101
|
Dávila B, Sánchez C, Fernández M, Cerecetto H, Lecot N, Cabral P, Glisoni R, González M. Selective Hypoxia‐Cytotoxin 7‐Fluoro‐2‐Aminophenazine 5,10‐Dioxide: Toward “Candidate‐to‐Drug” Stage in the Drug‐Development Pipeline. ChemistrySelect 2019. [DOI: 10.1002/slct.201902601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Belén Dávila
- Laboratorio de Química Orgánica MedicinalInstituto de Química Biológica. Facultad de CienciasUniversidad de la República. Iguá 4225 Montevideo 11400 Uruguay
| | - Carina Sánchez
- Laboratorio de Química Orgánica MedicinalInstituto de Química Biológica. Facultad de CienciasUniversidad de la República. Iguá 4225 Montevideo 11400 Uruguay
| | - Marcelo Fernández
- Laboratorio de Experimentación AnimalCentro de Investigaciones Nucleares. Facultad de CienciasUniversidad de la República. Mataojo 2055 Montevideo 11400 Uruguay
| | - Hugo Cerecetto
- Laboratorio de Química Orgánica MedicinalInstituto de Química Biológica. Facultad de CienciasUniversidad de la República. Iguá 4225 Montevideo 11400 Uruguay
- Área de RadiofarmaciaCentro de Investigaciones Nucleares. Facultad de CienciasUniversidad de la República. Mataojo 2055 Montevideo 11400 Uruguay
| | - Nicole Lecot
- Laboratorio de Química Orgánica MedicinalInstituto de Química Biológica. Facultad de CienciasUniversidad de la República. Iguá 4225 Montevideo 11400 Uruguay
- Laboratorio de Técnicas Nucleareas Aplicadas a Bioquímica y BiotecnologíaCentro de Investigaciones Nucleares. Facultad de CienciasUniversidad de la República. Mataojo 2055 Montevideo 11400 Uruguay
| | - Pablo Cabral
- Área de RadiofarmaciaCentro de Investigaciones Nucleares. Facultad de CienciasUniversidad de la República. Mataojo 2055 Montevideo 11400 Uruguay
| | - Romina Glisoni
- Departamento de Tecnología FarmacéuticaCátedra de Tecnología Farmacéutica II. CONICETInstituto de Nanobiotecnología (NANOBIOTEC). Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires
| | - Mercedes González
- Laboratorio de Química Orgánica MedicinalInstituto de Química Biológica. Facultad de CienciasUniversidad de la República. Iguá 4225 Montevideo 11400 Uruguay
| |
Collapse
|
102
|
Diethelm-Varela B, Ai Y, Liang D, Xue F. Nitrogen Mustards as Anticancer Chemotherapies: Historic Perspective, Current Developments and Future Trends. Curr Top Med Chem 2019; 19:691-712. [PMID: 30931858 DOI: 10.2174/1568026619666190401100519] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/11/2019] [Accepted: 02/25/2019] [Indexed: 12/30/2022]
Abstract
Nitrogen mustards, a family of DNA alkylating agents, marked the start of cancer pharmacotherapy. While traditionally characterized by their dose-limiting toxic effects, nitrogen mustards have been the subject of intense research efforts, which have led to safer and more effective agents. Even though the alkylating prodrug mustards were first developed decades ago, active research on ways to improve their selectivity and cytotoxic efficacy is a currently active topic of research. This review addresses the historical development of the nitrogen mustards, outlining their mechanism of action, and discussing the improvements on their therapeutic profile made through rational structure modifications. A special emphasis is made on discussing the nitrogen mustard prodrug category, with Cyclophosphamide (CPA) serving as the main highlight. Selected insights on the latest developments on nitrogen mustards are then provided, limiting such information to agents that preserve the original nitrogen mustard mechanism as their primary mode of action. Additionally, future trends that might follow in the quest to optimize these invaluable chemotherapeutic medications are succinctly suggested.
Collapse
Affiliation(s)
- Benjamin Diethelm-Varela
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Yong Ai
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
103
|
Mao X, McManaway S, Jaiswal JK, Hong CR, Wilson WR, Hicks KO. Schedule-dependent potentiation of chemotherapy drugs by the hypoxia-activated prodrug SN30000. Cancer Biol Ther 2019; 20:1258-1269. [PMID: 31131698 DOI: 10.1080/15384047.2019.1617570] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hypoxia-activated prodrugs (HAPs) are hypothesized to improve the therapeutic index of chemotherapy drugs that are ineffective against tumor cells in hypoxic microenvironments. SN30000 (CEN-209) is a benzotriazine di-N-oxide HAP that potentiates radiotherapy in preclinical models, but its combination with chemotherapy has not been explored. Here we apply multiple models (monolayers, multicellular spheroids and tumor xenografts) to identify promising SN30000/chemotherapy combinations (with chemotherapy drugs before, during or after SN30000 exposure). SN30000, unlike doxorubicin, cisplatin, gemcitabine or paclitaxel, was more active against cells in spheroids than monolayers by clonogenic assay. Combinations of SN30000 and chemotherapy drugs in HCT116/GFP and SiHa spheroids demonstrated hypoxia-and schedule-dependent potentiation of gemcitabine or doxorubicin in growth inhibition and clonogenic assays. Co-administration with SN30000 suppressed clearance of gemcitabine in NIH-III mice, likely due to SN30000-induced hypothermia which also modulated extravascular transport of gemcitabine in tumor tissue as assessed from its diffusion through HCT116 multicellular layer cultures. Despite these systemic effects, the same schedules that gave therapeutic synergy in spheroids (SN30000 3 h before or during gemcitabine, but not gemcitabine 3 h before SN30000) enhanced growth delay of HCT116 xenografts without increasing host toxicity. Identification of hypoxic and S-phase cells by immunohistochemistry and flow cytometry established that hypoxic cells initially spared by gemcitabine subsequently reoxygenate and re-enter the cell cycle, and that this repopulation is prevented by SN30000 only when administered with or before gemcitabine. This illustrates the value of spheroids in modeling tumor microenvironment-dependent drug interactions, and the potential of HAPs for overcoming hypoxia-mediated drug resistance.
Collapse
Affiliation(s)
- Xinjian Mao
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Sarah McManaway
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Jagdish K Jaiswal
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - Cho R Hong
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland , Auckland , New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, University of Auckland , Auckland , New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland , Auckland , New Zealand
| |
Collapse
|
104
|
Rumjanek VM, Maia RC, Salustiano EJ, Costa PR. Insights into the Biological Evaluation of Pterocarpanquinones and Carbapterocarpans with Anti-tumor Activity against MDR Leukemias. Anticancer Agents Med Chem 2019; 19:29-37. [DOI: 10.2174/1871520618666180420165128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 03/17/2018] [Accepted: 03/20/2018] [Indexed: 12/13/2022]
Abstract
In an attempt to find anticancer agents that could overcome multidrug resistance (MDR), two new
classes of modified isoflavonoids were designed and synthesized, and their effectiveness evaluated against a
vast array of tumor cell lines. Pterocarpanquinone (LQB-118) and 11a-aza-5-carbapterocarpan (LQB-223) were
the most promising. LQB-118 induced cell death, in vitro, in the µM range, to a number of human cancer cell
lines as well as to fresh tumor cells obtained from patients with acute or chronic myeloid leukemia, independent
on whether they exhibit the MDR phenotype or not. Furthermore, leukemic cells were more sensitive to LQB-
118 compared to cells from solid tumors. Given to mice, in vivo, LQB-118 affected the growth of melanoma,
Ehrlich carcinoma and prostate cancer cells. Conversely, no general toxicity was observed in vivo, by biochemical,
hematological, anatomical or histological parameters and toxicity in vitro against normal cells was low. The
process involved in tumor cell death seemed to vary according to cell type. Apoptosis was studied by externalization
of phosphatidylserine, DNA fragmentation, caspase-3 activation, reduced expression of XIAP and survivin,
ER stress, cytosolic calcium increase and mitochondrial membrane depolarization. Autophagy was also
evaluated inhibiting caspase-9, with no effect observed in beclin 1, whereas pre-treatment with rapamycin increased
cytotoxicity induced by LQB-118. In addition, LQB-118 increased ROS, inhibited NFκB nuclear translocation
and secretion of TNF-α, modulated microRNAs miR-9 and miR-21 and modified the cell cycle. Despite
being less studied, the cytotoxic effect of the 11a-aza-5-carbapterocarpan LQB-223 was present against several
tumor cell lines, including those with the MDR phenotype.
Collapse
Affiliation(s)
- Vivian M. Rumjanek
- Laboratorio de Imunologia Tumoral, Instituto de Bioquimica Médica Leopoldo de Meis (IBqM), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel C. Maia
- Laboratorio de Hemato-Oncologia Celular e Molecular, Programa de Hemato-Oncologia Molecular, Instituto Nacional de Cancer (INCA), Rio de Janeiro, Brazil
| | - Eduardo J. Salustiano
- Laboratorio de Imunologia Tumoral, Instituto de Bioquimica Médica Leopoldo de Meis (IBqM), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo R.R. Costa
- Laboratorio de Quimica Bio-organica (LQB), Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
105
|
Lee HH, Dickson BD, Stevenson RJ, Yang S, Tercel M. Optimised synthesis of a nitroCBI hypoxia-activated prodrug with substantial anticancer activity. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
106
|
Studies Towards Hypoxia-Activated Prodrugs of PARP Inhibitors. Molecules 2019; 24:molecules24081559. [PMID: 31010230 PMCID: PMC6514732 DOI: 10.3390/molecules24081559] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 01/21/2023] Open
Abstract
Poly(ADP-ribose)polymerase (PARP) inhibitors (PARPi) have recently been approved for the treatment of breast and ovarian tumors with defects in homologous recombination repair (HRR). Although it has been demonstrated that PARPi also sensitize HRR competent tumors to cytotoxic chemotherapies or radiotherapy, normal cell toxicity has remained an obstacle to their use in this context. Hypoxia-activated prodrugs (HAPs) provide a means to limit exposure of normal cells to active drug, thus adding a layer of tumor selectivity. We have investigated potential HAPs of model PARPi in which we attach a bioreducible “trigger” to the amide nitrogen, thereby blocking key binding interactions. A representative example showed promise in abrogating PARPi enzymatic activity in a biochemical assay, with a ca. 160-fold higher potency of benzyl phthalazinone 4 than the corresponding model HAP 5, but these N-alkylated compounds did not release the PARPi upon one-electron reduction by radiolysis. Therefore, we extended our investigation to include NU1025, a PARPi that contains a phenol distal to the core binding motif. The resulting 2-nitroimidazolyl ether provided modest abrogation of PARPi activity with a ca. seven-fold decrease in potency, but released the PARPi efficiently upon reduction. This investigation of potential prodrug approaches for PARPi has identified a useful prodrug strategy for future exploration.
