1
|
Scolaro T, Manco M, Pecqueux M, Amorim R, Trotta R, Van Acker HH, Van Haele M, Shirgaonkar N, Naulaerts S, Daniluk J, Prenen F, Varamo C, Ponti D, Doglioni G, Ferreira Campos AM, Fernandez Garcia J, Radenkovic S, Rouhi P, Beatovic A, Wang L, Wang Y, Tzoumpa A, Antoranz A, Sargsian A, Di Matteo M, Berardi E, Goveia J, Ghesquière B, Roskams T, Soenen S, Voets T, Manshian B, Fendt SM, Carmeliet P, Garg AD, DasGupta R, Topal B, Mazzone M. Nucleotide metabolism in cancer cells fuels a UDP-driven macrophage cross-talk, promoting immunosuppression and immunotherapy resistance. NATURE CANCER 2024; 5:1206-1226. [PMID: 38844817 PMCID: PMC11358017 DOI: 10.1038/s43018-024-00771-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 04/23/2024] [Indexed: 08/16/2024]
Abstract
Many individuals with cancer are resistant to immunotherapies. Here, we identify the gene encoding the pyrimidine salvage pathway enzyme cytidine deaminase (CDA) among the top upregulated metabolic genes in several immunotherapy-resistant tumors. We show that CDA in cancer cells contributes to the uridine diphosphate (UDP) pool. Extracellular UDP hijacks immunosuppressive tumor-associated macrophages (TAMs) through its receptor P2Y6. Pharmacologic or genetic inhibition of CDA in cancer cells (or P2Y6 in TAMs) disrupts TAM-mediated immunosuppression, promoting cytotoxic T cell entry and susceptibility to anti-programmed cell death protein 1 (anti-PD-1) treatment in resistant pancreatic ductal adenocarcinoma (PDAC) and melanoma models. Conversely, CDA overexpression in CDA-depleted PDACs or anti-PD-1-responsive colorectal tumors or systemic UDP administration (re)establishes resistance. In individuals with PDAC, high CDA levels in cancer cells correlate with increased TAMs, lower cytotoxic T cells and possibly anti-PD-1 resistance. In a pan-cancer single-cell atlas, CDAhigh cancer cells match with T cell cytotoxicity dysfunction and P2RY6high TAMs. Overall, we suggest CDA and P2Y6 as potential targets for cancer immunotherapy.
Collapse
Affiliation(s)
- Tommaso Scolaro
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marta Manco
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mathieu Pecqueux
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ricardo Amorim
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rosa Trotta
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Heleen H Van Acker
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Matthias Van Haele
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Niranjan Shirgaonkar
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Stefan Naulaerts
- Laboratory for Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jan Daniluk
- Laboratory of Ion Channel Research (LICR), VIB-KU Leuven Centre for Brain & Disease Research, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Fran Prenen
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Chiara Varamo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Donatella Ponti
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Medical-Surgical Sciences and Biotechnologies, University of Rome Sapienza, Latina, Italy
| | - Ginevra Doglioni
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ana Margarida Ferreira Campos
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Juan Fernandez Garcia
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Silvia Radenkovic
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven, Belgium
- Metabolomics Core Facility, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Pegah Rouhi
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Liwei Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Amalia Tzoumpa
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Ara Sargsian
- Translation Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Emanuele Berardi
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jermaine Goveia
- Unicle Biomedical Data Science, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Bart Ghesquière
- Metabolomics Core Facility, Center for Cancer Biology, VIB, Leuven, Belgium
- Metabolomics Core Facility, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tania Roskams
- Department of Imaging and Pathology, Translational Cell and Tissue Research, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Stefaan Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research (LICR), VIB-KU Leuven Centre for Brain & Disease Research, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Bella Manshian
- Translation Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory for Cell Stress & Immunity (CSI), Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ramanuj DasGupta
- Laboratory of Precision Oncology and Cancer Evolution, Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Baki Topal
- Department of Visceral Surgery, KU Leuven and University Hospitals Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Wang F, Liang L, Yu M, Wang W, Badar IH, Bao Y, Zhu K, Li Y, Shafi S, Li D, Diao Y, Efferth T, Xue Z, Hua X. Advances in antitumor activity and mechanism of natural steroidal saponins: A review of advances, challenges, and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155432. [PMID: 38518645 DOI: 10.1016/j.phymed.2024.155432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Cancer, the second leading cause of death worldwide following cardiovascular diseases, presents a formidable challenge in clinical settings due to the extensive toxic side effects associated with primary chemotherapy drugs employed for cancer treatment. Furthermore, the emergence of drug resistance against specific chemotherapeutic agents has further complicated the situation. Consequently, there exists an urgent imperative to investigate novel anticancer drugs. Steroidal saponins, a class of natural compounds, have demonstrated notable antitumor efficacy. Nonetheless, their translation into clinical applications has remained unrealized thus far. In light of this, we conducted a comprehensive systematic review elucidating the antitumor activity, underlying mechanisms, and inherent limitations of steroidal saponins. Additionally, we propose a series of strategic approaches and recommendations to augment the antitumor potential of steroidal saponin compounds, thereby offering prospective insights for their eventual clinical implementation. PURPOSE This review summarizes steroidal saponins' antitumor activity, mechanisms, and limitations. METHODS The data included in this review are sourced from authoritative databases such as PubMed, Web of Science, ScienceDirect, and others. RESULTS A comprehensive summary of over 40 steroidal saponin compounds with proven antitumor activity, including their applicable tumor types and structural characteristics, has been compiled. These steroidal saponins can be primarily classified into five categories: spirostanol, isospirostanol, furostanol, steroidal alkaloids, and cholestanol. The isospirostanol and cholestanol saponins are found to have more potent antitumor activity. The primary antitumor mechanisms of these saponins include tumor cell apoptosis, autophagy induction, inhibition of tumor migration, overcoming drug resistance, and cell cycle arrest. However, steroidal saponins have limitations, such as higher cytotoxicity and lower bioavailability. Furthermore, strategies to address these drawbacks have been proposed. CONCLUSION In summary, isospirostanol and cholestanol steroidal saponins demonstrate notable antitumor activity and different structural categories of steroidal saponins exhibit variations in their antitumor signaling pathways. However, the clinical application of steroidal saponins in cancer treatment still faces limitations, and further research and development are necessary to advance their potential in tumor therapy.
Collapse
Affiliation(s)
- Fengge Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR, PR China
| | - Ma Yu
- School of Life Science and Engineering, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang, 621010, Sichuan, PR China
| | - Wenjie Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Iftikhar Hussain Badar
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, PR China; Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Kai Zhu
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yanlin Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Saba Shafi
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Dangdang Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yongchao Diao
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany.
| | - Zheyong Xue
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| | - Xin Hua
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| |
Collapse
|
3
|
Kobuchi S, Morita A, Jonan S, Amagase K, Ito Y. Translational PK-PD/TD modeling of antitumor effects and peripheral neuropathy in gemcitabine and nab-paclitaxel chemotherapy from xenograft mice to patients for optimal dose and schedule. Cancer Chemother Pharmacol 2024; 93:365-379. [PMID: 38117301 DOI: 10.1007/s00280-023-04625-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023]
Abstract
PURPOSE Gemcitabine and nab-paclitaxel (GnP) treatment, the standard first-line chemotherapy for unresectable pancreatic cancer, often causes peripheral neuropathy (PN). To develop alternative dosing strategies to avoid severe PN, understanding the relationship between pharmacokinetics (PK) and pharmacodynamics/toxicodynamics (PD/TD) is necessary. We established a PK-PD/TD model of GnP treatment to develop an optimal dose schedule. METHODS A mouse xenograft model of human pancreatic cancer was generated to measure drug concentrations in the plasma and tumor, antitumor effects, and PN after GnP treatment. The Simeoni tumor growth inhibition model with tumor concentrations and empirical indirect response models were used for the PD and TD models, respectively. Clinical outcomes were predicted with reported population estimates of PK parameters in cancer patients. RESULTS The PK-PD/TD model simultaneously described the observed tumor volume and paw withdrawal frequency in the von Frey test. For the standard GnP regimen, the model predicted clinical overall response (75.1%), which was overestimated compared to that in a recent phase II study (42.1%) but lower than the observed disease control rate (96.5%). Model simulation showed that dose reduction to less than 40% GnP dose was not effective; a change of dose schedule from every week for 3 weeks to every 2 weeks was a more favorable approach than dose reduction to 60% every week. CONCLUSION The PK-PD/TD model-based translational approach provides a guide for optimal dose determination to avoid severe PN while maintaining antitumor effects during GnP chemotherapy. Further research is needed to enhance its applicability and potential for combination chemotherapy regimens.
Collapse
Affiliation(s)
- Shinji Kobuchi
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Atsuko Morita
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan
| | - Shizuka Jonan
- Laboratory of Pharmacology & Pharmacotherapeutics, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Kikuko Amagase
- Laboratory of Pharmacology & Pharmacotherapeutics, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Yukako Ito
- Department of Pharmacokinetics, Kyoto Pharmaceutical University, Kyoto, 607-8414, Japan.
| |
Collapse
|
4
|
Wang M, Qu K, Zhao P, Yin X, Meng Y, Herdewijn P, Liu C, Zhang L, Xia X. Synthesis and anticancer evaluation of acetylated-lysine conjugated gemcitabine prodrugs. RSC Med Chem 2023; 14:1572-1580. [PMID: 37593582 PMCID: PMC10429768 DOI: 10.1039/d3md00190c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/04/2023] [Indexed: 08/19/2023] Open
Abstract
Gemcitabine is an antimetabolite drug approved for the treatment of various cancers. However, its use is limited due to several issues such as stability, toxicity and drug resistance. Herein, we present the design and synthesis of a series of gemcitabine prodrugs with modifications on the 4-N-amino group by employing an acetylated l- or d-lysine moiety masked by different substitutions. Prodrugs 1-3 and 6-8 showed up to 2.4 times greater anticancer activity than gemcitabine in A549 lung cells, while they exhibited potent activity against BxPC-3 pancreatic cells with IC50 values in the range of 7-40 nM. Moreover, prodrugs 2-3 and 7-8 were found to be less potent against CTSL low expression Caco-2 cells and at least 69-fold less toxic towards human normal HEK-293T cells compared to gemcitabine, leading to improved selectivity and safety profiles. Further stability studies showed that representative prodrugs 2 and 7 exhibited enhanced metabolic stability in human plasma, human liver microsomes and cytidine deaminase. Prodrug 1 can be cleaved by tumor cell-enriched CTSL to release parent drug gemcitabine. Overall, these results demonstrated that acetylated lysine conjugated gemcitabine prodrugs could serve as promising leads for further evaluation as new anticancer drugs.
