Goldhoff
- Department of Pediatrics, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ren H, Tan X, Dong Y, Giese A, Chou TC, Rainov N, Yang B. Differential effect of imatinib and synergism of combination treatment with chemotherapeutic agents in malignant glioma cells. Basic Clin Pharmacol Toxicol 2009; 104:241-52. [PMID: 19159435 DOI: 10.1111/j.1742-7843.2008.00371.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Imatinib mesylate (STI571, Gleevec) is a signal transduction inhibitor and novel anti-cancer agent. It selectively inhibits aberrantly activated tyrosine kinases in malignant cells, for example, bcr-abl in leukaemia, platelet-derived growth factor receptor and stem cell factor receptor (c-Kit) in solid cancers including malignant glioma. However, recently published clinical studies with imatinib monotherapy in patients with malignant glioma demonstrated only very modest anti-tumour activity. The aim of this study was to investigate the biological activity of imatinib, its cellular mechanisms of action and its synergism with other chemotherapeutic agents in human malignant glioma cells in culture. Expression of PDGF/R and c-Kit was analyzed by RT-PCR. Proliferation was measured by MTT assays and drug synergy was assessed by the Chou-Talalay method. Cell cycle and apoptosis were analyzed by flow cytometry and migration by monolayer migration assays. Multi-immunoblot was performed on imatinib-treated and control malignant glioma cells. Results indicate that imatinib is more effective in inhibiting cell colony formation and migration rather than proliferation. Imatinib treatment caused cell cycle arrest of glioma cells in G0-G1 or G2/M, with significant elevation of a few cyclin-dependent kinases. Furthermore, imatinib acted synergistically with chemotherapy agents, such as the DNA alkylating agent, temozolomide, and riboneucleotide reductase inhibitors, for example, hydroxyurea at varied effective dose levels. In conclusion, imatinib exerts varied biological effects on malignant glioma cells in culture. Synergistic interaction of imatinib with chemotherapy agents may be related to cell cycle control mechanisms and could be potentially important in a clinical setting.
Collapse
Affiliation(s)
- Huan Ren
- Department of Immunology, Harbin Medical University, 150081 Harbin, China.
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Primary brain tumors account for less than 2% of all cancers in adults; however, they are often associated with neurologic morbidity and high mortality. Glioblastoma multiforme (GBM) has been a focus of new therapy development in neurooncology because it is the most common primary brain tumor in adults. Standard-of-care therapy for newly diagnosed GBM includes surgical resection, radiotherapy, and temozolomide, administered both during and after radiotherapy. However, most patients develop tumor recurrence or progression after this multimodality treatment. Repeat resection and stereotactic radiosurgery upon recurrence may improve outcome only in selected patients. Most salvage chemotherapies offer only palliation. Recent advances in our understanding of the molecular abnormalities of GBM have generated new therapeutic venues of molecularly targeted agents (designer drugs) against key components of cellular pathways critical for cancer initiation and maintenance. Such drugs may offer the potential advantage to increase therapeutic efficacy and decrease systemic toxicity compared with traditional cytotoxic agents. Nonetheless, first-generation targeted agents have failed to demonstrate survival benefits in unselected GBM patient populations. Several mechanisms of treatment failure of the first-generation designer drugs have been proposed, whereas new strategies have been developed to increase effectiveness of these agents. Here we will discuss the recent development and the strategies to optimize the effectiveness of designer therapy for GBM.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- Neuro-Oncology Program, Departments of Medicine (Neurology) and Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
21
|
Cytotoxic chemotherapeutic management of newly diagnosed glioblastoma multiforme. J Neurooncol 2008; 89:339-57. [PMID: 18712284 DOI: 10.1007/s11060-008-9615-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Accepted: 05/19/2008] [Indexed: 10/21/2022]
|
22
|
Abstract
BACKGROUND Malignant gliomas are amongst the most devastating and intractable of all cancers. The most common malignant glioma, glioblastoma multiforme (GBM), is associated with a median survival in the range of 12-15 months. Survival for patients with GBM has improved with the addition of temozolomide chemotherapy to post-operative radiotherapy. Further advances in the treatment of malignant glioma will hinge on the discovery of novel and likely targeted therapies with activity against these diseases. OBJECTIVE Review recent published experience using targeted therapeutics for malignant glioma. METHODS Key studies from a Medline review of targeted therapies for malignant glioma performed between 2000 and the present are summarised in this review. CONCLUSIONS Experience with targeted therapeutics for malignant glioma has been to date disappointing. These agents are generally well tolerated, but activity is limited. Novel therapeutics with activity against malignant gliomas must be identified to improve prognosis for patients with these diseases.
