1
|
Penketh PG, Williamson HS, Baumann RP, Shyam K. Design Strategy for the EPR Tumor-Targeting of 1,2-Bis(sulfonyl)-1-alkylhydrazines. Molecules 2021; 26:molecules26020259. [PMID: 33419160 PMCID: PMC7825511 DOI: 10.3390/molecules26020259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/30/2020] [Accepted: 01/01/2021] [Indexed: 02/04/2023] Open
Abstract
A design strategy for macromolecular prodrugs is described, that are expected to exhibit robust activity against most solid tumor types while resulting in minimal toxicities to normal tissues. This approach exploits the enhanced permeability, and retention (EPR) effect, and utilizes carefully engineered rate constants to selectively target tumor tissue with short-lived cytotoxic moieties. EPR based tumor accumulation (half-life ~ 15 h) is dependent upon the ubiquitous abnormal solid tumor capillary morphology and is expected to be independent of individual tumor cell genetic variability that leads to resistance to molecularly targeted agents. The macromolecular sulfonylhydrazine-based prodrugs hydrolyze spontaneously with long half-life values (~10 h to >300 h dependent upon their structure) resulting in the majority of the 1,2-bis(sulfonyl)-1-alkylhydrazines (BSHs) cytotoxic warhead being released only after tumor sequestration. The very short half-life (seconds) of the finally liberated BSHs localizes the cytotoxic stress to the tumor target site by allowing insufficient time for escape. Thus, short lifespan anticancer species are liberated, and exhibit their activity largely within the tumor target. The abnormal tumor cell membrane pH gradients favor the uptake of BSHs compared to that of normal cells, further enhancing their selectivity. The reliance on physicochemical/chemical kinetic parameters and the EPR effect is expected to reduce response variability, and the acquisition of resistance.
Collapse
Affiliation(s)
- Philip G. Penketh
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; (R.P.B.); (K.S.)
- Correspondence:
| | | | - Raymond P. Baumann
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; (R.P.B.); (K.S.)
| | - Krishnamurthy Shyam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA; (R.P.B.); (K.S.)
| |
Collapse
|
2
|
Penketh PG, Finch RA, Sauro R, Baumann RP, Ratner ES, Shyam K. pH-dependent general base catalyzed activation rather than isocyanate liberation may explain the superior anticancer efficacy of laromustine compared to related 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine prodrugs. Chem Biol Drug Des 2017. [PMID: 28636806 DOI: 10.1111/cbdd.13057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Laromustine (also known as cloretazine, onrigin, VNP40101M, 101M) is a prodrug of 90CE, a short-lived chloroethylating agent with anticancer activity. The short half-life of 90CE necessitates the use of latentiated prodrug forms for in vivo treatments. Alkylaminocarbonyl-based prodrugs such as laromustine exhibit significantly superior in vivo activity in several murine tumor models compared to analogs utilizing acyl, and alkoxycarbonyl latentiating groups. The alkylaminocarbonyl prodrugs possess two exclusive characteristics: (i) They are primarily unmasked by spontaneous base catalyzed elimination; and (ii) they liberate a reactive carbamoylating species. Previous speculations as to the therapeutic superiority of laromustine have focused upon the inhibition of enzymes by carbamoylation. We have investigated the therapeutic interactions of analogs with segregated chloroethylating and carbamoylating activities (singly and in combination) in the in vivo murine L1210 leukemia model. The combined treatment with chloroethylating and carbamoylating prodrugs failed to result in any synergism and produced a reduction in the therapeutic efficacy compared to the chloroethylating prodrug alone. Evidence supporting an alternative explanation for the superior tumor selectivity of laromustine is presented that is centered upon the high pH sensitivity of its base catalyzed activation, and the more alkaline intracellular pH values commonly found within tumor cells.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A Finch
- Department of Veterinary Sciences, The University of Texas MD Anderson Cancer Center, Bastrop, TX, USA
| | - Rachel Sauro
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Raymond P Baumann
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Elena S Ratner
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Krishnamurthy Shyam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
3
|
Shyam K, Penketh PG, Baumann RP, Finch RA, Zhu R, Zhu YL, Sartorelli AC. Antitumor sulfonylhydrazines: design, structure-activity relationships, resistance mechanisms, and strategies for improving therapeutic utility. J Med Chem 2015; 58:3639-71. [PMID: 25612194 DOI: 10.1021/jm501459c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
1,2-Bis(sulfonyl)-1-alkylhydrazines (BSHs) were conceived as more specific DNA guanine O-6 methylating and chloroethylating agents lacking many of the undesirable toxicophores contained in antitumor nitrosoureas. O(6)-Alkylguanine-DNA alkyltransferase (MGMT) is the sole repair protein for O(6)-alkylguanine lesions in DNA and has been reported to be absent in 5-20% of most tumor types. Many BSHs exhibit highly selective cytotoxicity toward cells deficient in MGMT activity. The development of clinically useful MGMT assays should permit the identification of tumors with this vulnerability and allow for the preselection of patient subpopulations with a high probability of responding. The BSH system is highly versatile, permitting the synthesis of many prodrug types with the ability to incorporate an additional level of tumor-targeting due to preferential activation by tumor cells. Furthermore, it may be possible to expand the spectrum of activity of these agents to include tumors with MGMT activity by combining them with tumor-targeted MGMT inhibitors.
Collapse
Affiliation(s)
- Krishnamurthy Shyam
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Philip G Penketh
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Raymond P Baumann
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Rick A Finch
- ‡Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, 650 Cool Water Drive, Bastrop, Texas 78602, United States
| | - Rui Zhu
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Yong-Lian Zhu
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Alan C Sartorelli
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| |
Collapse
|
4
|
Penketh PG, Patridge E, Shyam K, Baumann RP, Zhu R, Ishiguro K, Sartorelli AC. Influence of glutathione and glutathione S-transferases on DNA interstrand cross-link formation by 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine, the active anticancer moiety generated by laromustine. Chem Res Toxicol 2014; 27:1440-9. [PMID: 25012050 PMCID: PMC4137992 DOI: 10.1021/tx500197t] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
Prodrugs
of 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine
(90CE) are promising anticancer agents. The 90CE moiety is a readily
latentiated, short-lived (t1/2 ∼
30 s) chloroethylating agent that can generate high yields of oxophilic
electrophiles responsible for the chloroethylation of the O-6 position
of guanine in DNA. These guanine O-6 alkylations are believed to be
responsible for the therapeutic effects of 90CE and its prodrugs.
