1
|
A New Class of Uracil-DNA Glycosylase Inhibitors Active against Human and Vaccinia Virus Enzyme. Molecules 2021; 26:molecules26216668. [PMID: 34771075 PMCID: PMC8587785 DOI: 10.3390/molecules26216668] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/24/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022] Open
Abstract
Uracil-DNA glycosylases are enzymes that excise uracil bases appearing in DNA as a result of cytosine deamination or accidental dUMP incorporation from the dUTP pool. The activity of Family 1 uracil-DNA glycosylase (UNG) activity limits the efficiency of antimetabolite drugs and is essential for virulence in some bacterial and viral infections. Thus, UNG is regarded as a promising target for antitumor, antiviral, antibacterial, and antiprotozoal drugs. Most UNG inhibitors presently developed are based on the uracil base linked to various substituents, yet new pharmacophores are wanted to target a wide range of UNGs. We have conducted virtual screening of a 1,027,767-ligand library and biochemically screened the best hits for the inhibitory activity against human and vaccinia virus UNG enzymes. Although even the best inhibitors had IC50 ≥ 100 μM, they were highly enriched in a common fragment, tetrahydro-2,4,6-trioxopyrimidinylidene (PyO3). In silico, PyO3 preferably docked into the enzyme's active site, and in kinetic experiments, the inhibition was better consistent with the competitive mechanism. The toxicity of two best inhibitors for human cells was independent of the presence of methotrexate, which is consistent with the hypothesis that dUMP in genomic DNA is less toxic for the cell than strand breaks arising from the massive removal of uracil. We conclude that PyO3 may be a novel pharmacophore with the potential for development into UNG-targeting agents.
Collapse
|
2
|
Rahimian E, Amini A, Alikarami F, Pezeshki SMS, Saki N, Safa M. DNA repair pathways as guardians of the genome: Therapeutic potential and possible prognostic role in hematologic neoplasms. DNA Repair (Amst) 2020; 96:102951. [PMID: 32971475 DOI: 10.1016/j.dnarep.2020.102951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 11/30/2022]
Abstract
DNA repair pathways, which are also identified as guardians of the genome, protect cells from frequent damage that can lead to DNA breaks. The most deleterious types of damage are double-strand breaks (DSBs), which are repaired by homologous recombination (HR) and non-homologous end joining (NHEJ). Single strand breaks (SSBs) can be corrected through base excision repair (BER), nucleotide excision repair (NER), and mismatch repair (MMR). Failure to restore DNA lesions or inappropriately repaired DNA damage culminates in genomic instability and changes in the regulation of cellular functions. Intriguingly, particular mutations and translocations are accompanied by special types of leukemia. Besides, expression patterns of certain repair genes are altered in different hematologic malignancies. Moreover, analysis of mutations in key mediators of DNA damage repair (DDR) pathways, as well as investigation of their expression and function, may provide us with emerging biomarkers of response/resistance to treatment. Therefore, defective DDR pathways can offer a rational starting point for developing DNA repair-targeted drugs. In this review, we address genetic alterations and gene/protein expression changes, as well as provide an overview of DNA repair pathways.
