51
|
Collins SL, Saha J, Bouchez LC, Hammond EM, Conway SJ. Hypoxia-Activated, Small-Molecule-Induced Gene Expression. ACS Chem Biol 2018; 13:3354-3360. [PMID: 30451487 DOI: 10.1021/acschembio.8b00858] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hypoxia, a condition of reduced oxygen, occurs in a wide variety of biological contexts, including solid tumors and bacterial biofilms, which are relevant to human health. Consequently, the development of chemical tools to study hypoxia is vital. Here we report a hypoxia-activated, small-molecule-mediated gene expression system using a bioreductive prodrug of the inducer isopropyl 1-thio-β-d-galactopyranoside. As a proof-of-concept we have placed the production of a green fluorescent protein under the control of hypoxia. Our system has the potential to be extended to regulate the production of any given protein of choice.
Collapse
Affiliation(s)
- Sarah L Collins
- Department of Chemistry, Chemistry Research Laboratory , University of Oxford , Mansfield Road , Oxford OX1 3TA , U.K
- Cancer Research U.K./MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| | - Jaideep Saha
- Department of Chemistry, Chemistry Research Laboratory , University of Oxford , Mansfield Road , Oxford OX1 3TA , U.K
| | - Laure C Bouchez
- Chemical Biology and Therapeutics , Novartis Institutes for BioMedical Research , Fabrikstrasse 22 , 4054 Basel , Switzerland
| | - Ester M Hammond
- Cancer Research U.K./MRC Oxford Institute for Radiation Oncology, Department of Oncology , University of Oxford , Old Road Campus Research Building , Oxford OX3 7DQ , U.K
| | - Stuart J Conway
- Department of Chemistry, Chemistry Research Laboratory , University of Oxford , Mansfield Road , Oxford OX1 3TA , U.K
| |
Collapse
|
52
|
Abstract
The nitro group is considered to be a versatile and unique functional group in medicinal chemistry. Despite a long history of use in therapeutics, the nitro group has toxicity issues and is often categorized as a structural alert or a toxicophore, and evidence related to drugs containing nitro groups is rather contradictory. In general, drugs containing nitro groups have been extensively associated with mutagenicity and genotoxicity. In this context, efforts toward the structure-mutagenicity or structure-genotoxicity relationships have been undertaken. The current Perspective covers various aspects of agents that contain nitro groups, their bioreductive activation mechanisms, their toxicities, and approaches to combat their toxicity issues. In addition, recent advances in the field of anticancer, antitubercular and antiparasitic agents containing nitro groups, along with a patent survey on hypoxia-activated prodrugs containing nitro groups, are also covered.
Collapse
Affiliation(s)
- Kunal Nepali
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| |
Collapse
|
53
|
Jayaprakash P, Ai M, Liu A, Budhani P, Bartkowiak T, Sheng J, Ager C, Nicholas C, Jaiswal AR, Sun Y, Shah K, Balasubramanyam S, Li N, Wang G, Ning J, Zal A, Zal T, Curran MA. Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy. J Clin Invest 2018; 128:5137-5149. [PMID: 30188869 DOI: 10.1172/jci96268] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 08/30/2018] [Indexed: 12/23/2022] Open
Abstract
Despite the success of immune checkpoint blockade against melanoma, many "cold" tumors like prostate cancer remain unresponsive. We found that hypoxic zones were prevalent across preclinical prostate cancer and resisted T cell infiltration even in the context of CTLA-4 and PD-1 blockade. We demonstrated that the hypoxia-activated prodrug TH-302 reduces or eliminates hypoxia in these tumors. Combination therapy with this hypoxia-prodrug and checkpoint blockade cooperated to cure more than 80% of tumors in the transgenic adenocarcinoma of the mouse prostate-derived (TRAMP-derived) TRAMP-C2 model. Immunofluorescence imaging showed that TH-302 drives an influx of T cells into hypoxic zones, which were expanded by checkpoint blockade. Further, combination therapy reduced myeloid-derived suppressor cell density by more than 50%, and durably reduced the capacity of the tumor to replenish the granulocytic subset. Spontaneous prostate tumors in TRAMP transgenic mice, which completely resist checkpoint blockade, showed minimal adenocarcinoma tumor burden at 36 weeks of age and no evidence of neuroendocrine tumors with combination therapy. Survival of Pb-Cre4, Ptenpc-/-Smad4pc-/- mice with aggressive prostate adenocarcinoma was also significantly extended by this combination of hypoxia-prodrug and checkpoint blockade. Hypoxia disruption and T cell checkpoint blockade may sensitize some of the most therapeutically resistant cancers to immunotherapy.
Collapse
Affiliation(s)
- Priyamvada Jayaprakash
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Midan Ai
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Arthur Liu
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,University of Texas Health Science Center at Houston Graduate School of Biomedical Science, Houston, Texas, USA
| | - Pratha Budhani
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Todd Bartkowiak
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,University of Texas Health Science Center at Houston Graduate School of Biomedical Science, Houston, Texas, USA
| | - Jie Sheng
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Casey Ager
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,University of Texas Health Science Center at Houston Graduate School of Biomedical Science, Houston, Texas, USA
| | - Courtney Nicholas
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ashvin R Jaiswal
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yanqiu Sun
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Krishna Shah
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sadhana Balasubramanyam
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nan Li
- Department of Biostatistics and
| | - Guocan Wang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Anna Zal
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tomasz Zal
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,University of Texas Health Science Center at Houston Graduate School of Biomedical Science, Houston, Texas, USA
| | - Michael A Curran
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,University of Texas Health Science Center at Houston Graduate School of Biomedical Science, Houston, Texas, USA
| |
Collapse
|
54
|
Laubach JP, Liu CJ, Raje NS, Yee AJ, Armand P, Schlossman RL, Rosenblatt J, Hedlund J, Martin M, Reynolds C, Shain KH, Zackon I, Stampleman L, Henrick P, Rivotto B, Hornburg KTV, Dumke HJ, Chuma S, Savell A, Handisides DR, Kroll S, Anderson KC, Richardson PG, Ghobrial IM. A Phase I/II Study of Evofosfamide, A Hypoxia-activated Prodrug with or without Bortezomib in Subjects with Relapsed/Refractory Multiple Myeloma. Clin Cancer Res 2018; 25:478-486. [PMID: 30279233 DOI: 10.1158/1078-0432.ccr-18-1325] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 08/12/2018] [Accepted: 09/26/2018] [Indexed: 01/07/2023]
Abstract
PURPOSE The presence of hypoxia in the diseased bone marrow presents a new therapeutic target for multiple myeloma. Evofosfamide (formerly TH-302) is a 2-nitroimidazole prodrug of the DNA alkylator, bromo-isophosphoramide mustard, which is selectively activated under hypoxia. This trial was designed as a phase I/II study investigating evofosfamide in combination with dexamethasone, and in combination with bortezomib and dexamethasone in relapsed/refractory multiple myeloma. PATIENTS AND METHODS Fifty-nine patients initiated therapy, 31 received the combination of evofosfamide and dexamethasone, and 28 received the combination of evofosfamide, bortezomib, and dexamethasone. Patients were heavily pretreated with a median number of prior therapies of 7 (range: 2-15). All had previously received bortezomib and immunomodulators. The MTD, treatment toxicity, and efficacy were determined. RESULTS The MTD was established at 340 mg/m2 evofosfamide + dexamethasone with dose-limiting mucositis at higher doses. For the combination of evofosfamide, bortezomib, and dexamethasone, no patient had a dose-limiting toxicity (DLT) and the recommended phase II dose was established at 340 mg/m2. The most common ≥grade 3 adverse events (AE) were thrombocytopenia (25 patients), anemia (24 patients), neutropenia (15 patients), and leukopenia (9 patients). Skin toxicity was reported in 42 (71%) patients. Responses included 1 very good partial response (VGPR), 3 partial response (PR), 2 minor response (MR), 20 stable disease (SD), and 4 progressive disease (PD) for evofosfamide + dexamethasone and 1 complete response (CR), 2 PR, 1 MR, 18 SD, and 5 PD for evofosfamide + bortezomib + dexamethasone. Disease stabilization was observed in over 80% and this was reflective of the prolonged overall survival of 11.2 months. CONCLUSIONS Evofosfamide can be administered at 340 mg/m2 twice a week with or without bortezomib. Clinical activity has been noted in patients with heavily pretreated relapsed refractory multiple myeloma.
Collapse
Affiliation(s)
- Jacob P Laubach
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Chia-Jen Liu
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Noopur S Raje
- Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew J Yee
- Massachusetts General Hospital, Boston, Massachusetts
| | - Philippe Armand
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Robert L Schlossman
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Jacalyn Rosenblatt
- Blood Cancer Research Partnership (BCRP), Boston, Massachusetts.,Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Jacquelyn Hedlund
- Blood Cancer Research Partnership (BCRP), Boston, Massachusetts.,Maine Center For Cancer Medicine, Scarborough, Maine
| | - Michael Martin
- Blood Cancer Research Partnership (BCRP), Boston, Massachusetts.,The West Clinic, Memphis, Tennessee
| | - Craig Reynolds
- Blood Cancer Research Partnership (BCRP), Boston, Massachusetts.,Ocala Oncology Center, Ocala, Florida
| | | | - Ira Zackon
- Blood Cancer Research Partnership (BCRP), Boston, Massachusetts.,New York Oncology Hematology, Albany, New York
| | - Laura Stampleman
- Blood Cancer Research Partnership (BCRP), Boston, Massachusetts.,Pacific Cancer Care, Salinas, California
| | - Patrick Henrick
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Bradley Rivotto
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kalvis T V Hornburg
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Henry J Dumke
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Stacey Chuma
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Alexandra Savell
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | | | - Stew Kroll
- Threshold Pharmaceuticals, South San Francisco, California
| | - Kenneth C Anderson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Paul G Richardson
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. .,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| | - Irene M Ghobrial
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. .,Blood Cancer Research Partnership (BCRP), Boston, Massachusetts
| |
Collapse
|
55
|
Zhang Z, Ji S, Zhang B, Liu J, Qin Y, Xu J, Yu X. Role of angiogenesis in pancreatic cancer biology and therapy. Biomed Pharmacother 2018; 108:1135-1140. [PMID: 30372814 DOI: 10.1016/j.biopha.2018.09.136] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, and there is a close parallel between disease mortality and incidence. Malignancy is often diagnosed at an advanced stage due to the lack of early symptoms. For the majority of advanced or metastatic pancreatic cancer patients, therapeutic options are limited. Although several new chemotherapeutic regimens have been developed, the overall response rate remains low. Invasive tumour growth and distant metastasis require angiogenesis, a hallmark of cancer, and angiogenic inhibition is a valuable option for cancer therapy. Some anti-angiogenic drugs have been developed for cancer treatment. This review will focus on the role of angiogenesis and anti-angiogenic treatment strategies as well as combination therapy in pancreatic cancer. Translational information from recent molecular biology and animal studies is also summarized. Finally, the dosing schedule for bevacizumab with other chemotherapeutic protocols for pancreatic cancer treatment is discussed.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
56
|
Structure-activity relationship study of hypoxia-activated prodrugs for proteoglycan-targeted chemotherapy in chondrosarcoma. Eur J Med Chem 2018; 158:51-67. [DOI: 10.1016/j.ejmech.2018.08.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
|
57
|
Zhang Q, Jin C, Yu J, Lu W. Synthesis of New Branched 2-Nitroimidazole as a Hypoxia Sensitive Linker for Ligand-Targeted Drugs of Paclitaxel. ACS OMEGA 2018; 3:8813-8818. [PMID: 31459014 PMCID: PMC6644517 DOI: 10.1021/acsomega.8b01208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 07/24/2018] [Indexed: 06/10/2023]
Abstract
Because of the low selectivity and efficiency of normal antitumor agents, the strategy of ligand-targeted drugs was put forward. In this paper, we designed and synthesized a new bioreductive linker based on 2-nitroimidazole, which was used in three paclitaxel (PTX) prodrugs. The drug release mechanism via six-membered ring was demonstrated by chemical reduction and nitroreductase assay. Glucose and acetazolamide, which have been reported widely as ligands, were attached to compound 7 to afford Glu-PTX and AZO-PTX. The prodrugs were considerably stable in phosphate-buffered saline (pH 7.4) and plasma. What is more, PTX releasing could be triggered by nitroreductase rapidly. In in vitro cytotoxicity assay, the prodrugs exhibited moderate selectivity toward hypoxic tumor cells. We considered that the 2-nitroimidazole linker could accelerate the release of prodrugs under hypoxic condition. It was promising in the development of ligand-targeted drugs.