Thus, 90CE demonstrates high selectivity toward tumors with diminished
levels of O6-alkylguanine-DNA alkyltransferase
(MGMT), the resistance protein responsible for O6-alkylguanine repair. The formation of O6-(2-chloroethyl)guanine lesions ultimately leads to the generation
of highly cytotoxic 1-(N3-cytosinyl),-2-(N1-guaninyl)ethane DNA interstrand cross-links
via N1,O6-ethanoguanine
intermediates. The anticancer activity arising from this sequence
of reactions is thus identical to this component of the anticancer
activity of the clinically used chloroethylnitrosoureas. Herein, we
evaluate the ability of glutathione (GSH) and other low molecular
weight thiols, as well as GSH coupled with various glutathione S-transferase enzymes (GSTs) to attenuate the final yields
of cross-links generated by 90CE when added prior to or immediately
following the initial chloroethylation step to determine the major
point(s) of interaction. In contrast to studies utilizing BCNU as
a chloroethylating agent by others, GSH (or GSH/GST) did not appreciably
quench DNA interstrand cross-link precursors. While thiols alone offered
little protection at either alkylation step, the GSH/GST couple was
able to diminish the initial yields of cross-link precursors. 90CE
exhibited a very different GST isoenzyme susceptibility to that reported
for BCNU, this could have important implications in the relative resistance
of tumor cells to these agents. The protection afforded by GSH/GST
was compared to that produced by MGMT.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Pharmacology and Yale Cancer Center, Yale University School of Medicine , New Haven, Connecticut 06520, United States
| | | | | | | | | | | | | |
Collapse
|
5
|
Sun G, Zhao L, Fan T, Li S, Zhong R. Investigations on the Effect of O6-Benzylguanine on the Formation of dG-dC Interstrand Cross-Links Induced by Chloroethylnitrosoureas in Human Glioma Cells Using Stable Isotope Dilution High-Performance Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometry. Chem Res Toxicol 2014; 27:1253-62. [PMID: 24914620 DOI: 10.1021/tx500143b] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Guohui Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Tengjiao Fan
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Sisi Li
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, P. R. China
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100124, P. R. China
| |
Collapse
|
6
|
Penketh PG, Shyam K, Zhu R, Baumann RP, Ishiguro K, Sartorelli AC. Influence of phosphate and phosphoesters on the decomposition pathway of 1,2-bis(methylsulfonyl)-1-(2-chloroethyhydrazine (90CE), the active anticancer moiety generated by Laromustine, KS119, and KS119W. Chem Res Toxicol 2014; 27:818-33. [PMID: 24618018 PMCID: PMC4033638 DOI: 10.1021/tx500004y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Prodrugs of the short-lived chloroethylating
agent 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine
(90CE) and its methylating analogue 1,2-bis(methylsulfonyl)-1-(methyl)hydrazine
(KS90) are potentially useful anticancer agents. This class of agents
frequently yields higher ratios of therapeutically active oxophilic
electrophiles responsible for DNA O6-guanine
alkylations to other electrophiles with lower therapeutic relevance
than the nitrosoureas. This results in improved selectivity toward
tumors with diminished levels of O6-alkylguanine-DNA
alkyltransferase (MGMT), the resistance protein responsible for O6-alkylguanine repair. The formation of O6-(2-chloroethyl)guanine, which leads to the
formation of a DNA–DNA interstrand cross-link, accounts for
the bulk of the anticancer activity of 90CE prodrugs. Herein, we describe
a new decomposition pathway that is available to 90CE but not to its
methylating counterpart. This pathway appears to be subject to general/acid
base catalysis with phosphate (Pi), phosphomonoesters, and phosphodiesters,
being particularly effective. This pathway does not yield a chloroethylating
species and results in a major change in nucleophile preference since
thiophilic rather than oxophilic electrophiles are produced. Thus,
a Pi concentration dependent decrease in DNA–DNA interstand
cross-link formation was observed. Changes in 90CE decomposition products
but not alkylation kinetics occurred in the presence of Pi since the
prebranch point elimination of the N-1 methanesulfinate moiety remained
the rate-limiting step. The Pi catalyzed route is expected to dominate
at Pi and phosphoester concentrations totaling >25–35 mM.
In
view of the abundance of Pi and phosphoesters in cells, this pathway
may have important effects on agent toxicity, tumor selectivity, and
resistance to prodrugs of 90CE. Furthermore, it may be possible to
design analogues that diminish this thiophile-generating pathway,
which is likely superfluous at best and potentially detrimental to
the targeting of hypoxic regions where Pi concentrations can be significantly
elevated.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Pharmacology and Yale Cancer Center, Yale University School of Medicine , 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | | | | | | | | | | |
Collapse
|
7
|
Zhu R, Baumann RP, Penketh PG, Shyam K, Sartorelli AC. Hypoxia-selective O6-alkylguanine-DNA alkyltransferase inhibitors: design, synthesis, and evaluation of 6-(benzyloxy)-2-(aryldiazenyl)-9H-purines as prodrugs of O6-benzylguanine. J Med Chem 2013; 56:1355-9. [PMID: 23311288 PMCID: PMC3722860 DOI: 10.1021/jm301804p] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
O(6)-Alkylguanine-DNA alkyltransferase (AGT) is a DNA repair protein which removes alkyl groups from the O-6 position of guanine, thereby providing strong resistance to anticancer agents which alkylate this position. The clinical usefulness of these anticancer agents would be substantially augmented if AGT could be selectively inhibited in tumor tissue, without a corresponding depletion in normal tissue. We report the synthesis of a new AGT inhibitor (5c) which selectively depletes AGT in hypoxic tumor cells.