Collapse
|
107
|
Phillips RM, Loadman PM, Reddy G. Inactivation of apaziquone by haematuria: implications for the design of phase III clinical trials against non-muscle invasive bladder cancer. Cancer Chemother Pharmacol 2019; 83:1183-1189. [PMID: 30868237 PMCID: PMC6499894 DOI: 10.1007/s00280-019-03812-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/08/2019] [Indexed: 11/13/2022]
Abstract
Purpose Despite positive responses in phase II clinical trials, the bioreductive prodrug apaziquone failed to achieve statistically significant activity in non-muscle invasive bladder cancer in phase III trials. Apaziquone was administered shortly after transurethral resection and here we test the hypothesis that haematuria inactivates apaziquone. Methods HPLC analysis was used to determine the ability of human whole blood to metabolise apaziquone ex vivo. An in vitro model of haematuria was developed and the response of RT112 and EJ138 cells following a 1-h exposure to apaziquone was determined in the presence of urine plus or minus whole blood or lysed whole blood. Results HPLC analysis demonstrated that apaziquone is metabolised by human whole blood with a half-life of 78.6 ± 23.0 min. As a model for haematuria, incubation of cells in media containing up to 75% buffered (pH 7.4) urine and 25% whole blood was not toxic to cells for a 1-h exposure period. Whole blood (5% v/v) significantly (p < 0.01) reduced the potency of apaziquone in this experimental model. Lysed whole blood also significantly (p < 0.05) reduced cell growth, although higher concentrations were required to achieve an effect (15% v/v). Conclusions The results of this study demonstrate that haematuria can reduce the potency of apaziquone in this experimental model. These findings impact upon the design of further phase III clinical trials and strongly suggest that apaziquone should not be administered immediately after transurethral resection of non-muscle invasive bladder cancer when haematuria is common.
Collapse
Affiliation(s)
- Roger M Phillips
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK.
| | - Paul M Loadman
- Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, UK
| | - Guru Reddy
- Spectrum Pharmaceuticals Inc, Irvine, CA, USA
| |
Collapse
|
108
|
Shen X, Gates KS. Enzyme-Activated Generation of Reactive Oxygen Species from Heterocyclic N-Oxides under Aerobic and Anaerobic Conditions and Its Relevance to Hypoxia-Selective Prodrugs. Chem Res Toxicol 2019; 32:348-361. [PMID: 30817135 DOI: 10.1021/acs.chemrestox.9b00036] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Enzymatic one-electron reduction of heterocyclic N-oxides can lead to the intracellular generation of reactive oxygen species via several different chemical pathways. These reactions may be relevant to hypoxia-selective anticancer drugs, antimicrobial agents, and unwanted toxicity of heterocylic nitrogen compounds.
Collapse
|
109
|
Jackson RK, Liew LP, Hay MP. Overcoming Radioresistance: Small Molecule Radiosensitisers and Hypoxia-activated Prodrugs. Clin Oncol (R Coll Radiol) 2019; 31:290-302. [PMID: 30853148 DOI: 10.1016/j.clon.2019.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/12/2019] [Indexed: 12/25/2022]
Abstract
The role of hypoxia in radiation resistance is well established and many approaches to overcome hypoxia in tumours have been explored, with variable success. Two small molecule strategies for targeting hypoxia have dominated preclinical and clinical efforts. One approach has been the use of electron-affinic nitroheterocycles as oxygen-mimetic sensitisers. These agents are best exemplified by the 5-nitroimidazole nimorazole, which has limited use in conjunction with radiotherapy in head and neck squamous cell carcinoma. The second approach seeks to leverage tumour hypoxia as a tumour-specific address for hypoxia-activated prodrugs. These prodrugs are selectively activated by reductases under hypoxia to release cytotoxins, which in some instances may diffuse to kill surrounding oxic tumour tissue. A number of these hypoxia-activated prodrugs have been examined in clinical trial and the merits and shortcomings of recent examples are discussed. There has been an evolution from delivering DNA-interactive cytotoxins to molecularly targeted agents. Efforts to implement these strategies clinically continue today, but success has been elusive. Several issues have been identified that compromised these clinical campaigns. A failure to consider the extravascular transport and the micropharmacokinetic properties of the prodrugs has reduced efficacy. One key element for these 'targeted' approaches is the need to co-develop biomarkers to identify appropriate patients. Hypoxia-activated prodrugs require biomarkers for hypoxia, but also for appropriate activating reductases in tumours, as well as markers of intrinsic sensitivity to the released drug. The field is still evolving and changes in radiation delivery and the impact of immune-oncology will provide fertile ground for future innovation.
Collapse
Affiliation(s)
- R K Jackson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - L P Liew
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - M P Hay
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
110
|
O'Connor JPB, Robinson SP, Waterton JC. Imaging tumour hypoxia with oxygen-enhanced MRI and BOLD MRI. Br J Radiol 2019; 92:20180642. [PMID: 30272998 PMCID: PMC6540855 DOI: 10.1259/bjr.20180642] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/21/2018] [Accepted: 09/25/2018] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is known to be a poor prognostic indicator for nearly all solid tumours and also is predictive of treatment failure for radiotherapy, chemotherapy, surgery and targeted therapies. Imaging has potential to identify, spatially map and quantify tumour hypoxia prior to therapy, as well as track changes in hypoxia on treatment. At present no hypoxia imaging methods are available for routine clinical use. Research has largely focused on positron emission tomography (PET)-based techniques, but there is gathering evidence that MRI techniques may provide a practical and more readily translational alternative. In this review we focus on the potential for imaging hypoxia by measuring changes in longitudinal relaxation [R1; termed oxygen-enhanced MRI or tumour oxygenation level dependent (TOLD) MRI] and effective transverse relaxation [R2*; termed blood oxygenation level dependent (BOLD) MRI], induced by inhalation of either 100% oxygen or the radiosensitising hyperoxic gas carbogen. We explain the scientific principles behind oxygen-enhanced MRI and BOLD and discuss significant studies and their limitations. All imaging biomarkers require rigorous validation in order to translate into clinical use and the steps required to further develop oxygen-enhanced MRI and BOLD MRI into decision-making tools are discussed.
Collapse
Affiliation(s)
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | |
Collapse
|
111
|
Sharma A, Arambula JF, Koo S, Kumar R, Singh H, Sessler JL, Kim JS. Hypoxia-targeted drug delivery. Chem Soc Rev 2019; 48:771-813. [PMID: 30575832 PMCID: PMC6361706 DOI: 10.1039/c8cs00304a] [Citation(s) in RCA: 324] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hypoxia is a state of low oxygen tension found in numerous solid tumours. It is typically associated with abnormal vasculature, which results in a reduced supply of oxygen and nutrients, as well as impaired delivery of drugs. The hypoxic nature of tumours often leads to the development of localized heterogeneous environments characterized by variable oxygen concentrations, relatively low pH, and increased levels of reactive oxygen species (ROS). The hypoxic heterogeneity promotes tumour invasiveness, metastasis, angiogenesis, and an increase in multidrug-resistant proteins. These factors decrease the therapeutic efficacy of anticancer drugs and can provide a barrier to advancing drug leads beyond the early stages of preclinical development. This review highlights various hypoxia-targeted and activated design strategies for the formulation of drugs or prodrugs and their mechanism of action for tumour diagnosis and treatment.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Chemistry, Korea University, Seoul, 02841, Korea.
| | | | | | | | | | | | | |
Collapse
|
112
|
Liew LP, Singleton DC, Wong WW, Cheng GJ, Jamieson SMF, Hay MP. Hypoxia‐Activated Prodrugs of PERK Inhibitors. Chem Asian J 2019; 14:1238-1248. [DOI: 10.1002/asia.201801826] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/04/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Lydia P. Liew
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
| | - Dean C. Singleton
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
| | - Way W. Wong
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
| | - Gary J. Cheng
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
| | - Stephen M. F. Jamieson
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
| | - Michael P. Hay
- Auckland Cancer Society Research Centre, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of Auckland Private Bag 92019 Auckland New Zealand
- Maurice Wilkins Centre for Molecular BiodiscoveryUniversity of Auckland Symonds St Auckland 1010 New Zealand
| |
Collapse
|
113
|
Spiegelberg L, Houben R, Niemans R, de Ruysscher D, Yaromina A, Theys J, Guise CP, Smaill JB, Patterson AV, Lambin P, Dubois LJ. Hypoxia-activated prodrugs and (lack of) clinical progress: The need for hypoxia-based biomarker patient selection in phase III clinical trials. Clin Transl Radiat Oncol 2019; 15:62-69. [PMID: 30734002 PMCID: PMC6357685 DOI: 10.1016/j.ctro.2019.01.005] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 01/07/2023] Open
Abstract
Hypoxia-activated prodrugs have yielded promising results up to phase II trials. Implementation of hypoxia-activated prodrugs in the clinic has not been successful. Phase III clinical trials lack patient stratification based on tumor hypoxia status. Stratification will decrease the number of patients needed and increase success. Improvements in hypoxia-activated prodrug design can also increase success rates.
