1
|
Czemerska M, Robak T, Wierzbowska A. The efficacy of sapacitabine in treating patients with acute myeloid leukemia. Expert Opin Pharmacother 2018; 19:1835-1839. [PMID: 30222471 DOI: 10.1080/14656566.2018.1524875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) remains a poor prognosis hematological malignancy. The introduction of aggressive chemotherapy with allogeneic stem cell transplantation has resulted in improved clinical outcomes in younger patients. However, the treatment results in unfit elderly AML population remain disappointing. New strategies should be introduced to improve the prognosis in this group of patients. Areas covered: This review presents and discusses the mechanism of action, safety and efficacy of sapacitabine in AML patients. Expert opinion: Sapacitabine, a novel nucleoside analog, seemed to be a promising new agent for AML treatment. Its oral bioavailability and tolerable toxicity profile allow the drug to be used in an outpatient setting, especially in elderly unfit patients. Sapacitabine is known to have antileukemic activity in randomized clinical trials. In AML patients, sapacitabine monotherapy offered no advantage over low-intensity cytarabine treatment, and the combination of sapacitabine with decitabine was not significantly more effective than decitabine alone. However, the oral administration of sapacitabine allows it to be used in AML maintenance therapy.
Collapse
Affiliation(s)
- Magdalena Czemerska
- a Department of Hematology , Medical University of Lodz and Copernicus Memorial Hospital , Lodz , Poland
| | - Tadeusz Robak
- a Department of Hematology , Medical University of Lodz and Copernicus Memorial Hospital , Lodz , Poland
| | - Agnieszka Wierzbowska
- a Department of Hematology , Medical University of Lodz and Copernicus Memorial Hospital , Lodz , Poland
| |
Collapse
|
2
|
Two phase I, pharmacokinetic, and pharmacodynamic studies of DFP-10917, a novel nucleoside analog with 14-day and 7-day continuous infusion schedules. Invest New Drugs 2018; 37:76-86. [PMID: 29667134 DOI: 10.1007/s10637-018-0602-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/05/2018] [Indexed: 10/17/2022]
Abstract
Purpose DFP-10917 is a novel deoxycytidine analog with a unique mechanism of action. Brief exposure to high concentrations of DFP-10917 inhibits DNA polymerase resulting in S-phase arrest, while prolonged exposure to DFP-10917 at low concentration causes DNA fragmentation, G2/M-phase arrest, and apoptosis. DFP-10917 demonstrated activity in tumor xenografts resistant to other deoxycytidine analogs. Experimental design Two phase I studies assessed the safety, pharmacokinetic, pharmacodynamic and preliminary efficacy of DFP-10917. Patients with refractory solid tumors received DFP-10917 continuous infusion 14-day on/7-day off and 7-day on/7-day off. Enrollment required age > 18 years, ECOG Performance Status 0-2 and adequate organ function. Results 29 patients were dosed in both studies. In 14-day infusion, dose-limiting toxicities (DLT) consisting of febrile neutropenia and thrombocytopenia occurred at 4.0 mg/m2/day. At 3.0 mg/m2/day, 3 patients experienced neutropenia in cycle 2. The dose of 2.0 mg/m2/day was well tolerated in 6 patients. In 7-day infusion, grade 4 neutropenia was DLT at 4.0 mg/m2/day. The maximum tolerated dose was 3 mg/m2/day. Other toxicities included nausea, vomiting, diarrhea, neutropenia, and alopecia. Eight patients had stable disease for >12 weeks. Paired comet assays performed for 7 patients showed an increase in DNA strand breaks at day 8. Pharmacokinetic data showed dose-proportionality for steady-state concentration and AUC of DFP-10917 and its primary metabolite. Conclusion Continuous infusion of DFP-10917 is feasible and well tolerated with myelosuppression as main DLT. The recommended doses are 2.0 mg/m2/day and 3.0 mg/m2/day on the 14-day and 7-day continuous infusion schedules, respectively. Preliminary activity was suggested. Pharmacodynamic data demonstrate biological activity at the tested doses.