Collapse
Affiliation(s)
- Rui Zhu
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Raymond P. Baumann
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Philip G. Penketh
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Krishnamurthy Shyam
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Alan C. Sartorelli
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
8
|
Kim EY, Liu Y, Akintujoye OM, Shyam K, Grove TA, Sartorelli AC, Rockwell S. Preliminary studies with a new hypoxia-selective cytotoxin, KS119W, in vitro and in vivo. Radiat Res 2012; 178:126-37. [PMID: 22862779 DOI: 10.1667/rr2934.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Agents with selective toxicity to hypoxic cells have shown promise as adjuncts to radiotherapy. Our previous studies showed that the bioreductive alkylating agent KS119 had an extremely large differential toxicity to severely hypoxic and aerobic cells in cell culture, and was effective in killing the hypoxic cells of EMT6 mouse mammary tumors in vivo. However, the limited solubility of that compound precluded its development as an anticancer drug. Here we report our initial studies with KS119W, a water-soluble analog of KS119. The cytotoxicity of KS119W to EMT6 cells in vitro was similar to that of KS119, with both agents producing only minimal cytotoxicity to aerobic cells even after intensive treatments, while producing pronounced cytotoxicity to oxygen-deficient cells. This resulted in large differentials in the toxicities to hypoxic and aerobic cells (>1,000-fold at 10 μM). Low pH had only minimal effects on the cytotoxicity of KS119W. Under hypoxic conditions, EMT6 cells transfected to express high levels of either human or mouse versions of the repair protein O(6)-alkylguanine-DNA alkyltransferase, which is also known as O(6)-methylguanine DNA-methyltransferase, were much more resistant to KS119W than parental EMT6 cells lacking O(6)-alkylguanine-DNA alkyltransferase, confirming the importance of DNA O-6-alkylation to the cytotoxicity of this agent. Studies with EMT6 tumors in BALB/c Rw mice using both tumor cell survival and tumor growth delay assays showed that KS119W was effective as an adjunct to irradiation for the treatment of solid tumors in vivo, producing additive or supra-additive effects in most combination regimens for which the interactions could be evaluated. Our findings encourage additional preclinical studies to examine further the antineoplastic effects of KS119W alone and in combination with radiation, and to examine the pharmacology and toxicology of this new bioreductive alkylating agent so that its potential for clinical use as an adjuvant to radiotherapy can be evaluated.
Collapse
Affiliation(s)
- Eugene Y Kim
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut 06520-8040, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Penketh PG, Shyam K, Baumann RP, Ishiguro K, Patridge EV, Zhu R, Sartorelli AC. A strategy for selective O(6)-alkylguanine-DNA alkyltransferase depletion under hypoxic conditions. Chem Biol Drug Des 2012; 80:279-90. [PMID: 22553921 DOI: 10.1111/j.1747-0285.2012.01401.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cellular resistance to chemotherapeutics that alkylate the O-6 position of guanine residues in DNA correlates with their O(6)-alkylguanine-DNA alkyltransferase activity. In normal cells high [O(6)-alkylguanine-DNA alkyltransferase] is beneficial, sparing the host from toxicity, whereas in tumor cells high [O(6)-alkylguanine-DNA alkyltransferase] prevents chemotherapeutic response. Therefore, it is necessary to selectively inactivate O(6)-alkylguanine-DNA alkyltransferase in tumors. The oxygen-deficient compartment unique to solid tumors is conducive to reduction, and could be utilized to provide this selectivity. Therefore, we synthesized 2-nitro-6-benzyloxypurine, an analog of O(6)-benzylguanine in which the essential 2-amino group is replaced by a nitro moiety, and 2-nitro-6-benzyloxypurine is >2000-fold weaker than O(6)-benzylguanine as an O(6)-alkylguanine-DNA alkyltransferase inhibitor. We demonstrate oxygen concentration sensitive net reduction of 2-nitro-6-benzyloxypurine by cytochrome P450 reductase, xanthine oxidase, and EMT6, DU145, and HL-60 cells to yield O(6)-benzylguanine. We show that 2-nitro-6-benzyloxypurine treatment depletes O(6)-alkylguanine-DNA alkyltransferase in intact cells under oxygen-deficient conditions and selectively sensitizes cells to laromustine (an agent that chloroethylates the O-6 position of guanine) under oxygen-deficient but not normoxic conditions. 2-Nitro-6-benzyloxypurine represents a proof of concept lead compound; however, its facile reduction (E(1/2) - 177 mV versus Ag/AgCl) may result in excessive oxidative stress and/or the generation of O(6)-alkylguanine-DNA alkyltransferase inhibitors in normoxic regions in vivo.
Collapse
Affiliation(s)
- Philip G Penketh
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | | | | | | | | | |
Collapse
|
10
|
Nakatake M, Monte-Mor B, Debili N, Casadevall N, Ribrag V, Solary E, Vainchenker W, Plo I. JAK2(V617F) negatively regulates p53 stabilization by enhancing MDM2 via La expression in myeloproliferative neoplasms. Oncogene 2012; 31:1323-33. [PMID: 21785463 DOI: 10.1038/onc.2011.313] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 01/10/2023]
Abstract
JAK2(V617F) is a gain of function mutation that promotes cytokine-independent growth of myeloid cells and accounts for a majority of myeloproliferative neoplasms (MPN). Mutations in p53 are rarely found in these diseases before acute leukemia transformation, but this does not rule out a role for p53 deregulation in disease progression. Using Ba/F3-EPOR cells and ex vivo cultured CD34(+) cells from MPN patients, we demonstrate that expression of JAK2(V617F) affected the p53 response to DNA damage. We show that E3 ubiquitin ligase MDM2 accumulated in these cells, due to an increased translation of MDM2 mRNA. Accumulation of the La autoantigen, which interacts with MDM2 mRNA and promotes its translation, was responsible for the increase in MDM2 protein level and the subsequent degradation of p53 after DNA damage. Downregulation of La protein or cell treatment with nutlin-3, a MDM2 antagonist, restored the p53 response to DNA damage and the cytokine-dependence of Ba/F3-EPOR-JAK2(V617F) cells. Altogether, these data indicate that the JAK2(V617F) mutation affects p53 response to DNA damage through the upregulation of La antigen and accumulation of MDM2. They also suggest that p53 functional inactivation accounts for the cytokine hypersensitivity of JAK2(V617F) MPN and might have a role in disease progression.