Collapse
Affiliation(s)
- Mengmeng Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
| | - Kunyu Qu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
| | - Peipei Zhao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
| | - Xin Yin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
| | - Yiwei Meng
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven 3000 Leuven Belgium
| | - Chao Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan 250012 China
| | - Lixin Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
- State Key Laboratory of Bioreactor Engineering, and School of Biotechnology, East China University of Science and Technology (ECUST) Shanghai 200237 China
| | - Xuekui Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences) Jinan 250103 China
| |
Collapse
|
5
|
Sharma A, Bomzan P, Roy N, Dakua VK, Roy K, Barman A, Dey R, Chhetri A, Dewan R, Dutta A, Kumar A, Roy MN. Exploring the Inclusion Complex of an Anticancer Drug with β-Cyclodextrin for Reducing Cytotoxicity Toward the Normal Human Cell Line by an Experimental and Computational Approach. ACS OMEGA 2023; 8:29388-29400. [PMID: 37599964 PMCID: PMC10433473 DOI: 10.1021/acsomega.3c02783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/25/2023] [Indexed: 08/22/2023]
Abstract
The toxicity of any drug against normal cells is a health hazard for all humans. At present, health and disease researchers from all over the world are trying to synthesize designer drugs with diminished toxicity and side effects. The purpose of the present study is to enhance the bioavailability and biocompatibility of gemcitabine (GEM) by decreasing its toxicity and reducing deamination during drug delivery by incorporating it inside the hydrophobic cavity of β-cyclodextrin (β-CD) without affecting the drug ability of the parent compound (GEM). The newly synthesized inclusion complex (IC) was characterized by different physical and spectroscopic techniques, thereby confirming the successful incorporation of the GEM molecule into the nanocage of β-CD. The molecular docking study revealed the orientation of the GEM molecule into the β-CD cavity (-5.40 kcal/mol) to be stably posed for ligand binding. Photostability studies confirmed that the inclusion of GEM using β-CD could lead to better stabilization of GEM (≥96%) for further optical and clinical applications. IC (GEM-β-CD) and GEM exhibited effective antibacterial and antiproliferative activities without being metabolized in a dose-dependent manner. The CT-DNA analysis showed sufficiently strong IC (GEM-β-CD) binding (Ka = 8.1575 × 1010), and this interaction suggests that IC (GEM-β-CD) may possibly exert its biological effects by targeting nucleic acids in the host cell. The newly synthesized biologically active IC (GEM-β-CD), a derivative of GEM, has pharmaceutical development potentiality.
Collapse
Affiliation(s)
- Antara Sharma
- Department
of Chemistry, University of North Bengal, Darjeeling 734013, India
- Department
of Chemistry, St. Joseph’s College, Darjeeling 734104, India
| | - Pranish Bomzan
- Department
of Chemistry, Gorubathan Government College, Kalimpong 735231, India
| | - Niloy Roy
- Department
of Chemistry, University of North Bengal, Darjeeling 734013, India
| | - Vikas Kumar Dakua
- Department
of Chemistry, Alipurduar University, Alipurduar 736122, India
| | - Kanak Roy
- Department
of Chemistry, Alipurduar University, Alipurduar 736122, India
| | - Abhinath Barman
- Department
of Physics, Alipurduar University, Alipurduar 736122, India
| | - Rabindra Dey
- Department
of Chemistry, Cooch Behar College, Cooch Behar 736101, India
| | - Abhijit Chhetri
- Department
of Microbiology, St. Joseph’s College, Darjeeling 734104, India
| | - Rajani Dewan
- Department
of Chemistry, St. Joseph’s College, Darjeeling 734104, India
| | - Ankita Dutta
- Department
of Biotechnology, University of North Bengal, Darjeeling 734013, India
| | - Anoop Kumar
- Department
of Biotechnology, University of North Bengal, Darjeeling 734013, India
| | - Mahendra Nath Roy
- Department
of Chemistry, University of North Bengal, Darjeeling 734013, India
| |
Collapse
|
6
|
Persaud AK, Bernier MC, Massey MA, Agrawal S, Kaur T, Nayak D, Xie Z, Weadick B, Raj R, Hill K, Abbott N, Joshi A, Anabtawi N, Bryant C, Somogyi A, Cruz-Monserrate Z, Amari F, Coppola V, Sparreboom A, Baker SD, Unadkat JD, Phelps MA, Govindarajan R. Increased renal elimination of endogenous and synthetic pyrimidine nucleosides in concentrative nucleoside transporter 1 deficient mice. Nat Commun 2023; 14:3175. [PMID: 37264059 PMCID: PMC10235067 DOI: 10.1038/s41467-023-38789-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Concentrative nucleoside transporters (CNTs) are active nucleoside influx systems, but their in vivo roles are poorly defined. By generating CNT1 knockout (KO) mice, here we identify a role of CNT1 in the renal reabsorption of nucleosides. Deletion of CNT1 in mice increases the urinary excretion of endogenous pyrimidine nucleosides with compensatory alterations in purine nucleoside metabolism. In addition, CNT1 KO mice exhibits high urinary excretion of the nucleoside analog gemcitabine (dFdC), which results in poor tumor growth control in CNT1 KO mice harboring syngeneic pancreatic tumors. Interestingly, increasing the dFdC dose to attain an area under the concentration-time curve level equivalent to that achieved by wild-type (WT) mice rescues antitumor efficacy. The findings provide new insights into how CNT1 regulates reabsorption of endogenous and synthetic nucleosides in murine kidneys and suggest that the functional status of CNTs may account for the optimal action of pyrimidine nucleoside analog therapeutics in humans.
Collapse
Affiliation(s)
- Avinash K Persaud
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Matthew C Bernier
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael A Massey
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Life Sciences Education, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Shipra Agrawal
- Division of Nephrology & Hypertension, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tejinder Kaur
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Debasis Nayak
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhiliang Xie
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Brenna Weadick
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Kasey Hill
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Nicole Abbott
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Arnav Joshi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Nadeen Anabtawi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Claire Bryant
- Center for Clinical & Translational Research, Nationwide Children's Hospital, Columbus, OH, 43210, USA
| | - Arpad Somogyi
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Foued Amari
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Alex Sparreboom
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharyn D Baker
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, College of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Mitch A Phelps
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA.
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
7
|
Inkoom A, Ndemazie NB, Smith T, Frimpong E, Bulusu R, Poku R, Zhu X, Han B, Trevino J, Agyare E. Biological evaluation of novel gemcitabine analog in patient-derived xenograft models of pancreatic cancer. BMC Cancer 2023; 23:435. [PMID: 37179357 PMCID: PMC10182601 DOI: 10.1186/s12885-023-10928-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023] Open
Abstract
Gemcitabine (Gem) has been a standard first-line drug for pancreatic cancer (PCa) treatment; however, Gem's rapid metabolism and systemic instability (short half-life) limit its clinical outcome. The objective of this study was to modify Gem into a more stable form called 4-(N)-stearoyl-gemcitabine (4NSG) and evaluate its therapeutic efficacy in patient-derived xenograft (PDX) models from PCa of Black and White patients.Methods 4NSG was synthesized and characterized using nuclear magnetic resonance (NMR), elemental analysis, and high-performance liquid chromatography (HPLC). 4NSG-loaded solid lipid nanoparticles (4NSG-SLN) were developed using the cold homogenization technique and characterized. Patient-derived pancreatic cancer cell lines labeled Black (PPCL-192, PPCL-135) and White (PPCL-46, PPCL-68) were used to assess the in vitro anticancer activity of 4NSG-SLN. Pharmacokinetics (PK) and tumor efficacy studies were conducted using PDX mouse models bearing tumors from Black and White PCa patients.Results 4NSG was significantly stable in liver microsomal solution. The effective mean particle size (hydrodynamic diameter) of 4NSG-SLN was 82 ± 6.7 nm, and the half maximal inhibitory concentration (IC50) values of 4NSG-SLN treated PPCL-192 cells (9 ± 1.1 µM); PPCL-135 (11 ± 1.3 µM); PPCL-46 (12 ± 2.1) and PPCL-68 equaled to 22 ± 2.6 were found to be significantly lower compared to Gem treated PPCL-192 (57 ± 1.5 µM); PPCL-135 (56 ± 1.5 µM); PPCL-46 (56 ± 1.8 µM) and PPCL-68 (57 ± 2.4 µM) cells. The area under the curve (AUC), half-life, and pharmacokinetic clearance parameters for 4NSG-SLN were 3-fourfold higher than that of GemHCl. For in-vivo studies, 4NSG-SLN exhibited a two-fold decrease in tumor growth compared with GemHCl in PDX mice bearing Black and White PCa tumors.Conclusion 4NSG-SLN significantly improved the Gem's pharmacokinetic profile, enhanced Gem's systemic stability increased its antitumor efficacy in PCa PDX mice bearing Black and White patient tumors.
Collapse
Affiliation(s)
- Andriana Inkoom
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Nkafu Bechem Ndemazie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Taylor Smith
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Rosemary Poku
- College of Medicine, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA
| | - Bo Han
- Department of Surgery, Keck School of Medicine University of Southern California, Los Angeles, California, 90033, USA
| | - Jose Trevino
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA
- Department of Surgery, College of Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1415 South Martin Luther King Jr Blvd, Tallahassee, FL, 32307, USA.
| |
Collapse
|
8
|
Li Y, Liu Y, Chen Y, Wang K, Luan Y. Design, synthesis and antitumor activity study of a gemcitabine prodrug conjugated with a HDAC6 inhibitor. Bioorg Med Chem Lett 2022; 72:128881. [PMID: 35810950 DOI: 10.1016/j.bmcl.2022.128881] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/25/2022] [Accepted: 07/04/2022] [Indexed: 11/25/2022]
Abstract
Gemcitabine, as a first-line antitumor drug, has attracted extensive attention. However the occurrence of drug resistance limits its clinical utilization. In this paper, a gemcitabine prodrug GZ was designed and synthesized by conjugation of gemcitabine with a newly reported HDAC6 selective inhibitor pentadecanoic acid. GZ displayed high cytotoxicity to nine cancer cell lines with IC50 values in the low micromolar range. In vivo, GZ displayed superior antitumor activity to gemcitabine in a 4T1 tumor xenograft model without obvious pathological damage to important organs of mice. Our study showed that compound GZ is a potential gemcitabine prodrug, which is worthy of further antitumor activity exploration.
Collapse
Affiliation(s)
- Yongliang Li
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao, Shandong, China; Department of Pharmacology, School of Pharmacology, Qingdao University Medical College, Qingdao University, Qingdao, Shandong, China
| | - Yuanpeng Liu
- Department of Pharmacology, School of Basic Medicine, Qingdao University Medical College, Qingdao University, Qingdao, Shandong, China
| | - Yiran Chen
- Department of Pharmacology, School of Pharmacology, Qingdao University Medical College, Qingdao University, Qingdao, Shandong, China
| | - Kewei Wang
- Department of Pharmacology, School of Pharmacology, Qingdao University Medical College, Qingdao University, Qingdao, Shandong, China
| | - Yepeng Luan
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University Medical College, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
9
|
Matsumoto T, Masuo Y, Tanaka A, Kimura T, Ioroi T, Yamakawa T, Kitahara H, Kato Y. A physiologically based pharmacokinetic and pharmacodynamic model for disposition of FF-10832. Int J Pharm 2022; 627:122250. [DOI: 10.1016/j.ijpharm.2022.122250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/29/2022] [Accepted: 09/24/2022] [Indexed: 10/31/2022]
|
10
|
Pandit B, Royzen M. Recent Development of Prodrugs of Gemcitabine. Genes (Basel) 2022; 13:genes13030466. [PMID: 35328020 PMCID: PMC8954202 DOI: 10.3390/genes13030466] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/25/2022] [Accepted: 03/01/2022] [Indexed: 12/28/2022] Open
Abstract
Gemcitabine is a nucleoside analog that has been used widely as an anticancer drug for the treatment of a variety of conditions, including ovarian, bladder, non-small-cell lung, pancreatic, and breast cancer. However, enzymatic deamination, fast systemic clearance, and the emergence of chemoresistance have limited its efficacy. Different prodrug strategies have been explored in recent years, seeking to obtain better pharmacokinetic properties, efficacy, and safety. Different drug delivery strategies have also been employed, seeking to transform gemcitabine into a targeted medicine. This review will provide an overview of the recent developments in gemcitabine prodrugs and their effectiveness in treating cancerous tumors.