Collapse
|
3
|
Mccloskey J, Koprivnikar J, Faderl S. Sapacitabine in acute myelogenous leukemia. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1100992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4
|
Rashid M, Husain A, Shaharyar M, Mishra R, Hussain A, Afzal O. Design and synthesis of pyrimidine molecules endowed with thiazolidin-4-one as new anticancer agents. Eur J Med Chem 2014; 83:630-45. [PMID: 25010935 DOI: 10.1016/j.ejmech.2014.06.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 06/10/2014] [Accepted: 06/16/2014] [Indexed: 12/15/2022]
Abstract
Design and synthesis of new pyrimidine derivatives clubbed with thiazolidin-4-one from 4-(2-chlorophenyl)-6-(2,4-dichlorophenyl)pyrimidin-2-amine and their in vitro anticancer activities were screened at National Cancer Institute (NCI), USA against full NCI 60 cell lines. Compound 2 (NSC: 765735) exhibited remarkable growth inhibition at single dose (10 μM) and encourage chosen for broadcast at 10-fold dilutions of five different concentrations (0.01, 0.1, 1, 10 and 100 μM). The compound 2 was found better quality for Lung cancer cell line (HOP-92) by viewing growth inhibition (GI50 0.52) and no cytotoxicity seen (LC50 > 100). Molecular docking study was performed using Maestro 9.0 (Schrodinger Inc. USA) to provide binding mode into binding sites of CDK2. Compound 2 could be used as a lead compound for developing new potential anticancer agents.
Collapse
Affiliation(s)
- Mohd Rashid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India.
| | - Asif Husain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India.
| | - Mohammad Shaharyar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Ravinesh Mishra
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Afzal Hussain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), Hamdard Nagar, New Delhi 110062, India
| |
Collapse
|
5
|
Obata T, Suzuki Y, Ogawa N, Kurimoto I, Yamamoto H, Furuno T, Sasaki T, Tanaka M. Improvement of the Antitumor Activity of Poorly Soluble Sapacitabine (CS-682) by Using Soluplus® as a Surfactant. Biol Pharm Bull 2014; 37:802-7. [DOI: 10.1248/bpb.b13-00898] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tohru Obata
- Department of Bioorganic Chemistry, School of Pharmacy, Aichi Gakuin University
| | - Yuka Suzuki
- Department of Bioorganic Chemistry, School of Pharmacy, Aichi Gakuin University
| | - Noriko Ogawa
- Department of Pharmaceutical Engineering, School of Pharmacy, Aichi Gakuin University
| | - Ippei Kurimoto
- Department of Pharmaceutical Engineering, School of Pharmacy, Aichi Gakuin University
| | - Hiromitsu Yamamoto
- Department of Pharmaceutical Engineering, School of Pharmacy, Aichi Gakuin University
| | - Tadahide Furuno
- Department of Analytical Chemistry and Biophysics, School of Pharmacy, Aichi Gakuin University
| | - Takuma Sasaki
- Department of Bioorganic Chemistry, School of Pharmacy, Aichi Gakuin University
| | - Motohiro Tanaka
- Department of Bioorganic Chemistry, School of Pharmacy, Aichi Gakuin University
| |
Collapse
|
6
|
Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. Nat Rev Drug Discov 2013; 12:447-64. [PMID: 23722347 DOI: 10.1038/nrd4010] [Citation(s) in RCA: 823] [Impact Index Per Article: 74.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nucleoside analogues have been in clinical use for almost 50 years and have become cornerstones of treatment for patients with cancer or viral infections. The approval of several additional drugs over the past decade demonstrates that this family still possesses strong potential. Here, we review new nucleoside analogues and associated compounds that are currently in preclinical or clinical development for the treatment of cancer and viral infections, and that aim to provide increased response rates and reduced side effects. We also highlight the different approaches used in the development of these drugs and the potential of personalized therapy.