Collapse
Affiliation(s)
- Elahe Rahimian
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Amini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Alikarami
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA 19104, USA
| | - Seyed Mohammad Sadegh Pezeshki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Eads JR, Krishnamurthi SS, Saltzman J, Bokar JA, Savvides P, Meropol NJ, Gibbons J, Koon H, Sharma N, Rogers L, Pink JJ, Xu Y, Beumer JH, Riendeau J, Fu P, Gerson SL, Dowlati A. Phase I clinical trial of temozolomide and methoxyamine (TRC-102), an inhibitor of base excision repair, in patients with advanced solid tumors. Invest New Drugs 2020; 39:142-151. [PMID: 32556884 DOI: 10.1007/s10637-020-00962-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/11/2020] [Indexed: 11/26/2022]
Abstract
Temozolomide (TMZ) generates DNA adducts that are repaired by direct DNA and base excision repair mechanisms. Methoxyamine (MX, TRC-102) potentiates TMZ activity by binding to apurinic and apyrimidinic (AP) sites after removal of N3-methyladenine and N7-methylguanine, inhibiting site recognition of AP endonuclease. We conducted a phase I trial to determine the maximum tolerated dose and dose-limiting toxicities (DLTs) of intravenous MX when given with oral TMZ. Patients with advanced solid tumors and progression on standard treatment were enrolled to a standard 3 + 3 dose escalation trial assessing escalating doses of TMZ and MX. Tumor response was assessed per RECIST and adverse events (AEs) by CTCAEv3. Pharmacokinetics (PK) of MX and COMET assays on peripheral blood mononuclear cells were performed. 38 patients were enrolled-median age 59.5 years (38-76), mean number of cycles 2.9 [1-13]. No DLTs were observed. Cycle 1 grade 3 AEs included fatigue, lymphopenia, anemia, INR, leukopenia, neutropenia, allergic reaction, constipation, psychosis and paranoia. Cycle 2-13 grade 4 AEs included thrombocytopenia and confusion. A partial response was seen in 1 patient with a pancreatic neuroendocrine tumor (PNET) and six additional patients, each with different tumor types, demonstrated prolonged stable disease. MX PK was linear with dose and was not affected by concomitant TMZ. TMZ 200 mg/m2 daily × 5 may be safely administered with MX 150 mg/m2 intravenously once on day 1 with minimal toxicity. Further studies assessing this drug combination in select tumor types where temozolomide has activity may be warranted.
Collapse
Affiliation(s)
- Jennifer R Eads
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Smitha S Krishnamurthi
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Joel Saltzman
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Joseph A Bokar
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Panos Savvides
- St. Joseph's Hospital and Medical Center, University of Arizona Comprehensive Cancer Center, University of Arizona, Phoenix, AZ, USA
| | - Neal J Meropol
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
- Flatiron Health, New York, NY, USA
| | - Joseph Gibbons
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Henry Koon
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Neelesh Sharma
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Lisa Rogers
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - John J Pink
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Yan Xu
- Case Comprehensive Cancer Center, Cleveland State University, Cleveland, OH, USA
| | - Jan H Beumer
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Riendeau
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Pingfu Fu
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Stanton L Gerson
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA
| | - Afshin Dowlati
- Case Comprehensive Cancer Center, Case Western Reserve University, University Hospitals Seidman Cancer Center, 11100 Euclid Avenue, Lakeside 1200, Cleveland, OH, 44106, USA.
| |
Collapse
|
4
|
Mechetin GV, Endutkin AV, Diatlova EA, Zharkov DO. Inhibitors of DNA Glycosylases as Prospective Drugs. Int J Mol Sci 2020; 21:ijms21093118. [PMID: 32354123 PMCID: PMC7247160 DOI: 10.3390/ijms21093118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/22/2022] Open
Abstract
DNA glycosylases are enzymes that initiate the base excision repair pathway, a major biochemical process that protects the genomes of all living organisms from intrinsically and environmentally inflicted damage. Recently, base excision repair inhibition proved to be a viable strategy for the therapy of tumors that have lost alternative repair pathways, such as BRCA-deficient cancers sensitive to poly(ADP-ribose)polymerase inhibition. However, drugs targeting DNA glycosylases are still in development and so far have not advanced to clinical trials. In this review, we cover the attempts to validate DNA glycosylases as suitable targets for inhibition in the pharmacological treatment of cancer, neurodegenerative diseases, chronic inflammation, bacterial and viral infections. We discuss the glycosylase inhibitors described so far and survey the advances in the assays for DNA glycosylase reactions that may be used to screen pharmacological libraries for new active compounds.
Collapse
Affiliation(s)
- Grigory V. Mechetin
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
| | - Anton V. Endutkin
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
| | - Evgeniia A. Diatlova
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
| | - Dmitry O. Zharkov
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 8 Lavrentieva Ave., 630090 Novosibirsk, Russia; (G.V.M.); (A.V.E.); (E.A.D.)