Hypoxia-activated prodrugs (HAPs) are designed to specifically target the hypoxic cells of tumors, which are an important cause of treatment resistance to conventional therapies. Despite promising preclinical and clinical phase I and II results, the most important of which are described in this review, the implementation of hypoxia-activated prodrugs in the clinic has, so far, not been successful. The lack of stratification of patients based on tumor hypoxia status, which can vary widely, is sufficient to account for the failure of phase III trials. To fully exploit the potential of hypoxia-activated prodrugs, hypoxia stratification of patients is needed. Here, we propose a biomarker-stratified enriched Phase III study design in which only biomarker-positive (i.e. hypoxia-positive) patients are randomized between standard treatment and the combination of standard treatment with a hypoxia-activated prodrug. This implies the necessity of a Phase II study in which the biomarker or a combination of biomarkers will be evaluated. The total number of patients needed for both clinical studies will be far lower than in currently used randomize-all designs. In addition, we elaborate on the improvements in HAP design that are feasible to increase the treatment success rates.
Collapse
Affiliation(s)
- Linda Spiegelberg
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ruud Houben
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Raymon Niemans
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Dirk de Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ala Yaromina
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Theys
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Christopher P Guise
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Jeffrey B Smaill
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Adam V Patterson
- Translational Therapeutics Team, Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Philippe Lambin
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ludwig J Dubois
- Department of Precision Medicine, The M-Lab, GROW - School for Oncology and Developmental Biology, Maastricht Comprehensive Cancer Centre, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
114
|
Göttgens EL, Ostheimer C, Span PN, Bussink J, Hammond EM. HPV, hypoxia and radiation response in head and neck cancer. Br J Radiol 2019; 92:20180047. [PMID: 29493265 PMCID: PMC6435089 DOI: 10.1259/bjr.20180047] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/26/2018] [Accepted: 02/26/2018] [Indexed: 12/21/2022] Open
Abstract
Over the last decades, the incidence of human papilloma virus (HPV) positive head and neck squamous-cell carcinoma (HNSCC) has significantly increased. Infection with high-risk HPV types drives tumourigenesis through expression of the oncoproteins E6 and E7. Currently, the primary treatment of HNSCC consists of radiotherapy, often combined with platinum-based chemotherapeutics. One of the common features of HNSCC is the occurrence of tumour hypoxia, which impairs the efficacy of radiotherapy and is a negative prognostic factor. Therefore, it is important to detect and quantify the severity of hypoxia, as well as develop strategies to specifically target hypoxic tumours. HPV-positive tumours are remarkably radiosensitive compared to HPV-negative tumours and consequently the HPV-positive patients have a better prognosis. This provides an opportunity to elucidate mechanisms of radiation sensitivity, which may reveal targets for improved therapy for HPV-negative head and neck cancers. In this review, we will discuss the differences between HPV-positive and HPV-negative head and neck tumours and methods of hypoxia detection and targeting in these disease types. Particular emphasis will be placed on the mechanisms by which HPV infection impacts radiosensitivity.
Collapse
Affiliation(s)
- Eva-Leonne Göttgens
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Paul N Span
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jan Bussink
- Department of Radiation Oncology, Radiotherapy & OncoImmunology laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ester M Hammond
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
115
|
He Z, Dai Y, Li X, Guo D, Liu Y, Huang X, Jiang J, Wang S, Zhu G, Zhang F, Lin L, Zhu JJ, Yu G, Chen X. Hybrid Nanomedicine Fabricated from Photosensitizer-Terminated Metal-Organic Framework Nanoparticles for Photodynamic Therapy and Hypoxia-Activated Cascade Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1804131. [PMID: 30565431 DOI: 10.1002/smll.201804131] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/09/2018] [Indexed: 06/09/2023]
Abstract
During photodynamic therapy (PDT), severe hypoxia often occurs as an undesirable limitation of PDT owing to the O2 -consuming photodynamic process, compromising the effectiveness of PDT. To overcome this problem, several strategies aiming to improve tumor oxygenation are developed. Unlike these traditional approaches, an opposite method combining hypoxia-activated prodrug and PDT may provide a promising strategy for cancer synergistic therapy. In light of this, azido-/photosensitizer-terminated UiO-66 nanoscale metal-organic frameworks (UiO-66-H/N3 NMOFs) which serve as nanocarriers for the bioreductive prodrug banoxantrone (AQ4N) are engineered. Owing to the effective shielding of the nanoparticles, the stability of AQ4N is well preserved, highlighting the vital function of the nanocarriers. By virtue of strain-promoted azide-alkyne cycloaddition, the nanocarriers are further decorated with a dense PEG layer to enhance their dispersion in the physiological environment and improve their therapeutic performance. Both in vitro and in vivo studies reveal that the O2 -depleting PDT process indeed aggravates intracellular/tumor hypoxia that activates the cytotoxicity of AQ4N through a cascade process, consequently achieving PDT-induced and hypoxia-activated synergistic therapy. Benefiting from the localized therapeutic effect of PDT and hypoxia-activated cytotoxicity of AQ4N, this hybrid nanomedicine exhibits enhanced therapeutic efficacy with negligible systemic toxicity, making it a promising candidate for cancer therapy.
Collapse
Affiliation(s)
- Zhimei He
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Yunlu Dai
- Faculty of Health Sciences, University of Macau, Macau SAR, 999078, P. R. China
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Xiangli Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Dan Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Yijing Liu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Xiaolin Huang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Jingjing Jiang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Sheng Wang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Guizhi Zhu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Fuwu Zhang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Lisen Lin
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| |
Collapse
|
116
|
DiGiacomo JW, Gilkes DM. Therapeutic Strategies to Block the Hypoxic Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:141-157. [DOI: 10.1007/978-3-030-12734-3_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
117
|
An Intratumor Pharmacokinetic/Pharmacodynamic Model for the Hypoxia-Activated Prodrug Evofosfamide (TH-302): Monotherapy Activity is Not Dependent on a Bystander Effect. Neoplasia 2018; 21:159-171. [PMID: 30591421 PMCID: PMC6314220 DOI: 10.1016/j.neo.2018.11.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor hypoxia contributes to resistance to anticancer therapies. Hypoxia-activated prodrugs (HAPs) selectively target hypoxic cells and their activity can extend to well-oxygenated areas of tumors via diffusion of active metabolites. This type of bystander effect has been suggested to be responsible for the single agent activity of the clinical-stage HAP evofosfamide (TH-302) but direct evidence is lacking. To dissect the contribution of bystander effects to TH-302 activity, we implemented a Green's function pharmacokinetic (PK) model to simulate the spatial distribution of O2, TH-302 and its cytotoxic metabolites, bromo-isophosphoramide mustard (Br-IPM) and its dichloro derivative isophosphoramide mustard (IPM), in two digitized tumor microvascular networks. The model was parameterized from literature and experimentally, including measurement of diffusion coefficients of TH-302 and its metabolites in multicellular layer cultures. The latter studies demonstrate that Br-IPM and IPM cannot diffuse significantly from the cells in which they are generated, although evidence was obtained for diffusion of the hydroxylamine metabolite of TH-302. The spatially resolved PK model was linked to a pharmacodynamic (PD) model that describes cell killing probability at each point in the tumor microregion as a function of Br-IPM and IPM exposure. The resulting PK/PD model accurately predicted previously reported monotherapy activity of TH-302 in H460 tumors, without invoking a bystander effect, demonstrating that the notable single agent activity of TH-302 in tumors can be accounted for by significant bioreductive activation of TH-302 even in oxic regions, driven by the high plasma concentrations achievable with this well-tolerated prodrug.
Collapse
|
118
|
Duan XC, Yao X, Zhang S, Xu MQ, Hao YL, Li ZT, Zheng XC, Liu M, Li ZY, Li H, Wang JR, Feng ZH, Zhang X. Antitumor activity of the bioreductive prodrug 3-(2-nitrophenyl) propionic acid-paclitaxel nanoparticles (NPPA-PTX NPs) on MDA-MB-231 cells: in vitro and in vivo. Int J Nanomedicine 2018; 14:195-204. [PMID: 30636872 PMCID: PMC6307681 DOI: 10.2147/ijn.s186556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background 3-(2-Nitrophenyl) propionic acid-paclitaxel (NPPA-PTX) is a paclitaxel (PTX) bioreductive prodrug synthesized by our lab. We hypothesize that NPPA-PTX can self-assemble to form nanoparticles (NPs). Materials and methods In the present research, the theoretical partition coefficient (XlogP) and Hansen solubility parameters of NPPA-PTX were calculated. NPPA-PTX nanoparticles prepared by NPPA-PTX and DSPE-PEG (NPPA-PTX:DSPE-PEG =1:0.1, w/w) (NPPA-PTX@PEG NPs) were prepared and characterized. The cellular uptake, in vitro antitumor activity, in vivo targeting effect, tumor distribution, in vivo antitumor activity, and safety of NPPA-PTX@PEG NPs were investigated. Results Our results indicate that NPPA-PTX can self-assemble to form NPPA-PTX@PEG NPs. Both the cellular uptake and safety of NPPA-PTX@PEG NPs were higher than those of Taxol. NPPA-PTX@PEG NPs could target tumor tissues by a passive targeting effect. In tumor tissues, NPPA-PTX@PEG NPs could completely transform into active PTX. The in vivo antitumor activity of NPPA-PTX@PEG NPs was confirmed in MDA-MB-231 tumor-bearing nude mice. Conclusion The bioreductive prodrug NPPA-PTX could self-assemble to form NPs. The safety and antitumor activity of NPPA-PTX@PEG were confirmed in our in vitro and in vivo experiments. The NPPA-PTX@PEG NPs developed in this study could offer a new way of preparing bioreductive prodrug, self-assembled NPs suitable for antitumor therapy.
