101
|
Abstract
The presence of a microenvironment within most tumours containing regions of low oxygen tension or hypoxia has profound biological and therapeutic implications. Tumour hypoxia is known to promote the development of an aggressive phenotype, resistance to both chemotherapy and radiotherapy and is strongly associated with poor clinical outcome. Paradoxically, it is recognised as a high-priority target and one of the therapeutic strategies designed to eradicate hypoxic cells in tumours is a group of compounds known collectively as hypoxia-activated prodrugs (HAPs) or bioreductive drugs. These drugs are inactive prodrugs that require enzymatic activation (typically by 1 or 2 electron oxidoreductases) to generate cytotoxic species with selectivity for hypoxic cells being determined by (1) the ability of oxygen to either reverse or inhibit the activation process and (2) the presence of elevated expression of oxidoreductases in tumours. The concepts underpinning HAP development were established over 40 years ago and have been refined over the years to produce a new generation of HAPs that are under preclinical and clinical development. The purpose of this article is to describe current progress in the development of HAPs focusing on the mechanisms of action, preclinical properties and clinical progress of leading examples.
Collapse
|
102
|
Phillips RM. Targeting the hypoxic fraction of tumours using hypoxia-activated prodrugs. Cancer Chemother Pharmacol 2016; 77:441-57. [PMID: 26811177 PMCID: PMC4767869 DOI: 10.1007/s00280-015-2920-7] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/13/2015] [Indexed: 12/17/2022]
Abstract
The presence of a microenvironment within most tumours containing regions of low oxygen tension or hypoxia has profound biological and therapeutic implications. Tumour hypoxia is known to promote the development of an aggressive phenotype, resistance to both chemotherapy and radiotherapy and is strongly associated with poor clinical outcome. Paradoxically, it is recognised as a high-priority target and one of the therapeutic strategies designed to eradicate hypoxic cells in tumours is a group of compounds known collectively as hypoxia-activated prodrugs (HAPs) or bioreductive drugs. These drugs are inactive prodrugs that require enzymatic activation (typically by 1 or 2 electron oxidoreductases) to generate cytotoxic species with selectivity for hypoxic cells being determined by (1) the ability of oxygen to either reverse or inhibit the activation process and (2) the presence of elevated expression of oxidoreductases in tumours. The concepts underpinning HAP development were established over 40 years ago and have been refined over the years to produce a new generation of HAPs that are under preclinical and clinical development. The purpose of this article is to describe current progress in the development of HAPs focusing on the mechanisms of action, preclinical properties and clinical progress of leading examples.
Collapse
Affiliation(s)
- Roger M Phillips
- Department of Pharmacy, University of Huddersfield, Queensgate, Huddersfield, HD1 3DH, UK.
| |
Collapse
|
103
|
Liapis V, Zinonos I, Labrinidis A, Hay S, Ponomarev V, Panagopoulos V, Zysk A, DeNichilo M, Ingman W, Atkins GJ, Findlay DM, Zannettino ACW, Evdokiou A. Anticancer efficacy of the hypoxia-activated prodrug evofosfamide (TH-302) in osteolytic breast cancer murine models. Cancer Med 2016; 5:534-45. [PMID: 26749324 PMCID: PMC4799961 DOI: 10.1002/cam4.599] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/31/2022] Open
Abstract
Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, hypoxia offers treatment opportunities, exemplified by the development of compounds that target hypoxic regions within tumors. Evofosfamide (TH‐302) is a prodrug created by the conjugation of 2‐nitroimidazole to bromo‐isophosphoramide mustard (Br‐IPM). When evofosfamide is delivered to hypoxic regions, the DNA cross‐linking effector, Br‐IPM, is released. This study assessed the cytotoxic activity of evofosfamide in vitro and its antitumor activity against osteolytic breast cancer either alone or in combination with paclitaxel in vivo. A panel of human breast cancer cell lines were treated with evofosfamide under hypoxia and assessed for cell viability. Osteolytic MDA‐MB‐231‐TXSA cells were transplanted into the mammary fat pad, or into tibiae of mice, allowed to establish and treated with evofosfamide, paclitaxel, or both. Tumor burden was monitored using bioluminescence, and cancer‐induced bone destruction was measured using micro‐CT. In vitro, evofosfamide was selectively cytotoxic under hypoxic conditions. In vivo evofosfamide was tumor suppressive as a single agent and cooperated with paclitaxel to reduce mammary tumor growth. Breast cancer cells transplanted into the tibiae of mice developed osteolytic lesions. In contrast, treatment with evofosfamide or paclitaxel resulted in a significant delay in tumor growth and an overall reduction in tumor burden in bone, whereas combined treatment resulted in a significantly greater reduction in tumor burden in the tibia of mice. Evofosfamide cooperates with paclitaxel and exhibits potent tumor suppressive activity against breast cancer growth in the mammary gland and in bone.
Collapse
Affiliation(s)
- Vasilios Liapis
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Irene Zinonos
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Agatha Labrinidis
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Shelley Hay
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Vasilios Panagopoulos
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Aneta Zysk
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Mark DeNichilo
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| | - Wendy Ingman
- Discipline of Surgery, School of Medicine at The Queen Elizabeth Hospital, University of Adelaide, Woodville, South Australia, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Gerald J Atkins
- Discipline of Orthopaedics and Trauma, University of Adelaide, Adelaide, South Australia, Australia
| | - David M Findlay
- Discipline of Orthopaedics and Trauma, University of Adelaide, Adelaide, South Australia, Australia
| | - Andrew C W Zannettino
- School of Medical Sciences, Myeloma Research Laboratory Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Faculty of Health Science, University of Adelaide, Adelaide, South Australia, Australia
| | - Andreas Evdokiou
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, Woodville, South Australia, Australia
| |
Collapse
|
104
|
Sun JD, Liu Q, Ahluwalia D, Li W, Meng F, Wang Y, Bhupathi D, Ruprell AS, Hart CP. Efficacy and safety of the hypoxia-activated prodrug TH-302 in combination with gemcitabine and nab-paclitaxel in human tumor xenograft models of pancreatic cancer. Cancer Biol Ther 2016; 16:438-49. [PMID: 25679067 PMCID: PMC4623012 DOI: 10.1080/15384047.2014.1003005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumors often contain hypoxic regions resistant to chemo- and radiotherapy. TH-302 (T) is an investigational hypoxia-activated prodrug that selectively releases the DNA cross-linker bromo-isophosphoramide mustard under hypoxic conditions. This study evaluated the efficacy and safety profile of combining T with gemcitabine (G) and nab-paclitaxel (nP) in human pancreatic ductal adenocarcinoma (PDAC) xenograft models in mice. Antitumor activity of the G + nP + T triplet was assessed and compared with T-alone or the G + nP doublet in the Hs766t, MIA PaCa-2, PANC-1, and BxPC-3 PDAC xenograft models. Efficacy was assessed by tumor growth kinetic analysis. Body weight, blood cell counts, blood chemistry, and the von Frey neuropathy assay were analyzed to evaluate safety profiles. Pharmacodynamic changes after the treatment were determined by immunohistochemistry of cell proliferation, DNA damage, apoptosis, hypoxia, and tumor stroma density. The G + nP + T triplet exhibited enhanced efficacy compared with T-alone or the G + nP doublet. Compared with vehicle (V), G + nP induced body weight loss, reduced neutrophil and lymphocyte counts, increased the levels of liver function parameters, and induced neurotoxicity. However, when T was added to G + nP, there was no statistically increased impairment compared to G + nP. The triplet significantly increased DNA damage, apoptosis, and tumor necrosis. Furthermore, the triplet further inhibited cell proliferation and reduced stroma density and intratumoral hypoxia. The triplet combination of G + nP + T exhibited superior efficacy but additive toxicity was not evident compared to the G + nP doublet in this study. This study provides a translational rationale for combining G, nP, and T in the clinical setting to assess efficacy and safety. A Phase I clinical trial of the triplet combination is currently underway (NCT02047500).
Collapse
Key Words
- BW, body weight
- Br-IPM, a brominated analog of isophosphoramide mustard
- CAF, cancer-associated fibroblast
- CAIX, carbonic anhydrase IX
- CR, complete response
- EMT, epithelial to mesenchymal transition
- G, gemcitabine
- HF, hypoxic fraction
- ILS, increased life span
- MT, median time to reach the size of 1000 mm3
- MTD, maximum tolerated dose
- NF, necrotic fraction
- PDAC, pancreatic ductal adenocarcinoma
- T, TH-302
- TGD1000, tumor growth delay compared to Vehicle reaching the size of 1000 mm3
- TGI, tumor growth inhibition
- TH-302
- V, vehicle
- gemcitabine
- hypoxia
- hypoxia-activated prodrug
- nP, nab-paclitaxel
- nab-paclitaxel
- pancreatic cancer
- pharmacodynamics, biomarker
- smooth muscle actin
- xenograft
- α-SMA, α
Collapse
Affiliation(s)
- Jessica D Sun
- a Threshold Pharmaceuticals , South San Francisco , CA , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
105
|
Demkowicz S, Rachon J, Daśko M, Kozak W. Selected organophosphorus compounds with biological activity. Applications in medicine. RSC Adv 2016. [DOI: 10.1039/c5ra25446a] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The purpose of this article is to provide an overview of the latest applications of organophosphorus compounds (OPs) that exhibit biological activity.
