1
|
Critelli RM, Casari F, Borghi A, Serino G, Caporali C, Magistri P, Pecchi A, Shahini E, Milosa F, Di Marco L, Pivetti A, Lasagni S, Schepis F, De Maria N, Dituri F, Martínez-Chantar ML, Di Benedetto F, Giannelli G, Villa E. The Neoangiogenic Transcriptomic Signature Impacts Hepatocellular Carcinoma Prognosis and Can Be Triggered by Transarterial Chemoembolization Treatment. Cancers (Basel) 2024; 16:3549. [PMID: 39456643 PMCID: PMC11505901 DOI: 10.3390/cancers16203549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/05/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: We evaluated the relationship between the neoangiogenic transcriptomic signature (nTS) and clinical symptoms, treatment outcomes, and survival in hepatocellular carcinoma (HCC) patients. Methods: This study prospectively followed 328 patients in the derivation and 256 in the validation cohort (with a median follow-up of 31 and 22 months, respectively). The nTS was associated with disease presentation, treatments administered, and overall survival rates. Additionally, this study investigated how multiple treatments influenced changes in nTS status and alterations in microRNA expression. Results: The nTS was identified in 27.4% of patients, linked to aggressive features like multifocality and elevated alpha-fetoprotein (AFP), a pattern consistent with that of the validation cohort. Most patients in both cohorts received treatment for HCC. nTS+ patients had limited access to, and benefited less from, liver transplantation or radiofrequency ablation (RFA) compared to nTS- patients. By the end, 78.9% had died, with nTS- patients showing better median survival and response to treatments than their nTS+ counterparts, who had lower survival across all treatment types. Among those who received transarterial chemoembolization (TACE), 31.2% (21/80 patients after the initial treatment and another four following a second TACE) transitioned from an nTS- to an nTS+ status. This shift was associated with lower survival and alterations in microRNA expressions related to oncogenic pathways. Conclusions: The nTS markedly influences treatment eligibility and survival in patients with HCC. Notably, the nTS can develop after repeated TACE procedures, significantly impacting patient survival and altering oncogenic microRNA expression patterns. These findings highlight the critical role of the nTS in guiding treatment decisions and prognostication in HCC management.
Collapse
Affiliation(s)
- Rosina Maria Critelli
- Gastroenterology Unit, CHIMOMO Department, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.M.C.); (F.M.); (A.P.); (S.L.); (N.D.M.)
| | - Federico Casari
- Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.C.); (C.C.); (A.P.)
| | - Alberto Borghi
- Internal Medicine, Ospedale di Faenza, 48018 Faenza, Italy;
| | - Grazia Serino
- National Institute of Gastroenterology “IRCCS Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (G.S.); (E.S.); (F.D.); (G.G.)
| | - Cristian Caporali
- Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.C.); (C.C.); (A.P.)
| | - Paolo Magistri
- HPB Surgery and Liver Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Annarita Pecchi
- Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.C.); (C.C.); (A.P.)
| | - Endrit Shahini
- National Institute of Gastroenterology “IRCCS Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (G.S.); (E.S.); (F.D.); (G.G.)
| | - Fabiola Milosa
- Gastroenterology Unit, CHIMOMO Department, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.M.C.); (F.M.); (A.P.); (S.L.); (N.D.M.)
| | - Lorenza Di Marco
- Clinical and Experimental Medicine PhD Program, 41125 Modena, Italy;
| | - Alessandra Pivetti
- Gastroenterology Unit, CHIMOMO Department, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.M.C.); (F.M.); (A.P.); (S.L.); (N.D.M.)
| | - Simone Lasagni
- Gastroenterology Unit, CHIMOMO Department, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.M.C.); (F.M.); (A.P.); (S.L.); (N.D.M.)
| | - Filippo Schepis
- M.E.C. Dipartimental Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Nicola De Maria
- Gastroenterology Unit, CHIMOMO Department, University of Modena and Reggio Emilia, 41124 Modena, Italy; (R.M.C.); (F.M.); (A.P.); (S.L.); (N.D.M.)
| | - Francesco Dituri
- National Institute of Gastroenterology “IRCCS Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (G.S.); (E.S.); (F.D.); (G.G.)
| | - María Luz Martínez-Chantar
- Liver Disease Laboratory, Centre for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain;
- Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (CIBERehd), 28200 Madrid, Spain
| | - Fabrizio Di Benedetto
- HPB Surgery and Liver Transplant Unit, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology “IRCCS Saverio de Bellis”, Research Hospital, 70013 Castellana Grotte, Italy; (G.S.); (E.S.); (F.D.); (G.G.)