- Novosibirsk State University, 2 Pirogova St., 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-383-363-5187
| |
Collapse
|
5
|
Signaling Pathways, Chemical and Biological Modulators of Nucleotide Excision Repair: The Faithful Shield against UV Genotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4654206. [PMID: 31485292 PMCID: PMC6702832 DOI: 10.1155/2019/4654206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/10/2019] [Indexed: 12/28/2022]
Abstract
The continuous exposure of the human body's cells to radiation and genotoxic stresses leads to the accumulation of DNA lesions. Fortunately, our body has several effective repair mechanisms, among which is nucleotide excision repair (NER), to counteract these lesions. NER includes both global genome repair (GG-NER) and transcription-coupled repair (TC-NER). Deficiencies in the NER pathway underlie the development of several DNA repair diseases, such as xeroderma pigmentosum (XP), Cockayne syndrome (CS), and trichothiodystrophy (TTD). Deficiencies in GG-NER and TC-NER render individuals to become prone to cancer and neurological disorders, respectively. Therefore, NER regulation is of interest in fine-tuning these risks. Distinct signaling cascades including the NFE2L2 (NRF2), AHR, PI3K/AKT1, MAPK, and CSNK2A1 pathways can modulate NER function. In addition, several chemical and biological compounds have proven success in regulating NER's activity. These modulators, particularly the positive ones, could therefore provide potential treatments for genetic DNA repair-based diseases. Negative modulators, nonetheless, can help sensitize cells to killing by genotoxic chemicals. In this review, we will summarize and discuss the major upstream signaling pathways and molecules that could modulate the NER's activity.
Collapse
|
6
|
Targeting DNA repair with aphidicolin sensitizes primary chronic lymphocytic leukemia cells to purine analogs. Oncotarget 2018; 7:38367-38379. [PMID: 27223263 PMCID: PMC5122396 DOI: 10.18632/oncotarget.9525] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/04/2016] [Indexed: 11/25/2022] Open
Abstract
Purine analogs are among the most effective chemotherapeutic drugs for the treatment of chronic lymphocytic leukemia (CLL). However, chemoresistance and toxicity limit their clinical use. Here, we report that the DNA polymerase inhibitor aphidicolin, which displayed negligible cytotoxicity as a single agent in primary CLL cells, markedly synergizes with fludarabine and cladribine via enhanced apoptosis. Importantly, synergy was recorded regardless of CLL prognostic markers. At the molecular level, aphidicolin enhanced purine analog-induced phosphorylation of p53 and accumulation of γH2AX, consistent with increase in DNA damage. In addition, aphidicolin delayed γH2AX disappearance that arises after removal of purine analogs, suggesting that aphidicolin causes an increase in DNA damage by impeding DNA damage repair. Similarly, aphidicolin inhibited UV-induced DNA repair known to occur primarily through the nucleotide excision repair (NER) pathway. Finally, we showed that fludarabine induced nuclear import of XPA, an indispensable factor for NER, and that XPA silencing sensitized cell lines to undergo apoptosis in response to fludarabine. Together, our data indicate that aphidicolin potentiates the cytotoxicity of purine analogs by inhibiting a DNA repair pathway that involves DNA polymerases, most likely NER, and provide a rationale for manipulating it to therapeutic advantage.