Collapse
Affiliation(s)
| | | | | | - Wei Lu
- E-mail: . Fax: +86 21 62238771 (W.L.)
| |
Collapse
|
58
|
Jamieson SM, Tsai P, Kondratyev MK, Budhani P, Liu A, Senzer NN, Chiorean EG, Jalal SI, Nemunaitis JJ, Kee D, Shome A, Wong WW, Li D, Poonawala-Lohani N, Kakadia PM, Knowlton NS, Lynch CR, Hong CR, Lee TW, Grénman RA, Caporiccio L, McKee TD, Zaidi M, Butt S, Macann AM, McIvor NP, Chaplin JM, Hicks KO, Bohlander SK, Wouters BG, Hart CP, Print CG, Wilson WR, Curran MA, Hunter FW. Evofosfamide for the treatment of human papillomavirus-negative head and neck squamous cell carcinoma. JCI Insight 2018; 3:122204. [PMID: 30135316 PMCID: PMC6141174 DOI: 10.1172/jci.insight.122204] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/13/2018] [Indexed: 01/10/2023] Open
Abstract
Evofosfamide (TH-302) is a clinical-stage hypoxia-activated prodrug of a DNA-crosslinking nitrogen mustard that has potential utility for human papillomavirus (HPV) negative head and neck squamous cell carcinoma (HNSCC), in which tumor hypoxia limits treatment outcome. We report the preclinical efficacy, target engagement, preliminary predictive biomarkers and initial clinical activity of evofosfamide for HPV-negative HNSCC. Evofosfamide was assessed in 22 genomically characterized cell lines and 7 cell line-derived xenograft (CDX), patient-derived xenograft (PDX), orthotopic, and syngeneic tumor models. Biomarker analysis used RNA sequencing, whole-exome sequencing, and whole-genome CRISPR knockout screens. Five advanced/metastatic HNSCC patients received evofosfamide monotherapy (480 mg/m2 qw × 3 each month) in a phase 2 study. Evofosfamide was potent and highly selective for hypoxic HNSCC cells. Proliferative rate was a predominant evofosfamide sensitivity determinant and a proliferation metagene correlated with activity in CDX models. Evofosfamide showed efficacy as monotherapy and with radiotherapy in PDX models, augmented CTLA-4 blockade in syngeneic tumors, and reduced hypoxia in nodes disseminated from an orthotopic model. Of 5 advanced HNSCC patients treated with evofosfamide, 2 showed partial responses while 3 had stable disease. In conclusion, evofosfamide shows promising efficacy in aggressive HPV-negative HNSCC, with predictive biomarkers in development to support further clinical evaluation in this indication.
Collapse
Affiliation(s)
- Stephen M.F. Jamieson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Peter Tsai
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Maria K. Kondratyev
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Pratha Budhani
- Department of Immunology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Arthur Liu
- Department of Immunology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - E. Gabriela Chiorean
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana, USA
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington, USA
| | - Shadia I. Jalal
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, Indiana, USA
| | - John J. Nemunaitis
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Dennis Kee
- LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Avik Shome
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Way W. Wong
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Dan Li
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | | | - Purvi M. Kakadia
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Nicholas S. Knowlton
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Courtney R.H. Lynch
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Cho R. Hong
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Tet Woo Lee
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Reidar A. Grénman
- Department of Otolaryngology–Head and Neck Surgery, Turku University Hospital, Turku, Finland
| | - Laura Caporiccio
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Trevor D. McKee
- STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada
| | - Mark Zaidi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada
| | - Sehrish Butt
- STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrew M.J. Macann
- Department of Radiation Oncology, Auckland City Hospital, Auckland, New Zealand
| | - Nicholas P. McIvor
- Department of Otolaryngology–Head and Neck Surgery, Auckland City Hospital, Auckland, New Zealand
| | - John M. Chaplin
- Department of Otolaryngology–Head and Neck Surgery, Auckland City Hospital, Auckland, New Zealand
| | - Kevin O. Hicks
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Stefan K. Bohlander
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, 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
| | - Charles P. Hart
- Threshold Pharmaceuticals, South San Francisco, California, USA
| | - Cristin G. Print
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - William R. Wilson
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Michael A. Curran
- Department of Immunology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Francis W. Hunter
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| |
Collapse
|
59
|
Hong CR, Dickson BD, Jaiswal JK, Pruijn FB, Hunter FW, Hay MP, Hicks KO, Wilson WR. Cellular pharmacology of evofosfamide (TH-302): A critical re-evaluation of its bystander effects. Biochem Pharmacol 2018; 156:265-280. [PMID: 30134191 DOI: 10.1016/j.bcp.2018.08.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Evofosfamide (TH-302) is a clinical-stage hypoxia-activated prodrug with proven efficacy against hypoxic cells in preclinical tumour models. TH-302 is designed to release the DNA crosslinking agent bromo-isophosphoramide mustard (Br-IPM) when reduced in hypoxic tissue. Br-IPM is considered to diffuse locally from hypoxic regions, eliciting additional tumour cell killing, but the latter 'bystander effect' has not been demonstrated directly. Previous studies with multicellular co-cultures that included cells expressing the E. coli nitroreductase NfsA as a model TH-302 reductase have provided clear evidence of a bystander effect (which we confirm in the present study). However, NfsA is an oxygen-insensitive two-electron reductase that is not expected to generate the nitro radical intermediate that has been demonstrated to fragment to release Br-IPM. Here, we use mass spectrometry methods to characterise TH-302 metabolites generated by one-electron reduction (steady-state radiolysis by ionising radiation and cellular metabolism under hypoxia, including HCT116 cells that overexpress P450 oxidoreductase, POR) or by NfsA expressed in HCT116 cells under oxic conditions, and investigate the stability and cytotoxicity of these products. Br-IPM is shown to have very low cytotoxic potency when added to extracellular culture medium and to be rapidly converted to other hydrophilic products including dichloro-isophosphoramide mustard (IPM). Only traces of Br-IPM or IPM were detected in the extracellular medium when generated by cellular metabolism of TH-302. We identify, in NfsA-expressing cells, the hydroxylamine metabolite of TH-302, and downstream products resulting from rearrangement or hydration of the imidazole ring, and demonstrate that formation of these candidate bystander effect mediators is suppressed by hypoxia. This characterisation of the cellular pharmacology of TH-302 implies that bystander effects from hypoxic activation of TH-302 are unlikely to contribute to its anticancer activity.