Collapse
Affiliation(s)
- Xiao-Chuan Duan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Xin Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Shuang Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Yan-Li Hao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Zhan-Tao Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Xiu-Chai Zheng
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Man Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Jing-Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Zhen-Han Feng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China, .,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, People's Republic of China,
| |
Collapse
|
119
|
Nocentini A, Trallori E, Singh S, Lomelino CL, Bartolucci G, Di Cesare Mannelli L, Ghelardini C, McKenna R, Gratteri P, Supuran CT. 4-Hydroxy-3-nitro-5-ureido-benzenesulfonamides Selectively Target the Tumor-Associated Carbonic Anhydrase Isoforms IX and XII Showing Hypoxia-Enhanced Antiproliferative Profiles. J Med Chem 2018; 61:10860-10874. [PMID: 30433782 DOI: 10.1021/acs.jmedchem.8b01504] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human carbonic anhydrases (CA, EC, 4.2.1.1) IX and XII are overexpressed in cancer cells as adaptive response to hypoxia and acidic conditions characteristic of many tumors. In addition, hypoxia facilitates the activity of specific oxido-reductases that may be exploited to selectively activate bioreductive prodrugs. Here, new selective CA IX/XII inhibitors, as analogues of the antitumor phase II drug SLC-0111 are described, namely ureido-substituted benzenesulfonamides appended with a nitro-aromatic moiety to yield an antiproliferative action increased by hypoxia. These compounds were screened for the inhibition of the ubiquitous hCA I/II and the target hCA IX/XII. Six X-ray crystallographies with CA II and IX/mimic allowed for the rationalization of the compounds inhibitory activity. The effects of some such compounds on the viability of HT-29, MDA-MB-231, and PC-3 human cancer cell lines in both normoxic and hypoxic conditions were examined, providing the initiation toward the development of hypoxia-activated antitumor CAIs.
Collapse
Affiliation(s)
- Alessio Nocentini
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , 50019 Sesto Fiorentino Florence , Italy
| | - Elena Trallori
- Department of Biochemistry and Molecular Biology, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - Srishti Singh
- Department of NEUROFARBA-Pharmacology and Toxicology Section , University of Florence , 50019 Florence , Italy
| | - Carrie L Lomelino
- Department of NEUROFARBA-Pharmacology and Toxicology Section , University of Florence , 50019 Florence , Italy
| | - Gianluca Bartolucci
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , 50019 Sesto Fiorentino Florence , Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Biochemistry and Molecular Biology, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - Carla Ghelardini
- Department of Biochemistry and Molecular Biology, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - Robert McKenna
- Department of NEUROFARBA-Pharmacology and Toxicology Section , University of Florence , 50019 Florence , Italy
| | - Paola Gratteri
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , 50019 Sesto Fiorentino Florence , Italy
| | - Claudiu T Supuran
- NEUROFARBA Dept., Sezione di Scienze Farmaceutiche , Università degli Studi di Firenze , Via Ugo Schiff 6 , 50019 Sesto Fiorentino Florence , Italy
| |
Collapse
|
120
|
Hiraga T. Hypoxic Microenvironment and Metastatic Bone Disease. Int J Mol Sci 2018; 19:ijms19113523. [PMID: 30423905 PMCID: PMC6274963 DOI: 10.3390/ijms19113523] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 01/07/2023] Open
Abstract
Hypoxia is a common feature of solid tumors and is associated with an increased risk of metastasis and a poor prognosis. Recent imaging techniques revealed that bone marrow contains a quite hypoxic microenvironment. Low oxygen levels activate hypoxia signaling pathways such as hypoxia-inducible factors, which play critical roles in the key stages of metastatic dissemination including angiogenesis, epithelial-mesenchymal transition, invasion, maintenance of cancer stem cells, tumor cell dormancy, release of extracellular vesicles, and generation of pre-metastatic niches. Hypoxia also affects bone cells, such as osteoblasts and osteoclasts, and immune cells, which also act to support the development and progression of bone metastases. Paradoxically, hypoxia and related signaling molecules are recognized as high-priority therapeutic targets and many candidate drugs are currently under preclinical and clinical investigation. The present review focuses on our current knowledge of the potential roles of hypoxia in cancer metastasis to bone by considering the interaction between metastatic cancer cells and the bone microenvironment. Current therapeutic approaches targeting hypoxia are also described.
Collapse
Affiliation(s)
- Toru Hiraga
- Department of Histology and Cell Biology, Matsumoto Dental University, 1780 Gobara-Hirooka, Shiojiri, Nagano 399-0781, Japan.
| |
Collapse
|
121
|
Abstract
The nitro group is considered to be a versatile and unique functional group in medicinal chemistry. Despite a long history of use in therapeutics, the nitro group has toxicity issues and is often categorized as a structural alert or a toxicophore, and evidence related to drugs containing nitro groups is rather contradictory. In general, drugs containing nitro groups have been extensively associated with mutagenicity and genotoxicity. In this context, efforts toward the structure-mutagenicity or structure-genotoxicity relationships have been undertaken. The current Perspective covers various aspects of agents that contain nitro groups, their bioreductive activation mechanisms, their toxicities, and approaches to combat their toxicity issues. In addition, recent advances in the field of anticancer, antitubercular and antiparasitic agents containing nitro groups, along with a patent survey on hypoxia-activated prodrugs containing nitro groups, are also covered.
Collapse
Affiliation(s)
- Kunal Nepali
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| |
Collapse
|
122
|
Nunes AS, Barros AS, Costa EC, Moreira AF, Correia IJ. 3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs. Biotechnol Bioeng 2018; 116:206-226. [DOI: 10.1002/bit.26845] [Citation(s) in RCA: 309] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/30/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Ana S. Nunes
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Andreia S. Barros
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Elisabete C. Costa
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - André F. Moreira
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Ilídio J. Correia
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
- Departamento de Engenharia Química; Universidade de Coimbra, (CIEPQF); Coimbra Portugal
| |
Collapse
|
123
|
Valencia-Cervantes J, Huerta-Yepez S, Aquino-Jarquín G, Rodríguez-Enríquez S, Martínez-Fong D, Arias-Montaño JA, Dávila-Borja VM. Hypoxia increases chemoresistance in human medulloblastoma DAOY cells via hypoxia‑inducible factor 1α‑mediated downregulation of the CYP2B6, CYP3A4 and CYP3A5 enzymes and inhibition of cell proliferation. Oncol Rep 2018; 41:178-190. [PMID: 30320358 PMCID: PMC6278548 DOI: 10.3892/or.2018.6790] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023] Open
Abstract
Medulloblastomas are among the most frequently diagnosed pediatric solid tumors, and drug resistance remains as the principal cause of treatment failure. Hypoxia and the subsequent activation of hypoxia-inducible factor 1α (HIF-1α) are considered key factors in modulating drug antitumor effectiveness, but the underlying mechanisms in medulloblastomas have not yet been clearly understood. The aim of the present study was to determine whether hypoxia induces resistance to cyclophosphamide (CPA) and ifosfamide (IFA) in DAOY medulloblastoma cells, whether the mechanism is dependent on HIF-1α, and whether involves the modulation of the expression of cytochromes P450 (CYP)2B6, 3A4 and 3A5 and the control of cell proliferation. Monolayer cultures of DAOY medulloblastoma cells were exposed for 24 h to moderate (1% O2) or severe (0.1% O2) hypoxia, and protein expression was evaluated by immunoblotting. Cytotoxicity was studied with the MTT assay and by Annexin V/PI staining and flow cytometry. Cell proliferation was determined by the trypan-blue exclusion assay and cell cycle by propidium iodide staining and flow cytometry. Hypoxia decreased CPA and IFA cytotoxicity in medulloblastoma cells, which correlated with a reduction in the protein levels of CYP2B6, CYP3A4 and CYP3A5 and inhibition of cell proliferation. These responses were dependent on hypoxia-induced HIF-1α activation, as evidenced by chemical inhibition of its transcriptional activity with 2-methoxyestradiol (2-ME), which enhanced the cytotoxic activity of CPA and IFA and increased apoptosis. Our results indicate that by stimulating HIF-1α activity, hypoxia downregulates the expression of CYP2B6, CYP3A4 and CYP3A5, that in turn leads to decreased conversion of CPA and IFA into their active forms and thus to diminished cytotoxicity. These results support that the combination of HIF-1α inhibitors and canonical antineoplastic agents provides a potential therapeutic alternative against medulloblastoma.
Collapse
Affiliation(s)
- Jesús Valencia-Cervantes
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - Sara Huerta-Yepez
- Oncology Disease Research Unit, Children's Hospital of Mexico 'Federico Gomez', Mexico City 06720, Mexico
| | - Guillermo Aquino-Jarquín
- Laboratory of Research on Genomics, Genetics and Bioinformatics, Haemato‑Oncology Building, Children's Hospital of Mexico 'Federico Gomez', Mexico City 06720, Mexico
| | - Sara Rodríguez-Enríquez
- Department of Biochemistry,National Institute of Cardiology 'Ignacio Chavez', Mexico City 14080, Mexico
| | - Daniel Martínez-Fong
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | - José-Antonio Arias-Montaño
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City 07360, Mexico
| | | |
Collapse
|
124
|
Structure-activity relationship study of hypoxia-activated prodrugs for proteoglycan-targeted chemotherapy in chondrosarcoma. Eur J Med Chem 2018; 158:51-67. [DOI: 10.1016/j.ejmech.2018.08.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
|
125
|
Smith PJ, Darzynkiewicz Z, Errington RJ. Nuclear cytometry and chromatin organization. Cytometry A 2018; 93:771-784. [PMID: 30144297 DOI: 10.1002/cyto.a.23521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/25/2018] [Accepted: 06/13/2018] [Indexed: 12/18/2022]
Abstract
The nuclear-targeting chemical probe, for the detection and quantification of DNA within cells, has been a mainstay of cytometry-from the colorimetric Feulgen stain to smart fluorescent agents with tuned functionality. The level of nuclear structure and function at which the probe aims to readout, or indeed at which a DNA-targeted drug acts, is shadowed by a wide range of detection modalities and analytical methods. These methods are invariably limited in terms of the resolution attainable versus the volume occupied by targeted chromatin structures. The scalar challenge arises from the need to understand the extent and different levels of compaction of genomic DNA and how such structures can be re-modeled, reported, or even perturbed by both probes and drugs. Nuclear cytometry can report on the complex levels of chromatin order, disorder, disassembly, and even active disruption by probes and drugs. Nuclear probes can report defining features of clinical and therapeutic interest as in NETosis and other cell death processes. New cytometric approaches continue to bridge the scalar challenges of analyzing chromatin organization. Advances in super-resolution microscopy address the resolution and depth of analysis issues in cellular systems. Typical of recent insights into chromatin organization enabled by exploiting a DNA interacting probe is ChromEM tomography (ChromEMT). ChromEMT uses the unique properties of the anthraquinone-based cytometric dye DRAQ5™ to reveal that local and global 3D chromatin structures effect differences in compaction. The focus of this review is nuclear and chromatin cytometry, with linked reference to DNA targeting probes and drugs as exemplified by the anthracenediones.