Collapse
|
7
|
Myhrvold IK, Cremaschi A, Hermansen JU, Tjønnfjord GE, Munthe LA, Taskén K, Skånland SS. Single cell profiling of phospho-protein levels in chronic lymphocytic leukemia. Oncotarget 2018; 9:9273-9284. [PMID: 29507689 PMCID: PMC5823631 DOI: 10.18632/oncotarget.23949] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) has a high incidence and a steeply growing prevalence in the Western world. The heterogeneity of the disease necessitates individual mapping of biology and predicted drug response in each patient as basis for administration of tailored treatments. Cell signaling aberrations may serve as biological indicators for suitable therapy. By applying phospho-specific flow cytometry, we mapped basal and induced phosphorylation levels of 20 phospho-epitopes on proteins relevant to B-cell signaling in B cells from 22 CLL patients and 25 normal controls. The signaling response of the cytostatic drugs fludarabine, doxorubicin and vincristine was also investigated. CLL cells exerted similar or lower basal phosphorylation levels compared to normal B cells, with the exception of STAT3 (pY705) which was increased. Interestingly, STAT3 inhibitors normalized the STAT3 (pY705) level and reduced cell viability. Vincristine treatment significantly modulated phosphorylation levels in CLL cells, while no effect was observed in controls or after fludarabine or doxorubicin treatment. After BCR stimulation, CLL cells showed a tendency towards impaired phosphorylation levels, significant for several of the analyzed proteins. However, the level of Akt (pS473) was more potently induced in IgHV unmutated CLL (UM-CLL) patient samples and was significantly higher than in M-CLL samples. Importantly, the PI3Kδ inhibitor idelalisib potently reversed the effect of anti-IgM on Akt (pS473). Thus, signaling aberrations could be identified by phosphoflow cytometry and aberrant signaling could be normalized by small molecule drugs. This approach can identify relevant drug targets as well as drug effects in the individual patient.
Collapse
Affiliation(s)
- Ida K Myhrvold
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Centre for Inflammation Research, University of Oslo, Oslo, Norway.,K. G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Andrea Cremaschi
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
| | - Johanne U Hermansen
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Centre for Inflammation Research, University of Oslo, Oslo, Norway.,K. G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Geir E Tjønnfjord
- Department of Haematology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Centre for Immune Regulation, Department of Immunology, University of Oslo, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kjetil Taskén
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Centre for Inflammation Research, University of Oslo, Oslo, Norway.,K. G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Sigrid S Skånland
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Centre for Inflammation Research, University of Oslo, Oslo, Norway.,K. G. Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| |
Collapse
|
8
|
Caimi PF, Cooper BW, William BM, Dowlati A, Barr PM, Fu P, Pink J, Xu Y, Lazarus HM, de Lima M, Gerson SL. Phase I clinical trial of the base excision repair inhibitor methoxyamine in combination with fludarabine for patients with advanced hematologic malignancies. Oncotarget 2017; 8:79864-79875. [PMID: 29108368 PMCID: PMC5668101 DOI: 10.18632/oncotarget.20094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023] Open
Abstract
PURPOSE We determined the safety, pharmacokinetics, pharmacodynamics and recommended phase II dose of the base excision repair blocker methoxyamine combined with fludarabine. MATERIALS AND METHODS This was a phase I study with intravenous fludarabine (25 mg/m2, days 1-5), and methoxyamine (15 mg/m2-120 mg/m2, once). A maximum of six cycles were given. Adult patients with relapsed/refractory hematologic malignancies, excluding acute myeloid leukemia, were eligible. RESULTS Twenty patients were treated; diagnoses included CLL/SLL (n = 10), follicular lymphoma (n = 3), DLBCL (n = 3), mantle cell lymphoma (n = 1), anaplastic large cell lymphoma (n = 1) and plasma cell myeloma (n = 2). No DLTs were observed and dose escalation reached the maximum planned dose. Hematologic toxicity was frequent; most common grade 3-4 toxicities were lymphopenia (70%), neutropenia (60%), leukopenia (50%) and anemia (40%). Four patients achieved a partial remission and 8 achieved stable disease. The drug combination resulted in increased DNA damage measured with the Comet assay. CONCLUSIONS Methoxyamine combined with fludarabine was safe and well tolerated. Hematologic toxicity was comparable to single agent fludarabine. Activity appears to correlate with increased levels of DNA damage. Further studies will examine use of this combination of as part conditioning regimens of stem cell transplant and use of methoxyamine as fludarabine dose-sparing agent.