Collapse
|
11
|
Li X, Qian S, Zheng L, Yang B, He Q, Hu Y. A mechanism-based fluorescent probe for labeling O6-methylguanine-DNA methyltransferase in live cells. Org Biomol Chem 2012; 10:3189-91. [DOI: 10.1039/c2ob25231g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
12
|
Rockwell S, Liu Y, Seow HA, Ishiguro K, Baumann RP, Penketh PG, Shyam K, Akintujoye OM, Glazer PM, Sartorelli AC. Preclinical evaluation of Laromustine for use in combination with radiation therapy in the treatment of solid tumors. Int J Radiat Biol 2011; 88:277-85. [PMID: 22111842 DOI: 10.3109/09553002.2012.638359] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE These studies explored questions related to the potential use of Laromustine in the treatment of solid tumors and in combination with radiotherapy. MATERIALS AND METHODS The studies used mouse EMT6 cells (both parental and transfected with genes for O(6)-alkylguanine-DNA transferase [AGT]), repair-deficient human Fanconi Anemia C and Chinese hamster VC8 (BRCA2(-/-)) cells and corresponding control cells, and EMT6 tumors in mice assayed using cell survival and tumor growth assays. RESULTS Hypoxia during Laromustine treatment did not protect EMT6 cells or human fibroblasts from this agent. Rapidly proliferating EMT6 cells were more sensitive than quiescent cultures. EMT6 cells expressing mouse or human AGT, which removes O(6)-alkyl groups from DNA guanine, thereby protecting against G-C crosslink formation, increased resistance to Laromustine. Crosslink-repair-deficient Fanconi Anemia C and VC8 cells were hypersensitive to Laromustine, confirming the importance of crosslinks as lethal lesions. In vitro, Laromustine and radiation produced additive toxicities to EMT6 cells. Studies using tumor cell survival and tumor growth assays showed effects of regimens combining Laromustine and radiation that were compatible with additive or subadditive interactions. CONCLUSIONS The effects of Laromustine on solid tumors and with radiation are complex and are influenced by microenvironmental and proliferative heterogeneity within these malignancies.
Collapse
Affiliation(s)
- Sara Rockwell
- Department of Therapeutic Radiology, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06520-8040, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ballot C, Jendoubi M, Kluza J, Jonneaux A, Laine W, Formstecher P, Bailly C, Marchetti P. Regulation by survivin of cancer cell death induced by F14512, a polyamine-containing inhibitor of DNA topoisomerase II. Apoptosis 2011; 17:364-76. [DOI: 10.1007/s10495-011-0681-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
14
|
Zhu R, Liu MC, Luo MZ, Penketh PG, Baumann RP, Shyam K, Sartorelli AC. 4-nitrobenzyloxycarbonyl derivatives of O(6)-benzylguanine as hypoxia-activated prodrug inhibitors of O(6)-alkylguanine-DNA alkyltransferase (AGT), which produces resistance to agents targeting the O-6 position of DNA guanine. J Med Chem 2011; 54:7720-8. [PMID: 21955333 DOI: 10.1021/jm201115f] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A series of 4-nitrobenzyloxycarbonyl prodrug derivatives of O(6)-benzylguanine (O(6)-BG), conceived as prodrugs of O(6)-BG, an inhibitor of the resistance protein O(6)-alkylguanine-DNA alkyltransferase (AGT), were synthesized and evaluated for their ability to undergo bioreductive activation by reductase enzymes under oxygen deficiency. Three agents of this class, 4-nitrobenzyl (6-(benzyloxy)-9H-purin-2-yl)carbamate (1) and its monomethyl (2) and gem-dimethyl analogues (3), were tested for activation by reductase enzyme systems under oxygen deficient conditions. Compound 3, the most water-soluble of these agents, gave the highest yield of O(6)-BG following reduction of the nitro group trigger. Compound 3 was also evaluated for its ability to sensitize 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-[(methylamino)carbonyl]hydrazine (laromustine)-resistant DU145 human prostate carcinoma cells, which express high levels of AGT, to the cytotoxic effects of this agent under normoxic and oxygen deficient conditions. While 3 had little or no effect on laromustine cytotoxicity under aerobic conditions, significant enhancement occurred under oxygen deficiency, providing evidence for the preferential release of the AGT inhibitor O(6)-BG under hypoxia.
Collapse
Affiliation(s)
- Rui Zhu
- Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, New Haven, CT 06520-8066, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Pegg AE. Multifaceted roles of alkyltransferase and related proteins in DNA repair, DNA damage, resistance to chemotherapy, and research tools. Chem Res Toxicol 2011; 24:618-39. [PMID: 21466232 DOI: 10.1021/tx200031q] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
O(6)-Alkylguanine-DNA alkyltransferase (AGT) is a widely distributed, unique DNA repair protein that acts as a single agent to directly remove alkyl groups located on the O(6)-position of guanine from DNA restoring the DNA in one step. The protein acts only once, and its alkylated form is degraded rapidly. It is a major factor in counteracting the mutagenic, carcinogenic, and cytotoxic effects of agents that form such adducts including N-nitroso-compounds and a number of cancer chemotherapeutics. This review describes the structure, function, and mechanism of action of AGTs and of a family of related alkyltransferase-like proteins, which do not act alone to repair O(6)-alkylguanines in DNA but link repair to other pathways. The paradoxical ability of AGTs to stimulate the DNA-damaging ability of dihaloalkanes and other bis-electrophiles via the formation of AGT-DNA cross-links is also described. Other important properties of AGTs include the ability to provide resistance to cancer therapeutic alkylating agents, and the availability of AGT inhibitors such as O(6)-benzylguanine that might overcome this resistance is discussed. Finally, the properties of fusion proteins in which AGT sequences are linked to other proteins are outlined. Such proteins occur naturally, and synthetic variants engineered to react specifically with derivatives of O(6)-benzylguanine are the basis of a valuable research technique for tagging proteins with specific reagents.