Collapse
|
11
|
Slusarczyk M, Serpi M, Ghazaly E, Kariuki BM, McGuigan C, Pepper C. Single Diastereomers of the Clinical Anticancer ProTide Agents NUC-1031 and NUC-3373 Preferentially Target Cancer Stem Cells In Vitro. J Med Chem 2021; 64:8179-8193. [PMID: 34085825 DOI: 10.1021/acs.jmedchem.0c02194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A 3'-protected route toward the synthesis of the diastereomers of clinically active ProTides, NUC-1031 and NUC-3373, is described. The in vitro cytotoxic activities of the individual diastereomers were found to be similar to their diastereomeric mixtures. In the KG1a cell line, NUC-1031 and NUC-3373 have preferential cytotoxic effects on leukemic stem cells (LSCs). These effects were not diastereomer-specific and were not observed with the parental nucleoside analogues gemcitabine and FUDR, respectively. In addition, NUC-1031 preferentially targeted LSCs in primary AML samples and cancer stem cells in the prostate cancer cell line, LNCaP. Although the mechanism for this remains incompletely resolved, NUC-1031-treated cells showed increased levels of triphosphate in both LSC and bulk tumor fractions. As ProTides are not dependent on nucleoside transporters, it seems possible that the LSC targeting observed with ProTides may be caused, at least in part, by preferential accumulation of metabolized nucleos(t)ide analogues.
Collapse
Affiliation(s)
- Magdalena Slusarczyk
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Redwood Building, Cardiff CF10 3NB, U.K
| | - Michaela Serpi
- Cardiff University, School of Chemistry, Main Building, Park Place, Cardiff CF10 3AT, U.K
| | - Essam Ghazaly
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, U.K
| | - Benson M Kariuki
- Cardiff University, School of Chemistry, Main Building, Park Place, Cardiff CF10 3AT, U.K
| | - Christopher McGuigan
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Redwood Building, Cardiff CF10 3NB, U.K
| | - Chris Pepper
- Brighton and Sussex Medical School, University of Sussex, Medical Teaching Building, Brighton BN1 9PX, U.K
| |
Collapse
|
12
|
Design, synthesis, and evaluation of liver-specific gemcitabine prodrugs for potential treatment of hepatitis C virus infection and hepatocellular carcinoma. Eur J Med Chem 2021; 213:113135. [PMID: 33454548 DOI: 10.1016/j.ejmech.2020.113135] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 11/24/2022]
Abstract
Many successful anti-viral and anti-cancer drugs are nucleoside analogs, which disrupt RNA and/or DNA synthesis. Here, we present liver-specific prodrugs of the chemotherapy drug gemcitabine (2',2'-difluorodeoxycytidine) for the treatment of hepatitis C virus (HCV) infection and hepatocellular carcinoma. The prodrugs were synthesized by introducing aromatic functional moieties to the cytosine 4-NH2 group of gemcitabine via amide bonds. The chemical modification was designed to i) enable passive diffusion across cellular membrane, ii) protect the prodrugs from inactivating deamination by cellular enzymes, and iii) allow release of active gemcitabine after amide hydrolysis by high levels of carboxylesterases in the liver. We found that many of our prodrugs exhibited similar toxicity as gemcitabine toward liver- and kidney-derived cancer cell lines but were 24- to 620-fold less cytotoxic than gemcitabine in breast- and pancreas-derived cancer cells, respectively. The prodrugs also inhibited an HCV replicon with IC50 values ranging from 10 nM-1.7 μM. Moreover, many of the prodrugs had therapeutic index values of >10,000 and have synergetic effects when combined with other Food and Drug Administration-approved anti-HCV small molecule drugs. These characteristics support the development of gemcitabine prodrugs as liver-specific therapeutics.
Collapse
|
13
|
Shi Z, Song Q, Göstl R, Herrmann A. Mechanochemical activation of disulfide-based multifunctional polymers for theranostic drug release. Chem Sci 2020; 12:1668-1674. [PMID: 34163927 PMCID: PMC8179261 DOI: 10.1039/d0sc06054b] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Drug delivery systems responsive to physicochemical stimuli allow spatiotemporal control over drug activity to overcome limitations of systemic drug administration. Alongside, the non-invasive real-time tracking of drug release and uptake remains challenging as pharmacophore and reporter function are rarely unified within one molecule. Here, we present an ultrasound-responsive release system based on the mechanochemically induced 5-exo-trig cyclization upon scission of disulfides bearing cargo molecules attached via β-carbonate linker within the center of a water soluble polymer. In this bifunctional theranostic approach, we release one reporter molecule per drug molecule to quantitatively track drug release and distribution within the cell in real-time. We use N-butyl-4-hydroxy-1,8-naphthalimide and umbelliferone as fluorescent reporter molecules to accompany the release of camptothecin and gemcitabine as clinically employed anticancer agents. The generality of this approach paves the way for the theranostic release of a variety of probes and drugs by ultrasound. A theranostic approach for the mechanochemically induced release of drugs is presented to track drug release and uptake in real-time.![]()
Collapse
Affiliation(s)
- Zhiyuan Shi
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany .,Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Qingchuan Song
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany .,Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany
| | - Robert Göstl
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany
| | - Andreas Herrmann
- DWI - Leibniz Institute for Interactive Materials Forckenbeckstr. 50 52056 Aachen Germany .,Institute of Technical and Macromolecular Chemistry, RWTH Aachen University Worringerweg 1 52074 Aachen Germany.,Zernike Institute for Advanced Materials, University of Groningen Nijenborgh 4 9747 AG Groningen The Netherlands
| |
Collapse
|
14
|
Miao H, Chen X, Luan Y. Small Molecular Gemcitabine Prodrugs for Cancer Therapy. Curr Med Chem 2020; 27:5562-5582. [PMID: 31419928 DOI: 10.2174/0929867326666190816230650] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/22/2019] [Accepted: 07/09/2019] [Indexed: 02/04/2023]
Abstract
Gemcitabine as a pyrimidine nucleoside analog anticancer drug has high efficacy for a broad spectrum of solid tumors. Gemcitabine is activated within tumor cells by sequential phosphorylation carried out by deoxycytidine kinase to mono-, di-, and triphosphate nucleotides with the last one as the active form. But the instability, drug resistance and toxicity severely limited its utilization in clinics. In the field of medicinal chemistry, prodrugs have proven to be a very effective means for elevating drug stability and decrease undesirable side effects including the nucleoside anticancer drug such as gemcitabine. Many works have been accomplished in design and synthesis of gemcitabine prodrugs, majority of which were summarized in this review.
Collapse
Affiliation(s)
- He Miao
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Shandong Province, Qingdao, China
| | - Xuehong Chen
- Department of Pharmacology, College of Basic Medicine, Qingdao University, Shandong Province, Qingdao, China
| | - Yepeng Luan
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Shandong Province, Qingdao, China
| |
Collapse
|
15
|
Inkoom A, Ndemazie N, Affram K, Smith T, Zhu X, Underwood P, Krishnan S, Ofori E, Han B, Trevino J, Agyare E. Enhancing efficacy of gemcitabine in pancreatic patient-derived xenograft mouse models. Int J Pharm X 2020; 2:100056. [PMID: 33015617 PMCID: PMC7522377 DOI: 10.1016/j.ijpx.2020.100056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/07/2020] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
Gemcitabine (Gem), a nucleoside analog, is a preferred choice of treatment for pancreatic cancer (PCa) and often used in combination therapy against wide range of solid tumors. It is known to be rapidly inactivated in blood by cytidine deaminase. The objective of the study was to improve the systemic stability and anticancer activity of modified Gem termed 4-N-stearoylGem (4NSG) In this study, the IC50 values of 4NSG treated MiaPaCa-2 and primary pancreatic cancer (PPCL-46) cultures were significantly lower when compared with gemcitabine hydrochloride (GemHCl) treated cultures. In acute toxicity study, liver enzyme level of aspartate aminotransferase (AST) of the control mice was not significantly different from AST levels of 4NSG and GemHCl treated mice. However, alanine aminotransferase (ALT) level of control mice (67 ± 5 mUnits/mL) was significantly lower compared with ALT levels of GemHCl (232 ± 28 mUnits/mL) and that of 4NSG (172 ± 22 mUnits/mL) (p < 0.0001). More importantly, ALT level of 4NSG was lower than ALT level of GemHCl (p < 0.05). Although ALT levels were elevated, pathological images of liver and kidney tissues of control, GemHCl and 4NSG treated mice revealed no architectural changes and no significant change in mice weight was observed during treatment. The bioavailability (AUC) of 4NSG was 3-fold high and significantly inhibited the tumor growth as compared with equivalent dose of GemHCl. Immunohistochemical staining revealed that 4NSG significantly inhibited the expression vascular endothelial growth factor (VEGF) receptor. The study is unique because it established, for the first time, enhanced anticancer activity of 4NSG against pancreatic patient-derived xenograft (PDX) mouse model and PPCL-46 cells compared with Gem. 4SGN enhanced pharmacokinetic profile and improved the therapeutic efficacy of the standard-of-care Gem. Lastly, 4GSN showed a remarkable tumor growth inhibition and revealed significant antiangiogenic activity in 4GSN treated pancreatic PDX tumor.