Collapse
Affiliation(s)
- Paolo F. Caimi
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Brenda W. Cooper
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Basem M. William
- Division of Hematology. The Ohio State University Medical School, Columbus, Ohio, USA
| | - Afshin Dowlati
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Paul M. Barr
- Division of Hematology and Oncology, Department of Medicine, University of Rochester, Rochester, New York, USA
| | - Pingfu Fu
- Department of Biostatistics, Case Western Reserve University, Cleveland, Ohio, USA
| | - John Pink
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Yan Xu
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Hillard M. Lazarus
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Marcos de Lima
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| | - Stanton L. Gerson
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Oleinick NL, Biswas T, Patel R, Tao M, Patel R, Weeks L, Sharma N, Dowlati A, Gerson SL, Fu P, Zhang J, Machtay M. Radiosensitization of non-small-cell lung cancer cells and xenografts by the interactive effects of pemetrexed and methoxyamine. Radiother Oncol 2016; 121:335-341. [PMID: 27838149 DOI: 10.1016/j.radonc.2016.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 09/09/2016] [Accepted: 10/03/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE The anti-folate pemetrexed is a radiosensitizer. In pre-clinical models, pemetrexed is more effective along with the base-excision-repair inhibitor methoxyamine. We tested whether methoxyamine enhances pemetrexed-mediated radiosensitization of lung adenocarcinoma cells and xenografts. MATERIALS AND METHODS A549 and H1299 cells were evaluated for cell cycle distribution by flow cytometry, radiosensitization by clonogenic assay, and DNA repair by neutral comet assay and repair protein activation. H460 cells were included in some studies. Xenografts in nude mice received drug(s) and/or radiation, and tumor growth was monitored by caliper and in vivo toxicity by animal weight. RESULTS Exposure to pemetrexed/methoxyamine for 24 (H1299, H460) or 48 (A549)hours before irradiation resulted in accumulation of cells near the radiosensitive G1/S border; dose-enhancement factors of 1.62±0.19, 1.97±0.25, and 1.67±0.30, respectively; reduction of mean inactivation dose by 32%, 30%, and 46%, respectively; and significant reductions of SF2 and SF4 (p<0.05). Radiosensitization was associated with rapid DNA double-strand-break rejoining and increased levels of DNA-PKcs. Both tumor-growth rate and tumor-growth delay were significantly improved by adding methoxyamine to pemetrexed pre-irradiation (p<0.0001); no mice lost weight during treatment. CONCLUSIONS Addition of methoxyamine to pemetrexed and fractionated radiotherapy may improve outcome for patients with locally advanced non-squamous non-small-cell lung cancer.
Collapse
Affiliation(s)
- Nancy L Oleinick
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA.
| | - Tithi Biswas
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Rutulkumar Patel
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Department of Pharmacology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Mingfang Tao
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Ravi Patel
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Lachelle Weeks
- Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Neelesh Sharma
- Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Afshin Dowlati
- Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Stanton L Gerson
- Department of Medicine, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Pingfu Fu
- Department of Epidemiology and Biostatistics, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Junran Zhang
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| | - Mitchell Machtay
- Department of Radiation Oncology, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA; Case Comprehensive Cancer Center, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, USA
| |
Collapse
|
10
|
Hegde PM, Dutta A, Sengupta S, Mitra J, Adhikari S, Tomkinson AE, Li GM, Boldogh I, Hazra TK, Mitra S, Hegde ML. The C-terminal Domain (CTD) of Human DNA Glycosylase NEIL1 Is Required for Forming BERosome Repair Complex with DNA Replication Proteins at the Replicating Genome: DOMINANT NEGATIVE FUNCTION OF THE CTD. J Biol Chem 2015; 290:20919-20933. [PMID: 26134572 DOI: 10.1074/jbc.m115.642918] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Indexed: 12/21/2022] Open