Collapse
|
7
|
Advances in the development of nucleoside and nucleotide analogues for cancer and viral diseases. NATURE REVIEWS. DRUG DISCOVERY 2013. [PMID: 23722347 DOI: 10.1038/nrd4010]] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nucleoside analogues have been in clinical use for almost 50 years and have become cornerstones of treatment for patients with cancer or viral infections. The approval of several additional drugs over the past decade demonstrates that this family still possesses strong potential. Here, we review new nucleoside analogues and associated compounds that are currently in preclinical or clinical development for the treatment of cancer and viral infections, and that aim to provide increased response rates and reduced side effects. We also highlight the different approaches used in the development of these drugs and the potential of personalized therapy.
Collapse
|
8
|
Bone Marrow and Peripheral Blood AML Cells Are Highly Sensitive to CNDAC, the Active Form of Sapacitabine. Adv Hematol 2012; 2012:727683. [PMID: 23049558 PMCID: PMC3461608 DOI: 10.1155/2012/727683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 08/04/2012] [Indexed: 11/17/2022] Open
Abstract
Achieving improvements in survival and reducing relapse remains a challenge in acute myelogenous leukemia (AML) patients. This study evaluated the in vitro efficacy of the active form of novel agent sapacitabine, CNDAC, compared to current chemotherapeutic drugs Ara-C and mitoxantrone using two AML cell lines, HL-60 (promyelocytic) and THP-1 (monocytic), as well as bone marrow (BM) and peripheral blood (PB) cells collected from AML patients. Cell lines were exposed to compound for 3-6 days and primary cells for 4 days. The viability of primary cells was additionally evaluated 3, 7, and 31 days after removal of tested compound to determine the durability of the response. Our studies indicate that CNDAC and mitoxantrone have a greater impact on viability than ara-C in primary AML cells and AML cell lines. CNDAC is more effective at reducing viability and inducing apoptosis than ara-C at equivalent concentrations in the THP-1 cell line, which is defined as displaying resistance to ara-C. As sapacitabine has shown in vivo activity at clinically achievable doses, future studies are warranted to assess the potential for combining it with ara-C and/or mitoxantrone, with an emphasis on cells and patients insensitive to ara-C treatment.
Collapse
|
9
|
Liu XJ, Nowak B, Wang YQ, Plunkett W. Sapacitabine, the prodrug of CNDAC, is a nucleoside analog with a unique action mechanism of inducing DNA strand breaks. CHINESE JOURNAL OF CANCER 2012; 31:373-80. [PMID: 22739266 PMCID: PMC3777512 DOI: 10.5732/cjc.012.10077] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/16/2012] [Accepted: 05/24/2012] [Indexed: 01/05/2023]
Abstract
Sapacitabine is an orally bioavailable prodrug of the nucleoside analog 2'-C-cyano-2'-deoxy-1-β-D-arabino-pentofuranosylcytosine (CNDAC). Both the prodrug and active metabolite are in clinical trials for hematologic malignancies and/or solid tumors. CNDAC has a unique mechanism of action: after incorporation into DNA, it induces single-strand breaks (SSBs) that are converted into double-strand breaks (DSBs) when cells go through a second S phase. In our previous studies, we demonstrated that CNDAC-induced SSBs can be repaired by the transcription-coupled nucleotide excision repair pathway, whereas lethal DSBs are mainly repaired through homologous recombination. In the current work, we used clonogenic assays to compare the DNA damage repair mechanism of CNDAC with two other deoxycytidine analogs: cytarabine, which is used in hematologic malignacies, and gemcitabine, which shows activity in solid tumors. Deficiency in two Rad51 paralogs, Rad51D and XRCC3, greatly sensitized cells to CNDAC, but not to cytarabine or gemcitabine, indicating that homologous recombination is not a major mechanism for repairing damage caused by the latter two analogs. This study further suggests clinical activity and application of sapacitabine that is distinct from that of cytarabine or gemcitabine.