Collapse
Affiliation(s)
- Cho Rong Hong
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Benjamin D Dickson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Jagdish K Jaiswal
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Frederik B Pruijn
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Michael P Hay
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - William R Wilson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
60
|
Huang Y, Tian Y, Zhao Y, Xue C, Zhan J, Liu L, He X, Zhang L. Efficacy of the hypoxia-activated prodrug evofosfamide (TH-302) in nasopharyngeal carcinoma in vitro and in vivo. Cancer Commun (Lond) 2018; 38:15. [PMID: 29764490 PMCID: PMC5993153 DOI: 10.1186/s40880-018-0285-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Tumor hypoxia is considered an important factor in metastasis and disease relapse. Evofosfamide is a hypoxia-activated prodrug that selectively targets the hypoxic regions of solid tumors. As hypoxia-inducible factor-1α (HIF-1α) is overexpressed in nasopharyngeal carcinoma (NPC) tissues, we performed the present study to evaluate the efficacy profile of evofosfamide in NPC. METHODS We evaluated the efficacy of evofosfamide as a single agent or combined with cisplatin (DDP) in the NPC cell lines CNE-2, HONE-1 and HNE-1, and in nude mouse xenograft tumor models. RESULTS Evofosfamide exhibited hypoxia-selective cytotoxicity in NPC cell lines, with 50% inhibition concentration (IC50) values of 8.33 ± 0.75, 7.62 ± 0.67, and 0.31 ± 0.07 μmol/L under hypoxia in CNE-2, HONE-1 and HNE-1 cells, respectively. The sensitization ranged from ninefold to greater than 300-fold under hypoxia compared with normoxia controls. The combination of evofosfamide with DDP had a synergistic effect on cytotoxicity in the NPC cell lines by combination index values assessment. Cell cycle G2 phase was arrested after treated with 0.05 μmol/L evofosfamide under hypoxia. Histone H2AX phosphorylation (γH2AX) (a marker of DNA damage) expression increased while HIF-1α expression suppressed after evofosfamide treatment under hypoxic conditions. In the HNE-1 NPC xenograft models, evofosfamide exhibited antitumor activity both as a single agent and combined with DDP. Hypoxic regions in xenograft tissue were reduced after both evofosfamide monotherapy and combined therapy with DDP. CONCLUSIONS Our results present preclinical evidence for targeting the selective hypoxic portion of NPC by evofosfamide as a single agent and combined with DDP and provide rationale for the potential clinical application of evofosfamide for the treatment of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yan Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
| | - Ying Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
| | - Yuanyuan Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
| | - Cong Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
| | - Jianhua Zhan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
| | - Lin Liu
- Department of Medical Oncology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000 Guangdong P. R. China
| | - Xiaobo He
- Department of Radiation Oncology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000 Guangdong P. R. China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 Guangdong P. R. China
| |
Collapse
|
61
|
Therapeutic journery of nitrogen mustard as alkylating anticancer agents: Historic to future perspectives. Eur J Med Chem 2018; 151:401-433. [DOI: 10.1016/j.ejmech.2018.04.001] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/30/2018] [Accepted: 04/01/2018] [Indexed: 12/17/2022]
|
62
|
Nayab Rasool SK, Subramanyam C, Janakiramudu DB, Supraja P, Usha R, Raju CN. Convenient one-pot synthesis and biological evaluation of phosphoramidates and phosphonates containing heterocycles. PHOSPHORUS SULFUR 2018. [DOI: 10.1080/10426507.2018.1452229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- SK. Nayab Rasool
- Department of Chemistry, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - Ch. Subramanyam
- Department of Chemistry, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - D. B. Janakiramudu
- Department of Chemistry, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| | - P. Supraja
- Department of Biotechnology, Sri Padmavathi Mahila Visvavidyalayam, Tirupati, Andhra Pradesh, India
| | - R. Usha
- Department of Biotechnology, Sri Padmavathi Mahila Visvavidyalayam, Tirupati, Andhra Pradesh, India
| | - C. Naga Raju
- Department of Chemistry, Sri Venkateswara University, Tirupati, Andhra Pradesh, India
| |
Collapse
|
63
|
Haynes J, McKee TD, Haller A, Wang Y, Leung C, Gendoo DMA, Lima-Fernandes E, Kreso A, Wolman R, Szentgyorgyi E, Vines DC, Haibe-Kains B, Wouters BG, Metser U, Jaffray DA, Smith M, O'Brien CA. Administration of Hypoxia-Activated Prodrug Evofosfamide after Conventional Adjuvant Therapy Enhances Therapeutic Outcome and Targets Cancer-Initiating Cells in Preclinical Models of Colorectal Cancer. Clin Cancer Res 2018; 24:2116-2127. [PMID: 29476017 DOI: 10.1158/1078-0432.ccr-17-1715] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/21/2017] [Accepted: 02/19/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Cancer-initiating cells (C-IC) have been described in multiple cancer types, including colorectal cancer. C-ICs are defined by their capacity to self-renew, thereby driving tumor growth. C-ICs were initially thought to be static entities; however, recent studies have determined these cells to be dynamic and influenced by microenvironmental cues such as hypoxia. If hypoxia drives the formation of C-ICs, then therapeutic targeting of hypoxia could represent a novel means to target C-ICs.Experimental Design: Patient-derived colorectal cancer xenografts were treated with evofosfamide, a hypoxia-activated prodrug (HAP), in combination with 5-fluorouracil (5-FU) or chemoradiotherapy (5-FU and radiation; CRT). Treatment groups included both concurrent and sequential dosing regimens. Effects on the colorectal cancer-initiating cell (CC-IC) fraction were assessed by serial passage in vivo limiting dilution assays. FAZA-PET imaging was utilized as a noninvasive method to assess intratumoral hypoxia.Results: Hypoxia was sufficient to drive the formation of CC-ICs and colorectal cancer cells surviving conventional therapy were more hypoxic and C-IC-like. Using a novel approach to combination therapy, we show that sequential treatment with 5-FU or CRT followed by evofosfamide not only inhibits tumor growth of xenografts compared with 5-FU or CRT alone, but also significantly decreases the CC-IC fraction. Furthermore, noninvasive FAZA-PET hypoxia imaging was predictive of a tumor's response to evofosfamide.Conclusions: Our data demonstrate a novel means to target the CC-IC fraction by adding a HAP sequentially after conventional adjuvant therapy, as well as the use of FAZA-PET as a biomarker for hypoxia to identify tumors that will benefit most from this approach. Clin Cancer Res; 24(9); 2116-27. ©2018 AACR.
Collapse
Affiliation(s)
- Jennifer Haynes
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Trevor D McKee
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrew Haller
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yadong Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Cherry Leung
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Deena M A Gendoo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Antonija Kreso
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Robin Wolman
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Eva Szentgyorgyi
- Department of Pathology, University Health Network, Toronto, Ontario, Canada
| | - Douglass C Vines
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Bradly G Wouters
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Ur Metser
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Imaging, University of Toronto, Toronto, Ontario, Canada.,Techna Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - David A Jaffray
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.,STTARR Innovation Centre, University Health Network, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.,Techna Institute for the Advancement of Technology for Health, University Health Network, Toronto, Ontario, Canada
| | - Myles Smith
- Department of Surgery, The Royal Marsden Hospital and Institute of Cancer Research, London, United Kingdom
| | - Catherine A O'Brien
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
64
|
Vena F, Jia R, Esfandiari A, Garcia-Gomez JJ, Rodriguez-Justo M, Ma J, Syed S, Crowley L, Elenbaas B, Goodstal S, Hartley JA, Hochhauser D. MEK inhibition leads to BRCA2 downregulation and sensitization to DNA damaging agents in pancreas and ovarian cancer models. Oncotarget 2018; 9:11592-11603. [PMID: 29545922 PMCID: PMC5837749 DOI: 10.18632/oncotarget.24294] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/15/2018] [Indexed: 12/24/2022] Open
Abstract
Targeting the DNA damage response (DDR) in tumors with defective DNA repair is a clinically successful strategy. The RAS/RAF/MEK/ERK signalling pathway is frequently deregulated in human cancers. In this study, we explored the effects of MEK inhibition on the homologous recombination pathway and explored the potential for combination therapy of MEK inhibitors with DDR inhibitors and a hypoxia-activated prodrug. We studied effects of combining pimasertib, a selective allosteric inhibitor of MEK1/2, with olaparib, a small molecule inhibitor of poly (adenosine diphosphate [ADP]-ribose) polymerases (PARP), and with the hypoxia-activated prodrug evofosfamide in ovarian and pancreatic cancer cell lines. Apoptosis was assessed by Caspase 3/7 assay and protein expression was detected by immunoblotting. DNA damage response was monitored with γH2AX and RAD51 immunofluorescence staining. In vivo antitumor activity of pimasertib with evofosfamide were assessed in pancreatic cancer xenografts. We found that BRCA2 protein expression was downregulated following pimasertib treatment under hypoxic conditions. This translated into reduced homologous recombination repair demonstrated by levels of RAD51 foci. MEK inhibition was sufficient to induce formation of γH2AX foci, suggesting that inhibition of this pathway would impair DNA repair. When combined with olaparib or evofosfamide, pimasertib treatment enhanced DNA damage and increased apoptosis. The combination of pimasertib with evofosfamide demonstrated increased anti-tumor activity in BRCA wild-type Mia-PaCa-2 xenograft model, but not in the BRCA mutated BxPC3 model. Our data suggest that targeted MEK inhibition leads to impaired homologous recombination DNA damage repair and increased PARP inhibition sensitivity in BRCA-2 proficient cancers.
Collapse
Affiliation(s)
- Francesca Vena
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O’Gorman Building, University College London, London WC1E 6DD, UK
| | - Ruochen Jia
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O’Gorman Building, University College London, London WC1E 6DD, UK
| | - Arman Esfandiari
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O’Gorman Building, University College London, London WC1E 6DD, UK
| | - Juan J. Garcia-Gomez
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O’Gorman Building, University College London, London WC1E 6DD, UK
| | | | - Jianguo Ma
- EMD Serono Research and Development Institute, Billerica 01821, MA, USA
| | - Sakeena Syed
- EMD Serono Research and Development Institute, Billerica 01821, MA, USA
| | - Lindsey Crowley
- EMD Serono Research and Development Institute, Billerica 01821, MA, USA
| | - Brian Elenbaas
- EMD Serono Research and Development Institute, Billerica 01821, MA, USA
| | - Samantha Goodstal
- EMD Serono Research and Development Institute, Billerica 01821, MA, USA
| | - John A. Hartley
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O’Gorman Building, University College London, London WC1E 6DD, UK
| | - Daniel Hochhauser
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O’Gorman Building, University College London, London WC1E 6DD, UK
| |
Collapse
|
65
|
Cox TR, Erler JT, Rumney RMH. Established Models and New Paradigms for Hypoxia-Driven Cancer-Associated Bone Disease. Calcif Tissue Int 2018; 102:163-173. [PMID: 29098360 PMCID: PMC5805797 DOI: 10.1007/s00223-017-0352-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/19/2017] [Indexed: 12/16/2022]
Abstract
The five-year survival rate for primary bone cancers is ~ 70% while almost all cases of secondary metastatic bone cancer are terminal. Hypoxia, the deficiency of oxygen which occurs as the rate of tumour growth exceeds the supply of vascularisation, is a key promoter of tumour progression. Hypoxia-driven effects in the primary tumour are wide ranging including changes in gene expression, dysregulation of signalling pathways, resistance to chemotherapy, neovascularisation, increased tumour cell proliferation and migration. Paget's seed and soil theory states that for a metastasising tumour cell 'the seed' it requires the correct microenvironment 'soil' to colonise. Why and how metastasising tumour cells colonise the bone is a complex and intriguing problem. However, once present tumour cells are able to disrupt bone homeostasis through increasing osteoclast activity and downregulating osteoblast function. Osteoclast resorption releases growth factors from the bone matrix that subsequently contribute to the proliferation of invasive tumour cells creating the vicious cycle of bone loss and metastatic cancer progression. Recently, we have shown that hypoxia increases expression and release of lysyl oxidase (LOX) from primary mammary tumours, which in turn disrupts bone homeostasis to favour osteolytic degradation to create pre-metastatic niches in the bone microenvironment. We also demonstrated how treatment with bisphosphonates could block this cancer-induced bone remodelling and reduce secondary bone metastases. This review describes the roles of hypoxia in primary tumour progression to metastasis, with a focus on key signalling pathways and treatment options to reduce patient morbidity and increase survival.
Collapse
Affiliation(s)
- Thomas R Cox
- The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, 2010, Australia.
| | - Janine T Erler
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen (UCPH), Ole Maaløes Vej 5, 2200, Copenhagen, Denmark
| | - Robin M H Rumney
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
66
|
Takakusagi Y, Kishimoto S, Naz S, Matsumoto S, Saito K, Hart CP, Mitchell JB, Krishna MC. Radiotherapy Synergizes with the Hypoxia-Activated Prodrug Evofosfamide: In Vitro and In Vivo Studies. Antioxid Redox Signal 2018; 28:131-140. [PMID: 28741367 PMCID: PMC5725636 DOI: 10.1089/ars.2017.7106] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AIMS Evofosfamide (TH-302) is a hypoxia-activated prodrug (HAP) that releases the DNA-damaging bromo-isophosphoramide mustard (Br-IPM) moiety selectively under hypoxic conditions. Since solid tumors are known to have hypoxic regions, HAPs in combination with chemotherapy or radiotherapy (XRT) will be beneficial. We tested the oxygen dependence of release kinetics of Br-IPM using electron paramagnetic resonance (EPR) with spin trapping by monitoring redox cycling of the nitroimidazole moiety of TH-302, and oxygen dependence of TH-302 on in vitro cytotoxicity at different levels of hypoxia was also examined. Two tumor implants (SCCVII and HT29) in mice were studied. RESULTS TH-302 fragmentation to release Br-IPM was noticed at oxygen levels <76 mmHg, which increased with higher levels of hypoxia. Enhanced cellular cytotoxicity was also observed at oxygen levels <76 mmHg. In vivo pO2 imaging in the two tumor implants showed that the SCCVII tumor implant had higher level of hypoxia compared with the HT29 xenograft. TH-302 as a monotherapy in vivo showed modest effects in SCCVII implants and minimal effects in HT29 xenografts, whereas TH-302 in combination with ionizing radiation showed significant benefit in both tumor models. INNOVATION We examined the kinetics of redox cycling versus fragmentation of TH-302. The combination of oxygen-dependent XRT with TH-302 is effective even in tumors with significant hypoxia. CONCLUSIONS Imaging studies identifying the magnitude of hypoxia in tumors indicated that the responsiveness to TH-302 and the antitumor effect of TH-302 were enhanced by combining with XRT in both the TH-302-sensitive SCCVII tumor and -resistant HT29 tumor. Antioxid. Redox Signal. 28, 131-140.