Collapse
Affiliation(s)
- Andriana Inkoom
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, FL, United States of America
| | - Nkafu Ndemazie
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, FL, United States of America
| | - Kevin Affram
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, FL, United States of America
| | - Taylor Smith
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, FL, United States of America
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, FL, United States of America
| | - Patrick Underwood
- University of Florida Department of Surgery, Gainesville, FL, United States of America
| | | | - Edward Ofori
- College of Pharmacy, Chicago State University, Chicago, IL, United States of America
| | - Bo Han
- Department of Surgery, Keck School of Medicine University of Southern California, Los Angeles, United States of America
| | - Jose Trevino
- University of Florida Department of Surgery, Gainesville, FL, United States of America
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A & M University, Tallahassee, FL, United States of America
| |
Collapse
|
16
|
Thompson BR, Shi J, Zhu HJ, Smith DE. Pharmacokinetics of gemcitabine and its amino acid ester prodrug following intravenous and oral administrations in mice. Biochem Pharmacol 2020; 180:114127. [PMID: 32603666 DOI: 10.1016/j.bcp.2020.114127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/30/2022]
Abstract
Gemcitabine is an intravenously administered anti-cancer nucleoside analogue. Systemic exposure following oral administration of gemcitabine is limited by extensive first-pass metabolism via cytidine deaminase (CDA) and potentially by saturation of nucleoside transporter-mediated intestinal uptake. An amino acid ester prodrug of gemcitabine, 5'-l-valyl-gemcitabine (V-Gem), was previously shown to be a substrate of the intestinally expressed peptide transporter 1 (PEPT1) and stable against CDA-mediated metabolism. However, preliminary studies did not evaluate the in vivo oral performance of V-Gem as compared to parent drug. In the present study, we evaluated the pharmacokinetics and in vivo oral absorption of gemcitabine and V-Gem following intravenous and oral administrations in mice. These studies revealed that V-Gem undergoes rapid systemic elimination (half-life < 1 min) and has a low oral bioavailability (<1%). Most importantly, the systemic exposure of gemcitabine was not different following oral administration of equimolar doses of gemcitabine (gemcitabine bioavailability of 18.3%) and V-Gem (gemcitabine bioavailability of 16.7%). Single-pass intestinal perfusions with portal blood sampling in mice revealed that V-Gem undergoes extensive activation in intestinal epithelial cells and that gemcitabine undergoes first-pass metabolism in intestinal epithelial cells. Thus, formulation of gemcitabine as the prodrug V-Gem does not increase systemic gemcitabine exposure following oral dosing, due, in part, to the instability of V-Gem in intestinal epithelial cells.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jian Shi
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - David E Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
17
|
Kush P, Kaur M, Sharma M, Madan J, Kumar P, Deep A, Kim KH. Investigations of potent biocompatible metal-organic framework for efficient encapsulation and delivery of Gemcitabine: biodistribution, pharmacokinetic and cytotoxicity study. Biomed Phys Eng Express 2020; 6:025014. [DOI: 10.1088/2057-1976/ab73f7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
18
|
Biodistribution and Pharmacokinetic Study of Gemcitabine Hydrochloride Loaded Biocompatible Iron-Based Metal Organic Framework. J Inorg Organomet Polym Mater 2019. [DOI: 10.1007/s10904-019-01417-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
Thompson BR, Hu Y, Smith DE. Mechanisms of gemcitabine oral absorption as determined by in situ intestinal perfusions in mice. Biochem Pharmacol 2019; 168:57-64. [PMID: 31207211 DOI: 10.1016/j.bcp.2019.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/12/2019] [Indexed: 01/23/2023]
Abstract
Gemcitabine is a widely used chemotherapeutic drug that is administered via intravenous infusion due to a low oral bioavailability of only 10%. This low oral bioavailability is believed to be the result of gemcitabine's low intestinal permeability and oral absorption, followed by significant presystemic metabolism. In the present study, we sought to define the mechanisms of gemcitabine intestinal permeability, the potential for saturation of intestinal uptake, and the transporter(s) responsible for mediating the oral absorption of drug using in situ single-pass intestinal perfusions in mice. Concentration-dependent studies were performed for gemcitabine over 0.5-2000 μM, along with studies of 5 μM gemcitabine in a sodium-containing buffer ± thymidine (which can inhibit concentrative (i.e., CNT1 and CNT3) and equilibrative (i.e., ENT1 and ENT2) nucleoside transporters) or dilazep (which can inhibit ENT1 and ENT2), or in a sodium-free buffer (which can inhibit CNT1 and CNT3). Our findings demonstrated that gemcitabine was, in fact, a high-permeability drug in the intestine at low concentrations, that jejunal uptake of gemcitabine was saturable and mediated almost exclusively by nucleoside transporters, and that jejunal flux was mediated by both high-affinity, low-capacity (Km = 27.4 µM, Vmax = 3.6 pmol/cm2/s) and low-affinity, high-capacity (Km = 700 µM, Vmax = 35.9 pmol/cm2/s) transport systems. Thus, CNTs and ENTs at the apical membrane allow for gemcitabine uptake from the lumen to enterocyte, whereas ENTs at the basolateral membrane allow for gemcitabine efflux from the enterocyte to portal venous blood.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - Yongjun Hu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | - David E Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Synthesis of Gemcitabine-Threonine Amide Prodrug Effective on Pancreatic Cancer Cells with Improved Pharmacokinetic Properties. Molecules 2018; 23:molecules23102608. [PMID: 30314360 PMCID: PMC6222828 DOI: 10.3390/molecules23102608] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/02/2018] [Accepted: 10/10/2018] [Indexed: 12/24/2022] Open
Abstract
To investigate the amino acid transporter-based prodrug anticancer strategy further, several amino acid-conjugated amide gemcitabine prodrugs were synthesized to target amino acid transporters in pancreatic cancer cells. The structures of the synthesized amino acid-conjugated prodrugs were confirmed by 1H-NMR and LC-MS. The pancreatic cancer cells, AsPC1, BxPC-3, PANC-1 and MIAPaCa-2, appeared to overexpress the amino acid transporter LAT-1 by conventional RT-PCR. Among the six amino acid derivatives of gemcitabine, threonine derivative of gemcitabine (Gem-Thr) was more effective than free gemcitabine in the pancreatic cancer cells, BxPC-3 and MIAPaCa-2, respectively, in terms of anti-cancer effects. Furthermore, Gem-Thr was metabolically stable in PBS (pH 7.4), rat plasma and liver microsomal fractions. When Gem-Thr was administered to rats at 4 mg/kg i.v., Gem-Thr was found to be successfully converted to gemcitabine via amide bond cleavage. Moreover, the Gem-Thr showed the increased systemic exposure of formed gemcitabine by 1.83-fold, compared to free gemcitabine treatment, due to the significantly decreased total clearance (0.60 vs. 4.23 mL/min/kg), indicating that the amide prodrug approach improves the metabolic stability of gemcitabine in vivo. Taken together, the amino acid transporter-targeting gemcitabine prodrug, Gem-Thr, was found to be effective on pancreatic cancer cells and to offer an efficient potential means of treating pancreatic cancer with significantly better pharmacokinetic characteristics than gemcitabine.
Collapse
|
21
|
Zhang H, Wang K, Na K, Li D, Li Z, Zhao D, Zhong L, Wang M, Kou L, Luo C, Zhang H, Kan Q, Ding H, He Z, Sun J. Striking a Balance between Carbonate/Carbamate Linkage Bond- and Reduction-Sensitive Disulfide Bond-Bearing Linker for Tailored Controlled Release: In Situ Covalent-Albumin-Binding Gemcitabine Prodrugs Promote Bioavailability and Tumor Accumulation. J Med Chem 2018; 61:4904-4917. [DOI: 10.1021/acs.jmedchem.8b00293] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Huicong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Kuanglei Wang
- Wuyi University, Jiangmen, Guangdong 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, Guangdong 529080, P. R. China
| | - Kexin Na
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Dan Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Zhenbao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Dongyang Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Lu Zhong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Menglin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Longfa Kou
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Qiming Kan
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Huaiwei Ding
- School of Pharmaceutical and Engineering, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| |
Collapse
|
22
|
Abstract
Introduction Sickle cell disease (SCD) is an orphan disease in the United States, but is highly prevalent worldwide. Only two drugs, hydroxyurea and L-glutamine, are approved for this disease. With an improved understanding of the pathophysiology of SCD as well as the success of several recently approved drugs for other orphan diseases, there is an increased interest in the development of drugs for SCD. Areas covered This review summarizes published studies of drug therapies and ongoing trials of novel agents. Expert opinion The development of drugs with different mechanisms of action offers opportunities for combination and individualized therapy in SCD. In addition to acute pain crisis, the evaluation of other SCD-related complications, exercise capacity, patient reported outcomes and validated surrogate endpoints are necessary to advance drug development. It is important to involve sites in sub-Saharan Africa and India, which have the highest burden of SCD, in trials of novel therapies.
Collapse
Affiliation(s)
- Kenneth I Ataga
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC
| | - Payal C Desai
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, NC.,#Division of Hematology, The Ohio State University, Columbus, OH
| |
Collapse
|
23
|
Lavelle D, Engel JD, Saunthararajah Y. Fetal Hemoglobin Induction by Epigenetic Drugs. Semin Hematol 2018; 55:60-67. [PMID: 29958562 DOI: 10.1053/j.seminhematol.2018.04.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/13/2018] [Indexed: 12/11/2022]
Abstract
Fetal hemoglobin (HbF) inhibits the root cause of sickle pathophysiology, sickle hemoglobin polymerization. Individuals who naturally express high levels of HbF beyond infancy thus receive some protection from sickle complications. To mimic this natural genetic experiment using drugs, one guiding observation was that HbF is increased during recovery of bone marrow from extreme stress. This led to evaluation and approval of the cytotoxic (cell killing) drug hydroxyurea to treat sickle cell disease. Cytotoxic approaches are limited in potency and sustainability, however, since they require hematopoietic reserves sufficient to repeatedly mount recoveries from stress that destroys their counterparts, and such reserves are finite. HbF induction even by stress ultimately involves chromatin remodeling of the gene for HbF (HBG), therefore, a logical alternative approach is to directly inhibit epigenetic enzymes that repress HBG-implicated enzymes include DNA methyltransferase 1, histone deacetylases, lysine demethylase 1, protein arginine methyltransferase 5, euchromatic histone lysine methyltransferase 2 and chromodomain helicase DNA-binding protein 4. Clinical proof-of-principle that this alternative, noncytotoxic approach can generate substantial HbF and total hemoglobin increases has already been generated. Thus, with continued careful attention to fundamental biological and pharmacologic considerations (reviewed herein), there is potential that rational, molecular-targeted, safe and highly potent disease-modifying therapy can be realized for patients with sickle cell disease, with the accessibility and cost-effective properties needed for world-wide effect.
Collapse
Affiliation(s)
- Donald Lavelle
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, IL; Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL
| | | | - Yogen Saunthararajah
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
24
|
Zhou Y, Chang Q, Wang W, Zhang X, Zhou F, Sun J, Wang G, Peng Y. Sensitive analysis and pharmacokinetic study of a novel gemcitabine carbamate prodrug and its active metabolite gemcitabine in rats using LC-ESI-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1083:249-257. [PMID: 29554521 DOI: 10.1016/j.jchromb.2018.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/11/2018] [Accepted: 03/09/2018] [Indexed: 11/18/2022]
Abstract
FY363 is a new chemical entity of gemcitabine analog, which has been shown to have a significant inhibitory effect on cell proliferation in a variety of tumor cell lines in vitro. As a carbamate derivative, FY363 would be converted to the active metabolite gemcitabine through enzyme action in vivo. In order to clarify the exposure of FY363 prototype and its metabolite gemcitabine in vivo after administration of FY363, a sensitive and specific liquid chromatography tandem mass spectrometry (LC-MS/MS) was developed and validated to simultaneously determine FY363 and gemcitabine in rat plasma after liquid-liquid extraction with ethyl acetate. Chromatographic separation was achieved on a highly stable polar column of Synergi 4u Polar-RP 80A (4 μm, 4.6 × 250 mm) which has a unique ether - phenyl bonded phase. Gradient elution was accomplished with mobile phase system consisting of 5 mM ammonium formate buffer containing 0.1% formic acid and mixed organic solvents containing methanol-acetonitrile (3:2, v/v). Multiple reaction monitoring transitions were performed on triple quadrupole mass spectrometric detection in positive-ion mode with an electrospray ionization source. The calibration curves showed good linearity (r > 0.99) over the established concentration range of 1.0-1000 ng/mL both for FY363 and gemcitabine. The assay was validated to be selective, robust and reproducible. This well validated method was successfully applied to demonstrate the pharmacokinetic behavior and the metabolic transformation of FY363 in rats. Results revealed that about 20% of FY363 were converted into its active metabolite gemcitabine in rats by comparing the exposure of gemcitabine after the FY363 administration with that after direct gemcitabine administration at equimolar dose.
Collapse
Affiliation(s)
- Yi Zhou
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qingqing Chang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenjie Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaofang Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fang Zhou
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jianguo Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Ying Peng
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
25
|
Fluorinated nucleosides as an important class of anticancer and antiviral agents. Future Med Chem 2017; 9:1809-1833. [DOI: 10.4155/fmc-2017-0095] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Fluorine-containing nucleoside analogs (NAs) represent a significant class of the US FDA-approved chemotherapeutics widely used in the clinic. The incorporation of fluorine into drug-like agents modulates lipophilic, electronic and steric parameters, thus influencing pharmacodynamic and pharmacokinetic properties of drugs. Fluorine can block oxidative metabolism of drugs and the formation of undesired metabolites by changing H-bonding interactions. In this review, we focus our attention on chemical fluorination reagents and methods used in the NAs field, including positron emission tomography radiochemistry. We briefly discuss both the cellular biology and clinical properties of FDA-approved and fluorine-containing nucleoside/nucleotide analogs in development as well as common resistance mechanisms associated with their use. Finally, we emphasize pronucleotide strategies used to improve therapeutic outcome of NAs in the clinic.