Collapse
Affiliation(s)
- Xiao-Jun Liu
- Department of Experimental Therapeutics, University of Texas M. D. Anderson Cancer Center, Houston, TX 77054, USA
| | | | | | | |
Collapse
|
10
|
Abstract
INTRODUCTION Sapacitabine is an orally bioavailable nucleoside analog prodrug that is in clinical trials for hematologic malignancies and solid tumors. The active metabolite of sapacitabine, CNDAC (2'-C-cyano-2'-deoxy-1-β-D-arabino-pentofuranosylcytosine), exhibits the unique mechanism of action of causing single-strand breaks (SSBs) after incorporation into DNA, which are converted into double-strand breaks (DSBs) when cells enter a second S-phase. CNDAC-induced DSBs are predominantly repaired through homologous recombination (HR). Cells deficient in HR components are greatly sensitized to CNDAC. Therefore, sapacitabine could be specifically effective against tumors that are deficient in this repair pathway. AREAS COVERED This review summarizes results from supporting evidence for the mechanisms of action of sapacitabine, its preclinical activities and the current results of clinical trials in a variety of cancers. The novel action mechanism of sapacitabine is discussed, with a view to validate it as a chemotherapeutic drug targeting malignancies with defects in HR. EXPERT OPINION Knowledge of CNDAC mechanism identifies tumors that may be sensitized to sapacitabine, thus enabling a personalized treatment strategy. It also creates the opportunity to overcome resistance to current front-line therapies and identify synergistic interactions with known anticancer drugs. The results of such investigations may provide rationales for the design of sapacitabine-based clinical trials.
Collapse
Affiliation(s)
- Xiaojun Liu
- The University of Texas M.D. Anderson Cancer Center, Department of Experimental Therapeutics, Houston, TX, USA
| | - Hagop Kantarjian
- The University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - William Plunkett
- The University of Texas M.D. Anderson Cancer Center, Department of Experimental Therapeutics, Houston, TX, USA
- The University of Texas M.D. Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| |
Collapse
|
11
|
Su J, Chen X, Li Q, Yu Z, Guan X, Geng L, Bi K. Pharmacokinetics Study of the Antitumor Drug CYC-116 in Rat Plasma by Using LC–MS Analysis. Chromatographia 2012. [DOI: 10.1007/s10337-011-2175-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
12
|
Abstract
Hypomethylating agents such as 5-azacytidine or decitabine have been a major breakthrough in the treatment of patients with myelodysplastic syndromes (MDS). They have been shown to improve transfusion requirements and to change the natural history of the disease. However, with increasing cumulative clinical experience, it has become apparent that these agents are not curative and have their own shortcomings. There is a subgroup of patients who do not respond to frontline therapy and a large, growing cohort of patients that lose response or progress while on hypomethylating agent-based therapy. There are no standard treatment options in this arena and it is therefore a focus of significant research interest. Since the mechanisms of resistance to hypomethylating agents are not known, selection of therapy is largely empiric but must take into account the age, comorbidities, and performance status of the patient, as well as the characteristics of the disease at the time of treatment failure. Higher intensity approaches and allogeneic stem cell transplant can yield improved response rates and long-term disease control but should be limited to a selected cohort of patients who can tolerate the treatment-related morbidities. For the majority of patients who likely will be better candidates for lower intensity therapy, several novel, investigational approaches are becoming available. Among these are newer nucleoside analogues, inhibitors of protein tyrosine kinases, molecules that interact with redox signaling within the cell, immunotherapy approaches, and others. Patients with MDS whose disease has failed to respond to hypomethylating agent therapy should be referred for clinical trials when available. As we learn more about the patterns and mechanisms of failure, the next challenge will be to determine which therapies are suitable for each individual patient.