| | - Erica Villa
- M.E.C. Dipartimental Unit, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| |
Collapse
|
2
|
Rozbicki P, Oğuz E, Wolińska E, Türkan F, Cetin A, Branowska D. Synthesis and examination of 1,2,4-triazine-sulfonamide hybrids as potential inhibitory drugs: Inhibition effects on AChE and GST enzymes in silico and in vitro conditions. Arch Pharm (Weinheim) 2024; 357:e2400182. [PMID: 38771105 DOI: 10.1002/ardp.202400182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
The crucial functions of acetylcholinesterase (AChE) in neurotransmission and glutathione S-transferase (GST) in detoxification and cellular protection underscore their pivotal roles as key enzymes, essential for maintaining the integrity of neurological and cellular homeostasis. For this purpose, a series of 1,2,4-triazine-sulfonamide hybrids (3a-r) was successfully synthesized, and subsequently evaluated for their inhibitory effects on AChE and GST. The investigation was complemented by molecular docking studies and ADMET (Absorption, Distribution, Metabolism, Excretion, and Toxicity) predictions. The synthesized hybrids demonstrated significant promise in inhibiting both AChE and GST activities. Molecular docking analyses provided insights into the interactions between the compounds and the target enzymes, shedding light on potential binding modes and key amino acid residues involved. Furthermore, the study benefited from ADMET predictions, offering valuable information on the compounds' pharmacokinetic properties and potential toxicity. The promising results obtained from this comprehensive approach highlight the potential of these 1,2,4-triazine-sulfonamide hybrids as effective inhibitors of AChE and GST, paving the way for further development and optimization in the pursuit of novel therapeutic agents.
Collapse
Affiliation(s)
| | - Ercan Oğuz
- Department of Medical Services and Techniques, Health Services Vocational School, Igdır University, Igdır, Turkey
| | - Ewa Wolińska
- Institute of Chemical Sciences, University of Siedlce, Siedlce, Poland
| | - Fikret Türkan
- Department of Basic Sciences, Faculty of Dentistry, Igdır University, Igdır, Turkey
| | - Adnan Cetin
- Department of Chemistry, Faculty of Education, Van Yuzuncu Yil University, Van, Turkey
| | - Danuta Branowska
- Institute of Chemical Sciences, University of Siedlce, Siedlce, Poland
| |
Collapse
|
3
|
Zheng M, Xu H, Huang Y, Sun J, Zhang H, Lv Z, Liu Z, Tang Z, Chen X. Hypoxia-activated glutamine antagonist prodrug combined with combretastatin A4 nanoparticles for tumor-selective metabolic blockade. J Control Release 2024; 365:480-490. [PMID: 38040341 DOI: 10.1016/j.jconrel.2023.11.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
6-Diazo-5-oxo-L-norleucine (DON) is a potent glutamine antagonist with toxic side effects; in order to reduce these effects, multiple prodrugs have been designed. However, there are currently no reports of a DON prodrug with a defined mechanism to achieve high tumor selectivity. To improve the selective toxicity of DON to tumor cells while reducing systemic toxicity, a hypoxia-activated prodrug, termed HDON, was designed. HDON achieved remarkable tumor suppression of 76.4 ± 5.2% without leading to weight loss in an H22 murine liver cancer model with high hypoxia. Moreover, to augment the therapeutic efficacy of HDON, combretastatin A4 nanoparticles were used to aggravate tumor hypoxia of MC38 murine colon cancer and 4T1 murine breast cancer, activate HDON to DON, and stimulate a robust anti-tumor immune response while selectively killing in tumor cells in vivo, achieving significantly elevated tumor suppression rates of 98.3 ± 3.4% and 98.1 ± 3.1%, with cure rates of 80.0% and 20.0%, respectively.
Collapse
Affiliation(s)
- Mengfei Zheng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Hang Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yue Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jiali Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Honglei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education and Key Laboratory of Polymeric Materials & Application Technology of Hunan Province, Xiangtan University, Xiangtan 411105, China
| | - Zheng Lv
- The First Hospital of Jilin University, Changchun 130021, China
| | - Zhilin Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
4
|
Wang L, Yi S, Teng Y, Li W, Cai J. Role of the tumor microenvironment in the lymphatic metastasis of cervical cancer (Review). Exp Ther Med 2023; 26:486. [PMID: 37753293 PMCID: PMC10518654 DOI: 10.3892/etm.2023.12185] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/15/2023] [Indexed: 09/28/2023] Open
Abstract
Lymphatic metastasis is the primary type of cervical cancer metastasis and is associated with an extremely poor prognosis in patients. The tumor microenvironment primarily includes cancer-associated fibroblasts, tumor-associated macrophages, myeloid-derived suppressor cells, immune and inflammatory cells, and blood and lymphatic vascular networks, which can promote the establishment of lymphatic metastatic sites within immunosuppressive microenvironments or promote lymphatic metastasis by stimulating lymphangiogenesis and epithelial-mesenchymal transformation. As the most important feature of the tumor microenvironment, hypoxia plays an essential role in lymph node metastasis. In this review, the known mechanisms of hypoxia, and the involvement of stromal components and immune inflammatory cells in the tumor microenvironment of lymphatic metastasis of cervical cancer are discussed. Additionally, a summary of the clinical trials targeting the tumor microenvironment for the treatment of cervical cancer is provided, emphasizing the potential and challenges of immunotherapy.