Collapse
Affiliation(s)
- Sebastian Demkowicz
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Janusz Rachon
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Mateusz Daśko
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| | - Witold Kozak
- Department of Organic Chemistry
- Chemical Faculty
- Gdansk University of Technology
- 80-233 Gdansk
- Poland
| |
Collapse
|
106
|
Efficient Protocol for the Identification of Hypoxic Cell Radiosensitisers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 899:269-90. [DOI: 10.1007/978-3-319-26666-4_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
107
|
Nitroreductase gene-directed enzyme prodrug therapy: insights and advances toward clinical utility. Biochem J 2015; 471:131-53. [PMID: 26431849 DOI: 10.1042/bj20150650] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review examines the vast catalytic and therapeutic potential offered by type I (i.e. oxygen-insensitive) nitroreductase enzymes in partnership with nitroaromatic prodrugs, with particular focus on gene-directed enzyme prodrug therapy (GDEPT; a form of cancer gene therapy). Important first indications of this potential were demonstrated over 20 years ago, for the enzyme-prodrug pairing of Escherichia coli NfsB and CB1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide]. However, it has become apparent that both the enzyme and the prodrug in this prototypical pairing have limitations that have impeded their clinical progression. Recently, substantial advances have been made in the biodiscovery and engineering of superior nitroreductase variants, in particular development of elegant high-throughput screening capabilities to enable optimization of desirable activities via directed evolution. These advances in enzymology have been paralleled by advances in medicinal chemistry, leading to the development of second- and third-generation nitroaromatic prodrugs that offer substantial advantages over CB1954 for nitroreductase GDEPT, including greater dose-potency and enhanced ability of the activated metabolite(s) to exhibit a local bystander effect. In addition to forging substantial progress towards future clinical trials, this research is supporting other fields, most notably the development and improvement of targeted cellular ablation capabilities in small animal models, such as zebrafish, to enable cell-specific physiology or regeneration studies.
Collapse
|
108
|
Zhang L, Marrano P, Wu B, Kumar S, Thorner P, Baruchel S. Combined Antitumor Therapy with Metronomic Topotecan and Hypoxia-Activated Prodrug, Evofosfamide, in Neuroblastoma and Rhabdomyosarcoma Preclinical Models. Clin Cancer Res 2015; 22:2697-708. [DOI: 10.1158/1078-0432.ccr-15-1853] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/17/2015] [Indexed: 11/16/2022]
|
109
|
Ferguson M, Hingorani P, Gupta AA. Emerging molecular-targeted therapies in early-phase clinical trials and preclinical models. Am Soc Clin Oncol Educ Book 2015:420-4. [PMID: 23714564 DOI: 10.14694/edbook_am.2013.33.420] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Within the context of modern cooperative group trials, modification of standard cytotoxic chemotherapy has not improved survival in patients with rhabdomyosarcoma (RMS) over the last 30 years. There is need and interest to incorporate novel targeted anticancer agents into the treatment plans for children and adolescents with newly diagnosed RMS; however, targets directly driven by FOXO1 translocation remain elusive, and molecular events driving translocation negative tumors similarly remain ill-defined. Thus, alternate pathways driving the tumors require identification and targeting. Herein, we describe targeted therapies that could be of interest in RMS, but whose inclusion in clinical trials is thus far limited by scientific and regulatory criteria. Sorafenib, pazopanib, crizotinib, TH-302, aurora-kinase inhibitors, and anaplastic lymphoma kinase (ALK)/c-MET inhibitors will be discussed. The current preclinical and clinical data available, as well as limitations and challenges for each, will be outlined.
Collapse
Affiliation(s)
- Michael Ferguson
- From the Department of Hematology/Oncology, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN; Division of Hematology Oncology, Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ; Division of Hematology/Oncology, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
110
|
Mukherjee A, Rokita SE. Single Amino Acid Switch between a Flavin-Dependent Dehalogenase and Nitroreductase. J Am Chem Soc 2015; 137:15342-5. [PMID: 26616824 PMCID: PMC4684082 DOI: 10.1021/jacs.5b07540] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
A single
mutation within a flavoprotein is capable of switching
the catalytic activity of a dehalogenase into a nitroreductase.
This change in function correlates with a destabilization of the one-electron-reduced
flavin semiquinone that is differentially expressed in the nitro-FMN
reductase superfamily during redox cycling. The diversity of
function within such a superfamily therefore has the potential
to arise from rapid evolution, and its members should provide a convenient
basis for developing new catalysts with an altered specificity of
choice.
Collapse
Affiliation(s)
- Arnab Mukherjee
- Department of Chemistry, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - Steven E Rokita
- Department of Chemistry, Johns Hopkins University , Baltimore, Maryland 21218, United States
| |
Collapse
|
111
|
Penketh PG, Shyam K, Baumann RP, Ratner ES, Sartorelli AC. A simple and inexpensive method to control oxygen concentrations within physiological and neoplastic ranges. Anal Biochem 2015; 491:1-3. [PMID: 26361820 DOI: 10.1016/j.ab.2015.08.032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 11/29/2022]
Abstract
Traditional methods for regulating oxygen concentration ([O2]) in in vitro experiments over the range found in normal and tumor tissues require the use of expensive equipment to generate controlled gas atmospheres or the purchase of a range of gas cylinders with certified O2 percentages. Here we describe a simple and inexpensive enzymatic method for generating low, precise steady-state [O2] levels that are stable for several hours. This method is particularly applicable to the in vitro study of some classes of hypoxia-targeted antitumor prodrugs and bioreductively activated agents.
Collapse
Affiliation(s)
- P G Penketh
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - K Shyam
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - R P Baumann
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - E S Ratner
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT 06520, USA
| | - A C Sartorelli
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
112
|
Hunter FW, Young RJ, Shalev Z, Vellanki RN, Wang J, Gu Y, Joshi N, Sreebhavan S, Weinreb I, Goldstein DP, Moffat J, Ketela T, Brown KR, Koritzinsky M, Solomon B, Rischin D, Wilson WR, Wouters BG. Identification of P450 Oxidoreductase as a Major Determinant of Sensitivity to Hypoxia-Activated Prodrugs. Cancer Res 2015; 75:4211-23. [PMID: 26297733 DOI: 10.1158/0008-5472.can-15-1107] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/15/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Francis W Hunter
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard J Young
- Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Zvi Shalev
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ravi N Vellanki
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jingli Wang
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yongchuan Gu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand. AnQual Laboratories, School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Naveen Joshi
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sreevalsan Sreebhavan
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ilan Weinreb
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - David P Goldstein
- Department of Otolaryngology-Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Jason Moffat
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Troy Ketela
- Donnelly Centre and Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Kevin R Brown
- Donnelly Centre and Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Marianne Koritzinsky
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada. Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Solomon
- Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia. Department of Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Danny Rischin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia. Department of Medical Oncology, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - William R Wilson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Bradly G Wouters
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| |
Collapse
|
113
|
Cheng W, Yuan Y, Qiu N, Peng P, Sheng R, Hu Y. Identification of novel 4-anilinoquinazoline derivatives as potent EGFR inhibitors both under normoxia and hypoxia. Bioorg Med Chem 2015; 22:6796-805. [PMID: 25468044 DOI: 10.1016/j.bmc.2014.10.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/27/2014] [Accepted: 10/27/2014] [Indexed: 01/22/2023]
Abstract
A novel series of 4-anilinoquinazoline derivatives (19a-19t) were designed and synthesized through incorporation of the 2-nitroimidazole moiety into the 4-anilinoquinazoline scaffold of EGFR inhibitors. The most promising compound 19h displayed potent EGFR inhibitory activity with the IC50 value of 0.47 nM. It also strongly suppressed the proliferation of A549 and HT-29 cells with sub-micromolar IC50 values both under normoxia and hypoxia, which were several folds more potent than gefitinib and erlotinib. Further reductive mimic investigation revealed that 19h could be reductive activated under hypoxia and was fully consistent with the results of cell apoptotic assay and in vitro metabolism evaluation. Our results suggest that the incorporation of hypoxia-activated moiety into EGFR inhibitor scaffold might be a tractable strategy to overcome the tumor hypoxia.
Collapse
|
114
|
Lowery MA, O'Reilly EM. Novel Therapeutics for Pancreatic Adenocarcinoma. Hematol Oncol Clin North Am 2015; 29:777-87. [DOI: 10.1016/j.hoc.2015.04.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
115
|
Siemann DW, Horsman MR. Modulation of the tumor vasculature and oxygenation to improve therapy. Pharmacol Ther 2015; 153:107-24. [PMID: 26073310 DOI: 10.1016/j.pharmthera.2015.06.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment is increasingly recognized as a major factor influencing the success of therapeutic treatments and has become a key focus for cancer research. The progressive growth of a tumor results in an inability of normal tissue blood vessels to oxygenate and provide sufficient nutritional support to tumor cells. As a consequence the expanding neoplastic cell population initiates its own vascular network which is both structurally and functionally abnormal. This aberrant vasculature impacts all aspects of the tumor microenvironment including the cells, extracellular matrix, and extracellular molecules which together are essential for the initiation, progression and spread of tumor cells. The physical conditions that arise are imposing and manifold, and include elevated interstitial pressure, localized extracellular acidity, and regions of oxygen and nutrient deprivation. No less important are the functional consequences experienced by the tumor cells residing in such environments: adaptation to hypoxia, cell quiescence, modulation of transporters and critical signaling molecules, immune escape, and enhanced metastatic potential. Together these factors lead to therapeutic barriers that create a significant hindrance to the control of cancers by conventional anticancer therapies. However, the aberrant nature of the tumor microenvironments also offers unique therapeutic opportunities. Particularly interventions that seek to improve tumor physiology and alleviate tumor hypoxia will selectively impair the neoplastic cell populations residing in these environments. Ultimately, by combining such therapeutic strategies with conventional anticancer treatments it may be possible to bring cancer growth, invasion, and metastasis to a halt.