Collapse
Affiliation(s)
- Warren P Mason
- Princess Margaret Hospital, Department of Medicine, 610 University Avenue, Suite 18-717, Toronto, Ontario, M5G 2M9, Canada.
| |
Collapse
|
23
|
Chi A, Norden AD, Wen PY. Inhibition of angiogenesis and invasion in malignant gliomas. Expert Rev Anticancer Ther 2008; 7:1537-60. [PMID: 18020923 DOI: 10.1586/14737140.7.11.1537] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Malignant gliomas confer a dismal prognosis. As the molecular events that underlie tumor angiogenesis are elucidated, angiogenesis inhibition is emerging as a promising therapy for recurrent and newly diagnosed tumors. Data from animal studies suggest that angiogenesis inhibition may promote an invasive phenotype in tumor cells. This may represent an important mechanism of resistance to antiangiogenic therapies. Recent studies have begun to clarify the mechanisms by which glioma cells detach from the tumor mass, remodel the extracellular matrix and infiltrate normal brain. An array of potential therapeutic targets exists. Combination therapy with antiangiogenic and novel anti-invasion agents is a promising approach that may produce a synergistic antitumor effect and a survival benefit for patients with these devastating tumors.
Collapse
Affiliation(s)
- Andrew Chi
- Center for Neuro-Oncology, Dana-Farber/Brigham & Women's Cancer Center, Division of Neuro-Oncology, Department of Neurology, Brigham & Women's Hospital, SW430D, 44 Binney Street, Boston, MA 02115, USA.
| | | | | |
Collapse
|
24
|
Sathornsumetee S, Rich JN, Reardon DA. Diagnosis and treatment of high-grade astrocytoma. Neurol Clin 2008; 25:1111-39, x. [PMID: 17964028 DOI: 10.1016/j.ncl.2007.07.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High-grade astrocytomas include the most common adult central nervous system (CNS) tumor, glioblastoma multiforme, and anaplastic astrocytoma--a highly aggressive cancer with short median survival despite maximal multimodality therapy. Diagnosis is by clinical and radiographic findings confirmed by histopathology. Standard-of-care therapy includes surgical resection, radiotherapy, and temozolomide. Nearly all patients who have high-grade astrocytomas develop tumor recurrence or progression after this multimodality treatment. Two treatment challenges are molecular/genetic heterogeneity of tumors and limited CNS tumor delivery. It is probable that targeted therapies will be most effective in combination with one another or with cytotoxic therapies. This article discusses diagnosis and current treatment of high-grade astrocytomas.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- Division of Neurology, The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, DUMC 3624, Durham, NC 27710, USA
| | | | | |
Collapse
|
25
|
Abstract
✓ Although advances in surgery, radiation therapy, and stereotactic radiosurgery have significantly improved the treatment of meningiomas, there remains an important subset of patients whose tumors are refractory to conventional therapy. Treatment with traditional chemotherapeutic agents has provided minimal benefit. In this review, the role of targeted molecular therapies for recurrent or progressive meningiomas is discussed.