Abstract
The human DNA glycosylase NEIL1 was recently demonstrated to initiate prereplicative base excision repair (BER) of oxidized bases in the replicating genome, thus preventing mutagenic replication. A significant fraction of NEIL1 in cells is present in large cellular complexes containing DNA replication and other repair proteins, as shown by gel filtration. However, how the interaction of NEIL1 affects its recruitment to the replication site for prereplicative repair was not investigated. Here, we show that NEIL1 binarily interacts with the proliferating cell nuclear antigen clamp loader replication factor C, DNA polymerase δ, and DNA ligase I in the absence of DNA via its non-conserved C-terminal domain (CTD); replication factor C interaction results in ∼8-fold stimulation of NEIL1 activity. Disruption of NEIL1 interactions within the BERosome complex, as observed for a NEIL1 deletion mutant (N311) lacking the CTD, not only inhibits complete BER in vitro but also prevents its chromatin association and reduced recruitment at replication foci in S phase cells. This suggests that the interaction of NEIL1 with replication and other BER proteins is required for efficient repair of the replicating genome. Consistently, the CTD polypeptide acts as a dominant negative inhibitor during in vitro repair, and its ectopic expression sensitizes human cells to reactive oxygen species. We conclude that multiple interactions among BER proteins lead to large complexes, which are critical for efficient BER in mammalian cells, and the CTD interaction could be targeted for enhancing drug/radiation sensitivity of tumor cells.
Collapse
Affiliation(s)
- Pavana M Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Arijit Dutta
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030; Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Shiladitya Sengupta
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Joy Mitra
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Sanjay Adhikari
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030
| | - Alan E Tomkinson
- Department of Internal Medicine and University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico 87131
| | - Guo-Min Li
- Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536
| | - Istvan Boldogh
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Tapas K Hazra
- Departments of Internal Medicine, University of Texas Medical Branch, Galveston, Texas 77555
| | - Sankar Mitra
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030; Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555; Weill Medical College of Cornell University, New York, New York.
| | - Muralidhar L Hegde
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, Texas 77030; Weill Medical College of Cornell University, New York, New York; Houston Methodist Neurological Institute, Houston, Texas 77030.
| |
Collapse
|
11
|
Lee MW, Parker WB, Xu B. New insights into the synergism of nucleoside analogs with radiotherapy. Radiat Oncol 2013; 8:223. [PMID: 24066967 PMCID: PMC3851323 DOI: 10.1186/1748-717x-8-223] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/24/2013] [Indexed: 11/18/2022] Open
Abstract
Nucleoside analogs have been frequently used in combination with radiotherapy in the clinical setting, as it has long been understood that inhibition of DNA repair pathways is an important means by which many nucleoside analogs synergize. Recent advances in our understanding of the structure and function of deoxycytidine kinase (dCK), a critical enzyme required for the anti-tumor activity for many nucleoside analogs, have clarified the mechanistic role this kinase plays in chemo- and radio-sensitization. A heretofore unrecognized role of dCK in the DNA damage response and cell cycle machinery has helped explain the synergistic effect of these agents with radiotherapy. Since most currently employed nucleoside analogs are primarily activated by dCK, these findings lend fresh impetus to efforts focused on profiling and modulating dCK expression and activity in tumors. In this review we will briefly review the pharmacology and biochemistry of the major nucleoside analogs in clinical use that are activated by dCK. This will be followed by discussions of recent advances in our understanding of dCK activation via post-translational modifications in response to radiation and current strategies aimed at enhancing this activity in cancer cells.