Collapse
Affiliation(s)
- Dietmar W Siemann
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA.
| | - Michael R Horsman
- Department of Experimental Clinical Oncology, Aarhus University Hospital-NBG, Aarhus, Denmark
| |
Collapse
|
116
|
Hypoxia-activated chemotherapeutic TH-302 enhances the effects of VEGF-A inhibition and radiation on sarcomas. Br J Cancer 2015; 113:46-56. [PMID: 26010414 PMCID: PMC4647529 DOI: 10.1038/bjc.2015.186] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/23/2015] [Accepted: 04/29/2015] [Indexed: 12/13/2022] Open
Abstract
Background: Human sarcomas with a poor response to vascular endothelial growth factor-A (VEGF-A) inhibition and radiation therapy (RT) have upregulation of hypoxia-inducible factor 1α (HIF-1α) and HIF-1α target genes. This study examines the addition of the hypoxia-activated chemotherapy TH-302 to VEGF-A inhibition and RT (a.k.a. trimodality therapy). Methods: Trimodality therapy was examined in two xenograft models and in vitro in tumour endothelial cells and sarcoma cell lines. Results: In both mouse models, VEGF-A inhibition and radiation showed greater efficacy than either therapy alone in slowing sarcoma growth. When TH-302 was added, this trimodality therapy completely blocked tumour growth with tumours remaining dormant for over 3 months after cessation of therapy. Trimodality therapy caused 2.6- to 6.2-fold more endothelial cell-specific apoptosis than bimodality therapies, and microvessel density and HIF-1α activity were reduced to 11–13% and 13–20% of control, respectively. When trimodality therapy was examined in vitro, increases in DNA damage and apoptosis were much more pronounced in tumour endothelial cells compared with that in sarcoma cells, especially under hypoxia. Conclusions: The combination of TH-302, VEGF-A inhibition, and RT is highly effective in preclinical models of sarcoma and is associated with increased DNA damage and apoptosis in endothelial cells and decreased HIF-1α activity.
Collapse
|
117
|
Enhancement of hypoxia-activated prodrug TH-302 anti-tumor activity by Chk1 inhibition. BMC Cancer 2015; 15:422. [PMID: 25994202 PMCID: PMC4453293 DOI: 10.1186/s12885-015-1387-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/28/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The hypoxia-activated prodrug TH-302 is reduced at its nitroimidazole group and selectively under hypoxic conditions releases the DNA cross-linker bromo-isophosphoramide mustard (Br-IPM). Here, we have explored the effect of Chk1 inhibition on TH-302-mediated pharmacological activities. METHODS We employed in vitro cell viability, DNA damage, cellular signaling assays and the in vivo HT29 human tumor xenograft model to study the effect of Chk1inhibition on TH-302 antitumor activities. RESULTS TH-302 cytotoxicity is greatly enhanced by Chk1 inhibition in p53-deficient but not in p53-proficient human cancer cell lines. Chk1 inhibitors reduced TH-302-induced cell cycle arrest via blocking TH-302-induced decrease of phosphorylation of histone H3 and increasing Cdc2-Y15 phosphorylation. Employing the single-cell gel electrophoresis (comet) assay, we observed a potentiation of the TH-302 dependent tail moment. TH-302 induced γH2AX and apoptosis were also increased upon the addition of Chk1 inhibitor. Potentiation of TH-302 cytotoxicity by Chk1 inhibitor was only observed in cell lines proficient in, but not deficient in homology-directed DNA repair. We also show that combination treatment led to lowering of Rad51 expression levels as compared to either agent alone. In vivo data demonstrate that Chk1 inhibitor enhances TH-302 anti-tumor activity in p53 mutant HT-29 human tumor xenografts, supporting the hypothesis that these in vitro results can translate to enhanced in vivo efficacy of the combination. CONCLUSIONS TH-302-mediated in vitro and in vivo anti-tumor activities were greatly enhanced by the addition of Chk1 inhibitors. The preclinical data presented in this study support a new approach for the treatment of p53-deficient hypoxic cancers by combining Chk1 inhibitors with the hypoxia-activated prodrug TH-302.
Collapse
|
118
|
Borad MJ, Reddy SG, Bahary N, Uronis HE, Sigal D, Cohn AL, Schelman WR, Stephenson J, Chiorean EG, Rosen PJ, Ulrich B, Dragovich T, Del Prete SA, Rarick M, Eng C, Kroll S, Ryan DP. Randomized Phase II Trial of Gemcitabine Plus TH-302 Versus Gemcitabine in Patients With Advanced Pancreatic Cancer. J Clin Oncol 2015; 33:1475-81. [PMID: 25512461 PMCID: PMC4881365 DOI: 10.1200/jco.2014.55.7504] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
PURPOSE TH-302 is an investigational hypoxia-activated prodrug that releases the DNA alkylator bromo-isophosphoramide mustard in hypoxic settings. This phase II study (NCT01144455) evaluated gemcitabine plus TH-302 in patients with previously untreated, locally advanced or metastatic pancreatic cancer. PATIENTS AND METHODS Patients were randomly assigned 1:1:1 to gemcitabine (1,000 mg/m(2)), gemcitabine plus TH-302 240 mg/m(2) (G+T240), or gemcitabine plus TH-302 340 mg/m(2) (G+T340). Randomized crossover after progression on gemcitabine was allowed. The primary end point was progression-free survival (PFS). Secondary end points included overall survival (OS), tumor response, CA 19-9 response, and safety. RESULTS Two hundred fourteen patients (77% with metastatic disease) were enrolled between June 2010 and July 2011. PFS was significantly longer with gemcitabine plus TH-302 (pooled combination arms) compared with gemcitabine alone (median PFS, 5.6 v 3.6 months, respectively; hazard ratio, 0.61; 95% CI, 0.43 to 0.87; P = .005; median PFS for metastatic disease, 5.1 v 3.4 months, respectively). Median PFS times for G+T240 and G+T340 were 5.6 and 6.0 months, respectively. Tumor response was 12%, 17%, and 26% in the gemcitabine, G+T240, and G+T340 arms, respectively (G+T340 v gemcitabine, P = .04). CA 19-9 decrease was greater with G+T340 versus gemcitabine (-5,398 v -549 U/mL, respectively; P = .008). Median OS times for gemcitabine, G+T240, and G+T340 were 6.9, 8.7, and 9.2 months, respectively (P = not significant). The most common adverse events (AEs) were fatigue, nausea, and peripheral edema (frequencies similar across arms). Skin and mucosal toxicities (2% grade 3) and myelosuppression (55% grade 3 or 4) were the most common TH-302-related AEs but were not associated with treatment discontinuation. CONCLUSION PFS, tumor response, and CA 19-9 response were significantly improved with G+TH-302. G+T340 is being investigated further in the phase III MAESTRO study (NCT01746979).
Collapse
Affiliation(s)
- Mitesh J Borad
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA.
| | - Shantan G Reddy
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Nathan Bahary
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Hope E Uronis
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Darren Sigal
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Allen L Cohn
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - William R Schelman
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Joe Stephenson
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - E Gabriela Chiorean
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Peter J Rosen
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Brian Ulrich
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Tomislav Dragovich
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Salvatore A Del Prete
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Mark Rarick
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Clarence Eng
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - Stew Kroll
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| | - David P Ryan
- Mitesh J. Borad, Mayo Clinic, Scottsdale; Tomislav Dragovich, Arizona Cancer Center, Tucson, AZ; Shantan G. Reddy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA; Nathan Bahary, University of Pittsburgh Medical Center, Pittsburgh, PA; Hope E. Uronis, Duke University Medical Center, Durham, NC; Darren Sigal, Scripps Clinic, La Jolla; Peter J. Rosen, Disney Family Cancer Center, Burbank; Clarence Eng and Stew Kroll, Threshold Pharmaceuticals, South San Francisco, CA; Allen L. Cohn, Rocky Mountain Cancer Center, Denver, CO; William R. Schelman, University of Wisconsin Carbone Cancer Center, Madison, WI; Joe Stephenson Jr, Institute for Translational Oncology Research, Greenville, SC; E. Gabriela Chiorean, Indiana University Simon Cancer Center, Indianapolis, IN; Brian Ulrich, Texas Oncology, Wichita Falls, TX; Salvatore A. Del Prete, Hematology Oncology PC, Stamford, CT; Mark Rarick, Kaiser Permanente Northwest Region Oncology Hematology, Portland, OR; and David P. Ryan, Massachusetts General Hospital Cancer Center, Boston, MA
| |
Collapse
|
119
|
Han Y, Chen W, Kuang Y, Sun H, Wang Z, Peng X. UV-Induced DNA Interstrand Cross-Linking and Direct Strand Breaks from a New Type of Binitroimidazole Analogue. Chem Res Toxicol 2015; 28:919-26. [PMID: 25844639 DOI: 10.1021/tx500522r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Four novel photoactivated binitroimidazole prodrugs were synthesized. These agents produced DNA interstrand cross-links (ICLs) and direct strand breaks (DSB) upon UV irradiation, whereas no or very few DNA ICLs and DSBs were observed without UV treatment. Although these four molecules (1-4) contain the same binitroimidazole moiety, they bear four different leaving groups, which resulted in their producing different yields of DNA damage. Compound 4, with nitrogen mustard as a leaving group, showed the highest ICL yield. Surprisingly, compounds 1-3, without any alkylating functional group, also induced DNA ICL formation, although they did so with lower yields, which suggested that the binitroimidazole moiety released from UV irradiation of 1-3 is capable of cross-linking DNA. The DNA cross-linked products induced by these compounds were completely destroyed upon 1.0 M piperidine treatment at 90 °C (leading to cleavage at dG sites), which revealed that DNA cross-linking mainly occurred via alkylation of dGs. We proposed a possible mechanism by which alkylating agents were released from these compounds. HRMS and NMR analysis confirmed that free nitrogen mustards were generated by UV irradiation of 4. Suppression of DNA ICL and DSB formation by a radical trap, TEMPO, indicated the involvement of free radicals in the photo reactions of 3 and 4 with DNA. On the basis of these data, we propose that UV irradiation of compounds 1-4 generated a binitroimidazole intermediate that cross-links DNA. The higher ICL yield observed with 4 resulted from the amine effector nitrogen mustard released from UV irradiation.