Collapse
Affiliation(s)
- Anthony E Pegg
- Department of Cellular and Molecular Physiology, Milton S. Hershey Medical Center, Pennsylvania State University College of Medicine , Pennsylvania 17033, United States.
| |
Collapse
|
16
|
Alvarado Y, Swords R, Kelly KR, Giles FJ. Clinical activity of laromustine (Onrigin™) in hematologic malignancies. Expert Rev Hematol 2011; 2:481-8. [PMID: 21083013 DOI: 10.1586/ehm.09.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Laromustine (Onrigin™), formerly known as Cloretazine(®) (VNP40101M), belongs to a novel class of alkylating agents--the sulfonylhydrazines--and was selected for clinical development based on its broad anti-tumor activity in preclinical models. Laromustine is metabolized to yield 90CE and methylisocyanate, the former rapidly produces an alkylating, chloroethylating species, similar to the chloroethylating species generated by carmustine. However, several features distinguish laromustine from carmustine and possibly account for their biological differences in vitro and in vivo. The chloroethylating species responsible for laromustine's alkylator effect is relatively specific for guanine and forms a crosslink after incorporation into DNA. Laromustine has significant activity in both older patients with previously untreated acute myeloid leukemia or high-risk myelodysplastic syndrome, including those with very poor-risk disease, and in patients with relapsed disease. Further clinical studies are required with laromustine to evaluate its place as an anticancer agent in other hematological malignancies.
Collapse
Affiliation(s)
- Yesid Alvarado
- Institute for Drug Development, Cancer Therapy and Research Center at the UT Health Science Center, University of Texas, San Antonio, TX 78229, USA
| | | | | | | |
Collapse
|
17
|
Ishiguro K, Zhu YL, Shyam K, Penketh PG, Baumann RP, Sartorelli AC. Quantitative relationship between guanine O(6)-alkyl lesions produced by Onrigin™ and tumor resistance by O(6)-alkylguanine-DNA alkyltransferase. Biochem Pharmacol 2010; 80:1317-25. [PMID: 20654586 DOI: 10.1016/j.bcp.2010.07.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 07/07/2010] [Accepted: 07/08/2010] [Indexed: 11/16/2022]
Abstract
O(6)-Alkylguanine-DNA alkyltransferase (AGT) mediates tumor resistance to alkylating agents that generate guanine O(6)-chloroethyl (Onrigin™ and carmustine) and O(6)-methyl (temozolomide) lesions; however, the relative efficiency of AGT protection against these lesions and the degree of resistance to these agents that a given number of AGT molecules produces are unclear. Measured from differential cytotoxicity in AGT-ablated and AGT-intact HL-60 cells containing 17,000 AGT molecules/cell, AGT produced 12- and 24-fold resistance to chloroethylating (90CE) and methylating (KS90) analogs of Onrigin™, respectively. For 50% growth inhibition, KS90 and 90CE generated 5,600 O(6)-methylguanines/cell and ∼300 O(6)-chloroethylguanines/cell, respectively. AGT repaired O(6)-methylguanines until the AGT pool was exhausted, while its repair of O(6)-chloroethylguanines was incomplete due to progression of the lesions to AGT-irreparable interstrand DNA cross-links. Thus, the smaller number of O(6)-chloroethylguanine lesions needed for cytotoxicity accounted for the marked degree of resistance (12-fold) to 90CE produced by AGT. Transfection of human or murine AGT into AGT deficient transplantable tumor cells (i.e., EMT6, M109 and U251) generated transfectants expressing AGT ranging from 4,000 to 700,000 molecules/cell. In vitro growth inhibition assays using these transfectants treated with 90CE revealed that AGT caused a concentration dependent resistance up to a level of ∼10,000 AGT molecules/cell. This finding was corroborated by in vivo studies where expression of 4,000 and 10,000 murine AGT molecules/cell rendered EMT6 tumors partially and completely resistant to Onrigin™, respectively. These studies imply that the antitumor activity of Onrigin™ stems from guanine O(6)-chloroethylation and define the threshold concentration of AGT that negates its antineoplastic activity.
Collapse
Affiliation(s)
- Kimiko Ishiguro
- Department of Pharmacology, Cancer Center, Yale University School of Medicine, New Haven, CT 06520, United States
| | | | | | | | | | | |
Collapse
|
18
|
Ballot C, Kluza J, Martoriati A, Nyman U, Formstecher P, Joseph B, Bailly C, Marchetti P. Essential role of mitochondria in apoptosis of cancer cells induced by the marine alkaloid Lamellarin D. Mol Cancer Ther 2010; 8:3307-17. [PMID: 19952118 DOI: 10.1158/1535-7163.mct-09-0639] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lamellarin D, a potent cytotoxic marine alkaloid, exerts its antitumor action through two complementary pathways: a nuclear route via topoisomerase I inhibition and a mitochondrial targeting. The present study was designed to investigate the contribution of these two pathways for apoptosis in cancer cells. Lamellarin D promoted nuclear apoptosis in leukemia cells without prominent cell cycle arrest. Signals transmitted by lamellarin D initiated apoptosis via the intrinsic apoptotic pathway. The drug induced conformational activation of Bax and decreased the expression levels of antiapoptotic proteins Bcl-2 and cIAP2 in association with activation of caspase-9 and caspase-3. Upon lamellarin D exposure, Fas and Fas-L expression was not modified in leukemia cells. Moreover, leukemia cells deficient in caspase-8 or Fas-associated protein with death domain underwent apoptosis through the typical mitochondrial apoptotic cascade, indicating that cell death induced by lamellarin D was independent of the extrinsic apoptotic pathway. Lamellarin D also exerted a topoisomerase I-mediated DNA damage response resulting in H2AX phosphorylation, and the upregulation of the DNA repair protein Rad51 and of p53, as well as the phosphorylation of p53 at serine 15. However, lamellarin D killed efficiently mutated p53 or p53 null cancer cells, and sensitivity to lamellarin D was abrogated neither by cycloheximide nor in enucleated cells. Lamellarin D-induced cytochrome c release occurs independently of nuclear factors in a cell-free system. These results suggest that lamellarin D exerts its cytotoxic effects primarily by inducing mitochondrial apoptosis independently of nuclear signaling. Thus, lamellarin D constitutes a new proapoptotic agent that may bypass certain forms of apoptosis resistance that occur in tumor cells.