Collapse
Affiliation(s)
- Tapan M Kadia
- Department of Leukemia, University of Texas M.D. Anderson Cancer Center, Houston, USA.
| | | | | |
Collapse
|
13
|
Green SR, Choudhary AK, Fleming IN. Combination of sapacitabine and HDAC inhibitors stimulates cell death in AML and other tumour types. Br J Cancer 2010; 103:1391-9. [PMID: 20924380 PMCID: PMC2990608 DOI: 10.1038/sj.bjc.6605922] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Alternative treatments are needed for elderly patients with acute myeloid leukaemia, as the disease prognosis is poor and the current treatment is unsuitable for many patients. Methods: In this study, we investigated whether combining the nucleoside analogue sapacitabine with histone deacetylase (HDAC) inhibitors could be an effective treatment. Synergy and mode-of-action analysis were studied in cultured cell lines and the efficacy of the combination was confirmed in a xenograft model. Results: CNDAC (1-(2-C-cyano-2-deoxy-β-D-arabino-pentofuranosyl)-cytosine), the active component of sapacitabine, synergised with vorinostat in cell lines derived from a range of tumour types. Synergy was not dependent on a specific sequence of drug administration and was also observed when CNDAC was combined with an alternative HDAC inhibitor, valproate. Flow cytometry and western blot analysis confirmed that the combination induced a significant increase in apoptosis. Mode-of-action analysis detected changes in Bcl-xl, Mcl-1, Noxa, Bid and Bim, which are all regulators of the apoptotic process. The sapacitabine/vorinostat combination demonstrated significant benefit compared with the single-agent treatments in an MV4-11 xenograft, in the absence of any observed toxicity. Conclusion: Sapacitabine and HDAC inhibitors are an effective drug combination that is worthy of clinical exploration.
Collapse
Affiliation(s)
- S R Green
- Cyclacel Ltd., 1 James Lindsay Place, Dundee DD1 5JJ, UK.
| | | | | |
Collapse
|
14
|
Kantarjian H, Garcia-Manero G, O'Brien S, Faderl S, Ravandi F, Westwood R, Green SR, Chiao JH, Boone PA, Cortes J, Plunkett W. Phase I clinical and pharmacokinetic study of oral sapacitabine in patients with acute leukemia and myelodysplastic syndrome. J Clin Oncol 2010; 28:285-91. [PMID: 19933907 PMCID: PMC3340938 DOI: 10.1200/jco.2009.25.0209] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
PURPOSE Sapacitabine is an oral deoxycytidine nucleoside analog with a unique mechanism of action that is different from cytarabine. PATIENTS AND METHODS To define the dose-limiting toxicities (DLT) and maximum-tolerated dose (MTD) of sapacitabine given orally twice daily for 7 days every 3 to 4 weeks, or twice daily for 3 days for 2 weeks (days 1 through 3 and days 8 through 10) every 3 to 4 weeks, in refractory-relapse acute leukemia and myelodysplastic syndrome (MDS). A total of 47 patients were treated in the phase I study that used a classical 3 + 3 design. Sapacitabine was escalated from 75 to 375 mg twice daily for 7 days (n = 35) and from 375 to 475 mg twice daily for 3 days on days 1 through 3 and days 8 through 10. RESULTS The DLTs with both schedules were gastrointestinal. The MTDs were 375 mg twice daily for 7 days and 425 mg twice daily for 3 days on days 1 through 3 and days 8 through 10. The recommended phase II single-agent dose schedules were 325 mg twice daily for 7 days and 425 mg twice daily for 3 days on days 1 through 3 and days 8 through 10. Responses were observed in 13 patients (28%); four were complete responses, and nine were marrow complete responses. CONCLUSION Sapacitabine is a new, safely administered, oral deoxycytidine analog that has encouraging activity in leukemia and MDS. Phase II studies are ongoing.