Collapse
Affiliation(s)
- Yoichi Takakusagi
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland.,2 National Institutes for Quantum and Radiological Science and Technology , Chiba, Japan
| | - Shun Kishimoto
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland
| | - Sarwat Naz
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland
| | - Shingo Matsumoto
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland.,3 Graduate School of Information Science and Technology, Hokkaido University , Sapporo, Japan
| | - Keita Saito
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland
| | - Charles P Hart
- 4 Threshold Pharmaceuticals, Inc. , South San Francisco, California
| | - James B Mitchell
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland
| | - Murali C Krishna
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute , Bethesda, Maryland
| |
Collapse
|
67
|
Yoon C, Chang KK, Lee JH, Tap WD, Hart CP, Simon MC, Yoon SS. Multimodal targeting of tumor vasculature and cancer stem-like cells in sarcomas with VEGF-A inhibition, HIF-1α inhibition, and hypoxia-activated chemotherapy. Oncotarget 2018; 7:42844-42858. [PMID: 27374091 PMCID: PMC5189991 DOI: 10.18632/oncotarget.10212] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/07/2016] [Indexed: 01/08/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) inhibition with pazopanib is an approved therapy for sarcomas, but likely results in compensatory pathways such as upregulation of hypoxia inducible factor 1α (HIF-1α). In addition, cancer stem-like cells can preferentially reside in hypoxic regions of tumors and be resistant to standard chemotherapies. In this study, we hypothesized that the combination of VEGF-A inhibition, HIF-1α inhibition, and hypoxia-activated chemotherapy with evofosfamide would be an effective multimodal strategy. Multimodal therapy was examined in one genetically engineered and two xenograft mouse models of sarcoma. In all three models, multimodal therapy showed greater efficacy than any single agent therapy or bimodality therapy in blocking tumor growth. Even after cessation of therapy, tumors treated with multimodal therapy remained relatively dormant for up to 2 months. Compared to the next best bimodality therapy, multimodal therapy caused 2.8-3.3 fold more DNA damage, 1.5-2.7 fold more overall apoptosis, and 2.3-3.6 fold more endothelial cell-specific apoptosis. Multimodal therapy also decreased microvessel density and HIF-1α activity by 85-90% and 79-89%, respectively, compared to controls. Sarcomas treated with multimodal therapy had 95-96% depletion of CD133(+) cancer stem-like ells compared to control tumors. Sarcoma cells grown as spheroids to enrich for CD133(+) cancer stem-like cells were more sensitive than monolayer cells to multimodal therapy in terms of DNA damage and apoptosis, especially under hypoxic conditions. Thus multimodal therapy of sarcomas with VEGF-A inhibition, HIF-1α inhibition, and hypoxia-activated chemotherapy effectively blocks sarcoma growth through inhibition of tumor vasculature and cancer stem-like cells.
Collapse
Affiliation(s)
- Changhwan Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin K Chang
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Ho Lee
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sam S Yoon
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
68
|
Abstract
Evofosfamide, also formerly known as TH-302, is an investigational hypoxia-activated prodrug and is used to target cancerous cells under hypoxic conditions, which is a feature possessed by multiple solid tumors including pancreatic tumors. Gemcitabine, a cytotoxic agent, has for many years been the standard first-line treatment for metastatic pancreatic cancer in patients. In recent years, combination chemotherapeutic therapies have provided a new avenue for molecular targeting by increasing the probability of eliminating the cancer and minimizing the likelihood of resistance. We have evaluated multiple studies in an effort to shed light on an emerging prodrug, evofosfamide, which operates by selectively targeting the tumor hypoxic compartment. A web-based literature search was performed through PubMed and Google Scholar using the keywords 'evofosfamide', 'TH-302,' and 'pancreatic tumor.' Of the available results, 53 relevant studies were reviewed and summarized. Chemotherapeutic agents such as evofosfamide, which targets tumor hypoxia, are new agents against cancer cells. Current experience with these agents is limited as additional and longer prospective studies are needed to further evaluate the clinical efficacy and postmarketing safety profile.
Collapse
|
69
|
Paolicchi E, Gemignani F, Krstic-Demonacos M, Dedhar S, Mutti L, Landi S. Targeting hypoxic response for cancer therapy. Oncotarget 2017; 7:13464-78. [PMID: 26859576 PMCID: PMC4924654 DOI: 10.18632/oncotarget.7229] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/17/2016] [Indexed: 12/21/2022] Open
Abstract
Hypoxic tumor microenvironment (HTM) is considered to promote metabolic changes, oncogene activation and epithelial mesenchymal transition, and resistance to chemo- and radio-therapy, all of which are hallmarks of aggressive tumor behavior. Cancer cells within the HTM acquire phenotypic properties that allow them to overcome the lack of energy and nutrients supply within this niche. These phenotypic properties include activation of genes regulating glycolysis, glucose transport, acidosis regulators, angiogenesis, all of which are orchestrated through the activation of the transcription factor, HIF1A, which is an independent marker of poor prognosis. Moreover, during the adaptation to a HTM cancer cells undergo deep changes in mitochondrial functions such as “Warburg effect” and the “reverse Warburg effect”. This review aims to provide an overview of the characteristics of the HTM, with particular focus on novel therapeutic strategies currently in clinical trials, targeting the adaptive response to hypoxia of cancer cells.
Collapse
Affiliation(s)
- Elisa Paolicchi
- Genetics-Department of Biology, University of Pisa, Pisa, Italy
| | | | - Marija Krstic-Demonacos
- School of Environment and Life Sciences, College of Science and Technology, University of Salford, Salford, UK
| | - Shoukat Dedhar
- Department of Integrative Oncology, BC Cancer Research Centre, BC Cancer Agency and Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Luciano Mutti
- School of Environment and Life Sciences, College of Science and Technology, University of Salford, Salford, UK
| | - Stefano Landi
- Genetics-Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
70
|
Zhang W, Fan W, Zhou Z, Garrison J. Synthesis and Evaluation of Radiolabeled Phosphoramide Mustard with Selectivity for Hypoxic Cancer Cells. ACS Med Chem Lett 2017; 8:1269-1274. [PMID: 29259746 DOI: 10.1021/acsmedchemlett.7b00355] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 10/23/2017] [Indexed: 01/27/2023] Open
Abstract
Tumor hypoxia has been widely explored over the years as a diagnostic and therapeutic marker. Herein, we synthesized an alkyne functionalized version of evofosfamide, a hypoxia-selective prodrug. The purpose of this effort was to investigate if this novel 2-nitroimidazole phosphoramide nitrogen mustard (2-NIPAM) retained hypoxia selectivity and could be utilized in radiopharmaceutical development to significantly increase retention of conjugated agents in hypoxic cells. 2-NIPAM demonstrated good hypoxia selectivity with a 62- and 225-fold increase in cytotoxicity toward PC-3 and DU145 human prostate cancer cell lines, respectively, under hypoxic conditions. Radiolabeling of 2-NIPAM with 125I was accomplished through a Cu(I)-mediated azide-alkyne cycloaddition reaction. The 125I-conjugate demonstrated 13.6 and 17.8% lower efflux rates for DU145 and PC-3 cells, correspondingly, under hypoxic conditions, suggesting that the increased retention is likely due to the known intracellular trapping mechanism. In conclusion, these studies demonstrate the potential of 2-NIPAM in serving as a trapping agent for radiopharmaceutical development.
Collapse
Affiliation(s)
- Wenting Zhang
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Center
for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Wei Fan
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Center
for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Zhengyuan Zhou
- Department
of Radiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Jered Garrison
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department
of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Center
for Drug Delivery and Nanomedicine, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Eppley
Cancer Center, University of Nebraska Medical Center, 985950 Nebraska
Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
71
|
Hypoxia-activated prodrug enhances therapeutic effect of sunitinib in melanoma. Oncotarget 2017; 8:115140-115152. [PMID: 29383148 PMCID: PMC5777760 DOI: 10.18632/oncotarget.22944] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis is a critical step during tumor progression. Anti-angiogenic therapy has only provided modest benefits in delaying tumor progression despite its early promise in cancer treatment. It has been postulated that anti-angiogenic therapy may promote the emergence of a more aggressive cancer cell phenotype by generating increased tumor hypoxia—a well-recognized promoter of tumor progression. TH-302 is a 2-nitroimidazole triggered hypoxia-activated prodrug (HAP) which has been shown to selectively target the hypoxic tumor compartment and reduce tumor volume. Here, we show that melanoma cells grown under hypoxic conditions exhibit increased resistance to a wide variety of therapeutic agents in vitro and generate larger and more aggressive tumors in vivo than melanoma cells grown under normoxic conditions. However, hypoxic melanoma cells exhibit a pronounced sensitivity to TH-302 which is further enhanced by the addition of sunitinib. Short term sunitinib treatment fails to prolong the survival of melanoma bearing genetically engineered mice (Tyr::CreER; BRafCA;Ptenlox/lox) but increases tumor hypoxia. Long term TH-302 alone modestly prolongs the overall survival of melanoma bearing mice. Combination therapy of TH-302 with sunitinib further increases the survival of treated mice. These studies provide a translational rationale for combining hypoxic tumor cell targeted therapies with anti-angiogenics for treatment of melanoma.
Collapse
|
72
|
Anderson RF, Li D, Hunter FW. Antagonism in effectiveness of evofosfamide and doxorubicin through intermolecular electron transfer. Free Radic Biol Med 2017; 113:564-570. [PMID: 29111232 DOI: 10.1016/j.freeradbiomed.2017.10.385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/25/2017] [Accepted: 10/26/2017] [Indexed: 12/27/2022]
Abstract
Hypoxic cells pose a problem in anticancer chemotherapy, in which often drugs require oxygen as an electron acceptor to bring about the death of actively cycling cells. Bioreductive anticancer drugs, which are selectively activated in the hypoxic regions of tumours through enzymatic one-electron reduction, are being developed for combination with chemotherapy-, radiotherapy- and immunotherapy-containing regimens to kill treatment-resistant hypoxic cells. The most clinically-advanced bioreductive drug, evofosfamide (TH-302), which acts by releasing a DNA-crosslinking mustard, failed to extend overall survival in combination with doxorubicin, a topoisomerase II inhibitor, for advanced soft tissue sarcoma in a pivotal clinical trial. However, the reasons for the lack of additive efficacy with this combination are unknown. Here, we show that the radical anion of evofosfamide undergoes electron transfer to doxorubicin in kinetic competition to fragmentation of the radical anion, thus suppressing the release the cytotoxic mustard. This electron transfer process may account, at least in part, for the lack of overall survival improvement in the recent clinical trial. This study underlines the need to consider both redox and electron transfer chemistry when combining bioreductive prodrugs with other redox-active drugs in cancer treatment.