Collapse
Affiliation(s)
- Caroline Ballot
- INSERM U 837 Faculté de médecine, 1, place Verdun F- 59045 Lille Cedex France
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Schiller GJ, O'Brien SM, Pigneux A, DeAngelo DJ, Vey N, Kell J, Solomon S, Stuart RK, Karsten V, Cahill AL, Albitar MX, Giles FJ. Single-Agent Laromustine, A Novel Alkylating Agent, Has Significant Activity in Older Patients With Previously Untreated Poor-Risk Acute Myeloid Leukemia. J Clin Oncol 2010; 28:815-21. [DOI: 10.1200/jco.2009.24.2008] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose An international phase II study of laromustine (VNP40101M), a sulfonylhydrazine alkylating agent, was conducted in patients age 60 years or older with previously untreated poor-risk acute myeloid leukemia (AML). Patients and Methods Laromustine 600 mg/m2 was administered as a single 60-minute intravenous infusion. Patients were age 70 years or older or 60 years or older with at least one additional risk factor—unfavorable AML karyotype, Eastern Cooperative Oncology Group (ECOG) performance status (PS) of 2, and/or cardiac, pulmonary, or hepatic comorbidities. Results Eighty-five patients (median age, 72 years; range, 60 to 87 years) were treated. Poor-risk features included age 70 years or older, 78%; adverse karyotype, 47%; PS of 2, 41%; pulmonary disease, 77%; cardiac disease, 73%; and hepatic disease, 3%. Ninety-six percent of patients had at least two risk factors, and 39% had at least four risk factors. The overall response rate (ORR) was 32%, with 20 patients (23%) achieving complete response (CR) and seven (8%) achieving CR with incomplete platelet recovery (CRp). ORR was 20% in patients with adverse cytogenetics; 32% in those age 70 years or older; 32% in those with PS of 2; 32% in patients with baseline pulmonary dysfunction; 34% in patients with baseline cardiac dysfunction; and 27% in 33 patients with at least four risk factors. Twelve (14%) patients died within 30 days of receiving laromustine therapy. Median overall survival was 3.2 months, with a 1-year survival of 21%; the median duration of survival for those who achieved CR/CRp was 12.4 months, with a 1-year survival of 52%. Conclusion Laromustine has significant single-agent activity in elderly patients with poor-risk AML. Adverse events are predominantly myelosuppressive or respiratory. Response rates are consistent across a spectrum of poor-risk features.
Collapse
Affiliation(s)
- Gary J. Schiller
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Susan M. O'Brien
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Arnaud Pigneux
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Daniel J. DeAngelo
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Norbert Vey
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Jonathan Kell
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Scott Solomon
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Robert K. Stuart
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Verena Karsten
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Ann L. Cahill
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Maher X. Albitar
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| | - Francis J. Giles
- From the David Geffen School of Medicine at the University of California at Los Angeles, Los Angeles; Quest Diagnostics Nichols Institute, San Juan Capistrano, CA; The University of Texas M. D. Anderson Cancer Center, Houston; Cancer Therapy and Research Center at The University of Texas Health Science Center, San Antonio, TX; Hopital Haut Leveque, Bordeaux; Institut Paoli-Calmettes, Marseille, France; Dana-Farber Cancer Institute, Boston, MA; University Hospital of Wales, Cardiff, UK; Northside Hospital
| |
Collapse
|
20
|
Phase 3 randomized, placebo-controlled, double-blind study of high-dose continuous infusion cytarabine alone or with laromustine (VNP40101M) in patients with acute myeloid leukemia in first relapse. Blood 2009; 114:4027-33. [PMID: 19710500 DOI: 10.1182/blood-2009-06-229351] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Laromustine is a sulfonylhdrazine alkylator with significant antileukemia activity. An international, randomized (2:1), double-blind, placebo-controlled study was conducted to compare complete remission (CR) rates and overall survival (OS) in patients with first relapse acute myeloid leukemia (AML) treated with laromustine and high-dose cytarabine (HDAC) versus HDAC/placebo. Patients received 1.5 g/m(2) per day cytarabine continuous infusion for 3 days and laromustine 600 mg/m(2) (n = 177) or placebo (n = 86) on day 2. Patients in CR received consolidation with laromustine/HDAC or HDAC/placebo as per initial randomization. After interim analysis at 50% enrollment, the Data Safety Monitoring Board (DSMB) expressed concern that any advantage in CR would be compromised by the observed on-study mortality, and enrollment was held. The CR rate was significantly higher for the laromustine/HDAC group (35% vs 19%, P = .005). However, the 30-day mortality rate and median progression-free survival were significantly worse in this group compared with HDAC/placebo (11% vs 2%; P = .016; 54 days vs 34; P = .002). OS and median response durations were similar in both groups. Laromustine/HDAC induced significantly more CR than HDAC/placebo, but OS was not improved due to mortality associated with myelosuppression and its sequelae. The DSMB subsequently approved a revised protocol with laromustine dose reduction and recombinant growth factor support. The study was registered as NCT00112554 at http://www.clinicaltrials.gov.
Collapse
|
21
|
Giles FJ. Bendamustine and cloretazine: Alkylators with sharply contrasting activity in AML. Leuk Lymphoma 2009; 48:1064-6. [PMID: 17577766 DOI: 10.1080/10428190701332464] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Francis J Giles
- Division of Hematology and Medical Oncology, San Antonio, TX 78229, USA.
| |
Collapse
|
22
|
Inhibition of human DNA polymerase beta activity by the anticancer prodrug Cloretazine. Biochem Biophys Res Commun 2008; 378:419-23. [PMID: 19026985 DOI: 10.1016/j.bbrc.2008.11.042] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 11/11/2008] [Indexed: 11/18/2022]
Abstract
The antineoplastic prodrug Cloretazine exerts its cytotoxicity via a synergism between 2-chloroethylating and carbamoylating activities that are cogenerated upon activation in situ. Cloretazine is reported here to inhibit the nucleotidyl-transferase activity of purified human DNA polymerase beta (Pol beta), a principal enzyme of DNA base excision repair (BER). The 2-chloroethylating activity of Cloretazine alkylates DNA at the O(6) position of guanine bases resulting in 2-chloroethoxyguanine monoadducts, which further react to form cytotoxic interstrand DNA crosslinks. Alkylated DNA is often repaired via BER in vivo. Inhibition of the polymerase activity of Pol beta may account for some of the synergism between Cloretazine's two reactive subspecies in cytotoxicity assays. This inhibition was only observed using agents with carbamoylating activity. Furthermore, while therapeutically relevant concentrations of Cloretazine inhibited the polymerase activity of Pol beta, the enzyme's lyase activity, which may also participate in BER, was not significantly inhibited.