Collapse
Affiliation(s)
- Paul J Smith
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Zbigniew Darzynkiewicz
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, Valhalla, New York, 10595
| | - Rachel J Errington
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
126
|
Zhang XY, Elfarra AA. Toxicity mechanism-based prodrugs: glutathione-dependent bioactivation as a strategy for anticancer prodrug design. Expert Opin Drug Discov 2018; 13:815-824. [PMID: 30101640 DOI: 10.1080/17460441.2018.1508207] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION 6-Mercaptopurine (6-MP) and 6-thioguanine (6-TG), two anticancer drugs, have high systemic toxicity due to a lack of target specificity. Therefore, increasing target selectivity should improve drug safety. Areas covered: The authors examined the hypothesis that new prodrug designs based upon mechanisms of kidney-selective toxicity of trichloroethylene would reduce systemic toxicity and improve selectivity to kidney and tumor cells. Two approaches specifically were investigated. The first approach was based upon bioactivation of trichloroethylene-cysteine S-conjugate by renal cysteine S-conjugate β-lyases. The prodrugs obtained were kidney-selective but exhibited low turnover rates. The second approach was based on the toxic mechanism of trichloroethylene-cysteine S-conjugate sulfoxide, a Michael acceptor that undergoes rapid addition-elimination reactions with biological thiols. Expert opinion: Glutathione-dependent Michael addition-elimination reactions appear to be an excellent strategy to design highly efficient anticancer drugs. Targeting glutathione could be a promising approach for the development of anticancer prodrugs because cancer cells usually upregulate glutathione biosynthesis and/or glutathione S-transferases expression.
Collapse
Affiliation(s)
- Xin-Yu Zhang
- a Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Faculty of Public Health , Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - Adnan A Elfarra
- b Department of Comparative Biosciences and the Molecular and Environmental Toxicology Center , University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
127
|
Luan X, Guan Y, Liu H, Lu Q, Zhao M, Sun D, Lovell JF, Sun P, Chen H, Fang C. A Tumor Vascular-Targeted Interlocking Trimodal Nanosystem That Induces and Exploits Hypoxia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1800034. [PMID: 30128230 PMCID: PMC6097144 DOI: 10.1002/advs.201800034] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/06/2018] [Indexed: 05/04/2023]
Abstract
Vascular-targeted photodynamic therapy (VTP) is a recently approved strategy for treating solid tumors. However, the exacerbated hypoxic stress makes tumor eradication challenging with such a single modality approach. Here, a new graphene oxide (GO)-based nanosystem for rationally designed, interlocking trimodal cancer therapy that enables VTP using photosensitizer verteporfin (VP) (1) with codelivery of banoxantrone dihydrochloride (AQ4N) (2), a hypoxia-activated prodrug (HAP), and HIF-1α siRNA (siHIF-1α) (3) is reported. The VTP-induced aggravated hypoxia is highly favorable for AQ4N activation into AQ4 (a topoisomerase II inhibitor) for chemotherapy. However, the hypoxia-induced HIF-1α acts as a "hidden brake," through downregulating CYP450 (the dominant HAP-activating reductases), to substantially hinder AQ4N activation. siHIF-1α is rationally adopted to suppress the HIF-1α expression upon hypoxia and further enhance AQ4N activation. This trimodal nanosystem significantly delays the growth of PC-3 tumors in vivo compared to the control nanoparticles carrying VP, AQ4N, or siHIF-1α alone or their pairwise combinations. This multimodal nanoparticle design presents, the first example exploiting VTP to actively induce hypoxia for enhanced HAP activation. It is also revealed that HAP activation is still insufficient under hypoxia due to the hidden downregulation of the HAP-activating reductases (CYP450), and this can be well overcome by GO nanoparticle-mediated siHIF-1α intervention.
Collapse
Affiliation(s)
- Xin Luan
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of MichiganAnn ArborMI48105USA
- Institute of Interdisciplinary Integrative Biomedical ResearchShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Ying‐Yun Guan
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
- Department of PharmacyRuijin HospitalSJTU‐SM, 197 Rui Jin Er RoadShanghai200025China
| | - Hai‐Jun Liu
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
| | - Qin Lu
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
| | - Mei Zhao
- Department of PharmacyShanghai University of Medicine & Health Sciences279 Zhouzhu RoadShanghai201318China
| | - Duxin Sun
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of MichiganAnn ArborMI48105USA
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Peng Sun
- Department of General SurgeryShanghai Tongren HospitalSJTU‐SM, 1111 Xianxia RoadShanghai200336China
| | - Hong‐Zhuan Chen
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
- Institute of Interdisciplinary Integrative Biomedical ResearchShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Chao Fang
- Hongqiao International Institute of MedicineShanghai Tongren Hospital and Department of Pharmacology and Chemical BiologyInstitute of Medical SciencesShanghai Jiao Tong University School of Medicine (SJTU‐SM)280 South Chongqing RoadShanghai200025China
| |
Collapse
|
128
|
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| | - Amir Syahir
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| | - Syahida Ahmad
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
129
|
Sahu A, Choi WI, Tae G. Recent Progress in the Design of Hypoxia-Specific Nano Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| | - Won Il Choi
- Center for Convergence Bioceramic Materials; Convergence R&D Division; Korea Institute of Ceramic Engineering and Technology; 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu Cheongju Chungbuk 28160 Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| |
Collapse
|
130
|
Kumar S, Sun JD, Zhang L, Mokhtari RB, Wu B, Meng F, Liu Q, Bhupathi D, Wang Y, Yeger H, Hart C, Baruchel S. Hypoxia-Targeting Drug Evofosfamide (TH-302) Enhances Sunitinib Activity in Neuroblastoma Xenograft Models. Transl Oncol 2018; 11:911-919. [PMID: 29803017 PMCID: PMC6041570 DOI: 10.1016/j.tranon.2018.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 05/01/2018] [Accepted: 05/07/2018] [Indexed: 01/16/2023] Open
Abstract
Antiangiogenic therapy has shown promising results in preclinical and clinical trials. However, tumor cells acquire resistance to this therapy by gaining ability to survive and proliferate under hypoxia induced by antiangiogenic therapy. Combining antiangiogenic therapy with hypoxia-activated prodrugs can overcome this limitation. Here, we have tested the combination of antiangiogenic drug sunitinib in combination with hypoxia-activated prodrug evofosfamide in neuroblastoma. In vitro, neuroblastoma cell line SK-N-BE(2) was 40-folds sensitive to evofosfamide under hypoxia compared to normoxia. In IV metastatic model, evofosfamide significantly increased mice survival compared to the vehicle (P=.02). In SK-N-BE(2) subcutaneous xenograft model, we tested two different treatment regimens using 30 mg/kg sunitinib and 50 mg/kg evofosfamide. Here, sunitinib therapy when started along with evofosfamide treatment showed higher efficacy compared to single agents in subcutaneous SK-N-BE(2) xenograft model, whereas sunitinib when started 7 days after evofosfamide treatment did not have any advantage compared to treatment with either single agent. Immunofluorescence of tumor sections revealed higher number of apoptotic cells and hypoxic areas compared to either single agent when both treatments were started together. Treatment with 80 mg/kg sunitinib with 50 mg/kg evofosfamide was significantly superior to single agents in both xenograft and metastatic models. This study confirms the preclinical efficacy of sunitinib and evofosfamide in murine models of aggressive neuroblastoma. Sunitinib enhances the efficacy of evofosfamide by increasing hypoxic areas, and evofosfamide targets hypoxic tumor cells. Consequently, each drug enhances the activity of the other.
Collapse
Affiliation(s)
- Sushil Kumar
- Division of Hematology and Oncology, Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4; Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Room 2374, Toronto, Ontario, Canada, M5S 1A8
| | - Jessica D Sun
- Threshold Pharmaceuticals Inc., 170 Harbor Way # 300, South San Francisco, CA, USA, 94080
| | - Libo Zhang
- Division of Hematology and Oncology, Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4
| | - Reza Bayat Mokhtari
- Division of Hematology and Oncology, Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4; Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4
| | - Bing Wu
- Division of Hematology and Oncology, Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4
| | - Fanying Meng
- Threshold Pharmaceuticals Inc., 170 Harbor Way # 300, South San Francisco, CA, USA, 94080
| | - Qian Liu
- Threshold Pharmaceuticals Inc., 170 Harbor Way # 300, South San Francisco, CA, USA, 94080
| | - Deepthi Bhupathi
- Threshold Pharmaceuticals Inc., 170 Harbor Way # 300, South San Francisco, CA, USA, 94080
| | - Yan Wang
- Threshold Pharmaceuticals Inc., 170 Harbor Way # 300, South San Francisco, CA, USA, 94080
| | - Herman Yeger
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Room 2374, Toronto, Ontario, Canada, M5S 1A8; Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4
| | - Charles Hart
- Threshold Pharmaceuticals Inc., 170 Harbor Way # 300, South San Francisco, CA, USA, 94080
| | - Sylvain Baruchel
- Division of Hematology and Oncology, Hospital for Sick Children, 686 Bay St, Toronto, ON, Canada, M5G 0A4; Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King's College Circle, Room 2374, Toronto, Ontario, Canada, M5S 1A8.
| |
Collapse
|
131
|
Irigoyen M, García-Ruiz JC, Berra E. The hypoxia signalling pathway in haematological malignancies. Oncotarget 2018; 8:36832-36844. [PMID: 28415662 PMCID: PMC5482702 DOI: 10.18632/oncotarget.15981] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/27/2017] [Indexed: 12/25/2022] Open
Abstract
Haematological malignancies are tumours that affect the haematopoietic and the lymphatic systems. Despite the huge efforts to eradicate these tumours, the percentage of patients suffering resistance to therapies and relapse still remains significant. The tumour environment favours drug resistance of cancer cells, and particularly of cancer stem/initiating cells. Hypoxia promotes aggressiveness, metastatic spread and relapse in most of the solid tumours. Furthermore, hypoxia is associated with worse prognosis and resistance to conventional treatments through activation of the hypoxia-inducible factors. Haematological malignancies are not considered solid tumours, and therefore, the role of hypoxia in these diseases was initially presumed to be inconsequential. However, hypoxia is a hallmark of the haematopoietic niche. Here, we will review the current understanding of the role of both hypoxia and hypoxia-inducible factors in different haematological tumours.