Collapse
Affiliation(s)
- Michael W Lee
- Department of Medical Education, College of Medicine, University of Central Florida, 6850 Lake Nona Blvd,, Orlando, FL 32827, USA.
| | | | | |
Collapse
|
12
|
Khanra K, Bhattacharya C, Bhattacharyya N. Association of a newly identified variant of DNA polymerase beta (polβΔ63-123, 208-304) with the risk factor of ovarian carcinoma in India. Asian Pac J Cancer Prev 2013; 13:1999-2002. [PMID: 22901161 DOI: 10.7314/apjcp.2012.13.5.1999] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND DNA polymerase is a single-copy gene that is considered to be part of the DNA repair machinery in mammalian cells. The encoded enzyme is a key to the base excision repair (BER) pathway. It is evident that pol beta has mutations in various cancer samples, but little is known about ovarian cancer. AIM Identification of any variant form of polβ cDNA in ovarian carcinoma and determination of association between the polymorphism and ovarian cancer risk in Indian patients. We used 152 samples to isolate and perform RT-PCR and sequencing. RESULTS A variant of polymerase beta (deletion of exon 4-6 and 11-13, comprising of amino acid 63-123, and 208-304) is detected in heterozygous condition. The product size of this variant is 532 bp while wild type pol beta is 1 kb. Our study of association between the variant and the endometrioid type shows that it is a statistically significant factor for ovarian cancer [OR=31.9 (4.12-246.25) with p<0.001]. The association between variant and stage IV patients further indicated risk (χ2 value of 29.7, and OR value 6.77 with 95% CI values 3.3-13.86). The correlation study also confirms the association data (Pearson correlation values for variant/stage IV and variant/endometrioid of 0.44 and 0.39). CONCLUSION Individuals from this part of India with this type of variant may be at risk of stage IV, endometrioid type ovarian carcinoma.
Collapse
Affiliation(s)
- Kalyani Khanra
- Department of Biotechnology, Haldia Institute of Technology, West Bengal, India
| | | | | |
Collapse
|
13
|
A phase 1 study of TRC102, an inhibitor of base excision repair, and pemetrexed in patients with advanced solid tumors. Invest New Drugs 2012; 31:714-23. [PMID: 23054206 DOI: 10.1007/s10637-012-9876-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 08/29/2012] [Indexed: 10/27/2022]
Abstract
INTRODUCTION TRC102 potentiates the activity of cancer therapies that induce base excision repair (BER) including antimetabolite and alkylating agents. TRC102 rapidly and covalently binds to apurinic/apyrimidinic (AP) sites generated during BER, and TRC102-bound DNA causes topoisomerase II-dependent irreversible strand breaks and apoptosis. This study assessed the safety, maximum-tolerated dose (MTD), pharmacokinetics and pharmacodynamics of TRC102 alone and in combination with pemetrexed. PURPOSE Patients with advanced solid tumors received oral TRC102 daily for 4 days. Two weeks later, patients began standard-dose pemetrexed on day 1 in combination with oral TRC102 on days 1 to 4. The pemetrexed-TRC102 combination was repeated every 3 weeks until disease progression. METHODS Twenty-eight patients were treated with TRC102 at 15, 30, 60 or 100 mg/m(2)/d. The MTD was exceeded at 100 mg/m(2)/d due to grade 3 anemia in 50 % of patients. TRC102 exposure increased in proportion to dose with a mean t1/2 of 28 h. A pharmacodynamic assay confirmed that TRC102 binds to pemetrexed-induced AP sites at all doses studied. Stable disease or better was achieved in 15 of 25 patients evaluable for response (60 %), including one patient with recurrent metastatic oropharyngeal carcinoma that expressed high levels of thymidylate synthase, who achieved a partial response and was progression free for 14 months. CONCLUSIONS When administered with pemetrexed, the maximum tolerated dose of oral TRC102 is 60 mg/m(2)/d for 4 days. Randomized controlled studies are planned to evaluate the clinical benefit of adding TRC102 to pemetrexed and other agents that induce BER.