Collapse
|
23
|
Ishiguro K, Shyam K, Penketh PG, Sartorelli AC. Development of an O(6)-alkylguanine-DNA alkyltransferase assay based on covalent transfer of the benzyl moiety from [benzene-3H]O(6)-benzylguanine to the protein. Anal Biochem 2008; 383:44-51. [PMID: 18783719 DOI: 10.1016/j.ab.2008.08.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 08/12/2008] [Accepted: 08/13/2008] [Indexed: 11/15/2022]
Abstract
Although it is known that (i) O(6)-alkylguanine-DNA alkyltransferase (AGT) confers tumor cell resistance to guanine O(6)-targeting drugs such as cloretazine, carmustine, and temozolomide and that (ii) AGT levels in tumors are highly variable, measurement of AGT activity in tumors before treatment is not a routine clinical practice. This derives in part from the lack of a reliable clinical AGT assay; therefore, a simple AGT assay was devised based on transfer of radioactive benzyl residues from [benzene-3H]O(6)-benzylguanine ([3H]BG) to AGT. The assay involves incubation of intact cells or cell homogenates with [3H]BG and measurement of radioactivity in a 70% methanol precipitable fraction. Approximately 85% of AGT in intact cells was recovered in cell homogenates. Accuracy of the AGT assay was confirmed by examination of AGT levels by Western blot analysis with the exception of false-positive results in melanin-containing cells due to [3H]BG binding to melanin. Second-order kinetic constants for human and murine AGT were 1100 and 380 M(-1)s(-1), respectively. AGT levels in various human cell lines ranged from less than 500 molecules/cell (detection limit) to 45,000 molecules/cell. Rodent cell lines frequently lacked AGT expression, and AGT levels in rodent cells were much lower than in human cells.
Collapse
Affiliation(s)
- Kimiko Ishiguro
- Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
24
|
Gururangan S, Turner CD, Stewart CF, O'Shaughnessy M, Kocak M, Poussaint TY, Phillips PC, Goldman S, Packer R, Pollack IF, Blaney SM, Karsten V, Gerson SL, Boyett JM, Friedman HS, Kun LE. Phase I trial of VNP40101M (Cloretazine) in children with recurrent brain tumors: a pediatric brain tumor consortium study. Clin Cancer Res 2008; 14:1124-30. [PMID: 18281546 DOI: 10.1158/1078-0432.ccr-07-4242] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE VNP40101M (Cloretazine), a novel DNA alkylating agent, was evaluated in a phase I study in children with recurrent brain tumors. EXPERIMENTAL DESIGN VNP40101M was given i.v. daily for 5 consecutive days every 6 weeks for up to eight cycles. Dose escalation was done independently in patients stratified based on intensity of prior therapy (moderately pretreated, stratum I; heavily pretreated, stratum II). Correlative studies included pharmacokinetics and measurement of O(6)-alkylguanine-DNA alkyl transferase levels in peripheral blood mononuclear cells before and after treatment. RESULTS Forty-one eligible patients (stratum I, 19; stratum II, 22) were enrolled on this study. The dose-limiting toxicity in 35 evaluable patients was myelosuppression, which occurred in 4 of 16 patients in stratum I and 3 of 19 patients in stratum II. Pharmacokinetic studies showed a median terminal half-life of 30 min (range, 14-39.5). The maximum tolerated dose in stratum I and II were 45 and 30 mg/m(2)/d daily for 5 days every 6 weeks, respectively. Peripheral blood mononuclear cells alkylguanine alkyl transferase levels did not decrease significantly after VNP40101M treatment. Central imaging review confirmed that three patients had stable disease for a median of 45 weeks (range, 37-61+) after therapy. CONCLUSIONS The recommended dose of VNP40101M for phase II studies in children with brain tumors is 45 mg/m(2)/d in moderately pretreated and 30 mg/m(2)/d in heavily pretreated patients when administered for 5 consecutive days every 6 weeks.
Collapse
Affiliation(s)
- Sridharan Gururangan
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Lethality to leukemia cell lines of DNA interstrand cross-links generated by Cloretazine derived alkylating species. Leuk Res 2008; 32:1546-53. [PMID: 18479747 DOI: 10.1016/j.leukres.2008.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 03/04/2008] [Accepted: 03/07/2008] [Indexed: 11/22/2022]
Abstract
Cloretazine [1,2-bis(methylsulfonyl)-1-(2-chloroethyl)-2-[(methylamino)carbonyl]hydrazine; VNP40101M; 101M] is a relatively new prodrug with activity in elderly acute myelogenous leukemia (AML) patients. Its therapeutic action is due largely to the production of 1-(3-cytosinyl),2-(1-guanyl)ethane cross-links (G-C ethane cross-links) in DNA. The numbers of cross-links produced in three experimental leukemia lines (L1210, U937 and HL-60) were fewer than 10 per genome at their respective LC50 concentrations. Only 1 in approximately 20,000 90CE molecules produces a cross-link in the AGT (O6-alkylguanine-DNA alkyltransferase) negative L1210 and U937 cell lines and 1 in 400,000 in the AGT positive HL-60 cell line.