Collapse
Affiliation(s)
- Marta Irigoyen
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| | - Juan Carlos García-Ruiz
- Servicio de Hematología y Hemoterapia, BioCruces Health Research Institute, Hospital Universitario Cruces, Spain
| | - Edurne Berra
- Centro de Investigación Cooperativa en Biociencias CIC bioGUNE, Derio, Spain
| |
Collapse
|
132
|
Shen X, Laber CH, Sarkar U, Galazzi F, Johnson KM, Mahieu NG, Hillebrand R, Fuchs-Knotts T, Barnes CL, Baker GA, Gates KS. Exploiting the Inherent Photophysical Properties of the Major Tirapazamine Metabolite in the Development of Profluorescent Substrates for Enzymes That Catalyze the Bioreductive Activation of Hypoxia-Selective Anticancer Prodrugs. J Org Chem 2018; 83:3126-3131. [DOI: 10.1021/acs.joc.7b03035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
133
|
Karolak A, Rejniak KA. Micropharmacology: An In Silico Approach for Assessing Drug Efficacy Within a Tumor Tissue. Bull Math Biol 2018; 81:3623-3641. [PMID: 29423880 DOI: 10.1007/s11538-018-0402-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/30/2018] [Indexed: 12/25/2022]
Abstract
Systemic chemotherapy is one of the main anticancer treatments used for most kinds of clinically diagnosed tumors. However, the efficacy of these drugs can be hampered by the physical attributes of the tumor tissue, such as tortuous vasculature, dense and fibrous extracellular matrix, irregular cellular architecture, tumor metabolic gradients, and non-uniform expression of the cell membrane receptors. This can impede the transport of therapeutic agents to tumor cells in sufficient quantities. In addition, tumor microenvironments undergo dynamic spatio-temporal changes during tumor progression and treatment, which can also obstruct drug efficacy. To examine ways to improve drug delivery on a cell-to-tissue scale (single-cell pharmacology), we developed the microscale pharmacokinetics/pharmacodynamics (microPKPD) modeling framework. Our model is modular and can be adjusted to include only the mathematical equations that are crucial for a biological problem under consideration. This modularity makes the model applicable to a broad range of pharmacological cases. As an illustration, we present two specific applications of the microPKPD methodology that help to identify optimal drug properties. The hypoxia-activated drugs example uses continuous drug concentrations, diffusive-advective transport through the tumor interstitium, and passive transmembrane drug uptake. The targeted therapy example represents drug molecules as discrete particles that move by diffusion and actively bind to cell receptors. The proposed modeling approach takes into account the explicit tumor tissue morphology, its metabolic landscape and/or specific receptor distribution. All these tumor attributes can be assessed from patients' diagnostic biopsies; thus, the proposed methodology can be developed into a tool suitable for personalized medicine, such as neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Aleksandra Karolak
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Katarzyna A Rejniak
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA. .,Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
134
|
Cox TR, Erler JT, Rumney RMH. Established Models and New Paradigms for Hypoxia-Driven Cancer-Associated Bone Disease. Calcif Tissue Int 2018; 102:163-173. [PMID: 29098360 PMCID: PMC5805797 DOI: 10.1007/s00223-017-0352-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/19/2017] [Indexed: 12/16/2022]
Abstract
The five-year survival rate for primary bone cancers is ~ 70% while almost all cases of secondary metastatic bone cancer are terminal. Hypoxia, the deficiency of oxygen which occurs as the rate of tumour growth exceeds the supply of vascularisation, is a key promoter of tumour progression. Hypoxia-driven effects in the primary tumour are wide ranging including changes in gene expression, dysregulation of signalling pathways, resistance to chemotherapy, neovascularisation, increased tumour cell proliferation and migration. Paget's seed and soil theory states that for a metastasising tumour cell 'the seed' it requires the correct microenvironment 'soil' to colonise. Why and how metastasising tumour cells colonise the bone is a complex and intriguing problem. However, once present tumour cells are able to disrupt bone homeostasis through increasing osteoclast activity and downregulating osteoblast function. Osteoclast resorption releases growth factors from the bone matrix that subsequently contribute to the proliferation of invasive tumour cells creating the vicious cycle of bone loss and metastatic cancer progression. Recently, we have shown that hypoxia increases expression and release of lysyl oxidase (LOX) from primary mammary tumours, which in turn disrupts bone homeostasis to favour osteolytic degradation to create pre-metastatic niches in the bone microenvironment. We also demonstrated how treatment with bisphosphonates could block this cancer-induced bone remodelling and reduce secondary bone metastases. This review describes the roles of hypoxia in primary tumour progression to metastasis, with a focus on key signalling pathways and treatment options to reduce patient morbidity and increase survival.
Collapse
Affiliation(s)
- Thomas R Cox
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2010, Australia.
| | - Janine T Erler
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen (UCPH), Ole Maaløes Vej 5, 2200, Copenhagen, Denmark
| | - Robin M H Rumney
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
135
|
Perego P, Hempel G, Linder S, Bradshaw TD, Larsen AK, Peters GJ, Phillips RM. Cellular pharmacology studies of anticancer agents: recommendations from the EORTC-PAMM group. Cancer Chemother Pharmacol 2017; 81:427-441. [PMID: 29285635 DOI: 10.1007/s00280-017-3502-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/17/2017] [Indexed: 02/07/2023]
|
136
|
Anderson RF, Li D, Hunter FW. Antagonism in effectiveness of evofosfamide and doxorubicin through intermolecular electron transfer. Free Radic Biol Med 2017; 113:564-570. [PMID: 29111232 DOI: 10.1016/j.freeradbiomed.2017.10.385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 12/27/2022]
Abstract
Hypoxic cells pose a problem in anticancer chemotherapy, in which often drugs require oxygen as an electron acceptor to bring about the death of actively cycling cells. Bioreductive anticancer drugs, which are selectively activated in the hypoxic regions of tumours through enzymatic one-electron reduction, are being developed for combination with chemotherapy-, radiotherapy- and immunotherapy-containing regimens to kill treatment-resistant hypoxic cells. The most clinically-advanced bioreductive drug, evofosfamide (TH-302), which acts by releasing a DNA-crosslinking mustard, failed to extend overall survival in combination with doxorubicin, a topoisomerase II inhibitor, for advanced soft tissue sarcoma in a pivotal clinical trial. However, the reasons for the lack of additive efficacy with this combination are unknown. Here, we show that the radical anion of evofosfamide undergoes electron transfer to doxorubicin in kinetic competition to fragmentation of the radical anion, thus suppressing the release the cytotoxic mustard. This electron transfer process may account, at least in part, for the lack of overall survival improvement in the recent clinical trial. This study underlines the need to consider both redox and electron transfer chemistry when combining bioreductive prodrugs with other redox-active drugs in cancer treatment.
Collapse
Affiliation(s)
- Robert F Anderson
- Auckland Cancer Society Research Centre, Faculty of Health and Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; School of Chemical Sciences, Faculty of Science, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Dan Li
- Auckland Cancer Society Research Centre, Faculty of Health and Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, Faculty of Health and Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
137
|
Gao Y, Lin Y, Liu T, Chen H, Yang X, Tian C, Du L, Li M. Bioluminescent Probe for Tumor Hypoxia Detection via CYP450 Reductase in Living Animals. Anal Chem 2017; 89:12488-12493. [DOI: 10.1021/acs.analchem.7b03597] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yuqi Gao
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE),
School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Yuxing Lin
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE),
School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Tingting Liu
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE),
School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Hui Chen
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE),
School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Xiaofeng Yang
- School
of Chemistry and Chemical Engineering, University of Jinan, Jinan, Shandong 250022, China
| | - Chengsen Tian
- School
of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong 250200, China
| | - Lupei Du
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE),
School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
| | - Minyong Li
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE),
School of Pharmacy, Shandong University, Jinan, Shandong 250012, China
- State
Key Laboratory of Microbial Technology, Shandong University, Jinan, Shandong 250100, China
| |
Collapse
|
138
|
Nesbitt H, Worthington J, Errington RJ, Patterson LH, Smith PJ, McKeown SR, McKenna DJ. The unidirectional hypoxia-activated prodrug OCT1002 inhibits growth and vascular development in castrate-resistant prostate tumors. Prostate 2017; 77:1539-1547. [PMID: 28944496 DOI: 10.1002/pros.23434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/09/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND OCT1002 is a unidirectional hypoxia-activated prodrug (uHAP) OCT1002 that can target hypoxic tumor cells. Hypoxia is a common feature in prostate tumors and is known to drive disease progression and metastasis. It is, therefore, a rational therapeutic strategy to directly target hypoxic tumor cells in an attempt to improve treatment for this disease. Here we tested OCT1002 alone and in combination with standard-of-care agents in hypoxic models of castrate-resistant prostate cancer (CRPC). METHODS The effect of OCT1002 on tumor growth and vasculature was measured using murine PC3 xenograft and dorsal skin fold (DSF) window chamber models. The effects of abiraterone, docetaxel, and cabazitaxel, both singly and in combination with OCT1002, were also compared. RESULTS The hypoxia-targeting ability of OCT1002 effectively controls PC3 tumor growth. The effect was evident for at least 42 days after exposure to a single dose (30 mg/kg) and was comparable to, or better than, drugs currently used in the clinic. In DSF experiments OCT1002 caused vascular collapse in the PC3 tumors and inhibited the revascularization seen in controls. In this model OCT1002 also enhanced the anti-tumor effects of abiraterone, cabazitaxel, and docetaxel; an effect which was accompanied by a more prolonged reduction in tumor vasculature density. CONCLUSIONS These studies provide the first evidence that OCT1002 can be an effective agent in treating hypoxic, castrate-resistant prostate tumors, either singly or in combination with established chemotherapeutics for prostate cancer.