Collapse
Affiliation(s)
- Yanyan Han
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| | - Wenbing Chen
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| | - Yunyan Kuang
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| | - Huabing Sun
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| | - Zhiqiang Wang
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| | - Xiaohua Peng
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 3210 North Cramer Street, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
120
|
Saggar JK, Tannock IF. Chemotherapy Rescues Hypoxic Tumor Cells and Induces Their Reoxygenation and Repopulation-An Effect That Is Inhibited by the Hypoxia-Activated Prodrug TH-302. Clin Cancer Res 2015; 21:2107-14. [PMID: 25677696 DOI: 10.1158/1078-0432.ccr-14-2298] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/30/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE Chemotherapy targets rapidly proliferating tumor cells, but spares slowly proliferating hypoxic cells. We hypothesized that nutrition of hypoxic cells would improve in intervals between chemotherapy, and that hypoxic cells destined to die without treatment would survive and proliferate. EXPERIMENTAL DESIGN We therefore evaluated repopulation and reoxygenation following chemotherapy, and the effects of the hypoxia-activated prodrug TH-302 on these processes. Tumor-bearing mice were treated with doxorubicin or docetaxel ± TH-302. Pimonidazole (given concurrent with chemotherapy) and EF5 (given 24 to 120 hours later) identified hypoxic cells. Proliferation (Ki67) and oxygen status (EF5 uptake) of formerly hypoxic (pimo positive) cells were quantified by immunohistochemistry. RESULTS Chronically hypoxic cells had limited proliferation in control tumors. After chemotherapy, we observed reoxygenation and increased proliferation of previously hypoxic cells; these processes were inhibited by TH-302. CONCLUSIONS Chemotherapy leads to paradoxical sparing of hypoxic cells destined to die in solid tumors in absence of treatment, and their reoxygenation and proliferation: TH-302 inhibits these processes.
Collapse
Affiliation(s)
- Jasdeep K Saggar
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Ian F Tannock
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada. Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
| |
Collapse
|
121
|
Shyam K, Penketh PG, Baumann RP, Finch RA, Zhu R, Zhu YL, Sartorelli AC. Antitumor sulfonylhydrazines: design, structure-activity relationships, resistance mechanisms, and strategies for improving therapeutic utility. J Med Chem 2015; 58:3639-71. [PMID: 25612194 DOI: 10.1021/jm501459c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
1,2-Bis(sulfonyl)-1-alkylhydrazines (BSHs) were conceived as more specific DNA guanine O-6 methylating and chloroethylating agents lacking many of the undesirable toxicophores contained in antitumor nitrosoureas. O(6)-Alkylguanine-DNA alkyltransferase (MGMT) is the sole repair protein for O(6)-alkylguanine lesions in DNA and has been reported to be absent in 5-20% of most tumor types. Many BSHs exhibit highly selective cytotoxicity toward cells deficient in MGMT activity. The development of clinically useful MGMT assays should permit the identification of tumors with this vulnerability and allow for the preselection of patient subpopulations with a high probability of responding. The BSH system is highly versatile, permitting the synthesis of many prodrug types with the ability to incorporate an additional level of tumor-targeting due to preferential activation by tumor cells. Furthermore, it may be possible to expand the spectrum of activity of these agents to include tumors with MGMT activity by combining them with tumor-targeted MGMT inhibitors.
Collapse
Affiliation(s)
- Krishnamurthy Shyam
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Philip G Penketh
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Raymond P Baumann
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Rick A Finch
- ‡Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, 650 Cool Water Drive, Bastrop, Texas 78602, United States
| | - Rui Zhu
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Yong-Lian Zhu
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| | - Alan C Sartorelli
- †Department of Pharmacology and Developmental Therapeutics Program, Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8066, United States
| |
Collapse
|
122
|
Cheng W, Zhu S, Ma X, Qiu N, Peng P, Sheng R, Hu Y. Design, synthesis and biological evaluation of 6-(nitroimidazole-1H-alkyloxyl)-4-anilinoquinazolines as efficient EGFR inhibitors exerting cytotoxic effects both under normoxia and hypoxia. Eur J Med Chem 2015; 89:826-34. [DOI: 10.1016/j.ejmech.2014.11.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/27/2014] [Accepted: 11/05/2014] [Indexed: 11/16/2022]
|
123
|
O'Connor LJ, Cazares-Körner C, Saha J, Evans CNG, Stratford MRL, Hammond EM, Conway SJ. Efficient synthesis of 2-nitroimidazole derivatives and the bioreductive clinical candidate Evofosfamide (TH-302). Org Chem Front 2015. [DOI: 10.1039/c5qo00211g] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We describe an improved synthesis of the 2-nitroimidazole bioreductive group; this method is used to synthesise the anti-cancer drug Evofosfamide (TH-302.)
Collapse
Affiliation(s)
- Liam J. O'Connor
- Department of Chemistry
- Chemistry Research Laboratory
- University of Oxford
- Oxford
- UK
| | - Cindy Cazares-Körner
- Department of Chemistry
- Chemistry Research Laboratory
- University of Oxford
- Oxford
- UK
| | - Jaideep Saha
- Department of Chemistry
- Chemistry Research Laboratory
- University of Oxford
- Oxford
- UK
| | - Charles N. G. Evans
- Department of Chemistry
- Chemistry Research Laboratory
- University of Oxford
- Oxford
- UK
| | - Michael R. L. Stratford
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology
- Department of Oncology
- University of Oxford
- Oxford
- UK
| | - Ester M. Hammond
- Cancer Research UK/MRC Oxford Institute for Radiation Oncology
- Department of Oncology
- University of Oxford
- Oxford
- UK
| | - Stuart J. Conway
- Department of Chemistry
- Chemistry Research Laboratory
- University of Oxford
- Oxford
- UK
| |
Collapse
|
124
|
Bailey KM, Cornnell HH, Ibrahim-Hashim A, Wojtkowiak JW, Hart CP, Zhang X, Leos R, Martinez GV, Baker AF, Gillies RJ. Evaluation of the "steal" phenomenon on the efficacy of hypoxia activated prodrug TH-302 in pancreatic cancer. PLoS One 2014; 9:e113586. [PMID: 25532146 PMCID: PMC4273999 DOI: 10.1371/journal.pone.0113586] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/26/2014] [Indexed: 11/19/2022] Open
Abstract
Pancreatic ductal adenocarcinomas are desmoplastic and hypoxic, both of which are associated with poor prognosis. Hypoxia-activated prodrugs (HAPs) are specifically activated in hypoxic environments to release cytotoxic or cytostatic effectors. TH-302 is a HAP that is currently being evaluated in a Phase III clinical trial in pancreatic cancer. Using animal models, we show that tumor hypoxia can be exacerbated using a vasodilator, hydralazine, improving TH-302 efficacy. Hydralazine reduces tumor blood flow through the "steal" phenomenon, in which atonal immature tumor vasculature fails to dilate in coordination with normal vasculature. We show that MIA PaCa-2 tumors exhibit a "steal" effect in response to hydralazine, resulting in decreased tumor blood flow and subsequent tumor pH reduction. The effect is not observed in SU.86.86 tumors with mature tumor vasculature, as measured by CD31 and smooth muscle actin (SMA) immunohistochemistry staining. Combination therapy of hydralazine and TH-302 resulted in a reduction in MIA PaCa-2 tumor volume growth after 18 days of treatment. These studies support a combination mechanism of action for TH-302 with a vasodilator that transiently increases tumor hypoxia.
Collapse
Affiliation(s)
- Kate M. Bailey
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida 33612, United States of America
| | - Heather H. Cornnell
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Arig Ibrahim-Hashim
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Jonathan W. Wojtkowiak
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Charles P. Hart
- Threshold Pharmaceuticals, South San Francisco, California 94080, United States of America
| | - Xiaomeng Zhang
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Rafael Leos
- Arizona Cancer Center, Hematology/Oncology Section, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States of America
| | - Gary V. Martinez
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Amanda F. Baker
- Arizona Cancer Center, Hematology/Oncology Section, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States of America
| | - Robert J. Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
- * E-mail:
| |
Collapse
|
125
|
Li Y, Li P, Jin M, Jiang C, Gao Z. Docetaxel-encapsulating small-sized polymeric micelles with higher permeability and its efficacy on the orthotopic transplantation model of pancreatic ductal adenocarcinoma. Int J Mol Sci 2014; 15:23571-88. [PMID: 25526569 PMCID: PMC4284781 DOI: 10.3390/ijms151223571] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) elicits a dense stromal response that blocks vascular access because of pericyte coverage of vascular fenestrations. In this way, the PDAC stroma contributes to chemotherapy resistance, and the small-sized nanocarrier loaded with platinum has been adopted to address this problem which is not suitable for loading docetaxel (DTX). In the present study, we used the poly(d,l-lactide)-b-polyethylene glycol-methoxy (mPEG-b-PDLLA) to encapsulate DTX and got a small-sized polymeric micelle (SPM); meanwhile we functionalized the SPM’s surface with TAT peptide (TAT-PM) for a higher permeability. The diameters of both SPM and TAT-PM were in the range of 15–26 nm. In vitro experiments demonstrated that TAT-PM inhibited Capan-2 Luc PDAC cells growth more efficiently and induced more apoptosis compared to SPM and Duopafei. The in vivo therapeutic efficiencies of SPM and TAT-PM compared to free DTX was investigated on the orthotopic transplantation model of Capan-2 Luc. SPM exerted better therapeutic efficiency than free DTX, however, TAT-PM didn’t outperformed SPM. Overall, these results disclosed that SPM could represent a new therapeutic approach against pancreatic cancer, but its permeability to PDAC was not the only decisive factor.
Collapse
Affiliation(s)
- Yunfei Li
- State Key Laboratory of Bioactive Substance and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100050, China.
| | - Peiran Li
- Surgical Department, the Affiliated Hospital of Yanbian University, Yanji 133000, China.