Collapse
Affiliation(s)
- Robert F Anderson
- Auckland Cancer Society Research Centre, Faculty of Health and Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; School of Chemical Sciences, Faculty of Science, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| | - Dan Li
- Auckland Cancer Society Research Centre, Faculty of Health and Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Francis W Hunter
- Auckland Cancer Society Research Centre, Faculty of Health and Medical Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
73
|
Dragovich PS, Broccatelli F, Chen J, Fan P, Le H, Mao W, Pillow TH, Polson AG, Wai J, Xu Z, Yao H, Zhang D. Design, synthesis, and biological evaluation of pyrrolobenzodiazepine-containing hypoxia-activated prodrugs. Bioorg Med Chem Lett 2017; 27:5300-5304. [DOI: 10.1016/j.bmcl.2017.10.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/10/2017] [Accepted: 10/11/2017] [Indexed: 01/26/2023]
|
74
|
Rautio J, Kärkkäinen J, Sloan KB. Prodrugs – Recent approvals and a glimpse of the pipeline. Eur J Pharm Sci 2017; 109:146-161. [DOI: 10.1016/j.ejps.2017.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 01/12/2023]
|
75
|
Hajj C, Russell J, Hart CP, Goodman KA, Lowery MA, Haimovitz-Friedman A, Deasy JO, Humm JL. A Combination of Radiation and the Hypoxia-Activated Prodrug Evofosfamide (TH-302) is Efficacious against a Human Orthotopic Pancreatic Tumor Model. Transl Oncol 2017; 10:760-765. [PMID: 28778024 PMCID: PMC5538966 DOI: 10.1016/j.tranon.2017.06.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/07/2017] [Accepted: 06/12/2017] [Indexed: 12/31/2022] Open
Abstract
This study was designed to investigate the effect of single-dose radiation therapy (RT) in combination with evofosfamide (TH-302), a hypoxia-activated prodrug, in a pre-clinical model of pancreatic cancer. AsPC1 tumors were implanted orthotopically in the pancreas of nude mice. Tumors were treated with 15 Gy of RT, using a 1 cm diameter field, and delivered as a continuous arc. Image-guidance to center the field on the tumor was based on CT imaging with intraperitoneal contrast. Evofosfamide (100 mg/kg, i.p.) was administered 3 hours before RT. Tumor volumes were measured using ultrasound, and regrowth curves were plotted. Tumor hypoxia and cell proliferation were measured using pimonidazole and the thymidine analog EdU, respectively. In vitro clonogenic assays were performed. Tumors were shown to contain substantial areas of hypoxia, as calculated by percent pimonidazole staining. Evofosfamide was active in these tumors, as demonstrated by a significant reduction in uptake of the thymidine analog EdU. This effect was visible in oxygenated tissue, consistent with the previously reported bystander effects of evofosfamide. RT produced significant regrowth delay, as did evofosfamide. The combination of both agents produced a growth delay that was at least equal to the sum of the two treatments given separately. The improvement in tumor response when evofosfamide is combined with RT supports the hypothesis that hypoxia is a cause of radioresistance in high dose RT for pancreatic cancer. Assessing the efficacy and safety of stereotactic radiation treatment and evofosfamide is warranted in patients with locally advanced pancreatic cancer.
Collapse
|
76
|
A novel concept for tumour targeting with radiation: Inverse dose-painting or targeting the “Low Drug Uptake Volume”. Radiother Oncol 2017; 124:513-520. [DOI: 10.1016/j.radonc.2017.04.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/17/2017] [Accepted: 04/21/2017] [Indexed: 01/21/2023]
|
77
|
Constantinidou A, van der Graaf WTA. The fate of new fosfamides in phase III studies in advanced soft tissue sarcoma. Eur J Cancer 2017; 84:257-261. [PMID: 28841543 DOI: 10.1016/j.ejca.2017.07.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 07/25/2017] [Indexed: 01/13/2023]
Abstract
For decades, doxorubicin alone or in combination with ifosfamide has been used in advanced soft tissue sarcoma (STS). In 2014, a comparison of doxorubicin alone versus the combination with ifosfamide (in the randomised phase III EORTC 62012) showed no difference in overall survival (OS), but a difference in response and progression-free survival (PFS) were observed in favour of the combination but at the expense of increased toxicity. Newer fosfamides, with slightly different modes of action, and potentially less toxicity, namely evofosfamide and palifosfamide have recently been tested in randomised phase III clinical trials in STS. The TH CR-406/SARC021 (June 2017) and the PICASSO III (September 2016) studies compared doxorubicin, as the standard arm, to doxorubicin in combination with evofosfamide and palifosfamide, respectively. In both studies, the combination arm produced increased response rates but at the expense of higher toxicity. However, there was no difference in OS or PFS in favour of the combination. Importantly, the median OS of patients receiving standard of care, doxorubicin, in both studies appeared improved from 12.8 months (95.5% CI 10.5-14·III) in the EORTC 62012 to 16.9 months (95% CI 14.8 to 22.9) in PICASSO III and 19.0 months (95% CI 16.2-22.4) in TH CR-406/SARC021. The results of these three randomised phase III studies highlight several critical issues related to the design and conduct of such trials in STS. We discuss these issues aiming to contribute to the ongoing debate about the optimal approach to perform clinical research in STS.
Collapse
|
78
|
Liapis V, Zysk A, DeNichilo M, Zinonos I, Hay S, Panagopoulos V, Shoubridge A, Difelice C, Ponomarev V, Ingman W, Atkins GJ, Findlay DM, Zannettino ACW, Evdokiou A. Anticancer efficacy of the hypoxia-activated prodrug evofosfamide is enhanced in combination with proapoptotic receptor agonists against osteosarcoma. Cancer Med 2017; 6:2164-2176. [PMID: 28799237 PMCID: PMC5603834 DOI: 10.1002/cam4.1115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/21/2017] [Accepted: 03/31/2017] [Indexed: 11/18/2022] Open
Abstract
Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, hypoxia also leads to treatment opportunities as demonstrated by the development of compounds that target regions of hypoxia within tumors. Evofosfamide is a hypoxia‐activated prodrug that is created by linking the hypoxia‐seeking 2‐nitroimidazole moiety to the cytotoxic bromo‐isophosphoramide mustard (Br‐IPM). When evofosfamide is delivered to hypoxic regions of tumors, the DNA cross‐linking toxin, Br‐IPM, is released leading to cell death. This study assessed the anticancer efficacy of evofosfamide in combination with the Proapoptotic Receptor Agonists (PARAs) dulanermin and drozitumab against human osteosarcoma in vitro and in an intratibial murine model of osteosarcoma. Under hypoxic conditions in vitro, evofosfamide cooperated with dulanermin and drozitumab, resulting in the potentiation of cytotoxicity to osteosarcoma cells. In contrast, under the same conditions, primary human osteoblasts were resistant to treatment. Animals transplanted with osteosarcoma cells directly into their tibiae developed mixed osteosclerotic/osteolytic bone lesions and consequently developed lung metastases 3 weeks post cancer cell transplantation. Tumor burden in the bone was reduced by evofosfamide treatment alone and in combination with drozitumab and prevented osteosarcoma‐induced bone destruction while also reducing the growth of pulmonary metastases. These results suggest that evofosfamide may be an attractive therapeutic agent, with strong anticancer activity alone or in combination with either drozitumab or dulanermin against osteosarcoma.
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
| | - Aneta Zysk
- 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
- Vascular Biology and Cell Trafficking Laboratory, Centre for Cancer Biology, University of South Australia, Adelaide, 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
| | - 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
| | - Alexandra Shoubridge
- Discipline of Surgery, Breast Cancer Research Unit, Basil Hetzel Institute and Centre for Personalised Cancer Medicine, University of Adelaide, Woodville, South Australia, Australia
| | - Christopher Difelice
- 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, New York
| | - Wendy Ingman
- Discipline of Surgery, School of Medicine at The Queen Elizabeth Hospital, University of Adelaide, Woodville, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, 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, 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
|
79
|
Tap WD, Papai Z, Van Tine BA, Attia S, Ganjoo KN, Jones RL, Schuetze S, Reed D, Chawla SP, Riedel RF, Krarup-Hansen A, Toulmonde M, Ray-Coquard I, Hohenberger P, Grignani G, Cranmer LD, Okuno S, Agulnik M, Read W, Ryan CW, Alcindor T, Del Muro XFG, Budd GT, Tawbi H, Pearce T, Kroll S, Reinke DK, Schöffski P. Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2017; 18:1089-1103. [PMID: 28651927 PMCID: PMC7771354 DOI: 10.1016/s1470-2045(17)30381-9] [Citation(s) in RCA: 214] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 01/18/2023]
Abstract
BACKGROUND Evofosfamide is a hypoxia-activated prodrug of bromo-isophosphoramide mustard. We aimed to assess the benefit of adding evofosfamide to doxorubicin as first-line therapy for advanced soft-tissue sarcomas. METHODS We did this international, open-label, randomised, phase 3, multicentre trial (TH CR-406/SARC021) at 81 academic or community investigational sites in 13 countries. Eligible patients were aged 15 years or older with a diagnosis of an advanced unresectable or metastatic soft-tissue sarcoma, of intermediate or high grade, for which no standard curative therapy was available, an Eastern Cooperative Oncology Group performance status of 0-1, and measurable disease by Response Evaluation Criteria in Solid Tumors version 1.1. Patients were randomly assigned (1:1) to receive doxorubicin alone (75 mg/m2 via bolus injection administered over 5-20 min or continuous intravenous infusion for 6-96 h on day 1 of every 21-day cycle for up to six cycles) or doxorubicin (given via the same dose procedure) plus evofosfamide (300 mg/m2 intravenously for 30-60 min on days 1 and 8 of every 21-day cycle for up to six cycles). After six cycles of treatment, patients in the single-drug doxorubicin group were followed up expectantly whereas patients with stable or responsive disease in the combination group were allowed to continue with evofosfamide monotherapy until documented disease progression. A web-based central randomisation with block sizes of two and four was stratified by extent of disease, doxorubicin administration method, and previous systemic therapy. Patients and investigators were not masked to treatment assignment. The primary endpoint was overall survival, analysed in the intention-to-treat population. Safety analyses were done in all patients who received any amount of study drug. This study was registered with ClinicalTrials.gov, number NCT01440088. FINDINGS Between Sept 26, 2011, and Jan 22, 2014, 640 patients were enrolled and randomly assigned to a treatment group (317 to doxorubicin plus evofosfamide and 323 to doxorubicin alone), all of whom were included in the intention-to-treat analysis. The overall survival endpoint was not reached (hazard ratio 1·06, 95% CI 0·88-1·29; p=0·527), with a median overall survival of 18·4 months (95% CI 15·6-22·1) with doxorubicin plus evofosfamide versus 19·0 months (16·2-22·4) with doxorubicin alone. The most common grade 3 or worse adverse events in both groups were haematological, including anaemia (150 [48%] of 313 patients in the doxorubicin plus evofosfamide group vs 65 [21%] of 308 in the doxorubicin group), neutropenia (47 [15%] vs 92 [30%]), febrile neutropenia (57 [18%] vs 34 [11%]), leucopenia (22 [7%] vs 17 [6%]), decreased neutrophil count (31 [10%] vs 41 [13%]), and decreased white blood cell count (39 [13%] vs 33 [11%]). Grade 3-4 thrombocytopenia was more common in the combination group (45 [14%]) than in the doxorubicin alone group (four [1%]), as was grade 3-4 stomatitis (26 [8%] vs seven [2%]). Serious adverse events were reported in 145 (46%) of 313 patients in the combination group and 99 (32%) of 308 in the doxorubicin alone group. Five (2%) patients died from treatment-related causes in the combination group (sepsis [n=2], septic shock [n=1], congestive cardiac failure [n=1], and unknown cause [n=1]) versus one (<1%) patient in the doxorubicin alone group (lactic acidosis [n=1]). INTERPRETATION The addition of evofosfamide to doxorubicin as first-line therapy did not improve overall survival compared with single-drug doxorubicin in patients with locally advanced, unresectable, or metastatic soft-tissue sarcomas and so this combination cannot be recommended in this setting. FUNDING Threshold Pharmaceuticals.