Collapse
|
26
|
Sathornsumetee S, Reardon DA, Desjardins A, Quinn JA, Vredenburgh JJ, Rich JN. Molecularly targeted therapy for malignant glioma. Cancer 2007; 110:13-24. [PMID: 17520692 DOI: 10.1002/cncr.22741] [Citation(s) in RCA: 222] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Malignant gliomas are relatively uncommon but lethal cancers. Despite recent research efforts in cancer therapy, the prognosis of patients with malignant gliomas has remained dismal. Understanding the molecular pathogenesis of glioma may lead to a rational development of new therapies. Despite the genetic heterogeneity of malignant gliomas, common aberrations in the signaling elements of the growth and survival pathways are found. New treatments have emerged to target molecules in these signaling pathways with the goal to increase specific efficacy and minimize toxicity. Monoclonal antibodies and low molecular-weight kinase inhibitors are the most common classes of agents in targeted cancer treatment. Most clinical trials of these agents as monotherapies have failed to demonstrate survival benefit in unselected malignant glioma patient populations. Several mechanisms of treatment failure have been demonstrated. In response, multitargeted kinase inhibitors and combinations of single-targeted kinase inhibitors have been developed to overcome therapeutic resistance. In addition, multimodality combinations of targeted agents with radiation, chemotherapy, or immunotherapy/vaccines may enhance treatment efficacy. Future development of these agents will require advances in discovery and validation of new molecular targets, improvement of therapeutic delivery, and identification of correlative biomarkers. Novel clinical trial designs and endpoints may increase the efficiency of new drug evaluation. In this review, the authors discussed the current understanding of molecular pathogenesis and the development of molecularly targeted therapies in malignant glioma.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | |
Collapse
|
27
|
Carapancea M, Cosaceanu D, Budiu R, Kwiecinska A, Tataranu L, Ciubotaru V, Alexandru O, Banita M, Pisoschi C, Bäcklund ML, Lewensohn R, Dricu A. Dual targeting of IGF-1R and PDGFR inhibits proliferation in high-grade gliomas cells and induces radiosensitivity in JNK-1 expressing cells. J Neurooncol 2007; 85:245-54. [PMID: 17568996 DOI: 10.1007/s11060-007-9417-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 05/17/2007] [Indexed: 02/08/2023]
Abstract
Increased expression and activation of receptor tyrosine kinases frequently occur in human brain tumors, mediating a variety of growth-promoting pathways and leading to radioresistance; however, little is known about their motogenic potency relative to one another. In this study, we found co-expression of Insulin like growth factor-1 receptor (IGF-1R) and platelet derived growth factor receptor (PDGFR) in two high-grade gliomas (HGG) cell lines 18 and 38. Dual targeting of IGF-1R and PDGFR increased cell death in both 18 and 38 cell lines in comparison to inhibition of either receptor alone. In addition, co-inhibition of IGF-1R and PDGFR increased radiosensitivity in 18 cells but failed to intensify the effect of radiation in 38 cells. In HGG cells, radiation-induced cell death has been connected to the activation of c-Jun-NH2-terminal kinase-1 (JNK1). We found that JNK1 was weakly expressed in 38 cells while it had an elevated expression in 18 cells. Exposure to ionizing radiation induced JNK1 activation only in 18 cells without affecting the protein activity in 38 cells. These results suggest that in 18 cell line radiation-activated JNK1 may provide an anti-proliferative signaling, parallel to receptors co-targeting. To test this hypothesis, HGG cells were treated with dominant negative JNK1 (dnJNK1) and the response to radiation was assayed in presence or absence of receptors co-inhibition. Indeed dnJNK protected 18 cells against gamma-irradiation-induced cell death. dnJNK treatment did not influence radiation response of the 38 cell line, which expressed low levels of JNK1. In conclusion we found that IGF-1R and PDGFR co-inhibition caused an increased cell death in two HGG cell line and induced the radiosensitization of the JNK1 expressing cell line.
Collapse
Affiliation(s)
- Mia Carapancea
- Department of Oncology-Pathology, Cancer Center Karolinska and Radiumhemmet Karolinska Institute/University Hospital, R8:00, Stockholm 171 76, Sweden
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Advances in the understanding of glioma pathogenesis have led to increasing interest in the development of targeted molecular agents, and especially kinase inhibitors, for treatment of malignant gliomas. Protein kinases are a large family of enzymes that function as key regulators of cellular signaling pathways governing diverse functions, such as cell proliferation, growth, differentiation, invasion, angiogenesis and apoptosis in malignant gliomas. Preliminary clinical results with kinase inhibitors suggest that they are generally well-tolerated but have shown only modest activity. However, valuable information was obtained from these early clinical trials that will help the future development of these agents. This article reviews the important protein kinases in malignant gliomas, summarizes the existing clinical development of kinase inhibitors and discusses strategies to improve their effectiveness.