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100050, China.
| | - Changgao Jiang
- Surgical Department, the Affiliated Hospital of Yanbian University, Yanji 133000, China.
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
126
|
Liapis V, Labrinidis A, Zinonos I, Hay S, Ponomarev V, Panagopoulos V, DeNichilo M, Ingman W, Atkins GJ, Findlay DM, Zannettino ACW, Evdokiou A. Hypoxia-activated pro-drug TH-302 exhibits potent tumor suppressive activity and cooperates with chemotherapy against osteosarcoma. Cancer Lett 2014; 357:160-169. [PMID: 25444931 DOI: 10.1016/j.canlet.2014.11.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/07/2014] [Accepted: 11/11/2014] [Indexed: 12/19/2022]
Abstract
Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, tumor hypoxia also offers treatment opportunities, exemplified by the development compounds that target hypoxic regions within tumors. TH-302 is a pro-drug created by the conjugation of 2-nitroimidazole to bromo-isophosphoramide (Br-IPM). When TH-302 is delivered to regions of hypoxia, Br-IPM, the DNA cross linking toxin, is released. In this study we assessed the cytotoxic activity of TH-302 against osteosarcoma cells in vitro and evaluated its anticancer efficacy as a single agent, and in combination with doxorubicin, in an orthotopic mouse model of human osteosarcoma (OS). In vitro, TH-302 was potently cytotoxic to osteosarcoma cells selectively under hypoxic conditions, whereas primary normal human osteoblasts were protected. Animals transplanted with OS cells directly into their tibiae and left untreated developed mixed osteolytic/osteosclerotic bone lesions and subsequently developed lung metastases. TH-302 reduced tumor burden in bone and cooperated with doxorubicin to protect bone from osteosarcoma induced bone destruction, while it also reduced lung metastases. TH-302 may therefore be an attractive therapeutic agent with strong activity as a single agent and in combination with chemotherapy against OS.
Collapse
Affiliation(s)
- Vasilios Liapis
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia
| | - Agatha Labrinidis
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia
| | - Irene Zinonos
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia
| | - Shelley Hay
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Vasilios Panagopoulos
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia
| | - Mark DeNichilo
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia
| | - Wendy Ingman
- Discipline of Surgery, Haematology - Oncology, Breast Biology Cancer Unit, Basil Hetzel Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Gerald J Atkins
- Discipline of Orthopaedics and Trauma, University of Adelaide, Adelaide, South Australia, Australia
| | - David M Findlay
- Discipline of Orthopaedics and Trauma, University of Adelaide, Adelaide, South Australia, Australia
| | - Andrew C W Zannettino
- School of Medical Sciences, Myeloma Research Laboratory Cancer Theme, South Australian Health and Medical Research Institute (SAHMRI), Faculty of Health Science, University of Adelaide, Australia
| | - Andreas Evdokiou
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide Woodville, South Australia, Australia.
| |
Collapse
|
127
|
Takakusagi Y, Matsumoto S, Saito K, Matsuo M, Kishimoto S, Wojtkowiak JW, DeGraff W, Kesarwala AH, Choudhuri R, Devasahayam N, Subramanian S, Munasinghe JP, Gillies RJ, Mitchell JB, Hart CP, Krishna MC. Pyruvate induces transient tumor hypoxia by enhancing mitochondrial oxygen consumption and potentiates the anti-tumor effect of a hypoxia-activated prodrug TH-302. PLoS One 2014; 9:e107995. [PMID: 25254649 PMCID: PMC4177858 DOI: 10.1371/journal.pone.0107995] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/18/2014] [Indexed: 01/15/2023] Open
Abstract
Background TH-302 is a hypoxia-activated prodrug (HAP) of bromo isophosphoramide mustard that is selectively activated within hypoxic regions in solid tumors. Our recent study showed that intravenously administered bolus pyruvate can transiently induce hypoxia in tumors. We investigated the mechanism underlying the induction of transient hypoxia and the combination use of pyruvate to potentiate the anti-tumor effect of TH-302. Methodology/Results The hypoxia-dependent cytotoxicity of TH-302 was evaluated by a viability assay in murine SCCVII and human HT29 cells. Modulation in cellular oxygen consumption and invivo tumor oxygenation by the pyruvate treatment was monitored by extracellular flux analysis and electron paramagnetic resonance (EPR) oxygen imaging, respectively. The enhancement of the anti-tumor effect of TH-302 by pyruvate treatment was evaluated by monitoring the growth suppression of the tumor xenografts inoculated subcutaneously in mice. TH-302 preferentially inhibited the growth of both SCCVII and HT29 cells under hypoxic conditions (0.1% O2), with minimal effect under aerobic conditions (21% O2). Basal oxygen consumption rates increased after the pyruvate treatment in SCCVII cells in a concentration-dependent manner, suggesting that pyruvate enhances the mitochondrial respiration to consume excess cellular oxygen. In vivo EPR oxygen imaging showed that the intravenous administration of pyruvate globally induced the transient hypoxia 30 min after the injection in SCCVII and HT29 tumors at the size of 500–1500 mm3. Pretreatment of SCCVII tumor bearing mice with pyruvate 30 min prior to TH-302 administration, initiated with small tumors (∼550 mm3), significantly delayed tumor growth. Conclusions/Significance Our invitro and invivo studies showed that pyruvate induces transient hypoxia by enhancing mitochondrial oxygen consumption in tumor cells. TH-302 therapy can be potentiated by pyruvate pretreatment if started at the appropriate tumor size and oxygen concentration.
Collapse
Affiliation(s)
- Yoichi Takakusagi
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Shingo Matsumoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Keita Saito
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Masayuki Matsuo
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jonathan W. Wojtkowiak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - William DeGraff
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Aparna H. Kesarwala
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Rajani Choudhuri
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Sankaran Subramanian
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jeeva P. Munasinghe
- National Institute of Neurological Diseases and Stroke, Bethesda, Maryland, United States of America
| | - Robert J. Gillies
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, United States of America
| | - James B. Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Charles P. Hart
- Threshold Pharmaceuticals, South San Francisco, California, United States of America
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
128
|
Hunter FW, Hsu HL, Su J, Pullen SM, Wilson WR, Wang J. Dual targeting of hypoxia and homologous recombination repair dysfunction in triple-negative breast cancer. Mol Cancer Ther 2014; 13:2501-14. [PMID: 25193512 DOI: 10.1158/1535-7163.mct-14-0476] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy with poor clinical outcome and few validated drug targets. Two prevalent features of TNBC, tumor hypoxia and derangement of homologous recombination (HR) repair, are potentially exploitable for therapy. This study investigated whether hypoxia-activated prodrugs (HAP) of DNA-damaging cytotoxins may inhibit growth of TNBC by simultaneously addressing these two targets. We measured in vitro activity of HAP of DNA breakers (tirapazamine, SN30000) and alkylators (TH-302, PR-104, SN30548) in TNBC cell lines and isogenic models, and related this to measures of HR repair and expression of prodrug-activating enzymes. Antitumor activity of HAP was examined in isogenic BRCA2-knockout xenograft models and compared with platinum chemotherapy. All five HAP selectively inhibited growth of TNBC cell lines under hypoxia. Sensitivity to HAP was not strongly associated with BRCA1 genotype. However, HAP sensitivity was enhanced by suppression of HR (assessed by radiation-induced RAD51 focus formation) when BRCA1 and PALB2 were knocked down in a common (MDA-MB-231) background. Furthermore, knockout of BRCA2 markedly sensitized DLD-1 cells to the clinical nitrogen mustard prodrugs TH-302 and PR-104 and significantly augmented sterilization of clonogens by these agents in xenografts, both as monotherapy and in combination with radiotherapy, but had less effect on activity of the benzotriazine di-N-oxide SN30000. PR-104 monotherapy was more effective than cisplatin at inhibiting growth of BRCA2-knockout tumors at equitoxic doses. This study demonstrates the potential for HAP of nitrogen mustards to simultaneously exploit hypoxia and HR defects in tumors, with translational implications for TNBC and other HR-deficient malignancies.
Collapse
Affiliation(s)
- Francis W Hunter
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Huai-Ling Hsu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jiechuang Su
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Susan M Pullen
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Jingli Wang
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
129
|
Muz B, de la Puente P, Azab F, Luderer M, Azab AK. The role of hypoxia and exploitation of the hypoxic environment in hematologic malignancies. Mol Cancer Res 2014; 12:1347-54. [PMID: 25158954 DOI: 10.1158/1541-7786.mcr-14-0028] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor hypoxia is a well-described phenomenon during the progression of solid tumors affecting cell signaling pathways and cell metabolism; however, its role in hematologic malignancies has not been given the same attention in the literature. Therefore, this review focuses on the comparative differences between solid and hematologic malignancies with emphasis on the role of hypoxia during tumorigenesis and progression. In addition, contribution of the bone marrow and angiogenic environment are also discussed. Insight is provided into the role of hypoxia in metastatic spread, stemness, and drug resistance in hematologic conditions. Finally, emerging therapeutic strategies such as small-molecule prodrugs and hypoxia-inducible factor (HIF) targeting approaches are outlined to combat hypoxic cells and/or adaptive mechanisms in the treatment of hematologic malignancies.