Collapse
Affiliation(s)
- William D Tap
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA.
| | - Zsuzsanna Papai
- Allami Egeszsegugyi Kozpont (State Health Center), Budapest, Hungary
| | | | | | - Kristen N Ganjoo
- Stanford University Department of Medicine Division of Oncology, Stanford, CA, USA
| | - Robin L Jones
- University of Washington Cancer Center/Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Scott Schuetze
- University of Michigan Cancer Center, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | | - Lee D Cranmer
- University of Arizona Cancer Center, Seattle, WA, USA
| | | | - Mark Agulnik
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - William Read
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | | | | | | | | | - Hussein Tawbi
- University of Pittsburgh Medical Center, Houston, TX, USA
| | | | - Stew Kroll
- Threshold Pharmaceuticals, South San Francisco, CA, USA
| | - Denise K Reinke
- Sarcoma Alliance for Research through Collaboration, Ann Arbor, MI, USA
| | | |
Collapse
|
80
|
Ao X, Bright SA, Taylor NC, Elmes RBP. 2-Nitroimidazole based fluorescent probes for nitroreductase; monitoring reductive stress in cellulo. Org Biomol Chem 2017; 15:6104-6108. [PMID: 28715020 DOI: 10.1039/c7ob01406f] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Two 2-nitroimidazole-1,8-naphthalimide conjugates, 1 and 2, have been synthesised as fluorescence probes for the detection of reductive stress in HeLa cells. The 4-substituted derivative 1 was shown to act as a highly sensitive and selective substrate for nitroreductase where it exhibited a clear blue to green ratiometric fluorescence response visible to the naked eye. Moreover, biological studies demonstrated 1 could be activated in cellulo where the impact of reductive stress was easily monitored using confocal microscopy and flow cytommetry.
Collapse
Affiliation(s)
- X Ao
- Department of Chemistry, Maynooth University, National, University of Ireland, Maynooth, Co. Kildare, Ireland.
| | - S A Bright
- Trinity Biomedical Sciences Institute (TBSI), Trinity, College Dublin, Dublin 2, Ireland
| | - N C Taylor
- Trinity Biomedical Sciences Institute (TBSI), Trinity, College Dublin, Dublin 2, Ireland
| | - R B P Elmes
- Department of Chemistry, Maynooth University, National, University of Ireland, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
81
|
Engel C, Brügmann G, Lambing S, Mühlenbeck LH, Marx S, Hagen C, Horváth D, Goldeck M, Ludwig J, Herzner AM, Drijfhout JW, Wenzel D, Coch C, Tüting T, Schlee M, Hornung V, Hartmann G, Van den Boorn JG. RIG-I Resists Hypoxia-Induced Immunosuppression and Dedifferentiation. Cancer Immunol Res 2017; 5:455-467. [PMID: 28468914 DOI: 10.1158/2326-6066.cir-16-0129-t] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 02/16/2017] [Accepted: 04/25/2017] [Indexed: 12/19/2022]
Abstract
A hypoxic tumor microenvironment is linked to poor prognosis. It promotes tumor cell dedifferentiation and metastasis and desensitizes tumor cells to type-I IFN, chemotherapy, and irradiation. The cytoplasmic immunoreceptor retinoic acid-inducible gene-I (RIG-I) is ubiquitously expressed in tumor cells and upon activation by 5'-triphosphate RNA (3pRNA) drives the induction of type I IFN and immunogenic cell death. Here, we analyzed the impact of hypoxia on the expression of RIG-I in various human and murine tumor and nonmalignant cell types and further investigated its function in hypoxic murine melanoma. 3pRNA-inducible RIG-I-expression was reduced in hypoxic melanoma cells compared with normoxic controls, a phenomenon that depended on the hypoxia-associated transcription factor HIF1α. Still, RIG-I functionality was conserved in hypoxic melanoma cells, whereas responsiveness to recombinant type-I IFN was abolished, due to hypoxia-induced loss of type I IFN receptor expression. Likewise, RIG-I activation in hypoxic melanoma cells, but not exposure to recombinant IFNα, provoked melanocyte antigen-specific CD8+ T-cell and NK-cell attack. Scavenging of hypoxia-induced reactive oxygen species by vitamin C restored the inducible expression of RIG-I under hypoxia in vitro, boosted in vitro anti-melanoma NK- and CD8+ T-cell attack, and augmented 3pRNA antitumor efficacy in vivo These results demonstrate that RIG-I remains operational under hypoxia and that RIG-I function is largely insensitive to lower cell surface expression of the IFNα receptor. RIG-I function could be fortified under hypoxia by the combined use of 3pRNA with antioxidants. Cancer Immunol Res; 5(6); 455-67. ©2017 AACR.
Collapse
Affiliation(s)
- Christina Engel
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Grethe Brügmann
- Institute for Molecular Medicine, University Hospital Bonn, Bonn, Germany
| | - Silke Lambing
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Larissa H Mühlenbeck
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Samira Marx
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Christian Hagen
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Dorottya Horváth
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Marion Goldeck
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Janos Ludwig
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Anna-Maria Herzner
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Jan W Drijfhout
- Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniela Wenzel
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christoph Coch
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Thomas Tüting
- Department for Dermatology and Allergy, University Hospital Bonn, Bonn, Germany.,Department of Dermatology, University of Magdeburg, Magdeburg, Germany
| | - Martin Schlee
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Veit Hornung
- Institute for Molecular Medicine, University Hospital Bonn, Bonn, Germany.,Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, München, Germany
| | - Gunther Hartmann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Jasper G Van den Boorn
- Institute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
82
|
Hydrogen peroxide activated quinone methide precursors with enhanced DNA cross-linking capability and cytotoxicity towards cancer cells. Eur J Med Chem 2017; 133:197-207. [PMID: 28388522 DOI: 10.1016/j.ejmech.2017.03.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/24/2017] [Accepted: 03/22/2017] [Indexed: 11/21/2022]
Abstract
Quinone methide (QM) formation induced by endogenously generated H2O2 is attractive for biological and biomedical applications. To overcome current limitations due to low biological activity of H2O2-activated QM precursors, we are introducing herein several new arylboronates with electron donating substituents at different positions of benzene ring and/or different neutral leaving groups. The reaction rate of the arylboronate esters with H2O2 and subsequent bisquinone methides formation and DNA cross-linking was accelerated with the application of Br as a leaving group instead of acetoxy groups. Additionally, a donating group placed meta to the nascent exo-methylene group of the quinone methide greatly improves H2O2-induced DNA interstrand cross-link formation as well as enhances the cellular activity. Multiple donating groups decrease the stability and DNA cross-linking capability, which lead to low cellular activity. A cell-based screen demonstrated that compounds 2a and 5a with a OMe or OH group dramatically inhibited the growth of various tissue-derived cancer cells while normal cells were less affected. Induction of H2AX phosphorylation by these compounds in CLL lymphocytes provide evidence for a correlation between cell death and DNA damage. The compounds presented herein showed potent anticancer activities and selectivity, which represent a novel scaffold for anticancer drug development.
Collapse
|
83
|
Jin C, Zhang Q, Lu W. Synthesis and biological evaluation of hypoxia-activated prodrugs of SN-38. Eur J Med Chem 2017; 132:135-141. [PMID: 28350997 DOI: 10.1016/j.ejmech.2017.03.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/01/2017] [Accepted: 03/21/2017] [Indexed: 12/21/2022]
Abstract
We designed new hypoxia-activated prodrugs by conjugating (1-methyl-2-nitro-1H-imidazol-5-yl)methanol with 7-ethyl-10-hydroxy camptothecin (SN-38). Initially, we improved the method of multi-gram scale synthesis of (1-methyl-2-nitro-1H-imidazol-5-yl)methanol, which increased the yield to 42% compared to 8% by the original synthesis method. The improved method was used to synthesize evofosfamide (TH-302) and hypoxia-activated prodrugs of SN-38. Two different linkages between (1-methyl-2-nitro-1H-imidazol-5-yl)methanol and SN-38 were evaluated that afforded different hypoxia-selectivity and toxicity. Compound 16 (IOS), containing an ether linkage, was considered to be a promising hypoxia-selective antitumor agent.
Collapse
Affiliation(s)
- Chen Jin
- School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Qiumeng Zhang
- School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China
| | - Wei Lu
- School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, PR China.
| |
Collapse
|
84
|
Winn BA, Shi Z, Carlson GJ, Wang Y, Nguyen BL, Kelly EM, Ross RD, Hamel E, Chaplin DJ, Trawick ML, Pinney KG. Bioreductively activatable prodrug conjugates of phenstatin designed to target tumor hypoxia. Bioorg Med Chem Lett 2017; 27:636-641. [PMID: 28007448 PMCID: PMC5319644 DOI: 10.1016/j.bmcl.2016.11.093] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/30/2016] [Accepted: 11/30/2016] [Indexed: 01/17/2023]
Abstract
A variety of solid tumor cancers contain significant regions of hypoxia, which provide unique challenges for targeting by potent anticancer agents. Bioreductively activatable prodrug conjugates (BAPCs) represent a promising strategy for therapeutic intervention. BAPCs are designed to be biologically inert until they come into contact with low oxygen tension, at which point reductase enzyme mediated cleavage releases the parent anticancer agent in a tumor-specific manner. Phenstatin is a potent inhibitor of tubulin polymerization, mimicking the chemical structure and biological activity of the natural product combretastatin A-4. Synthetic approaches have been established for nitrobenzyl, nitroimidazole, nitrofuranyl, and nitrothienyl prodrugs of phenstatin incorporating nor-methyl, mono-methyl, and gem-dimethyl variants of the attached nitro compounds. A series of BAPCs based on phenstatin have been prepared by chemical synthesis and evaluated against the tubulin-microtubule protein system. In a preliminary study using anaerobic conditions, the gem-dimethyl nitrothiophene and gem-dimethyl nitrofuran analogues were shown to undergo efficient enzymatic cleavage in the presence of NADPH cytochrome P450 oxidoreductase. Each of the eleven BAPCs evaluated in this study demonstrated significantly reduced inhibitory activity against tubulin in comparison to the parent anti-cancer agent phenstatin (IC50=1.0μM). In fact, the majority of the BAPCs (seven of the eleven analogues) were not inhibitors of tubulin polymerization (IC50>20μM), which represents an anticipated (and desirable) attribute for these prodrugs, since they are intended to be biologically inactive prior to enzyme-mediated cleavage to release phenstatin.