Collapse
Affiliation(s)
- Andrew S Chi
- Center For Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Shields-Warren 430D, 44 Binney Street, Boston, MA 02115, USA
| | | |
Collapse
|
29
|
Abstract
BACKGROUND Gliomas are the most common type of primary brain tumor. Nearly two-thirds of gliomas are highly malignant lesions that account for a disproportionate share of brain tumor-related morbidity and mortality. Despite recent advances, two-year survival for glioblastoma with optimal therapy is less than 30%. Even among patients with low-grade gliomas that confer a relatively good prognosis, treatment is almost never curative. REVIEW SUMMARY Surgery and radiation have been the mainstays of therapy for most glioma patients, but temozolomide chemotherapy has recently been proven to prolong overall survival in patients with glioblastoma. Intriguing data suggests that activity of O6-methylguanine-DNA methyltransferase (MGMT), in tumor cells may predict responsiveness to temozolomide and other alkylating agents. Novel treatment approaches, especially targeted molecular therapies against critical components of glioma signaling pathways, appear promising in preliminary studies. Optimal treatment for patients with low-grade gliomas has yet to be determined. Advances in oligodendroglioma biology have identified loss of chromosomes 1p and 19q as powerful indicators of a favorable prognosis. These same changes may predict response to chemotherapy. CONCLUSIONS Though the prognosis for many patients with gliomas is poor, the last decade produced a number of important advances, some of which have translated directly into survival benefits. Rapid progress in the field of glioma molecular biology continues to identify therapeutic targets and provide hope for the future of this challenging disease.
Collapse
Affiliation(s)
- Andrew D Norden
- Division of Cancer Neurology, Department of Neurology, Brigham and Women's Hospital and Center For Neuro-Oncology, Dana Farber Brigham and Women's Cancer Center, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
30
|
Paraskevakou G, Allen C, Nakamura T, Zollman P, James CD, Peng KW, Schroeder M, Russell SJ, Galanis E. Epidermal growth factor receptor (EGFR)-retargeted measles virus strains effectively target EGFR- or EGFRvIII expressing gliomas. Mol Ther 2007; 15:677-86. [PMID: 17299404 DOI: 10.1038/sj.mt.6300105] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
A retargeted measles virus strain MV-GFP-H(AA)-scEGFR was generated by engineering the MV-NSe Edmonston vaccine strain to incorporate both CD46 (Y481A) and signaling lymphocyte activation molecule (SLAM) (R533A) ablating mutations in the hemagglutinin protein in combination with the display of a single-chain antibody against epidermal growth factor receptor (EGFR) at the C terminus of hemagglutinin. The unmodified MV-GFP virus was used as a positive control. Specificity of the EGFR retargeted virus was demonstrated in non-permissive Chinese hamster ovary (CHO) cells stably transfected to express either the natural receptors CD46 or SLAM or the target receptors EGFR and EGFRvIII. In vitro, the retargeted virus had potent antitumor activity against EGFR- or EGFRvIII-overexpressing primary glioblastoma multi-forme (GBM) cell lines that was comparable to the activity of the unmodified MV-GFP virus. Intratumoral administration of MV-GFP-H(AA)-scEGFRvIII in orthotopic GBM12 xenografts resulted in tumor regression, as demonstrated by bioluminescence imaging and significant prolongation of survival, that was comparable to the effect of the unmodified strain. In contrast to MV-GFP, central nervous system administration of the targeted MV-GFP-H(AA)-scEGFR virus in measles replication-permissive Ifnar(ko) CD46 transgenic mice resulted in no neurotoxicity. In conclusion, EGFR-retargeted measles virus strains have comparable therapeutic efficacy to the unmodified virus in glioma cells overexpressing EGFR or EGFRvIII in vivo and in vitro, and improved therapeutic index, a finding with potential translational implications in glioma virotherapy.
Collapse
|
31
|
Narin O, Drappatz J, Doherty LM, Wen PY, Kesari S. Cerebrospinal Fluid Spread of Anaplastic Glioma. J Clin Oncol 2007; 25:596-7. [PMID: 17290068 DOI: 10.1200/jco.2006.09.3096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Ozden Narin
- Dana-Farber/Brigham and Women's Cancer Center, Center for Neuro-Oncology, Boston, MA, USA
| | | | | | | | | |
Collapse
|
32
|
Sathornsumetee S, Vredenburgh KA, Lattimore KP, Rich JN. Malignant glioma drug discovery – targeting protein kinases. Expert Opin Drug Discov 2007; 2:1-17. [DOI: 10.1517/17460441.2.1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|