Collapse
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Micah Luderer
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, Missouri
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, St. Louis, Missouri.
| |
Collapse
|
130
|
Johnson K, Parsons ZD, Barnes CL, Gates KS. Toward hypoxia-selective DNA-alkylating agents built by grafting nitrogen mustards onto the bioreductively activated, hypoxia-selective DNA-oxidizing agent 3-amino-1,2,4-benzotriazine 1,4-dioxide (tirapazamine). J Org Chem 2014; 79:7520-31. [PMID: 25029663 PMCID: PMC4136725 DOI: 10.1021/jo501252p] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Indexed: 12/14/2022]
Abstract
Tirapazamine (3-amino-1,2,4-benzotriazine 1,4-dioxide) is a heterocyclic di-N-oxide that undergoes enzymatic deoxygenation selectively in the oxygen-poor (hypoxic) cells found in solid tumors to generate a mono-N-oxide metabolite. This work explored the idea that the electronic changes resulting from the metabolic deoxygenation of tirapazamine analogues might be exploited to activate a DNA-alkylating species selectively in hypoxic tissue. Toward this end, tirapazamine analogues bearing nitrogen mustard units were prepared. In the case of the tirapazamine analogue 18a bearing a nitrogen mustard unit at the 6-position, it was found that removal of the 4-oxide from the parent di-N-oxide to generate the mono-N-oxide analogue 17a did indeed cause a substantial increase in reactivity of the mustard unit, as measured by hydrolysis rates and DNA-alkylation yields. Hammett sigma values were measured to quantitatively assess the magnitude of the electronic changes induced by metabolic deoxygenation of the 3-amino-1,2,4-benzotriazine 1,4-dioxide heterocycle. The results provide evidence that the 1,2,4-benzotiazine 1,4-dioxide unit can serve as an oxygen-sensing prodrug platform for the selective unmasking of bioactive agents in hypoxic cells.
Collapse
Affiliation(s)
- Kevin
M. Johnson
- Departments of Chemistry and Biochemistry, University of Missouri, 125 Chemistry Building, Columbia, Missouri 65211, United
States
| | - Zachary D. Parsons
- Departments of Chemistry and Biochemistry, University of Missouri, 125 Chemistry Building, Columbia, Missouri 65211, United
States
| | - Charles L. Barnes
- Departments of Chemistry and Biochemistry, University of Missouri, 125 Chemistry Building, Columbia, Missouri 65211, United
States
| | - Kent S. Gates
- Departments of Chemistry and Biochemistry, University of Missouri, 125 Chemistry Building, Columbia, Missouri 65211, United
States
| |
Collapse
|
131
|
Lou Q, Ji L, Zhong W, Li S, Yu S, Li Z, Meng X. Synthesis and cytotoxicity evaluation of naphthalimide derived N-mustards. Molecules 2014; 19:8803-19. [PMID: 24968335 PMCID: PMC6271267 DOI: 10.3390/molecules19078803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/10/2014] [Accepted: 06/17/2014] [Indexed: 12/23/2022] Open
Abstract
A series of N-mustards, which was conjugated to mono- or bis-naphthalimides with a flexible amine link, were synthesized and evaluated for cytotoxicity against five cancer cell lines (HCT-116, PC-3, U87 MG, Hep G2 and SK-OV-3). Several compounds displayed better activities than the control compound amonafide. Further evaluations by fluorescence spectroscopy studies and DNA-interstrand cross-linking assays revealed that the derivatives showed both alkylating and intercalating properties. Among the derivatives, the bis-naphthalimide N-mustard derivative 11b was found to exhibit the highest cytotoxic activity and DNA cross-linking ability. Both 11b and 7b induce HCT-116 cell apoptosis by S phase arrest.
Collapse
Affiliation(s)
- Qinghua Lou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Liyan Ji
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Wenhe Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Shasha Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Siwang Yu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Zhongjun Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
132
|
Guise CP, Mowday AM, Ashoorzadeh A, Yuan R, Lin WH, Wu DH, Smaill JB, Patterson AV, Ding K. Bioreductive prodrugs as cancer therapeutics: targeting tumor hypoxia. CHINESE JOURNAL OF CANCER 2014; 33:80-6. [PMID: 23845143 PMCID: PMC3935009 DOI: 10.5732/cjc.012.10285] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 02/28/2013] [Accepted: 04/26/2013] [Indexed: 11/11/2022]
Abstract
Hypoxia, a state of low oxygen, is a common feature of solid tumors and is associated with disease progression as well as resistance to radiotherapy and certain chemotherapeutic drugs. Hypoxic regions in tumors, therefore, represent attractive targets for cancer therapy. To date, five distinct classes of bioreactive prodrugs have been developed to target hypoxic cells in solid tumors. These hypoxia-activated prodrugs, including nitro compounds, N-oxides, quinones, and metal complexes, generally share a common mechanism of activation whereby they are reduced by intracellular oxidoreductases in an oxygen-sensitive manner to form cytotoxins. Several examples including PR-104, TH-302, and EO9 are currently undergoing phase II and phase III clinical evaluation. In this review, we discuss the nature of tumor hypoxia as a therapeutic target, focusing on the development of bioreductive prodrugs. We also describe the current knowledge of how each prodrug class is activated and detail the clinical progress of leading examples.
Collapse
Affiliation(s)
- Christopher P Guise
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Oliveira FM, Barbosa LCA, Ismail FMD. The diverse pharmacology and medicinal chemistry of phosphoramidates – a review. RSC Adv 2014. [DOI: 10.1039/c4ra01454e] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Promising examples of the phosphoramidates, which possess antiviral, antitumor, antibacterial, antimalarial and anti-protozoal as well as enzyme inhibitor activity are reviewed.
Collapse
Affiliation(s)
| | - Luiz C. A. Barbosa
- Department of Chemistry
- Universidade Federal de Viçosa
- Viçosa, Brazil
- Department of Chemistry
- Universidade Federal de Minas Gerais
| | - Fyaz M. D. Ismail
- Medicinal Chemistry Research Group
- Institute for Health Research
- School of Pharmacy & Biomolecular Sciences
- Liverpool John Moores University
- Liverpool, UK
| |
Collapse
|
134
|
Hypoxia-Directed Drug Strategies to Target the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:111-45. [DOI: 10.1007/978-1-4614-5915-6_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
135
|
Abstract
In the past 10 years, we have developed a new approach to the development of a clinically accurate rodent model for human cancer based on our invention of surgical orthotopic implantation (SOI). The SOI models have been described in approx. 70 publications and in 4 patents.*SOI allows human tumors of all the major types of human cancer to reproduce clinical like tumor growth and metastasis in the transplanted rodents. The major features of the SOI models are reviewed here and also compared to transgenic mouse models of cancer.
Collapse
|
136
|
Histological advantages of the tumor graft: a murine model involving transplantation of human pancreatic cancer tissue fragments. Pancreas 2013; 42:1275-82. [PMID: 24152953 DOI: 10.1097/mpa.0b013e318296f866] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES Experimental data based on cell line-derived xenograft models (cell xenograft) seldom reproduce the clinical situation, and therefore we demonstrated here the superiority of a murine model involving transplantation of human pancreatic cancer tissue fragments (tumor graft), focusing on the histological features and drug delivery characteristics. METHODS Tumor pieces from 10 pancreatic cancer patients were transplanted into SCID (severe combined immunodeficient) mice. Histological characteristics of tumor grafts, including morphology, desmoplastic reaction, and vascularization, were compared with those of cell xenografts. Drug delivery was evaluated by quantifying the concentrations of injected drug, and the results were compared with its histological features. RESULTS Eight of the 10 transplanted tumors successfully engrafted. Histological comparisons between tumor grafts and cell xenografts revealed the following: the amount of stroma was more (22.9% ± 11.8% vs 10.8% ± 5.4%; P < 0.05), vessel-cancer cell distance was longer (35.3 ± 39.0 vs 3.9 ± 3.1 μm; P < 0.001), and microvessel density was lower (6.8 ± 1.9 vs 10.8 ± 2.1 vessels/0.4 mm(2); P < 0.05) in tumor grafts. Drug concentrations in tumor grafts were lower than those in cell xenografts (3.3 ± 1.2 vs 6.0±0.2 μg/mL; P = 0.003), and the differences were correlated with the histological differences. CONCLUSIONS Pancreatic tumor grafts better reproduce the histological nature of clinical cancer and thus provide a more realistic model that is applicable for pharmacokinetic studies.
Collapse
|
137
|
Benito J, Zeng Z, Konopleva M, Wilson WR. Targeting hypoxia in the leukemia microenvironment. Int J Hematol Oncol 2013; 2:279-288. [PMID: 24490034 DOI: 10.2217/ijh.13.32] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The bone marrow (BM) microenvironment regulates survival and maintenance of normal hematopoietic stem cells. Within the endosteal niche, hypoxia has an essential role in maintenance of the primitive quiescent hematopoietic stem cell. We and others have demonstrated that in the context of hematologic malignancies the BM is highly hypoxic, and that progression of the disease is associated with expansion of hypoxic niches and stabilization of the oncogenic HIF-1α. This review will provide an overview of the normal and leukemic BM microenvironment with a special emphasis on pathological hypoxia including the development of hypoxia-activated prodrugs and their applicability in hematological malignancies.