Collapse
|
26
|
Molokie R, Lavelle D, Gowhari M, Pacini M, Krauz L, Hassan J, Ibanez V, Ruiz MA, Ng KP, Woost P, Radivoyevitch T, Pacelli D, Fada S, Rump M, Hsieh M, Tisdale JF, Jacobberger J, Phelps M, Engel JD, Saraf S, Hsu LL, Gordeuk V, DeSimone J, Saunthararajah Y. Oral tetrahydrouridine and decitabine for non-cytotoxic epigenetic gene regulation in sickle cell disease: A randomized phase 1 study. PLoS Med 2017; 14:e1002382. [PMID: 28880867 PMCID: PMC5589090 DOI: 10.1371/journal.pmed.1002382] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 08/03/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Sickle cell disease (SCD), a congenital hemolytic anemia that exacts terrible global morbidity and mortality, is driven by polymerization of mutated sickle hemoglobin (HbS) in red blood cells (RBCs). Fetal hemoglobin (HbF) interferes with this polymerization, but HbF is epigenetically silenced from infancy onward by DNA methyltransferase 1 (DNMT1). METHODS AND FINDINGS To pharmacologically re-induce HbF by DNMT1 inhibition, this first-in-human clinical trial (NCT01685515) combined 2 small molecules-decitabine to deplete DNMT1 and tetrahydrouridine (THU) to inhibit cytidine deaminase (CDA), the enzyme that otherwise rapidly deaminates/inactivates decitabine, severely limiting its half-life, tissue distribution, and oral bioavailability. Oral decitabine doses, administered after oral THU 10 mg/kg, were escalated from a very low starting level (0.01, 0.02, 0.04, 0.08, or 0.16 mg/kg) to identify minimal doses active in depleting DNMT1 without cytotoxicity. Patients were SCD adults at risk of early death despite standard-of-care, randomized 3:2 to THU-decitabine versus placebo in 5 cohorts of 5 patients treated 2X/week for 8 weeks, with 4 weeks of follow-up. The primary endpoint was ≥ grade 3 non-hematologic toxicity. This endpoint was not triggered, and adverse events (AEs) were not significantly different in THU-decitabine-versus placebo-treated patients. At the decitabine 0.16 mg/kg dose, plasma concentrations peaked at approximately 50 nM (Cmax) and remained elevated for several hours. This dose decreased DNMT1 protein in peripheral blood mononuclear cells by >75% and repetitive element CpG methylation by approximately 10%, and increased HbF by 4%-9% (P < 0.001), doubling fetal hemoglobin-enriched red blood cells (F-cells) up to approximately 80% of total RBCs. Total hemoglobin increased by 1.2-1.9 g/dL (P = 0.01) as reticulocytes simultaneously decreased; that is, better quality and efficiency of HbF-enriched erythropoiesis elevated hemoglobin using fewer reticulocytes. Also indicating better RBC quality, biomarkers of hemolysis, thrombophilia, and inflammation (LDH, bilirubin, D-dimer, C-reactive protein [CRP]) improved. As expected with non-cytotoxic DNMT1-depletion, platelets increased and neutrophils concurrently decreased, but not to an extent requiring treatment holds. As an early phase study, limitations include small patient numbers at each dose level and narrow capacity to evaluate clinical benefits. CONCLUSION Administration of oral THU-decitabine to patients with SCD was safe in this study and, by targeting DNMT1, upregulated HbF in RBCs. Further studies should investigate clinical benefits and potential harms not identified to date. TRIAL REGISTRATION ClinicalTrials.gov, NCT01685515.
Collapse
Affiliation(s)
- Robert Molokie
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Donald Lavelle
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Michel Gowhari
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Michael Pacini
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Lani Krauz
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Johara Hassan
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Vinzon Ibanez
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Maria A. Ruiz
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| | - Kwok Peng Ng
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Philip Woost
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Tomas Radivoyevitch
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Daisy Pacelli
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Sherry Fada
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Matthew Rump
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Matthew Hsieh
- Molecular and Clinical Hematology Section, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John F. Tisdale
- Molecular and Clinical Hematology Section, National Institutes of Health, Bethesda, Maryland, United States of America
| | - James Jacobberger
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Mitch Phelps
- College of Pharmacy, The Ohio State University, Columbus, Ohio, United States of America
| | - James Douglas Engel
- Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Santhosh Saraf
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Lewis L. Hsu
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Victor Gordeuk
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Joseph DeSimone
- Department of Medicine, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, United States of America
| | - Yogen Saunthararajah
- Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
27
|
Matsumoto J, Kiesel BF, Parise RA, Guo J, Taylor S, Huang M, Eiseman JL, Ivy SP, Kunos C, Chu E, Beumer JH. LC-MS/MS assay for the quantitation of the ribonucleotide reductase inhibitor triapine in human plasma. J Pharm Biomed Anal 2017; 146:154-160. [PMID: 28881312 DOI: 10.1016/j.jpba.2017.08.036] [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] [Received: 06/29/2017] [Revised: 08/18/2017] [Accepted: 08/28/2017] [Indexed: 10/18/2022]
Abstract
The ribonucleotide reductase inhibitor and radiosensitizer triapine (3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP), NSC 663249) is clinically being evaluated via the intravenous (IV) route for the treatment of cervical and vulvar cancer in combination with primary cisplatin chemoradiation. The need for a 2-h infusion and frequent administration of triapine is logistically challenging, prompting us to pursue oral (PO) administration. In support of the clinical trial investigating oral triapine in combination with chemoradiation, we developed and validated a novel LC-MS/MS assay for the quantification of triapine in 50μL human plasma. After protein precipitation, chromatographic separation of the supernatant was achieved with a Shodex ODP2 column and an isocratic acetonitrile-water mobile phase with 10% ammonium acetate. Detection with an ABI 4000 mass spectrometer utilized electrospray positive mode ionization. The assay was linear from 3 to 3,000ng/mL and proved to be accurate (97.1-103.1%) and precise (<7.4% CV), and met the U.S. FDA guidance for bioanalytical method validation. This LC-MS/MS assay will be an essential tool to further define the pharmacokinetics and oral bioavailability of triapine.
Collapse
Affiliation(s)
- Julia Matsumoto
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States; School of Pharmaceutical Sciences, Sao Paulo State University, Araraquara, SP, Brazil
| | - Brian F Kiesel
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Robert A Parise
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Jianxia Guo
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States
| | - Sarah Taylor
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Medicine, University of Pittsburgh-Magee Women's Hospital, Pittsburgh, PA, United States
| | - Marilyn Huang
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Medicine, University of Pittsburgh-Magee Women's Hospital, Pittsburgh, PA, United States
| | - Julie L Eiseman
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States; Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - S Percy Ivy
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, United States
| | - Charles Kunos
- Investigational Drug Branch, Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, United States
| | - Edward Chu
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States; Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jan H Beumer
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, United States; Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
28
|
Phase I study of veliparib in combination with gemcitabine. Cancer Chemother Pharmacol 2017; 80:631-643. [PMID: 28770300 DOI: 10.1007/s00280-017-3409-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Veliparib (ABT-888) is an oral PARP inhibitor expected to increase gemcitabine activity. This phase I determined the maximal tolerable dose (MTD), dose-limiting toxicities (DLT), antitumor activity, pharmacokinetics (PK), and pharmacodynamics (PD) of veliparib combined with gemcitabine. METHODS Patients with advanced solid tumors received veliparib (10-40-mg PO BID) on chemotherapy weeks with gemcitabine 500-750-mg/m2 IV on days 1, 8, and 15 (28-day cycle), or on days 1 and 8 (21-day cycle). The MTD, DLT, adverse events, PK, and PD were evaluated. RESULTS Eleven patients were enrolled on the 28-day schedule. The 28-day schedule was considered intolerable and amended to a 21-day schedule, with 20 patients enrolled. Grade ≥ 3 adverse events were myelosuppression-related. The MTD was determined to be 750-mg/m2 gemcitabine IV on days 1 and 8- and 20-mg PO veliparib BID days 1-14 on a 21-day schedule. Of 27 patients evaluable for response, 3 had PR and 15 had SD. There was no evidence of any major drug-drug interaction, and PK parameter values for veliparib, gemcitabine, and dFdU were as expected. Analysis of PBMCs showed evidence of PARP inhibition and DNA damage associated with therapy. CONCLUSIONS Gemcitabine at 750-mg/m2 IV on days 1 and 8 combined with veliparib at a dose of 20-mg PO BID days 1-14 on a 21-day schedule is relatively well-tolerated, with manageable, expected toxicities. Clinical responses were observed in a pretreated population of patients, suggesting that this combination should be further evaluated in the phase II setting.
Collapse
|
29
|
Zhang Z, Yang E, Hu C, Cheng H, Chen CY, Huang D, Wang R, Zhao Y, Rong L, Vignuzzi M, Shen H, Shen L, Chen ZW. Cell-Based High-Throughput Screening Assay Identifies 2',2'-Difluoro-2'-deoxycytidine Gemcitabine as a Potential Antipoliovirus Agent. ACS Infect Dis 2017; 3:45-53. [PMID: 27733043 DOI: 10.1021/acsinfecdis.6b00116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As we approach the global eradication of circulating wild-type polioviruses (PV), vaccination with oral poliovirus vaccine (OPV) has led to the emergence of circulating vaccine-derived poliovirus (cVDPV) and vaccine-associated paralytic poliomyelitis (VAPP). Complete cessation of all poliovirus infections may require stopping use of OPV and formulating improved vaccines and new antiviral drugs. Currently, no licensed drugs are available to treat chronically infected poliovirus excretors. Here, we created a modified PV expressing Gaussia Luciferase (Sb-Gluc) and developed a cell-based high-throughput screening (HTS) antiviral assay. Using the validated HTS assay, we screened the FDA-approved drug library of compounds and identified candidate agents capable of inhibiting PV replication. We then characterized antipoliovirus activity for the best hit, gemcitabine, a nucleoside analogue used in tumor chemotherapy. We found that gemcitabine inhibited PV Mahoney replication with an IC50 of 0.3 μM. It completely protected HeLa cells from PV-induced cytopathic effects at 25 μM, without detectable toxicity for cell viability. Furthermore, a gemcitabine metabolite directly inhibited the ability of PV RNA polymerase to synthesize or elongate PV RNA. Because PV RNA polymerase is somehow conserved among species in the Picornaviridae family, gemcitabine may be further developed as an attractive broad-spectrum antiviral for PV and others.
Collapse
Affiliation(s)
- Zhuoran Zhang
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Enzhuo Yang
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
- Unit of anti-tuberculosis immunity, CAS
Key Laboratory of Molecular Virology and Immunology, Institut Pasteur
of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chunmiao Hu
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Han Cheng
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Crystal Y. Chen
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Dan Huang
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Richard Wang
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Yue Zhao
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Lijun Rong
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Marco Vignuzzi
- Viral Populations and Pathogenesis Unit, CNRS UMR 3569, Institut Pasteur, 25-28 rue du Dr. Roux, 75724 Paris cedex
15, France
| | - Hongbo Shen
- Unit of anti-tuberculosis immunity, CAS
Key Laboratory of Molecular Virology and Immunology, Institut Pasteur
of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ling Shen
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
| | - Zheng W. Chen
- Department of Microbiology and Immunology and Center for Primate
Biomedical Research, University of Illinois College of Medicine, 909 South Wolcott Avenue, E704, M/C790, Chicago, Illinois 60612, United States
- Institut Pasteur of Shanghai, Shanghai 200031, China
| |
Collapse
|
30
|
DeZern AE, Zeidan AM, Barnard J, Hand W, Al Ali N, Brown F, Zimmerman C, Roboz GJ, Garcia-Manero G, Steensma DP, Komrokji RS, Sekeres MA. Differential response to hypomethylating agents based on sex: a report on behalf of the MDS Clinical Research Consortium (MDS CRC). Leuk Lymphoma 2016; 58:1325-1331. [PMID: 27774847 DOI: 10.1080/10428194.2016.1246726] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
First-line therapy for higher-risk myelodysplastic syndromes (MDS) includes decitabine (DAC) or azacitidine (AZA). Variables have not identified differential response rates between these. We assessed the influence of patient sex on outcomes including overall survival (OS) in 642 patients with higher-risk MDS treated with AZA or DAC. DAC-treated patients (35% of females, 31% of males) had marginally better OS than AZA-treated patients (p = .043), (median OS of 18.7 months versus 16.4 months), but the difference varied strongly by sex. Female patients treated with DAC had a longer median OS (21.1 months, 95% CI: 16.0-28.0) than female patients treated with AZA (13.2 months, 95% CI: 11.0-15.9; p = .0014), while for males there was no significant difference between HMAs (median OS 18.3 months with DAC versus 17.9 months for AZA, p = .59). The biological reason for this variability is unclear, but may be a consequence of differences in cytidine deaminase activity between men and women.