Collapse
|
14
|
Casorelli I, Bossa C, Bignami M. DNA damage and repair in human cancer: molecular mechanisms and contribution to therapy-related leukemias. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2012; 9:2636-57. [PMID: 23066388 PMCID: PMC3447578 DOI: 10.3390/ijerph9082636] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 06/12/2012] [Accepted: 07/02/2012] [Indexed: 12/12/2022]
Abstract
Most antitumour therapies damage tumour cell DNA either directly or indirectly. Without repair, damage can result in genetic instability and eventually cancer. The strong association between the lack of DNA damage repair, mutations and cancer is dramatically demonstrated by a number of cancer-prone human syndromes, such as xeroderma pigmentosum, ataxia-telangiectasia and Fanconi anemia. Notably, DNA damage responses, and particularly DNA repair, influence the outcome of therapy. Because DNA repair normally excises lethal DNA lesions, it is intuitive that efficient repair will contribute to intrinsic drug resistance. Unexpectedly, a paradoxical relationship between DNA mismatch repair and drug sensitivity has been revealed by model studies in cell lines. This suggests that connections between DNA repair mechanism efficiency and tumour therapy might be more complex. Here, we review the evidence for the contribution of carcinogenic properties of several drugs as well as of alterations in specific mechanisms involved in drug-induced DNA damage response and repair in the pathogenesis of therapy-related cancers.
Collapse
Affiliation(s)
- Ida Casorelli
- Azienda Ospedaliera Sant’Andrea, Via di Grottarossa 1035-1039, Roma 00189, Italy;
| | - Cecilia Bossa
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, Roma 00161, Italy;
| | - Margherita Bignami
- Department of Environment and Primary Prevention, Istituto Superiore di Sanità, Viale Regina Elena 299, Roma 00161, Italy;
- Author to whom correspondence should be addressed; ; Tel.: +39-6-49901-2355; Fax: +39-6-49901-3650
| |
Collapse
|
15
|
Bulgar AD, Weeks LD, Miao Y, Yang S, Xu Y, Guo C, Markowitz S, Oleinick N, Gerson SL, Liu L. Removal of uracil by uracil DNA glycosylase limits pemetrexed cytotoxicity: overriding the limit with methoxyamine to inhibit base excision repair. Cell Death Dis 2012; 3:e252. [PMID: 22237209 PMCID: PMC3270269 DOI: 10.1038/cddis.2011.135] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 11/09/2011] [Accepted: 11/09/2011] [Indexed: 01/03/2023]
Abstract
Uracil DNA glycosylase (UDG) specifically removes uracil bases from DNA, and its repair activity determines the sensitivity of the cell to anticancer agents that are capable of introducing uracil into DNA. In the present study, the participation of UDG in the response to pemetrexed-induced incorporation of uracil into DNA was studied using isogenic human tumor cell lines with or without UDG (UDG(+/+)/UDG(-/-)). UDG(-/-) cells were very sensitive to pemetrexed. Cell killing by pemetrexed was associated with genomic uracil accumulation, stalled DNA replication, and catastrophic DNA strand breaks. By contrast, UDG(+/+) cells were >10 times more resistant to pemetrexed due to the rapid removal of uracil from DNA by UDG and subsequent repair of the resultant AP sites (abasic sites) via the base excision repair (BER). The resistance to pemetrexed in UDG(+/+) cells could be reversed by the addition of methoxyamine (MX), which binds to AP sites and interrupts BER pathway. Furthermore, MX-bound AP sites induced cell death was related to their cytotoxic effect of dual inactivation of UDG and topoisomerase IIα, two genes that are highly expressed in lung cancer cells in comparison with normal cells. Thus, targeting BER-based therapy exhibits more selective cytotoxicity on cancer cells through a synthetic lethal mechanism.
Collapse
Affiliation(s)
- A D Bulgar
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - L D Weeks
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - Y Miao
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - S Yang
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Y Xu
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
- Department of Chemistry, Cleveland State University, Cleveland, OH, USA
| | - C Guo
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - S Markowitz
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
- Department of Chemistry, Cleveland State University, Cleveland, OH, USA
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA
| | - N Oleinick
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH, USA
| | - S L Gerson
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - L Liu
- Department of Medicine, Division of Hematology/Oncology, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 2103 Cornell Road, Cleveland, OH 44106, USA
| |
Collapse
|