Collapse
Affiliation(s)
- Juliana Benito
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston TX 77030, USA
| | - Zhihong Zeng
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston TX 77030, USA
| | - Marina Konopleva
- Division of Cancer Medicine - Unit 448, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston TX 77030, USA
| | - William R Wilson
- Auckland Cancer Society Research Centre, University of Auckland, Private Bag 92019, Auckland, New Zealand
| |
Collapse
|
138
|
Hu J, Van Valckenborgh E, Xu D, Menu E, De Raeve H, De Bruyne E, De Bryune E, Xu S, Van Camp B, Handisides D, Hart CP, Vanderkerken K. Synergistic induction of apoptosis in multiple myeloma cells by bortezomib and hypoxia-activated prodrug TH-302, in vivo and in vitro. Mol Cancer Ther 2013; 12:1763-73. [PMID: 23832122 DOI: 10.1158/1535-7163.mct-13-0123] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recently, we showed that hypoxia is a critical microenvironmental factor in multiple myeloma, and that the hypoxia-activated prodrug TH-302 selectively targets hypoxic multiple myeloma cells and improves multiple disease parameters in vivo. To explore approaches for sensitizing multiple myeloma cells to TH-302, we evaluated in this study the antitumor effect of TH-302 in combination with the clinically used proteasome inhibitor bortezomib. First, we show that TH-302 and bortezomib synergistically induce apoptosis in multiple myeloma cell lines in vitro. Second, we confirm that this synergism is related to the activation of caspase cascades and is mediated by changes of Bcl-2 family proteins. The combination treatment induces enhanced cleavage of caspase-3/8/9 and PARP, and therefore triggers apoptosis and enhances the cleavage of proapoptotic BH3-only protein BAD and BID as well as the antiapoptotic protein Mcl-1. In particular, TH-302 can abrogate the accumulation of antiapoptotic Mcl-1 induced by bortezomib, and decreases the expression of the prosurvival proteins Bcl-2 and Bcl-xL. Furthermore, we found that the induction of the proapoptotic BH3-only proteins PUMA (p53-upregulated modulator of apoptosis) and NOXA is associated with this synergism. In response to the genotoxic and endoplasmic reticulum stresses by TH-302 and bortezomib, the expression of PUMA and NOXA were upregulated in p53-dependent and -independent manners. Finally, in the murine 5T33MMvv model, we showed that the combination of TH-302 and bortezomib can improve multiple disease parameters and significantly prolong the survival of diseased mice. In conclusion, our studies provide a rationale for clinical evaluation of the combination of TH-302 and bortezomib in patients with multiple myeloma.
Collapse
Affiliation(s)
- Jinsong Hu
- Corresponding Author: Karin Vanderkerken, Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel, Laarbeeklaan 103, B-1090 Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Rajapakse A, Linder C, Morrison RD, Sarkar U, Leigh ND, Barnes CL, Daniels JS, Gates KS. Enzymatic conversion of 6-nitroquinoline to the fluorophore 6-aminoquinoline selectively under hypoxic conditions. Chem Res Toxicol 2013; 26:555-63. [PMID: 23488987 DOI: 10.1021/tx300483z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
There is substantial interest in small molecules that can be used to detect or kill the hypoxic (low oxygen) cells found in solid tumors. Nitroaryl moieties are useful components in the design of hypoxia-selective imaging agents and prodrugs because one-electron reductases can convert the nitroaryl group to nitroso, hydroxylamino, and amino metabolites selectively under low oxygen conditions. Here, we describe the in vitro, cell free metabolism of a pro-fluorescent substrate, 6-nitroquinoline (1) under both aerobic and hypoxic conditions. Both LC-MS and fluorescence spectroscopic analyses provided evidence that the one-electron reducing enzyme system, xanthine/xanthine oxidase, converted the nonfluorescent parent compound 1 to the known fluorophore 6-aminoquinoline (2) selectively under hypoxic conditions. The presumed intermediate in this reduction process, 6-hydroxylaminoquinoline (6), is fluorescent and can be efficiently converted by xanthine/xanthine oxidase to 2 only under hypoxic conditions. This finding provides evidence for multiple oxygen-sensitive steps in the enzymatic conversion of nitroaryl compounds to the corresponding amino derivatives. In a side reaction that is separate from the bioreductive metabolism of 1, xanthine oxidase converted 1 to 6-nitroquinolin-2(1H)-one (5). These studies may enable the use of 1 as a fluorescent substrate for the detection and profiling of one-electron reductases in cell culture or biopsy samples. In addition, the compound may find use as a fluorogenic probe for the detection of hypoxia in tumor models. The occurrence of side products such as 5 in the enzymatic bioreduction of 1 underscores the importance of metabolite identification in the characterization of hypoxia-selective probes and drugs that employ nitroaryl units as oxygen sensors.
Collapse
Affiliation(s)
- Anuruddha Rajapakse
- Department of Chemistry, University of Missouri , 125 Chemistry Building, Columbia, Missouri 65211, United States
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Abstract
A prodrug is a compound that has negligible, or lower, activity against a specified pharmacological target than one of its major metabolites. Prodrugs can be used to improve drug delivery or pharmacokinetics, to decrease toxicity, or to target the drug to specific cells or tissues. Ester and phosphate hydrolysis are widely used in prodrug design because of their simplicity, but such approaches are relatively ineffective for targeting drugs to specific sites. The activation of prodrugs by the cytochrome P450 system provides a highly versatile approach to prodrug design that is particularly adaptable for targeting drug activation to the liver, to tumors or to hypoxic tissues.
Collapse
Affiliation(s)
- Paul R Ortiz de Montellano
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California, San Francisco, CA 94158-2517, USA.
| |
Collapse
|
141
|
Somaiah N, von Mehren M. New drugs and combinations for the treatment of soft-tissue sarcoma: a review. Cancer Manag Res 2012; 4:397-411. [PMID: 23226072 PMCID: PMC3514064 DOI: 10.2147/cmar.s23257] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Sarcomas are a heterogeneous group of solid tumors arising from either soft tissues or bone, accounting for approximately 1% of all cancers in adults. Management of these diseases has changed little over the past 10 years, with the exception of treatment of gastrointestinal stromal tumors. Reasons for this stagnation include multiple histologies commonly grouped together in clinical trials limiting the understanding of benefit of treatment and limited investigation of molecular targeted therapies. More recently, advances in molecular pathogenesis, the advent of novel and targeted therapeutics, and increasing collaborations between sarcoma investigators has helped move the field forward in the right direction. Here, we review the recent data on novel agents tested for the management of adult soft-tissue sarcomas, excluding gastrointestinal stromal tumors.
Collapse
Affiliation(s)
- Neeta Somaiah
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret von Mehren
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
142
|
Hoffman RM. Orthotopic mouse models expressing fluorescent proteins for cancer drug discovery. Expert Opin Drug Discov 2012; 5:851-66. [PMID: 22823260 DOI: 10.1517/17460441.2010.510129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IMPORTANCE OF THE FIELD Currently used rodent tumor models, including transgenic tumor models, or subcutaneously growing human tumors in immunodeficient mice, do not sufficiently represent clinical cancer, especially with regard to metastasis and drug sensitivity. AREAS COVERED IN THIS REVIEW To obtain clinically accurate models, we have developed the technique of surgical orthotopic implantation (SOI) to transplant histologically intact fragments of human cancer, including tumors taken directly from the patient, to the corresponding organ of immunodeficient rodents. SOI allows the growth and metastatic potential of the transplanted tumors to be expressed and reflects clinical cancer of all types. Effective drugs can be discovered and evaluated in the SOI models utilizing human tumor cell lines and patient tumors. Visualization of many aspects of cancer initiation and progression in vivo has been achieved with fluorescent proteins. Tumors and metastases in the SOI models that express fluorescent proteins can be visualized noninvasively in intact animals, greatly facilitating drug discovery. WHAT THE READER WILL GAIN This review will provide information on the imageable mouse models of cancer that are clinically relevant, especially regarding metastasis and their use for drug discovery and evaluation. TAKE HOME MESSAGE SOI mouse models of cancer reproduce the features of clinical cancer.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., 7917 Ostrow Street, San Diego, CA 92111, USA +1 858 654 2555 ; +1 858 268 4175 ;
| |
Collapse
|
143
|
Gillies RJ, Verduzco D, Gatenby RA. Evolutionary dynamics of carcinogenesis and why targeted therapy does not work. Nat Rev Cancer 2012; 12:487-93. [PMID: 22695393 PMCID: PMC4122506 DOI: 10.1038/nrc3298] [Citation(s) in RCA: 474] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
All malignant cancers, whether inherited or sporadic, are fundamentally governed by Darwinian dynamics. The process of carcinogenesis requires genetic instability and highly selective local microenvironments, the combination of which promotes somatic evolution. These microenvironmental forces, specifically hypoxia, acidosis and reactive oxygen species, are not only highly selective, but are also able to induce genetic instability. As a result, malignant cancers are dynamically evolving clades of cells living in distinct microhabitats that almost certainly ensure the emergence of therapy-resistant populations. Cytotoxic cancer therapies also impose intense evolutionary selection pressures on the surviving cells and thus increase the evolutionary rate. Importantly, the principles of Darwinian dynamics also embody fundamental principles that can illuminate strategies for the successful management of cancer.
Collapse
Affiliation(s)
- Robert J Gillies
- Department of Cancer Physiology and Biophysics, H. Lee Moffitt Cancer Center and Research Institute, Tampa FL 33602, USA.
| | | | | |
Collapse
|
144
|
Alama A, Orengo AM, Ferrini S, Gangemi R. Targeting cancer-initiating cell drug-resistance: a roadmap to a new-generation of cancer therapies? Drug Discov Today 2012; 17:435-42. [DOI: 10.1016/j.drudis.2011.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 02/04/2011] [Indexed: 01/20/2023]
|
145
|
Imaging biomarkers to monitor response to the hypoxia-activated prodrug TH-302 in the MiaPaCa2 flank xenograft model. Magn Reson Imaging 2012; 30:1002-9. [PMID: 22554971 DOI: 10.1016/j.mri.2012.02.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 10/15/2011] [Accepted: 02/29/2012] [Indexed: 12/21/2022]
Abstract
TH-302, a hypoxia-activated anticancer prodrug, was evaluated for antitumor activity and changes in dynamic contrast-enhanced (DCE) and diffusion-weighted (DW) magnetic resonance imaging (MRI) in a mouse model of pancreatic cancer. TH-302 monotherapy resulted in a significant delay in tumor growth compared to vehicle-treated controls. TH-302 treatment was also associated with a significant decrease in the volume transfer constant (K(trans)) compared to vehicle-treated controls 1 day following the first dose measured using DCE-MRI. This early decrease in K(trans) following the first dose as measured is consistent with selective killing of the hypoxic fraction of cells which are associated with enhanced expression of hypoxia inducible transcription factor-1 alpha that regulates expression of permeability and perfusion factors including vascular endothelial growth factor-A. No changes were observed in DW-MRI following treatment with TH-302, which may indicate that this technique is not sensitive enough to detect changes in small hypoxic fractions of the tumor targeted by TH-302. These results suggest that changes in tumor permeability and/or perfusion may be an early imaging biomarker for response to TH-302 therapy.