Collapse
Affiliation(s)
- Hagop Kantarjian
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom.,Corresponding author: Hagop Kantarjian, MD, The University of Texas M. D. Anderson Cancer Center, Department of Leukemia, Unit 428, 1515 Holcombe Blvd, Houston, TX 77030; e-mail:
| | - Guillermo Garcia-Manero
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Susan O'Brien
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Stefan Faderl
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Farhad Ravandi
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Robert Westwood
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Simon R. Green
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Judy H. Chiao
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Patricia A. Boone
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - Jorge Cortes
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| | - William Plunkett
- From the Departments of Leukemia and Experimental Therapeutics, The University of Texas M. D. Anderson Cancer Center, Houston, TX; and Cyclacel, Dundee, United Kingdom
| |
Collapse
|
15
|
Parker WB. Enzymology of purine and pyrimidine antimetabolites used in the treatment of cancer. Chem Rev 2009; 109:2880-93. [PMID: 19476376 DOI: 10.1021/cr900028p] [Citation(s) in RCA: 388] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- William B Parker
- Southern Research Institute, 2000 Ninth Avenue, South Birmingham, Alabama 35205, USA.
| |
Collapse
|
16
|
Asai T, Miyazawa S, Maeda N, Hatanaka K, Katanasaka Y, Shimizu K, Shuto S, Oku N. Antineovascular therapy with angiogenic vessel-targeted polyethyleneglycol-shielded liposomal DPP-CNDAC. Cancer Sci 2008; 99:1029-33. [PMID: 18294280 PMCID: PMC11159210 DOI: 10.1111/j.1349-7006.2008.00758.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Revised: 12/29/2007] [Accepted: 01/07/2008] [Indexed: 11/29/2022] Open
Abstract
Causing damage to angiogenic vessels is a promising approach for cancer chemotherapy. The present study is a codification of a designed liposomal drug delivery system (DDS) for antineovascular therapy (ANET) with 2'-C-cyano-2'-deoxy-1-beta-D-arabino-pentofuranosylcytosine (CNDAC). The authors have previously reported that liposomalized 5'-O-dipalmitoylphosphatidyl CNDAC (DPP-CNDAC), a phospholipid derivative of the novel antitumor nucleoside CNDAC, is quite useful for ANET. DPP-CNDAC liposomes modified with APRPG, a peptide having affinity toward angiogenic vessels, efficiently suppressed tumor growth by damaging angiogenic endothelial cells. In the present study, the authors masked the hydrophilic moiety of DPP-CNDAC, namely, CNDAC, on the liposomal surface with APRPG-polyethyleneglycol (PEG) conjugate to improve the availability of DPP-CNDAC liposomes. The use of the APRPG-PEG conjugate attenuated the negative zeta-potential of the DPP-CNDAC liposomes and reduced the agglutinability of them in the presence of serum. These effects improved the blood level of DPP-CNDAC liposomes in colon 26 NL-17 tumor-bearing BALB/c male mice, resulting in enhanced accumulation of them in the tumor. Laser scanning microscopic observations indicated that APRPG-PEG-modified DPP-CNDAC liposomes (LipCNDAC/APRPG-PEG) colocalized with angiogenic vessels and strongly induced apoptosis of tumor cells, whereas PEG-modified DPP-CNDAC liposomes (LipCNDAC/PEG) did not. In fact, LipCNDAC/APRPG-PEG suppressed the tumor growth more strongly compared to LipCNDAC/PEG and increased significantly the life span of the mice. The present study is a good example of an effective liposomal DDS for ANET that is characterized by: (i) phospholipid derivatization of a certain anticancer drug to suit the liposomal formulation; (ii) PEG-shielding for masking undesirable properties of the drug on the liposomal surface; and (iii) active targeting to angiogenic endothelial cells using a specific probe.
Collapse
Affiliation(s)
- Tomohiro Asai
- Department of Medical Biochemistry and Global COE, University of Shizuoka School of Pharmaceutical Sciences, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | | | | | | | | | | | | | | |
Collapse
|