Collapse
Affiliation(s)
- Blake A Winn
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - Zhe Shi
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - Graham J Carlson
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - Yifan Wang
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - Benson L Nguyen
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - Evan M Kelly
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - R David Ross
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - David J Chaplin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States; Mateon Therapeutics, Inc., 701 Gateway Boulevard, Suite 210, South San Francisco, CA 94080, United States
| | - Mary L Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States.
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798-7348, United States.
| |
Collapse
|
85
|
Lin S, Li Y, Zheng Y, Luo L, Sun Q, Ge Z, Cheng T, Li R. Design, synthesis and biological evaluation of quinazoline–phosphoramidate mustard conjugates as anticancer drugs. Eur J Med Chem 2017; 127:442-458. [DOI: 10.1016/j.ejmech.2016.12.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/26/2016] [Accepted: 12/26/2016] [Indexed: 11/26/2022]
|
86
|
Molecular targeting of hypoxia in radiotherapy. Adv Drug Deliv Rev 2017; 109:45-62. [PMID: 27771366 DOI: 10.1016/j.addr.2016.10.002] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 10/02/2016] [Accepted: 10/15/2016] [Indexed: 12/21/2022]
Abstract
Hypoxia (low O2) is an essential microenvironmental driver of phenotypic diversity in human solid cancers. Hypoxic cancer cells hijack evolutionarily conserved, O2- sensitive pathways eliciting molecular adaptations that impact responses to radiotherapy, tumor recurrence and patient survival. In this review, we summarize the radiobiological, genetic, epigenetic and metabolic mechanisms orchestrating oncogenic responses to hypoxia. In addition, we outline emerging hypoxia- targeting strategies that hold promise for individualized cancer therapy in the context of radiotherapy and drug delivery.
Collapse
|
87
|
Jin C, Zhang Q, Lu W. Selective turn-on near-infrared fluorescence probe for hypoxic tumor cell imaging. RSC Adv 2017. [DOI: 10.1039/c7ra01466j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In this study, we designed a new selective turn on near-infrared fluorescence probe by conjugating (1-methyl-2-nitro-1H-imidazol-5-yl)methanol to DCPO with ether linkage for hypoxic tumor cell imaging.
Collapse
Affiliation(s)
- Chen Jin
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
- P. R. China
| | - Qiumeng Zhang
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
- P. R. China
| | - Wei Lu
- School of Chemistry and Molecular Engineering
- East China Normal University
- Shanghai 200062
- P. R. China
- State Key Laboratory of Fine Chemicals
| |
Collapse
|
88
|
Erkan M, Kurtoglu M, Kleeff J. The role of hypoxia in pancreatic cancer: a potential therapeutic target? Expert Rev Gastroenterol Hepatol 2016; 10:301-16. [PMID: 26560854 DOI: 10.1586/17474124.2016.1117386] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
One of the key factors that correlates with poor survival of patients with pancreatic cancer is the extent of hypoxic areas within the tumor tissue. The adaptation of pancreatic cancer cells to limited oxygen delivery promotes the induction of an invasive and treatment-resistant phenotype, triggering metastases at an early stage of tumor development, which resist in most cases adjuvant therapies following tumor resection. In this article, the authors summarize the evidence demonstrating the significance of hypoxia in pancreatic cancer pathogenesis and discuss the possible hypoxia-induced mechanisms underlying its aggressive nature. We then conclude with promising strategies that target hypoxia-adapted pancreatic cancer cells.
Collapse
Affiliation(s)
- Mert Erkan
- a Department of Surgery , Koç University School of Medicine , Istanbul , Turkey
| | - Metin Kurtoglu
- b Department of Oncology , Koç University School of Medicine , Istanbul , Turkey
| | - Jorg Kleeff
- c Department of Surgery , The Royal Liverpool and Broadgreen University Hospitals , Liverpool , UK.,d Department of General-, Visceral- and Pediatric Surgery , University Hospital Düsseldorf, Heinrich Heine University Düsseldorf , Düsseldorf , Germany
| |
Collapse
|
89
|
Design and Synthesis of Vandetanib Derivatives Containing Nitroimidazole Groups as Tyrosine Kinase Inhibitors in Normoxia and Hypoxia. Molecules 2016; 21:molecules21121693. [PMID: 27983649 PMCID: PMC6273768 DOI: 10.3390/molecules21121693] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/04/2016] [Accepted: 12/06/2016] [Indexed: 01/27/2023] Open
Abstract
Sixteen novel epidermal growth factor receptor (EGFR)/vascular endothelial growth factor (VEGF)-2 inhibitors (nitroimidazole-substituted 4-anilinoquinazoline derivatives (16a–p)) were designed and prepared via the introduction of a nitroimidazole group in the piperidine side chain and modification on the aniline moiety of vandetanib. Preliminary biological tests showed that comparing with vandetanib, some target compounds exhibited excellent EGFR inhibitory activities and anti-proliferative over A549/H446 cells in hypoxia. Meanwhile, several of the above compounds demonstrated better bioactivity than vandetanib in VEGF gene expression inhibition. Owing to the excellent IC50 value (1.64 μmol/L), the inhibition ratios of 16f over A549 and H446 cells were 62.01% and 59.86% at the concentration of 0.5 μM in hypoxia, respectively. All of these results indicated that 16f was a potential cancer therapeutic agent in hypoxia and was worthy of further development.
Collapse
|
90
|
|
91
|
Karnthaler-Benbakka C, Groza D, Koblmüller B, Terenzi A, Holste K, Haider M, Baier D, Berger W, Heffeter P, Kowol CR, Keppler BK. Targeting a Targeted Drug: An Approach Toward Hypoxia-Activatable Tyrosine Kinase Inhibitor Prodrugs. ChemMedChem 2016; 11:2410-2421. [PMID: 27706901 PMCID: PMC6151264 DOI: 10.1002/cmdc.201600417] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Indexed: 01/09/2023]
Abstract
Tyrosine kinase inhibitors (TKIs), which have revolutionized cancer therapy over the past 15 years, are limited in their clinical application due to serious side effects. Therefore, we converted two approved TKIs (sunitinib and erlotinib) into 2-nitroimidazole-based hypoxia-activatable prodrugs. Kinetics studies showed very different stabilities over 24 h; however, fast reductive activation via E. coli nitroreductase could be confirmed for both panels. The anticancer activity and signaling inhibition of the compounds against various human cancer cell lines were evaluated in cell culture. These data, together with molecular docking simulations, revealed distinct differences in the impact of structural modifications on drug binding to the enzymes: whereas the catalytic pocket of the epidermal growth factor receptor (EGFR) accepted all new erlotinib derivatives, the vascular endothelial growth factor receptor (VEGFR)-inhibitory potential in the case of the sunitinib prodrugs was dramatically diminished by derivatization. In line, hypoxia dependency of ERK signaling inhibition was observed with the sunitinib prodrugs, while oxygen levels had no impact on the activity of the erlotinib derivatives. Overall, proof of principle could be shown for this concept, and the results obtained are an important basis for the future development of tyrosine kinase inhibitor prodrugs.
Collapse
Affiliation(s)
| | - Diana Groza
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
| | - Bettina Koblmüller
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
| | - Alessio Terenzi
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Straße 42, 1090 Wien (Austria)
- Research Platform “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, (Austria)
| | - Katharina Holste
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
| | - Melanie Haider
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
| | - Dina Baier
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
- Research Platform “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, (Austria)
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8A, 1090 Wien (Austria)
- Research Platform “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, (Austria)
| | - Christian R. Kowol
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Straße 42, 1090 Wien (Austria)
- Research Platform “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, (Austria)
| | - Bernhard K. Keppler
- Institute of Inorganic Chemistry, University of Vienna, Waehringer Straße 42, 1090 Wien (Austria)
- Research Platform “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, (Austria)
| |
Collapse
|
92
|
Stokes AM, Hart CP, Quarles CC. Hypoxia Imaging With PET Correlates With Antitumor Activity of the Hypoxia-Activated Prodrug Evofosfamide (TH-302) in Rodent Glioma Models. Tomography 2016; 2:229-237. [PMID: 27752544 PMCID: PMC5065246 DOI: 10.18383/j.tom.2016.00259] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
High-grade gliomas are often characterized by hypoxia, which is associated with both poor long-term prognosis and therapy resistance. The adverse role hypoxia plays in treatment resistance and disease progression has led to the development of hypoxia imaging methods and hypoxia-targeted treatments. Here, we determined the tumor hypoxia and vascular perfusion characteristics of 2 rat orthotopic glioma models using 18-fluoromisonidozole positron emission tomography. In addition, we determined tumor response to the hypoxia-activated prodrug evofosfamide (TH-302) in these rat glioma models. C6 tumors exhibited more hypoxia and were less perfused than 9L tumors. On the basis of these differences in their tumor hypoxic burden, treatment with evofosfamide resulted in 4- and 2-fold decreases in tumor growth rates of C6 and 9L tumors, respectively. This work shows that imaging methods sensitive to tumor hypoxia and perfusion are able to predict response to hypoxia-targeted agents. This has implications for improved patient selection, particularly in clinical trials, for treatment with hypoxia-activated cytotoxic prodrugs, such as evofosfamide.
Collapse
Affiliation(s)
- Ashley M. Stokes
- Institute of Imaging Science, Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, Tennessee
- Department of Imaging Research, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and
| | - Charles P. Hart
- Threshold Pharmaceuticals Inc., South San Francisco, California
| | - C. Chad Quarles
- Institute of Imaging Science, Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, Tennessee
- Department of Imaging Research, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona; and
| |
Collapse
|
93
|
Patel A, Sant S. Hypoxic tumor microenvironment: Opportunities to develop targeted therapies. Biotechnol Adv 2016; 34:803-812. [PMID: 27143654 PMCID: PMC4947437 DOI: 10.1016/j.biotechadv.2016.04.005] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/13/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023]
Abstract
In recent years, there has been great progress in the understanding of tumor biology and its surrounding microenvironment. Solid tumors create regions with low oxygen levels, generally termed as hypoxic regions. These hypoxic areas offer a tremendous opportunity to develop targeted therapies. Hypoxia is not a random by-product of the cellular milieu due to uncontrolled tumor growth; rather it is a constantly evolving participant in overall tumor growth and fate. This article reviews current trends and recent advances in drug therapies and delivery systems targeting hypoxia in the tumor microenvironment. In the first part, we give an account of important physicochemical changes and signaling pathways activated in the hypoxic microenvironment. This is then followed by various treatment strategies including hypoxia-sensitive signaling pathways and approaches to develop hypoxia-targeted drug delivery systems.