Collapse
|
146
|
Rajapakse A, Gates KS. Hypoxia-selective, enzymatic conversion of 6-nitroquinoline into a fluorescent helicene: pyrido[3,2-f]quinolino[6,5-c]cinnoline 3-oxide. J Org Chem 2012; 77:3531-7. [PMID: 22417220 DOI: 10.1021/jo3004748] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Regions of low oxygen concentration (hypoxia) occur in both normal human physiology and under pathophysiological conditions. Fluorescent probes for the direct imaging of cellular hypoxia could be useful tools that complement radiochemical imaging and immunohistochemical staining methods. In this work, we set out to characterize the hypoxia-selective enzymatic metabolism of a simple nitroaryl probe, 6-nitroquinoline (1). We envisioned that this compound might undergo hypoxia-selective, bioreductive conversion to the fluorescent product, 6-aminoquinoline (2). The probe 1 was, indeed, converted to a fluorescent product selectively under hypoxic conditions by the one-electron reducing enzyme NADPH:cytochrome P450 reductase. However, inspection of the fluorescence spectrum and LC-MS analysis of the reaction mixture revealed that the expected product 2 was not formed. Rather, the 63-fold increase in fluorescence emission at 445 nm resulting from the hypoxic metabolism of 1 was due to formation of the azoxy-helicene product, pyrido[3,2-f]quinolino[6,5-c]cinnoline 3-oxide (4). The generation of 4 involves an unusual biaryl bond formation under reductive conditions. The mechanism of this process remains uncertain but could proceed via combination of a nitroaryl radical anion with a neutral nitrosoaryl radical, followed by tautomerization and intramolecular condensation between the resulting hydroxylamine and nitroso functional groups. Bioreductive metabolism of nitroaryl compounds represents a promising strategy for the selective delivery of cytotoxic agents and fluorescent markers to hypoxic tissue, but the results described here provide an important glimpse of the chemical complexity that can be associated with the enzymatic one-electron reduction of nitroaryl compounds.
Collapse
Affiliation(s)
- Anuruddha Rajapakse
- Department of Chemistry, University of Missouri, 125 Chemistry Building, Columbia, Missouri 65211, USA
| | | |
Collapse
|
147
|
Liu Q, Sun JD, Wang J, Ahluwalia D, Baker AF, Cranmer LD, Ferraro D, Wang Y, Duan JX, Ammons WS, Curd JG, Matteucci MD, Hart CP. TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules. Cancer Chemother Pharmacol 2012; 69:1487-98. [PMID: 22382881 DOI: 10.1007/s00280-012-1852-8] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 02/13/2012] [Indexed: 01/12/2023]
Abstract
PURPOSE Subregional hypoxia is a common feature of tumors and is recognized as a limiting factor for the success of radiotherapy and chemotherapy. TH-302, a hypoxia-activated prodrug selectively targeting hypoxic regions of solid tumors, delivers a cytotoxic warhead to the tumor, while maintaining relatively low systemic toxicity. The antitumor activity, different dosing sequences, and dosing regimens of TH-302 in combination with commonly used conventional chemotherapeutics were investigated in human tumor xenograft models. METHODS Seven chemotherapeutic drugs (docetaxel, cisplatin, pemetrexed, irinotecan, doxorubicin, gemcitabine, and temozolomide) were tested in combination with TH-302 in eleven human xenograft models, including non-small cell lung cancer (NSCLC), colon cancer, prostate cancer, fibrosarcoma, melanoma, and pancreatic cancer. RESULTS The antitumor activity of docetaxel, cisplatin, pemetrexed, irinotecan, doxorubicin, gemcitabine, and temozolomide was increased when combined with TH-302 in nine out of eleven models tested. Administration of TH-302 2-8 h prior to the other chemotherapeutics yielded superior efficacy versus other sequences tested. Simultaneous administration of TH-302 and chemotherapeutics increased toxicity versus schedules with dosing separations. In a dosing optimization study, TH-302 administered daily at 50 mg/kg intraperitoneally for 5 days per week in the H460 NSCLC model showed the optimal response with minimal toxicity. CONCLUSIONS TH-302 enhances the activity of a wide range of conventional anti-neoplastic agents in a broad panel of in vivo xenograft models. These data highlight in vivo effects of schedule and order of drug administration in regimen efficacy and toxicity and have relevance to the design of human regimens incorporating TH-302.
Collapse
Affiliation(s)
- Qian Liu
- Threshold Pharmaceuticals, 170 Harbor Way, Suite 300, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Abstract
The metabolism and excretion of a hypoxically activating prodrug for the treatment of cancer, TH-302, were studied in beagle dogs following intravenous administration of 20 mg/kg (14)C-TH-302. TH-302 was extensively metabolized with total recovery of 75.1%, with 47.5% and 25.3% excreted through the urine and through the bile into the feces, respectively. The three TH-302 metabolites in plasma were: DM7, a conjugate of TH-302 with glutathione replacing a bromine atom; DM5, a hydrolysis product of DM7 with loss of the glutamic acid moiety; and DM6, a hydrolysis product of DM5 with loss of the glycine moiety. DM6 and TH-302 were the major radioactive components in plasma and accounted for 69.8% and 27.3% of the total AUC, respectively. The major metabolite in urine was DM6, which accounted for 22.7% of the administered dose. Two other metabolites identified in urine were: DM3, a dicysteine conjugate of TH-302; and DM4, which was formed by hydrolysis and loss of the 1-methyl-2-nitro-imidazol-5-yl methoxy moiety, followed by oxidation on the cysteinyl ethylamine moiety. DM1 and DM2 in urine accounted for 6.50 and 7.76% of administered dose and were not identified. DM1 was the only fecal metabolite. Further investigations are required to completely characterize the metabolism of TH-302.
Collapse
Affiliation(s)
- Donald Jung
- Threshold Pharmaceuticals, Inc., South San Francisco, CA 94080, USA.
| | | | | | | | | |
Collapse
|
149
|
Sun JD, Liu Q, Wang J, Ahluwalia D, Ferraro D, Wang Y, Duan JX, Ammons WS, Curd JG, Matteucci MD, Hart CP. Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibits tumor growth in preclinical models of cancer. Clin Cancer Res 2012; 18:758-70. [PMID: 22184053 DOI: 10.1158/1078-0432.ccr-11-1980] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Tumor hypoxia underlies treatment failure and yields a more aggressive, invasive, and metastatic cancer phenotype. TH-302 is a 2-nitroimidazole triggered hypoxia-activated prodrug of the cytotoxin bromo-isophosphoramide mustard (Br-IPM). The purpose of this study is to characterize the antitumor activity of TH-302 and investigate its selective targeting of the hypoxic cells in human tumor xenograft models. EXPERIMENTAL DESIGN Antitumor efficacy was assessed by tumor growth kinetics or by clonogenic survival of isolated cells after tumor excision. Hypoxic fractions (HF) were determined by immunohistochemistry and morphometrics of pimonidazole staining. Tumor hypoxia levels were manipulated by exposing animals to different oxygen concentration breathing conditions. The localization and kinetics of TH-302 induced DNA damage was determined by γH2AX immunohistochemistry. RESULTS TH-302 antitumor activity was dose-dependent and correlated with total drug exposure. Correlation was found between antitumor activity and tumor HF across 11 xenograft models. Tumor-bearing animals breathing 95% O(2) exhibited attenuated TH-302 efficacy, with whereas those breathing 10% O(2) exhibited enhanced TH-302 efficacy, both compared with air (21% O(2)) breathing. TH-302 treatment resulted in a reduction in the volume of the HF 48 hours after dosing and a corresponding increase in the necrotic fraction. TH-302 induced DNA damage as measured by γH2AX was initially only present in the hypoxic regions and then radiated to the entire tumor in a time-dependent manner, consistent with TH-302 having a "bystander effect." CONCLUSIONS The results show that TH-302 has broad antitumor activity and selectively targets hypoxic tumor tissues.
Collapse
Affiliation(s)
- Jessica D Sun
- Threshold Pharmaceuticals, South San Francisco, California 94080, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Bailey KM, Wojtkowiak JW, Hashim AI, Gillies RJ. Targeting the metabolic microenvironment of tumors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 65:63-107. [PMID: 22959024 DOI: 10.1016/b978-0-12-397927-8.00004-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The observation of aerobic glycolysis by tumor cells in 1924 by Otto Warburg, and subsequent innovation of imaging glucose uptake by tumors in patients with PET-CT, has incited a renewed interest in the altered metabolism of tumors. As tumors grow in situ, a fraction of it is further away from their blood supply, leading to decreased oxygen concentrations (hypoxia), which induces the hypoxia response pathways of HIF1α, mTOR, and UPR. In normal tissues, these responses mitigate hypoxic stress and induce neoangiogenesis. In tumors, these pathways are dysregulated and lead to decreased perfusion and exacerbation of hypoxia as a result of immature and chaotic blood vessels. Hypoxia selects for a glycolytic phenotype and resultant acidification of the tumor microenvironment, facilitated by upregulation of proton transporters. Acidification selects for enhanced metastatic potential and reduced drug efficacy through ion trapping. In this review, we provide a comprehensive summary of preclinical and clinical drugs under development for targeting aerobic glycolysis, acidosis, hypoxia and hypoxia response pathways. Hypoxia and acidosis can be manipulated, providing further therapeutic benefit for cancers that feature these common phenotypes.
Collapse
Affiliation(s)
- Kate M Bailey
- Department of Imaging and Metabolism, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | |
Collapse
|