Collapse
Affiliation(s)
- Amy E DeZern
- a Department of Oncology , Sidney Kimmel Cancer Center , Baltimore , MD , USA
| | - Amer M Zeidan
- b Yale Cancer Center, Yale University , New Haven , CT , USA
| | - John Barnard
- c Leukemia Program, Cleveland Clinic Taussig Cancer Institute , Cleveland , OH , USA
| | - Wesley Hand
- a Department of Oncology , Sidney Kimmel Cancer Center , Baltimore , MD , USA
| | - Najla Al Ali
- d Hematologic Malignancies, H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Francis Brown
- e Adult Leukemia Program, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Cassie Zimmerman
- c Leukemia Program, Cleveland Clinic Taussig Cancer Institute , Cleveland , OH , USA
| | - Gail J Roboz
- f Weill Medical College of Cornell University, Leukemia Program , New York , NY , USA
| | | | - David P Steensma
- e Adult Leukemia Program, Department of Medical Oncology , Dana-Farber Cancer Institute , Boston , MA , USA
| | - Rami S Komrokji
- d Hematologic Malignancies, H. Lee Moffitt Cancer Center , Tampa , FL , USA
| | - Mikkael A Sekeres
- c Leukemia Program, Cleveland Clinic Taussig Cancer Institute , Cleveland , OH , USA
| | | |
Collapse
|
31
|
Poulin P, Chen YH, Ding X, Gould SE, Hop CE, Messick K, Oeh J, Liederer BM. Prediction of Drug Distribution in Subcutaneous Xenografts of Human Tumor Cell Lines and Healthy Tissues in Mouse: Application of the Tissue Composition-Based Model to Antineoplastic Drugs. J Pharm Sci 2015; 104:1508-21. [DOI: 10.1002/jps.24336] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 12/05/2014] [Accepted: 12/12/2014] [Indexed: 12/20/2022]
|
32
|
Clouser CL, Bonnac L, Mansky LM, Patterson SE. Characterization of permeability, stability and anti-HIV-1 activity of decitabine and gemcitabine divalerate prodrugs. Antivir Chem Chemother 2014; 23:223-30. [PMID: 23994876 DOI: 10.3851/imp2682] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2013] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Over 25 drugs have been approved for the treatment of HIV-1 replication. All but one of these drugs is delivered as an oral medication. Previous studies have demonstrated that two drugs, decitabine and gemcitabine, have potent anti-HIV-1 activities and can work together in synergy to reduce HIV-1 infectivity via lethal mutagenesis. For their current indications, decitabine and gemcitabine are delivered intravenously. METHODS As an initial step towards the clinical translation of these drugs for the treatment of HIV-1 infection, we synthesized decitabine and gemcitabine prodrugs in order to increase drug permeability, which has generally been shown to correlate with increased bioavailability in vivo. In the present study we investigated the permeability, stability and anti-HIV-1 activity of decitabine and gemcitabine prodrugs and selected the divalerate esters of each as candidates for further investigation. RESULTS Our results provide the first demonstration of divalerate prodrugs of decitabine and gemcitabine that are readily permeable, stable and possess anti-HIV-1 activity. CONCLUSIONS These observations predict improved oral availability of decitabine and gemcitabine, and warrant further study of their ability to reduce HIV-1 infectivity in vivo.
Collapse
Affiliation(s)
- Christine L Clouser
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN, USA.
| | | | | | | |
Collapse
|
33
|
Bera A, VenkataSubbaRao K, Manoharan MS, Hill P, Freeman JW. A miRNA signature of chemoresistant mesenchymal phenotype identifies novel molecular targets associated with advanced pancreatic cancer. PLoS One 2014; 9:e106343. [PMID: 25184537 PMCID: PMC4153643 DOI: 10.1371/journal.pone.0106343] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/06/2014] [Indexed: 01/24/2023] Open
Abstract
In this study a microRNA (miRNA) signature was identified in a gemcitabine resistant pancreatic ductal adenocarcinoma (PDAC) cell line model (BxPC3-GZR) and this signature was further examined in advanced PDAC tumor specimens from The Cancer Genome Atlas (TCGA) database. BxPC3-GZR showed a mesenchymal phenotype, expressed high levels of CD44 and showed a highly significant deregulation of 17 miRNAs. Based on relevance to cancer, a seven-miRNA signature (miR-100, miR-125b, miR-155, miR-21, miR-205, miR-27b and miR-455-3p) was selected for further studies. A strong correlation was observed for six of the seven miRNAs in 43 advanced tumor specimens compared to normal pancreas tissue. To assess the functional relevance we initially focused on miRNA-125b, which is over-expressed in both the BxPC3-GZR model and advanced PDAC tumor specimens. Knockdown of miRNA-125b in BxPC3-GZR and Panc-1 cells caused a partial reversal of the mesenchymal phenotype and enhanced response to gemcitabine. Moreover, RNA-seq data from each of 40 advanced PDAC tumor specimens from the TCGA data base indicate a negative correlation between expression of miRNA-125b and five of six potential target genes (BAP1, BBC3, NEU1, BCL2, STARD13). Thus far, two of these target genes, BBC3 and NEU1, that are tumor suppressor genes but not yet studied in PDAC, appear to be functional targets of miR-125b since knockdown of miR125b caused their up regulation. These miRNAs and their molecular targets may serve as targets to enhance sensitivity to chemotherapy and reduce metastatic spread.
Collapse
Affiliation(s)
- Alakesh Bera
- Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Kolaparthi VenkataSubbaRao
- Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Muthu Saravanan Manoharan
- Research and Development, Audie Murphy Veterans Administration Hospital, San Antonio, Texas, United States of America
| | - Ping Hill
- Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - James W. Freeman
- Department of Medicine, Division of Hematology and Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Cancer Therapy and Research Center, Experimental and Developmental Therapeutics Program, San Antonio, Texas, United States of America
- Research and Development, Audie Murphy Veterans Administration Hospital, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
34
|
Bapiro TE, Frese KK, Courtin A, Bramhall JL, Madhu B, Cook N, Neesse A, Griffiths JR, Tuveson DA, Jodrell DI, Richards FM. Gemcitabine diphosphate choline is a major metabolite linked to the Kennedy pathway in pancreatic cancer models in vivo. Br J Cancer 2014; 111:318-25. [PMID: 24874484 PMCID: PMC4102943 DOI: 10.1038/bjc.2014.288] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/15/2014] [Accepted: 04/30/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The modest benefits of gemcitabine (dFdC) therapy in patients with pancreatic ductal adenocarcinoma (PDAC) are well documented, with drug delivery and metabolic lability cited as important contributing factors. We have used a mouse model of PDAC: KRAS(G12D); p53(R172H); pdx-Cre (KPC) that recapitulates the human disease to study dFdC intra-tumoural metabolism. METHODS LC-MS/MS and NMR were used to measure drug and physiological analytes. Cytotoxicity was assessed by the Sulphorhodamine B assay. RESULTS In KPC tumour tissue, we identified a new, Kennedy pathway-linked dFdC metabolite (gemcitabine diphosphate choline (GdPC)) present at equimolar amounts to its precursor, the accepted active metabolite gemcitabine triphosphate (dFdCTP). Utilising additional subcutaneous PDAC tumour models, we demonstrated an inverse correlation between GdPC/dFdCTP ratios and cytidine triphosphate (CTP). In tumour homogenates in vitro, CTP inhibited GdPC formation from dFdCTP, indicating competition between CTP and dFdCTP for CTP:phosphocholine cytidylyltransferase (CCT). As the structure of GdPC precludes entry into cells, potential cytotoxicity was assessed by stimulating CCT activity using linoleate in KPC cells in vitro, leading to increased GdPC concentration and synergistic growth inhibition after dFdC addition. CONCLUSIONS GdPC is an important element of the intra-tumoural dFdC metabolic pathway in vivo.
Collapse
Affiliation(s)
- T E Bapiro
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - K K Frese
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - A Courtin
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - J L Bramhall
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - B Madhu
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - N Cook
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - A Neesse
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - J R Griffiths
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - D A Tuveson
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, USA
| | - D I Jodrell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| | - F M Richards
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Box 278, Robinson Way, Cambridge CB2 0RE, UK
| |
Collapse
|
35
|
Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: an open-label, multicenter phase 1b study. Target Oncol 2014; 10:111-23. [DOI: 10.1007/s11523-014-0320-2] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/09/2014] [Indexed: 10/25/2022]
|
36
|
Terse P, Engelke K, Chan K, Ling Y, Sharpnack D, Saunthararajah Y, Covey JM. Subchronic oral toxicity study of decitabine in combination with tetrahydrouridine in CD-1 mice. Int J Toxicol 2014; 33:75-85. [PMID: 24639139 PMCID: PMC4001115 DOI: 10.1177/1091581814524994] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Decitabine (5-aza-2'-deoxycytidine; DAC) in combination with tetrahydrouridine (THU) is a potential oral therapy for sickle cell disease and β-thalassemia. A study was conducted in mice to assess safety of this combination therapy using oral gavage of DAC and THU administered 1 hour prior to DAC on 2 consecutive days/week for up to 9 weeks followed by a 28-day recovery to support its clinical trials up to 9-week duration. Tetrahydrouridine, a competitive inhibitor of cytidine deaminase, was used in the combination to improve oral bioavailability of DAC. Doses were 167 mg/kg THU followed by 0, 0.2, 0.4, or 1.0 mg/kg DAC; THU vehicle followed by 1.0 mg/kg DAC; or vehicle alone. End points evaluated were clinical observations, body weights, food consumption, clinical pathology, gross/histopathology, bone marrow micronuclei, and toxicokinetics. There were no treatment-related effects noticed on body weight, food consumption, serum chemistry, or urinalysis parameters. Dose- and gender-dependent changes in plasma DAC levels were observed with a Cmax within 1 hour. At the 1 mg/kg dose tested, THU increased DAC plasma concentration (∼ 10-fold) as compared to DAC alone. Severe toxicity occurred in females receiving high-dose 1 mg/kg DAC + THU, requiring treatment discontinuation at week 5. Severity and incidence of microscopic findings increased in a dose-dependent fashion; findings included bone marrow hypocellularity (with corresponding hematologic changes and decreases in white blood cells, red blood cells, hemoglobin, hematocrit, reticulocytes, neutrophils, and lymphocytes), thymic/lymphoid depletion, intestinal epithelial apoptosis, and testicular degeneration. Bone marrow micronucleus analysis confirmed bone marrow cytotoxicity, suppression of erythropoiesis, and genotoxicity. Following the recovery period, a complete or trend toward resolution of these effects was observed. In conclusion, the combination therapy resulted in an increased sensitivity to DAC toxicity correlating with DAC plasma levels, and females are more sensitive compared to their male counterparts.