Collapse
Affiliation(s)
- Akhil Patel
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
94
|
Oliveira FM, Barbosa LC, Fernandes SA, Lage MR, Carneiro JWDM, Kabeshov MA. Evaluation of some density functional methods for the estimation of hydrogen and carbon chemical shifts of phosphoramidates. COMPUT THEOR CHEM 2016. [DOI: 10.1016/j.comptc.2016.06.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
95
|
Duran R, Mirpour S, Pekurovsky V, Ganapathy-Kanniappan S, Brayton CF, Cornish TC, Gorodetski B, Reyes J, Chapiro J, Schernthaner RE, Frangakis C, Lin M, Sun JD, Hart CP, Geschwind JF. Preclinical Benefit of Hypoxia-Activated Intra-arterial Therapy with Evofosfamide in Liver Cancer. Clin Cancer Res 2016; 23:536-548. [PMID: 27440271 DOI: 10.1158/1078-0432.ccr-16-0725] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/03/2016] [Accepted: 07/12/2016] [Indexed: 12/24/2022]
Abstract
PURPOSE To evaluate safety and characterize anticancer efficacy of hepatic hypoxia-activated intra-arterial therapy (HAIAT) with evofosfamide in a rabbit model. EXPERIMENTAL DESIGN VX2-tumor-bearing rabbits were assigned to 4 intra-arterial therapy (IAT) groups (n = 7/group): (i) saline (control); (ii) evofosfamide (Evo); (iii) doxorubicin-lipiodol emulsion followed by embolization with 100-300 μm beads (conventional, cTACE); or (iv) cTACE and evofosfamide (cTACE + Evo). Blood samples were collected pre-IAT and 1, 2, 7, and 14 days post-IAT. A semiquantitative scoring system assessed hepatocellular damage. Tumor volumes were segmented on multidetector CT (baseline, 7/14 days post-IAT). Pathologic tumor necrosis was quantified using manual segmentation on whole-slide images. Hypoxic fraction (HF) and compartment (HC) were determined by pimonidazole staining. Tumor DNA damage, apoptosis, cell proliferation, endogenous hypoxia, and metabolism were quantified (γ-H2AX, Annexin V, caspase-3, Ki-67, HIF1α, VEGF, GAPDH, MCT4, and LDH). RESULTS cTACE + Evo showed a similar profile of liver enzymes elevation and pathologic scores compared with cTACE. Neither hematologic nor renal toxicity were observed. Animals treated with cTACE + Evo demonstrated smaller tumor volumes, lower tumor growth rates, and higher necrotic fractions compared with cTACE. cTACE + Evo resulted in a marked reduction in the HF and HC. Correlation was observed between decreases in HF or HC and tumor necrosis. cTACE + Evo promoted antitumor effects as evidenced by increased expression of γ-H2AX, apoptotic biomarkers, and decreased cell proliferation. Increased HIF1α/VEGF expression and tumor glycolysis supported HAIAT. CONCLUSIONS HAIAT achieved a promising step towards the locoregional targeting of tumor hypoxia. The favorable toxicity profile and enhanced anticancer effects of evofosfamide in combination with cTACE pave the way towards clinical trials in patients with liver cancer. Clin Cancer Res; 23(2); 536-48. ©2016 AACR.
Collapse
Affiliation(s)
- Rafael Duran
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Sahar Mirpour
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Vasily Pekurovsky
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Shanmugasundaram Ganapathy-Kanniappan
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Toby C Cornish
- Department of Pathology, Division of Gastrointestinal and Liver Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Boris Gorodetski
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Juvenal Reyes
- Department of Radiation Oncology and Molecular Radiation Sciences, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Julius Chapiro
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Rüdiger E Schernthaner
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Vascular and Interventional Radiology, The Johns Hopkins Hospital, Baltimore, Maryland
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Constantine Frangakis
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - MingDe Lin
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
- U/S Imaging and Interventions (UII), Philips Research North America, Cambridge, Massachusetts
| | - Jessica D Sun
- Threshold Pharmaceuticals, South San Francisco, California
| | - Charles P Hart
- Threshold Pharmaceuticals, South San Francisco, California
| | - Jean-François Geschwind
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
96
|
Ikeda Y, Hisano H, Nishikawa Y, Nagasaki Y. Targeting and Treatment of Tumor Hypoxia by Newly Designed Prodrug Possessing High Permeability in Solid Tumors. Mol Pharm 2016; 13:2283-9. [PMID: 27187083 DOI: 10.1021/acs.molpharmaceut.6b00011] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tumor hypoxia, which is associated with poor prognosis in cancer, is known to lead to resistance to radiotherapy and anticancer chemotherapy. Impaired drug penetration in hypoxic regions has been recognized as an essential barrier to drug development in solid tumors. Here, we propose novel hypoxia-activated prodrugs, which drastically improved the penetration property of commonly used anticancer drugs in the hypoxic region. In this design, conventional anticancer drugs were modified with 2-nitroimidazole derivatives. The most important point of this study was that the prodrug designed formed a 6-membered cyclic structure to allow liberation of the active drug in the hypoxic region. This design markedly increased the selectivity of the hypoxia-targeted prodrug, resulting in significant reduction of adverse effects in the normoxic region. In vitro studies confirmed the selective activation under hypoxic conditions. In vivo studies showed drastic reduction of adverse effects associated with conventional anticancer drugs and improvement of the survival rate of mice. Immunofluorescence analyses confirmed that the designed prodrug had a tendency to localize at the hypoxic region, in contrast to conventional anticancer drugs, which localize only at the normoxic region.
Collapse
Affiliation(s)
- Yutaka Ikeda
- Department of Materials Science, Master's School of Medical Sciences, University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| | - Hikaru Hisano
- Department of Materials Science, Master's School of Medical Sciences, University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University , Midorigaoka Higashi-2-jyo 1-1-1, Asahikawa, Hokkaido 078-8510, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Master's School of Medical Sciences, University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan.,Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba , Tennodai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan.,Satellite Laboratory, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), University of Tsukuba , Tennoudai 1-1-1, Tsukuba 305-8573, Japan
| |
Collapse
|
97
|
Hypoxia-Sensitive Materials for Biomedical Applications. Ann Biomed Eng 2016; 44:1931-45. [DOI: 10.1007/s10439-016-1578-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 02/23/2016] [Indexed: 10/22/2022]
|
98
|
Sun JD, Liu Q, Ahluwalia D, Ferraro DJ, Wang Y, Jung D, Matteucci MD, Hart CP. Comparison of hypoxia-activated prodrug evofosfamide (TH-302) and ifosfamide in preclinical non-small cell lung cancer models. Cancer Biol Ther 2016; 17:371-80. [PMID: 26818215 DOI: 10.1080/15384047.2016.1139268] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Evofosfamide (TH-302) is a hypoxia-activated prodrug of the cytotoxin bromo-isophosphoramide. In hypoxic conditions Br-IPM is released and alkylates DNA. Ifosfamide is a chloro-isophosphoramide prodrug activated by hepatic Cytochrome P450 enzymes. Both compounds are used for the treatment of cancer. Ifosfamide has been approved by the FDA while evofosfamide is currently in the late stage of clinical development. The purpose of this study is to compare efficacy and safety profile of evofosfamide and ifosfamide in preclinical non-small cell lung cancer H460 xenograft models. Immunocompetent CD-1 mice and H460 tumor-bearing immunocompromised nude mice were used to investigate the safety profile. The efficacy of evofosfamide or ifosfamide, alone, and in combination with docetaxel or sunitinib was compared in ectopic and intrapleural othortopic H460 xenograft models in animals exposed to ambient air or different oxygen concentration breathing conditions. At an equal body weight loss level, evofosfamide showed greater or comparable efficacy in both ectopic and orthotopic H460 xenograft models. Evofosfamide, but not ifosfamide, exhibited controlled oxygen concentration breathing condition-dependent antitumor activity. However, at an equal body weight loss level, ifosfamide yielded severe hematologic toxicity when compared to evofosfamide, both in monotherapy and in combination with docetaxel. At an equal hematoxicity level, evofosfamide showed superior antitumor activity. These results indicate that evofosfamide shows superior or comparable efficacy and a favorable safety profile when compared to ifosfamide in preclinical human lung carcinoma models. This finding is consistent with multiple clinical trials of evofosfamide as a single agent, or in combination therapy, which demonstrated both anti-tumor activity and safety profile without severe myelosuppression.
Collapse
Affiliation(s)
- Jessica D Sun
- a Threshold Pharmaceuticals , South San Francisco , CA , USA
| | - Qian Liu
- a Threshold Pharmaceuticals , South San Francisco , CA , USA
| | | | | | - Yan Wang
- a Threshold Pharmaceuticals , South San Francisco , CA , USA
| | - Don Jung
- a Threshold Pharmaceuticals , South San Francisco , CA , USA
| | | | - Charles P Hart
- a Threshold Pharmaceuticals , South San Francisco , CA , USA
| |
Collapse
|
99
|
Chand S, O'Hayer K, Blanco FF, Winter JM, Brody JR. The Landscape of Pancreatic Cancer Therapeutic Resistance Mechanisms. Int J Biol Sci 2016; 12:273-82. [PMID: 26929734 PMCID: PMC4753156 DOI: 10.7150/ijbs.14951] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer (pancreatic ductal adenocarcinoma, PDA) is infamously moving to the top of the list as one of the most lethal cancers with an overall 5 year survival rate of 7%. Multiple genomic-based and molecular characterization studies of PDA specimens and established animal models have provided the field with multiple targets and a progression model of this disease. Still, to date, the best therapeutic options are surgery and combination cytotoxic therapies. In general, even in the best case scenario (i.e., an early stage diagnosis and a response to a specific therapy), most of these fortunate patients' PDA cells acquire or exert resistance mechanisms and eventually kill the patient. Herein, we touch on a growing field of investigation that focuses on PDA cell therapeutic resistance mechanisms. We examine extrinsic elements (i.e., the tumor microenvironment, hypoxia) to the intrinsic processes within the cell (i.e., post-transcriptional gene regulation and somatic mutations) that are important for therapeutic efficacy and resistance. Even as better targeted and personalized approaches move through the clinical trial pipeline the discussed resistance mechanisms will most likely play a role in the management of this deadly disease.
Collapse
Affiliation(s)
- Saswati Chand
- 1. Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center
| | - Kevin O'Hayer
- 1. Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center;; 2. Department of Medical Oncology, and the; 3. Department of Pharmacology & Experimental Therapeutics, Division of Clinical Pharmacology, Thomas Jefferson University, Philadelphia PA
| | - Fernando F Blanco
- 1. Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center;; 3. Department of Pharmacology & Experimental Therapeutics, Division of Clinical Pharmacology, Thomas Jefferson University, Philadelphia PA
| | - Jordan M Winter
- 1. Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center
| | - Jonathan R Brody
- 1. Department of Surgery, The Jefferson Pancreas, Biliary, and Related Cancer Center
| |
Collapse
|
100
|
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
|