Collapse
|
26
|
Giles F, Rizzieri D, Karp J, Vey N, Ravandi F, Faderl S, Khan KD, Verhoef G, Wijermans P, Advani A, Roboz G, Kantarjian H, Bilgrami SFA, Ferrant A, Daenen SMGJ, Karsten V, Cahill A, Albitar M, Mufti G, O'Brien S. Cloretazine (VNP40101M), a Novel Sulfonylhydrazine Alkylating Agent, in Patients Age 60 Years or Older With Previously Untreated Acute Myeloid Leukemia. J Clin Oncol 2007; 25:25-31. [PMID: 17146105 DOI: 10.1200/jco.2006.07.0961] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposeCloretazine (VNP40101M) is a sulfonylhydrazine alkylating agent with significant antileukemia activity. A multicenter phase II study of cloretazine was conducted in patients 60 years of age or older with previously untreated acute myeloid leukemia (AML) or high-risk myelodysplastic syndrome (MDS).Patients and MethodsCloretazine 600 mg/m2was administered as a single intravenous infusion. Patients were stratified by age, performance score, cytogenetic risk category, type of AML, and comorbidity.ResultsOne hundred four patients, median age 72 years (range, 60 to 84 years), were treated on study. Performance status was 2 in 31 patients (30%) and no patient had a favorable karyotype. Forty-seven patients (45%) had cardiac disease, 25 patients (24%) had hepatic disease, and 19 patients (18%) had pulmonary disease, defined as per the Hematopoietic Cell Transplantation–Specific Comorbidity Index, at study entry. The overall response rate was 32%, with 29 patients (28%) achieving complete response (CR) and four patients (4%) achieving CR with incomplete platelet recovery. Response rates in 44 de novo AML patients, 45 secondary AML patients, and 15 high-risk MDS patients were 50%, 11%, and 40%, respectively. Response by cytogenetic risk category was 39% in 56 patients with intermediate cytogenetic risk and 24% in 46 patients with unfavorable cytogenetic risk. Nineteen (18%) patients died within 30 days of receiving cloretazine therapy. Median overall survival was 94 days, with a 1-year survival of 14%; the median duration of survival was 147 days, with a 1-year survival of 28% for those who achieved CR.ConclusionCloretazine has significant activity and modest extramedullary toxicity in elderly patients with AML or high-risk MDS. Response rates remain consistent despite increasing age and comorbidity.
Collapse
Affiliation(s)
- Francis Giles
- The University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ishiguro K, Seow HA, Penketh PG, Shyam K, Sartorelli AC. Mode of action of the chloroethylating and carbamoylating moieties of the prodrug cloretazine. Mol Cancer Ther 2006; 5:969-76. [PMID: 16648568 PMCID: PMC2680221 DOI: 10.1158/1535-7163.mct-05-0532] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cloretazine is an antitumor sulfonylhydrazine prodrug that generates both chloroethylating and carbamoylating species. The cytotoxic potency of these species was analyzed in L1210 leukemia cells using analogues with chloroethylating or carbamoylating function only. Clonogenic assays showed that the chloroethylating-only agent 1,2-bis(methylsulfonyl)-1-(2-chloroethyl)hydrazine (90CE) produced marked differential cytotoxicity against wild-type and O6-alkylguanine-DNA alkyltransferase-transfected L1210 cells (LC10, 1.4 versus 31 micromol/L), indicating that a large portion of the cytotoxicity was due to alkylation of DNA at the O-6 position of guanine. Consistent with the concept that O-6 chloroethylation of DNA guanine progresses to interstrand cross-links, the comet assay, in which DNA cross-links were measured by a reduction in DNA migration induced by strand breaks, showed that cloretazine and 90CE, but not the carbamoylating-only agent 1,2-bis(methylsulfonyl)-1-[(methylamino)carbonyl]hydrazine (101MDCE), produced DNA cross-links and that cloretazine caused more DNA cross-links than 90CE at equimolar concentrations. Cell cycle analyses showed that 90CE and 101MDCE at concentrations of 5 and 80 micromol/L, respectively, produced similar degrees of G2-M arrest. 90CE produced selective inhibition of DNA synthesis after overnight incubation, whereas 101MDCE caused rapid and nonselective inhibition of RNA, DNA, and protein syntheses. Both 90CE and 101MDCE induced phosphorylation of histone H2AX, albeit with distinct kinetics. These results indicate that (a) differential expression of O6-alkylguanine-DNA alkyltransferase in tumor and host cells seems to be responsible for tumor selectivity exerted by cloretazine; (b) 101MDCE enhances DNA cross-linking activity; and (c) 90CE induces cell death at concentrations lower than those causing alterations in the cell cycle and macromolecular syntheses.
Collapse
Affiliation(s)
- Kimiko Ishiguro
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
28
|
Giles F, Verstovsek S, Faderl S, Vey N, Karp J, Roboz G, Khan KD, Cooper M, Bilgrami SFA, Ferrant A, Daenen S, Karsten V, Cahill A, Albitar M, Kantarjian H, O'Brien S, Feldman E. A phase II study of cloretazine (VNP40101M), a novel sulfonylhydrazine alkylating agent, in patients with very high risk relapsed acute myeloid leukemia. Leuk Res 2006; 30:1591-5. [PMID: 16574225 DOI: 10.1016/j.leukres.2006.02.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2006] [Revised: 02/10/2006] [Accepted: 02/14/2006] [Indexed: 10/24/2022]
Abstract
Cloretazine (VNP40101M) is a sulfonylhydrazine alkylating agent with significant anti-leukemia activity. A multicenter phase II study of cloretazine was conducted in patients with first relapse of acute myeloid leukemia (AML) following an initial complete remission (CR) of less than 12 months. Cloretazine was given as a single intravenous infusion at a dose of 600 mg/m(2). Fifty-three patients (median age 62 years (18-84), 41 of 44 (93%) evaluable with intermediate or high risk cytogenetics, 32 (60%) with initial CR durations < or =6 months) were treated on study. Two patients (4%) achieved a second CR. Five (9%) patients died within 30 days of receiving cloretazine therapy. Median overall survival (2.3 months) in the study cohort was directly comparable to that of 233 matched patients treated with other single agents. The study cloretazine regimen had minimal activity in a very high risk subset of patients with relapsed AML.
Collapse
|