Collapse
Affiliation(s)
- Pramod Terse
- Division of Pre-Clinical Innovations, National Center for Advancing Translational Sciences, Bethesda, MD, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Ferraris D, Duvall B, Delahanty G, Mistry B, Alt J, Rojas C, Rowbottom C, Sanders K, Schuck E, Huang KC, Redkar S, Slusher BB, Tsukamoto T. Design, Synthesis, and Pharmacological Evaluation of Fluorinated Tetrahydrouridine Derivatives as Inhibitors of Cytidine Deaminase. J Med Chem 2014; 57:2582-8. [DOI: 10.1021/jm401856k] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dana Ferraris
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| | - Bridget Duvall
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| | - Greg Delahanty
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| | - Bipin Mistry
- Eisai Inc., Baltimore, Maryland 21224, United States
| | - Jesse Alt
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| | - Camilo Rojas
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| | | | | | - Edgar Schuck
- Eisai Inc., Andover, Massachusetts 01810, United States
| | | | - Sanjeev Redkar
- Astex Pharmaceuticals, Inc., Dublin, California 94568, United States
| | - Barbara B. Slusher
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| | - Takashi Tsukamoto
- Eisai Inc., Baltimore, Maryland 21224, United States
- Brain
Science Institute and Department of Neurology, Johns Hopkins University, 855 North Wolfe Street, Suite 231, Baltimore, Maryland 21205, United States
| |
Collapse
|
38
|
Slusarczyk M, Lopez MH, Balzarini J, Mason M, Jiang WG, Blagden S, Thompson E, Ghazaly E, McGuigan C. Application of ProTide technology to gemcitabine: a successful approach to overcome the key cancer resistance mechanisms leads to a new agent (NUC-1031) in clinical development. J Med Chem 2014; 57:1531-42. [PMID: 24471998 DOI: 10.1021/jm401853a] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gemcitabine is a nucleoside analogue commonly used in cancer therapy but with limited efficacy due to a high susceptibility to cancer cell resistance. The addition of a phosphoramidate motif to the gemcitabine can protect it against many of the key cancer resistance mechanisms. We have synthesized a series of gemcitabine phosphoramidate prodrugs and screened for cytostatic activity in a range of different tumor cell lines. Among the synthesized compounds, one in particular (NUC-1031, 6f) was shown to be potent in vitro. Importantly, compared with gemcitabine, 6f activation was significantly less dependent on deoxycytidine kinase and on nucleoside transporters, and it was resistant to cytidine deaminase-mediated degradation. Moreover, 6f showed a significant reduction in tumor volumes in vivo in pancreatic cancer xenografts. The ProTide 6f is now in clinical development with encouraging efficacy signals in a Phase I/II study, which strongly supports the ProTide approach to generate promising new anticancer agents.
Collapse
Affiliation(s)
- Magdalena Slusarczyk
- Cardiff School of Pharmacy & Pharmaceutical Sciences, Cardiff University , King Edward VII Avenue, Cardiff CF10 3NB, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Neesse A, Frese KK, Bapiro TE, Nakagawa T, Sternlicht MD, Seeley TW, Pilarsky C, Jodrell DI, Spong SM, Tuveson DA. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proc Natl Acad Sci U S A 2013; 110:12325-30. [PMID: 23836645 PMCID: PMC3725120 DOI: 10.1073/pnas.1300415110] [Citation(s) in RCA: 214] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is characterized by abundant desmoplasia and poor tissue perfusion. These features are proposed to limit the access of therapies to neoplastic cells and blunt treatment efficacy. Indeed, several agents that target the PDA tumor microenvironment promote concomitant chemotherapy delivery and increased antineoplastic response in murine models of PDA. Prior studies could not determine whether chemotherapy delivery or microenvironment modulation per se were the dominant features in treatment response, and such information could guide the optimal translation of these preclinical findings to patients. To distinguish between these possibilities, we used a chemical inhibitor of cytidine deaminase to stabilize and thereby artificially elevate gemcitabine levels in murine PDA tumors without disrupting the tumor microenvironment. Additionally, we used the FG-3019 monoclonal antibody (mAb) that is directed against the pleiotropic matricellular signaling protein connective tissue growth factor (CTGF/CCN2). Inhibition of cytidine deaminase raised the levels of activated gemcitabine within PDA tumors without stimulating neoplastic cell killing or decreasing the growth of tumors, whereas FG-3019 increased PDA cell killing and led to a dramatic tumor response without altering gemcitabine delivery. The response to FG-3019 correlated with the decreased expression of a previously described promoter of PDA chemotherapy resistance, the X-linked inhibitor of apoptosis protein. Therefore, alterations in survival cues following targeting of tumor microenvironmental factors may play an important role in treatment responses in animal models, and by extension in PDA patients.
Collapse
Affiliation(s)
- Albrecht Neesse
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Department of Gastroenterology, Endocrinology, and Metabolism, Philipps University Marburg, 35043 Marburg, Germany
| | - Kristopher K. Frese
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Tashinga E. Bapiro
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Tomoaki Nakagawa
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | | | | | - Christian Pilarsky
- Department of General, Thoracic, and Vascular Surgery, University Hospital Carl Gustav Carus, Technical University Dresden, 01307 Dresden, Germany; and
| | - Duncan I. Jodrell
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Department of Oncology, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | | | - David A. Tuveson
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| |
Collapse
|
40
|
Singh S, Chitkara D, Kumar V, Behrman SW, Mahato RI. miRNA profiling in pancreatic cancer and restoration of chemosensitivity. Cancer Lett 2013; 334:211-20. [DOI: 10.1016/j.canlet.2012.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 09/26/2012] [Accepted: 10/07/2012] [Indexed: 12/13/2022]
|
41
|
Ebrahem Q, Mahfouz RZ, Ng KP, Saunthararajah Y. High cytidine deaminase expression in the liver provides sanctuary for cancer cells from decitabine treatment effects. Oncotarget 2013; 3:1137-45. [PMID: 23087155 PMCID: PMC3717944 DOI: 10.18632/oncotarget.597] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We document for the first time that sanctuary in an organ which expresses high levels of the enzyme cytidine deaminase (CDA) is a mechanism of cancer cell resistance to cytidine analogues. This mechanism could explain why historically, cytidine analogues have not been successful chemotherapeutics against hepatotropic cancers, despite efficacy in vitro. Importantly, this mechanism of resistance can be readily reversed, without increasing toxicity to sensitive organs, by combining cytidine analogue with an inhibitor of cytidine deaminase (tetrahydrouridine). Specifically, CDA rapidly metabolizes cytidine analogues into inactive uridine counterparts. Hence, to determine if sheltering/protection of cancer cells in organs which express high levels of CDA (e.g., liver) is a mechanism of resistance, we utilized a murine xenotransplant model of myeloid cancer that is sensitive to epigenetic therapeutic effects of the cytidine analogue decitabine in vitro and hepato-tropic in vivo. Treatment of tumor-bearing mice with decitabine (subcutaneous 0.2mg/kg 2X/week) doubled median survival and significantly decreased extra-hepatic tumor burden, but hepatic tumor burden remained substantial, to which the animals eventually succumbed. Combining a clinically-relevant inhibitor of CDA (tetrahydrouridine) with a lower dose of decitabine (subcutaneous 0.1mg/kg 2X/week) markedly decreased liver tumor burden without blood count or bone marrow evidence of myelotoxicity, and with further improvement in survival. In conclusion, sanctuary in a CDA-rich organ is a mechanism by which otherwise susceptible cancer cells can resist the effects of decitabine epigenetic therapy. This protection can be reversed without increasing myelotoxicity by combining tetrahydrouridine with a lower dose of decitabine.
Collapse
Affiliation(s)
- Quteba Ebrahem
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | |
Collapse
|
42
|
Dasari M, Acharya AP, Kim D, Lee S, Lee S, Rhea J, Molinaro R, Murthy N. H-gemcitabine: a new gemcitabine prodrug for treating cancer. Bioconjug Chem 2012; 24:4-8. [PMID: 23237198 DOI: 10.1021/bc300095m] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
In this report, we present a new strategy for targeting chemotherapeutics to tumors, based on targeting extracellular DNA. A gemcitabine prodrug was synthesized, termed H-gemcitabine, which is composed of Hoechst conjugated to gemcitabine. H-gemcitabine has low toxicity because it is membrane-impermeable; however, it still has high tumor efficacy because of its ability to target gemcitabine to E-DNA in tumors. We demonstrate here that H-gemcitabine has a wider therapeutic window than free gemcitabine.
Collapse
Affiliation(s)
- Madhuri Dasari
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Parker H. Petit Institute for Bioengineering and Bioscience, 315 Ferst Drive, Atlanta, GA 30332, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kahramanoğullari O, Fantaccini G, Lecca P, Morpurgo D, Priami C. Algorithmic modeling quantifies the complementary contribution of metabolic inhibitions to gemcitabine efficacy. PLoS One 2012; 7:e50176. [PMID: 23239976 PMCID: PMC3519828 DOI: 10.1371/journal.pone.0050176] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 10/22/2012] [Indexed: 01/19/2023] Open
Abstract
Gemcitabine (2,2-difluorodeoxycytidine, dFdC) is a prodrug widely used for treating various carcinomas. Gemcitabine exerts its clinical effect by depleting the deoxyribonucleotide pools, and incorporating its triphosphate metabolite (dFdC-TP) into DNA, thereby inhibiting DNA synthesis. This process blocks the cell cycle in the early S phase, eventually resulting in apoptosis. The incorporation of gemcitabine into DNA takes place in competition with the natural nucleoside dCTP. The mechanisms of indirect competition between these cascades for common resources are given with the race for DNA incorporation; in clinical studies dedicated to singling out mechanisms of resistance, ribonucleotide reductase (RR) and deoxycytidine kinase (dCK) and human equilibrative nucleoside transporter1 (hENT1) have been associated to efficacy of gemcitabine with respect to their roles in the synthesis cascades of dFdC-TP and dCTP. However, the direct competition, which manifests itself in terms of inhibitions between these cascades, remains to be quantified. We propose an algorithmic model of gemcitabine mechanism of action, verified with respect to independent experimental data. We performed in silico experiments in different virtual conditions, otherwise difficult in vivo, to evaluate the contribution of the inhibitory mechanisms to gemcitabine efficacy. In agreement with the experimental data, our model indicates that the inhibitions due to the association of dCTP with dCK and the association of gemcitabine diphosphate metabolite (dFdC-DP) with RR play a key role in adjusting the efficacy. While the former tunes the catalysis of the rate-limiting first phosphorylation of dFdC, the latter is responsible for depletion of dCTP pools, thereby contributing to gemcitabine efficacy with a dependency on nucleoside transport efficiency. Our simulations predict the existence of a continuum of non-efficacy to high-efficacy regimes, where the levels of dFdC-TP and dCTP are coupled in a complementary manner, which can explain the resistance to this drug in some patients.