Collapse
Affiliation(s)
- Heather Nesbitt
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| | | | - Rachel J Errington
- School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
- BioStatus Ltd, Shepshed, Leicestershire, United Kingdom
| | | | - Paul J Smith
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Stephanie R McKeown
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
- OncoTherics Ltd, Shepshed, Leicestershire, United Kingdom
| | - Declan J McKenna
- Biomedical Sciences, Research Institute, University of Ulster, Cromore Road, Coleraine, Northern Ireland, United Kingdom
| |
Collapse
|
139
|
Shi Y, Oeh J, Hitz A, Hedehus M, Eastham-Anderson J, Peale FV, Hamilton P, O'Brien T, Sampath D, Carano RAD. Monitoring and Targeting Anti-VEGF Induced Hypoxia within the Viable Tumor by 19F-MRI and Multispectral Analysis. Neoplasia 2017; 19:950-959. [PMID: 28987998 PMCID: PMC5635323 DOI: 10.1016/j.neo.2017.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 01/21/2023] Open
Abstract
The effect of anti-angiogenic agents on tumor oxygenation has been in question for a number of years, where both increases and decreases in tumor pO2 have been observed. This dichotomy in results may be explained by the role of vessel normalization in the response of tumors to anti-angiogenic therapy, where anti-angiogenic therapies may initially improve both the structure and the function of tumor vessels, but more sustained or potent anti-angiogenic treatments will produce an anti-vascular response, producing a more hypoxic environment. The first goal of this study was to employ multispectral (MS) 19F–MRI to noninvasively quantify viable tumor pO2 and evaluate the ability of a high dose of an antibody to vascular endothelial growth factor (VEGF) to produce a strong and prolonged anti-vascular response that results in significant tumor hypoxia. The second goal of this study was to target the anti-VEGF induced hypoxic tumor micro-environment with an agent, tirapazamine (TPZ), which has been designed to target hypoxic regions of tumors. These goals have been successfully met, where an antibody that blocks both murine and human VEGF-A (B20.4.1.1) was found by MS 19F–MRI to produce a strong anti-vascular response and reduce viable tumor pO2 in an HM-7 xenograft model. TPZ was then employed to target the anti-VEGF-induced hypoxic region. The combination of anti-VEGF and TPZ strongly suppressed HM-7 tumor growth and was superior to control and both monotherapies. This study provides evidence that clinical trials combining anti-vascular agents with hypoxia-activated prodrugs should be considered to improved efficacy in cancer patients.
Collapse
Affiliation(s)
- Yunzhou Shi
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, CA
| | - Jason Oeh
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Anna Hitz
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Maj Hedehus
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, CA
| | | | - Franklin V Peale
- Department of Pathology, Genentech Inc., South San Francisco, CA
| | - Patricia Hamilton
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Thomas O'Brien
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Deepak Sampath
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Richard A D Carano
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, CA.
| |
Collapse
|
140
|
Proteoglycan-targeting applied to hypoxia-activated prodrug therapy in chondrosarcoma: first proof-of-concept. Oncotarget 2017; 8:95824-95840. [PMID: 29221170 PMCID: PMC5707064 DOI: 10.18632/oncotarget.21337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 07/09/2017] [Indexed: 01/22/2023] Open
Abstract
Due to its abundant chondrogenic matrix and hypoxic tissue, chondrosarcoma is chemo- and radio-resistant. Our group has developed a proteoglycan targeting strategy by using a quaternary ammonium (QA) function as a carrier of DNA alkylating agents to chondrosarcoma environment. Here, we assessed the relevance of this strategy applied to hypoxia-activated prodrugs, by conjugating a QA to 2-nitroimidazole phosphoramidate. This derivative, named as 8-QA, was evaluated respectively to its non-QA equivalent and to a QA-conjugated but non-hypoxia activated. Firstly binding to aggrecan was confirmed from dissociation constant determined by Surface Plasmon Resonance. In vitro, in HEMC-SS chondrosarcoma cells cultured in monolayer and in spheroids, 8-QA showed higher cytotoxic activity in hypoxia versus normoxia, and led to a strong accumulation of cells in S phase and apoptosis. In vivo, a HEMC-SS xenograft model was implanted on SCID mice and characterized for hypoxia by photoacoustic imaging as well as proteoglycan content. When HEMC-SS bearing mice were given 8-QA at 47 μmol/kg according to a q4d x 6 schedule, a significant 62.1% inhibition of tumor growth was observed, without associated hematological side effects. Mechanistic studies of treated tumors highlighted decrease in mitotic index associated to increase in both p21 and p53S15 markers. Interestingly, 8-QA treatment induced an increase of DNA damages as measured by γH2AX predominantly found in pimonidazole-positive hypoxic areas. These preclinical results are the first to demonstrate the interest of addressing hypoxia-activated prodrugs selectively to proteoglycan of chondrogenic tumor tissue, as a promising therapeutic strategy.
Collapse
|
141
|
Gu Y, Chang TTA, Wang J, Jaiswal JK, Edwards D, Downes NJ, Liyanage HDS, Lynch CRH, Pruijn FB, Hickey AJR, Hay MP, Wilson WR, Hicks KO. Reductive Metabolism Influences the Toxicity and Pharmacokinetics of the Hypoxia-Targeted Benzotriazine Di-Oxide Anticancer Agent SN30000 in Mice. Front Pharmacol 2017; 8:531. [PMID: 28848445 PMCID: PMC5554537 DOI: 10.3389/fphar.2017.00531] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/28/2017] [Indexed: 12/23/2022] Open
Abstract
3-(3-Morpholinopropyl)-7,8-dihydro-6H-indeno[5,6-e][1,2,4]triazine 1,4-dioxide (SN30- 000), an analog of the well-studied bioreductive prodrug tirapazamine (TPZ), has improved activity against hypoxic cells in tumor xenografts. However, little is known about its biotransformation in normal tissues. Here, we evaluate implications of biotransformation of SN30000 for its toxicokinetics in NIH-III mice. The metabolite profile demonstrated reduction to the 1-N-oxide (M14), oxidation of the morpholine side-chain (predominantly to the alkanoic acid M18) and chromophore, and subsequent glucuronidation. Plasma pharmacokinetics of SN30000 and its reduced metabolites was unaffected by the presence of HT29 tumor xenografts, indicating extensive reduction in normal tissues. This bioreductive metabolism, as modeled by hepatic S9 preparations, was strongly inhibited by oxygen indicating that it proceeds via the one-electron (radical) intermediate previously implicated in induction of DNA double strand breaks and cytotoxicity by SN30000. Plasma pharmacokinetics of SN30000 and M14 (but not M18) corresponded closely to the timing of reversible acute clinical signs (reduced mobility) and marked hypothermia (rectal temperature drop of ∼8°C at nadir following the maximum tolerated dose). Similar acute toxicity was elicited by dosing with TPZ or M14, although M14 did not induce the kidney and lung histopathology caused by SN30000. M14 also lacked antiproliferative potency in hypoxic cell cultures. In addition M14 showed much slower redox cycling than SN30000 in oxic cultures. Thus a non-bioreductive mechanism, mediated through M14, appears to be responsible for the acute toxicity of SN30000 while late toxicities are consistent with DNA damage resulting from its one-electron reduction. A two-compartment pharmacokinetic model, in which clearance of SN30000 is determined by temperature-dependent bioreductive metabolism to M14, was shown to describe the non-linear PK of SN30000 in mice. This study demonstrates the importance of non-tumor bioreductive metabolism in the toxicology and pharmacokinetics of benzotriazine di-oxides designed to target tumor hypoxia.
Collapse
Affiliation(s)
- Yongchuan Gu
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Tony T-A Chang
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Jingli Wang
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Jagdish K Jaiswal
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - David Edwards
- Cancer Research Centre for Drug Development, Cancer Research UK (CRUK)London, United Kingdom
| | | | - H D Sarath Liyanage
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Courtney R H Lynch
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Frederik B Pruijn
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Anthony J R Hickey
- School of Biological Sciences, The University of AucklandAuckland, New Zealand
| | - Michael P Hay
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - William R Wilson
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| | - Kevin O Hicks
- Experimental Therapeutics Group, Auckland Cancer Society Research Centre, School of Medical Sciences, The University of AucklandAuckland, New Zealand
| |
Collapse
|
142
|
Cascioferro S, Parrino B, Spanò V, Carbone A, Montalbano A, Barraja P, Diana P, Cirrincione G. An overview on the recent developments of 1,2,4-triazine derivatives as anticancer compounds. Eur J Med Chem 2017; 142:328-375. [PMID: 28851503 DOI: 10.1016/j.ejmech.2017.08.009] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 02/06/2023]
Abstract
The synthesis, the antitumor activity, the SAR and, whenever described, the possible mode of action of 1,2,4-triazine derivatives, their N-oxides, N,N'-dioxides as well as the benzo- and hetero-fused systems are reported. Herein are treated derivatives disclosed to literature from the beginning of this century up to 2016. Among the three possible triazine isomers, 1,2,4-triazines are the most studied ones and many derivatives having remarkable antitumor activity have been reported in the literature and also patented reaching advanced phases of clinical trials.
Collapse
Affiliation(s)
- Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy.
| |
Collapse
|
143
|
Qiu GZ, Jin MZ, Dai JX, Sun W, Feng JH, Jin WL. Reprogramming of the Tumor in the Hypoxic Niche: The Emerging Concept and Associated Therapeutic Strategies. Trends Pharmacol Sci 2017; 38:669-686. [DOI: 10.1016/j.tips.2017.05.002] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/06/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023]
|
144
|
Phillips RM, Hendriks HR, Sweeney JB, Reddy G, Peters GJ. Efficacy, pharmacokinetic and pharmacodynamic evaluation of apaziquone in the treatment of non-muscle invasive bladder cancer. Expert Opin Drug Metab Toxicol 2017. [PMID: 28637373 DOI: 10.1080/17425255.2017.1341490] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Apaziquone (also known as EO9 and QapzolaTM) is a prodrug that is activated to DNA damaging species by oxidoreductases (particularly NQO1) and has the ability to kill aerobic and/or hypoxic cancer cells. Areas covered: Whilst its poor pharmacokinetic properties contributed to its failure in phase II clinical trials when administered intravenously, these properties were ideal for loco-regional therapies. Apaziquone demonstrated good anti-cancer activity against non-muscle invasive bladder cancer (NMIBC) when administered intravesically to marker lesions and was well tolerated with no systemic side effects. However, phase III clinical trials did not reach statistical significance for the primary endpoint of 2-year recurrence in apaziquone over placebo although improvements were observed. Post-hoc analysis of the combined study data did indicate a significant benefit for patients treated with apaziquone, especially when the instillation of apaziquone was given 30 min or more after surgery. A further phase III study is ongoing to test the hypotheses generated in the unsuccessful phase III studies conducted to date. Expert opinion: Because of its specific pharmacological properties, Apaziquone is excellently suited for local therapy such as NMIBC. Future studies should include proper biomarkers.
Collapse
Affiliation(s)
- R M Phillips
- a Department of Pharmacy , University of Huddersfield , Huddersfield , UK.,b Department of Chemical Sciences , University of Huddersfield , UK
| | - H R Hendriks
- c Hendriks Pharmaceutical Consulting , Purmerend , The Netherlands
| | - J B Sweeney
- a Department of Pharmacy , University of Huddersfield , Huddersfield , UK.,b Department of Chemical Sciences , University of Huddersfield , UK
| | - G Reddy
- d Spectrum Pharmaceuticals Inc , Irvine , CA , USA
| | - G J Peters
- e Department of Medical Oncology , VU University Medical Center , Amsterdam , The Netherlands
| |
Collapse
|
145
|
Ding C, Li Z. A review of drug release mechanisms from nanocarrier systems. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:1440-1453. [DOI: 10.1016/j.msec.2017.03.130] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 03/15/2017] [Accepted: 03/17/2017] [Indexed: 12/16/2022]
|
146
|
Tercel M, Lee HH, Mehta SY, Youte Tendoung JJ, Bai SY, Liyanage HDS, Pruijn FB. Influence of a Basic Side Chain on the Properties of Hypoxia-Selective Nitro Analogues of the Duocarmycins: Demonstration of Substantial Anticancer Activity in Combination with Irradiation or Chemotherapy. J Med Chem 2017. [PMID: 28644035 DOI: 10.1021/acs.jmedchem.7b00563] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A new series of nitro analogues of the duocarmycins was prepared and evaluated for hypoxia-selective anticancer activity. The compounds incorporate 13 different amine-containing side chains designed to bind in the minor groove of DNA while spanning a wide range of base strength from pKa 9.64 to 5.24. The most favorable in vitro properties were associated with strongly basic side chains, but the greatest in vivo antitumor activity was found for compounds containing a weakly basic morpholine. This applies to single-agent activity and for activity in combination with irradiation or chemotherapy (gemcitabine or docetaxel). In combination with a single dose of γ irradiation 50 at 42 μmol/kg eliminated detectable clonogens in some SiHa cervical carcinoma xenografts, and in combination with gemcitabine using a well-tolerated multidose schedule, the same compound caused regression of all treated A2780 ovarian tumor xenografts. In the latter experiment, three of seven animals receiving the combination treatment were completely tumor free at day 100.
Collapse
Affiliation(s)
- Moana Tercel
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Ho H Lee
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Sunali Y Mehta
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Jean-Jacques Youte Tendoung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Sally Y Bai
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - H D Sarath Liyanage
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| | - Frederik B Pruijn
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland , Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
147
|
Xiong A, Liu Y. Targeting Hypoxia Inducible Factors-1α As a Novel Therapy in Fibrosis. Front Pharmacol 2017; 8:326. [PMID: 28611671 PMCID: PMC5447768 DOI: 10.3389/fphar.2017.00326] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/16/2017] [Indexed: 02/05/2023] Open
Abstract
Fibrosis, characterized by increased extracellular matrix (ECM) deposition, and widespread vasculopathy, has the prominent trait of chronic hypoxia. Hypoxia inducible factors-1α (HIF-1α), a key transcriptional factor in response to this chronic hypoxia, is involved in fibrotic disease, such as Systemic sclerosis (SSc). The implicated function of HIF-1α in fibrosis include stimulation of excessive ECM, vascular remodeling, and futile angiogenesis with further exacerbation of chronic hypoxia and deteriorate pathofibrogenesis. This review will focus on the molecular biological behavior of HIF-1α in regulating progressive fibrosis. Better understanding of the role for HIF-1α-regulated pathways in fibrotic disease will accelerate development of novel therapeutic strategies that target HIF-1α. Such new therapeutic strategies may be particularly effective for treatment of the prototypic, multisystem fibrotic, autoimmune disease SSc.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan UniversityChengdu, China
| |
Collapse
|
148
|
Yamazaki H, Lai YC, Tateno M, Setoguchi A, Goto-Koshino Y, Endo Y, Nakaichi M, Tsujimoto H, Miura N. Hypoxia-activated prodrug TH-302 decreased survival rate of canine lymphoma cells under hypoxic condition. PLoS One 2017; 12:e0177305. [PMID: 28489881 PMCID: PMC5425042 DOI: 10.1371/journal.pone.0177305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 04/25/2017] [Indexed: 12/21/2022] Open
Abstract
We tested the hypotheses that hypoxic stimulation enhances growth potentials of canine lymphoma cells by activating hypoxia-inducible factor 1α (HIF-1α), and that the hypoxia-activated prodrug (TH-302) inhibits growth potentials in the cells. We investigated how hypoxic culture affects the growth rate, chemoresistance, and invasiveness of canine lymphoma cells and doxorubicin (DOX)-resistant lymphoma cells, and influences of TH-302 on survival rate of the cells under hypoxic conditions. Our results demonstrated that hypoxic culture upregulated the expression of HIF-1α and its target genes, including ATP-binding cassette transporter B1 (ABCB1), ATP-binding cassette transporter G2 (ABCG2), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and survivin, and enhanced the growth rate, DOX resistance, and invasiveness of the cells. Additionally, TH-302 decreased the survival rate of the cells under hypoxic condition. Our studies suggest that hypoxic stimulation may advance the tumorigenicity of canine lymphoma cells, favoring malignant transformation. Therefore, the data presented may contribute to the development of TH-302-based hypoxia-targeting therapies for canine lymphoma.
Collapse
Affiliation(s)
- Hiroki Yamazaki
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima, Japan
| | - Yu-Chang Lai
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima, Japan
| | - Morihiro Tateno
- Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima, Japan
| | | | - Yuko Goto-Koshino
- Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Yasuyuki Endo
- Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima, Japan
| | - Munekazu Nakaichi
- Laboratory of Veterinary Radiology, Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yoshida, Yamaguchi, Japan
| | - Hajime Tsujimoto
- Department of Veterinary Internal Medicine, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Naoki Miura
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Korimoto, Kagoshima, Japan
- * E-mail:
| |
Collapse
|
149
|
Jahanban-Esfahlan R, de la Guardia M, Ahmadi D, Yousefi B. Modulating tumor hypoxia by nanomedicine for effective cancer therapy. J Cell Physiol 2017; 233:2019-2031. [PMID: 28198007 DOI: 10.1002/jcp.25859] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
Hypoxia, a characteristic feature of tumors, is indispensable to tumor angiogenesis, metastasis, and multi drug resistance. Hypoxic avascular regions, deeply embedded inside the tumors significantly hinder delivery of therapeutic agents. The low oxygen tension results in resistance to the current applied anti-cancer therapeutics including radiotherapy, chemotherapy, and photodynamic therapy, the efficacy of which is firmly tied to the level of tumor oxygen supply. However, emerging data indicate that nanocarriers/nanodrugs can offer substantial benefits to improve the efficacy of current therapeutics, through modulation of tumor hypoxia. This review aims to introduce the most recent advances made in nanocarrier mediated targeting of tumor hypoxia. The first part is dedicated to the approaches by which nanocarriers could be designed to target/leverage hypoxia. These approaches include i) inhibiting Hypoxia Inducer Factor (HIF-1α); ii) hypoxia activated prodrugs/linkers; and iii) obligate anaerobe mediated targeting of tumor hypoxia. The second part, details novel nanosystems proposed to modulate tumor hypoxia through tumor oxygenation. These methods seek to lessen tumor hypoxia through vascular normalization, or reoxygenation therapy. The reoxygenation of tumor could be accomplished by: i) generation of oxygen filled nanocarriers; ii) natural/artificial oxygen nanocarriers; and iii) oxygen generators. The efficacy of each approach and their potential in cancer therapy is further discussed.
Collapse
Affiliation(s)
- Rana Jahanban-Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Delshad Ahmadi
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Targeting Therapy Research Group, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
150
|
Xu S, Yao H, Pei L, Hu M, Li D, Qiu Y, Wang G, Wu L, Yao H, Zhu Z, Xu J. Design, synthesis, and biological evaluation of NAD(P)H: Quinone oxidoreductase (NQO1)-targeted oridonin prodrugs possessing indolequinone moiety for hypoxia-selective activation. Eur J Med Chem 2017; 132:310-321. [DOI: 10.1016/j.ejmech.2017.03.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/22/2017] [Indexed: 02/07/2023]
|