Collapse
Affiliation(s)
- Ozan Kahramanoğullari
- The Microsoft Research-University of Trento Centre for Computational and Systems Biology, Rovereto (Trento), Italy.
| | | | | | | | | |
Collapse
|
44
|
Hao WH, Wang JJ, Hsueh SP, Hsu PJ, Chang LC, Hsu CS, Hsu KY. In vitro and in vivo studies of pharmacokinetics and antitumor efficacy of D07001-F4, an oral gemcitabine formulation. Cancer Chemother Pharmacol 2012; 71:379-88. [PMID: 23143189 DOI: 10.1007/s00280-012-2017-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 10/21/2012] [Indexed: 12/01/2022]
Abstract
PURPOSE The chemotherapy agent gemcitabine is currently administered intravenously because the drug has poor oral bioavailability. In order to assess the pharmacokinetics and antitumor activity of D07001-F4, a new self-microemulsifying oral drug delivery system preparation of gemcitabine, this study was performed to compare the effect of D07001-F4 with administered gemcitabine in vitro and in vivo. METHODS D07001-F4 pharmacokinetics was examined by evaluation of in vitro deamination of D07001-F4 and gemcitabine hydrochloride by recombinant human cytidine deaminase (rhCDA) and in vivo evaluation of D07001-F4 pharmacokinetics in mice. Antitumor activity was evaluated by comparing the effect of D07001-F4 and gemcitabine hydrochloride in inhibiting growth in nine cancer cell lines and by examining the effect of D07001-F4 and gemcitabine in two xenograft tumor models in mice. RESULTS In vitro deamination of D07001-F4 by rhCDA was 3.3-fold slower than deamination of gemcitabine hydrochloride. Growth inhibition by D07001-F4 of 7 of the 8 cancer cell lines was increased compared with that seen with gemcitabine hydrochloride, and D07001-F4 inhibited the growth of pancreatic and colon cancer xenografts. In vivo pharmacokinetics showed the oral bioavailability of D07001-F4 to be 34%. CONCLUSIONS D07001-F4 was effective against several cancer types, was metabolized more slowly than gemcitabine hydrochloride, and exhibited enhanced oral bioavailability.
Collapse
Affiliation(s)
- Wei-Hua Hao
- InnoPharmax Inc, 9F, No 22, Lane 478, Rueiguang Rd, Neihu District, Taipei 11492, Taiwan
| | | | | | | | | | | | | |
Collapse
|
45
|
Funamizu N, Lacy CR, Fujita K, Furukawa K, Misawa T, Yanaga K, Manome Y. Tetrahydrouridine inhibits cell proliferation through cell cycle regulation regardless of cytidine deaminase expression levels. PLoS One 2012; 7:e37424. [PMID: 22616006 PMCID: PMC3353937 DOI: 10.1371/journal.pone.0037424] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 04/21/2012] [Indexed: 12/24/2022] Open
Abstract
Tetrahydrouridine (THU) is a well characterized and potent inhibitor of cytidine deaminase (CDA). Highly expressed CDA catalyzes and inactivates cytidine analogues, ultimately contributing to increased gemcitabine resistance. Therefore, a combination therapy of THU and gemcitabine is considered to be a potential and promising treatment for tumors with highly expressed CDA. In this study, we found that THU has an alternative mechanism for inhibiting cell growth which is independent of CDA expression. Three different carcinoma cell lines (MIAPaCa-2, H441, and H1299) exhibited decreased cell proliferation after sole administration of THU, while being unaffected by knocking down CDA. To investigate the mechanism of THU-induced cell growth inhibition, cell cycle analysis using flow cytometry was performed. This analysis revealed that THU caused an increased rate of G1-phase occurrence while S-phase occurrence was diminished. Similarly, Ki-67 staining further supported that THU reduces cell proliferation. We also found that THU regulates cell cycle progression at the G1/S checkpoint by suppressing E2F1. As a result, a combination regimen of THU and gemcitabine might be a more effective therapy than previously believed for pancreatic carcinoma since THU works as a CDA inhibitor, as well as an inhibitor of cell growth in some types of pancreatic carcinoma cells.
Collapse
Affiliation(s)
- Naotake Funamizu
- Department of Molecular Cell Biology, Institute of DNA Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Frese KK, Neesse A, Cook N, Bapiro TE, Lolkema MP, Jodrell DI, Tuveson DA. nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. Cancer Discov 2012; 2:260-269. [PMID: 22585996 PMCID: PMC4866937 DOI: 10.1158/2159-8290.cd-11-0242] [Citation(s) in RCA: 332] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Nanoparticle albumin-bound (nab)-paclitaxel, an albumin-stabilized paclitaxel formulation, demonstrates clinical activity when administered in combination with gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma (PDA). The limited availability of patient tissue and exquisite sensitivity of xenografts to chemotherapeutics have limited our ability to address the mechanistic basis of this treatment regimen. Here, we used a mouse model of PDA to show that the coadministration of nab-paclitaxel and gemcitabine uniquely demonstrates evidence of tumor regression. Combination treatment increases intratumoral gemcitabine levels attributable to a marked decrease in the primary gemcitabine metabolizing enzyme, cytidine deaminase. Correspondingly, paclitaxel reduced the levels of cytidine deaminase protein in cultured cells through reactive oxygen species-mediated degradation, resulting in the increased stabilization of gemcitabine. Our findings support the concept that suboptimal intratumoral concentrations of gemcitabine represent a crucial mechanism of therapeutic resistance in PDA and highlight the advantages of genetically engineered mouse models in preclinical therapeutic trials. SIGNIFICANCE This study provides mechanistic insight into the clinical cooperation observed between gemcitabine and nab-paclitaxel in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Kristopher K. Frese
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
| | - Albrecht Neesse
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
- Department of Gastroenterology, Endocrinology and Metabolism, Philipps University Marburg, Baldingerstr, 35043 Marburg, Germany
| | - Natalie Cook
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Tashinga E. Bapiro
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Martijn P. Lolkema
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Duncan I. Jodrell
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - David A. Tuveson
- Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge, UK
- Department of Oncology, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Lavelle D, Vaitkus K, Ling Y, Ruiz MA, Mahfouz R, Ng KP, Negrotto S, Smith N, Terse P, Engelke KJ, Covey J, Chan KK, Desimone J, Saunthararajah Y. Effects of tetrahydrouridine on pharmacokinetics and pharmacodynamics of oral decitabine. Blood 2012; 119:1240-7. [PMID: 22160381 PMCID: PMC3277356 DOI: 10.1182/blood-2011-08-371690] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 12/05/2011] [Indexed: 12/24/2022] Open
Abstract
The deoxycytidine analog decitabine (DAC) can deplete DNA methyl-transferase 1 (DNMT1) and thereby modify cellular epigenetics, gene expression, and differentiation. However, a barrier to efficacious and accessible DNMT1-targeted therapy is cytidine deaminase, an enzyme highly expressed in the intestine and liver that rapidly metabolizes DAC into inactive uridine counterparts, severely limiting exposure time and oral bioavailability. In the present study, the effects of tetrahydrouridine (THU), a competitive inhibitor of cytidine deaminase, on the pharmacokinetics and pharmacodynamics of oral DAC were evaluated in mice and nonhuman primates. Oral administration of THU before oral DAC extended DAC absorption time and widened the concentration-time profile, increasing the exposure time for S-phase-specific depletion of DNMT1 without the high peak DAC levels that can cause DNA damage and cytotoxicity. THU also decreased interindividual variability in pharmacokinetics seen with DAC alone. One potential clinical application of DNMT1-targeted therapy is to increase fetal hemoglobin and treat hemoglobinopathy. Oral THU-DAC at a dose that would produce peak DAC concentrations of less than 0.2μM administered 2×/wk for 8 weeks to nonhuman primates was not myelotoxic, hypomethylated DNA in the γ-globin gene promoter, and produced large cumulative increases in fetal hemoglobin. Combining oral THU with oral DAC changes DAC pharmacology in a manner that may facilitate accessible noncytotoxic DNMT1-targeted therapy.
Collapse
Affiliation(s)
- Donald Lavelle
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Saunthararajah Y, Triozzi P, Rini B, Singh A, Radivoyevitch T, Sekeres M, Advani A, Tiu R, Reu F, Kalaycio M, Copelan E, Hsi E, Lichtin A, Bolwell B. p53-Independent, normal stem cell sparing epigenetic differentiation therapy for myeloid and other malignancies. Semin Oncol 2012; 39:97-108. [PMID: 22289496 PMCID: PMC3655437 DOI: 10.1053/j.seminoncol.2011.11.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cytotoxic chemotherapy for acute myeloid leukemia (AML) usually produces only temporary remissions, at the cost of significant toxicity and risk for death. One fundamental reason for treatment failure is that it is designed to activate apoptosis genes (eg, TP53) that may be unavailable because of mutation or deletion. Unlike deletion of apoptosis genes, genes that mediate cell cycle exit by differentiation are present in myelodysplastic syndrome (MDS) and AML cells but are epigenetically repressed: MDS/AML cells express high levels of key lineage-specifying transcription factors. Mutations in these transcription factors (eg, CEBPA) or their cofactors (eg., RUNX1) affect transactivation function and produce epigenetic repression of late-differentiation genes that antagonize MYC. Importantly, this aberrant epigenetic repression can be redressed clinically by depleting DNA methyltransferase 1 (DNMT1, a central component of the epigenetic network that mediates transcription repression) using the deoxycytidine analogue decitabine at non-cytotoxic concentrations. The DNMT1 depletion is sufficient to trigger upregulation of late-differentiation genes and irreversible cell cycle exit by p53-independent differentiation mechanisms. Fortuitously, the same treatment maintains or increases self-renewal of normal hematopoietic stem cells, which do not express high levels of lineage-specifying transcription factors. The biological rationale for this approach to therapy appears to apply to cancers other than MDS/AML also. Decitabine or 5-azacytidine dose and schedule can be rationalized to emphasize this mechanism of action, as an alternative or complement to conventional apoptosis-based oncotherapy.
Collapse
Affiliation(s)
- Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jansen RS, Rosing H, Schellens JHM, Beijnen JH. Deoxyuridine analog nucleotides in deoxycytidine analog treatment: secondary active metabolites? Fundam Clin Pharmacol 2011; 25:172-85. [PMID: 20199587 DOI: 10.1111/j.1472-8206.2010.00823.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Deoxycytidine analogs (dCa's) are nucleosides widely used in anticancer and anti (retro) viral therapies. Intracellularly phosphorylated dCa anabolites are considered to be their main active metabolites. This article reviews the literature on the formation and pharmacological activity of deaminated dCa nucleotides. Most dCa's are rapidly deaminated into deoxyuridine analogs (dUa's) which are only slowly phosphorylated and therefore relatively inactive. dUa nucleotides are, however, also formed via deamination of dCa monophosphates by deoxycytidine monophosphate deaminase (dCMPD). dUa-monophosphates can interact with thymidylate synthase (TS), whereas dUa-triphosphates are incorporated into nucleic acids and interfere with polymerases. Administration of dCa's as monophosphate prodrugs or co-administration of the cytidine deaminase inhibitor tetrahydrouridine (THU) does not prevent dUa nucleotide formation which is, on the other hand, influenced by the dose and dCMPD activity. Taken together, these observations show that the formation of dUa nucleotides is a common phenomenon in treatment with dCa's and these compounds may play a role in treatment outcome. We conclude that more attention should be given to these relatively unknown, but potentially important metabolites.
Collapse
Affiliation(s)
- Robert S Jansen
- Department of Pharmacy & Pharmacology, Slotervaart Hospital/The Netherlands Cancer Institute, Louwesweg 6, 1066 EC Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
50
|
Hodge L, Taub M, Tracy T. Effect of its deaminated metabolite, 2′,2′-difluorodeoxyuridine, on the transport and toxicity of gemcitabine in HeLa cells. Biochem Pharmacol 2011; 81:950-6. [DOI: 10.1016/j.bcp.2011.01.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Revised: 01/19/2011] [Accepted: 01/20/2011] [Indexed: 11/28/2022]
|