Collapse
Affiliation(s)
- Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shuyan Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yun Teng
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine; Key Laboratory of Clinical In Vitro Diagnostic Techniques of Zhejiang Province; Institute of Laboratory Medicine, Zhejiang University, Hangzhou, Zhejiang 310000, P.R. China
| | - Wenhan Li
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
5
|
Vanhaezebrouck IF, Scarpelli ML. Companion Animals as a Key to Success for Translating Radiation Therapy Research into the Clinic. Cancers (Basel) 2023; 15:3377. [PMID: 37444487 PMCID: PMC10341092 DOI: 10.3390/cancers15133377] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Many successful preclinical findings fail to be replicated during translation to human studies. This leads to significant resources being spent on large clinical trials, and in some cases, promising therapeutics not being pursued due to the high costs of clinical translation. These translational failures emphasize the need for improved preclinical models of human cancer so that there is a higher probability of successful clinical translation. Companion-animal cancers offer a potential solution. These cancers are more similar to human cancer than other preclinical models, with a natural evolution over time, genetic alterations, intact immune system, and a permanent adaptation to the microenvironment. These advantages have led pioneers in veterinary radiation oncology to aid human medicine by elucidating basic principles of radiation biology. More recently, the veterinary and human radiation oncology fields have increasingly collaborated to achieve advancements in education, radiotherapy techniques, and trial networks. This review describes these advancements, including significant prior research findings and the evolution of the veterinary radiation oncology discipline. It concludes by describing how companion-animal models can help shape the future of human radiotherapy. Taken as a whole, this review suggests companion-animal cancers may become widely used for preclinical radiotherapy research.
Collapse
Affiliation(s)
| | - Matthew L. Scarpelli
- School of Health Sciences, Purdue University, 550 W Stadium Ave, West Lafayette, IN 47907, USA;
| |
Collapse
|
6
|
Alden RS, Kamran MZ, Bashjawish BA, Simone BA. Glutamine metabolism and radiosensitivity: Beyond the Warburg effect. Front Oncol 2022; 12:1070514. [PMID: 36465373 PMCID: PMC9712788 DOI: 10.3389/fonc.2022.1070514] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 06/03/2024] Open
Abstract
Mounting data suggest that cancer cell metabolism can be utilized therapeutically to halt cell proliferation, metastasis and disease progression. Radiation therapy is a critical component of cancer treatment in curative and palliative settings. The use of metabolism-based therapeutics has become increasingly popular in combination with radiotherapy to overcome radioresistance. Over the past year, a focus on glutamine metabolism in the setting of cancer therapy has emerged. In this mini-review, we discuss several important ways (DNA damage repair, oxidative stress, epigenetic modification and immune modulation) glutamine metabolism drives cancer growth and progression, and present data that inhibition of glutamine utilization can lead to radiosensitization in preclinical models. Future research is needed in the clinical realm to determine whether glutamine antagonism is a feasible synergistic therapy that can be combined with radiotherapy.
Collapse
Affiliation(s)
| | | | | | - Brittany A. Simone
- Radiation Oncology Department, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
7
|
Zhou R, Zhao D, Beeraka NM, Wang X, Lu P, Song R, Chen K, Liu J. Novel Implications of Nanoparticle-Enhanced Radiotherapy and Brachytherapy: Z-Effect and Tumor Hypoxia. Metabolites 2022; 12:943. [PMID: 36295845 PMCID: PMC9612299 DOI: 10.3390/metabo12100943] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 10/29/2023] Open
Abstract
Radiotherapy and internal radioisotope therapy (brachytherapy) induce tumor cell death through different molecular signaling pathways. However, these therapies in cancer patients are constrained by dose-related adverse effects and local discomfort due to the prolonged exposure to the surrounding tissues. Technological advancements in nanotechnology have resulted in synthesis of high atomic elements such as nanomaterials, which can be used as radiosensitizers due to their photoelectric characteristics. The aim of this review is to elucidate the effects of novel nanomaterials in the field of radiation oncology to ameliorate dose-related toxicity through the application of ideal nanoparticle-based radiosensitizers such as Au (gold), Bi (bismuth), and Lu (Lutetium-177) for enhancing cytotoxic effects of radiotherapy via the high-Z effect. In addition, we discuss the role of nanoparticle-enhanced radiotherapy in alleviating tumor hypoxia through the nanodelivery of genes/drugs and other functional anticancer molecules. The implications of engineered nanoparticles in preclinical and clinical studies still need to be studied in order to explore potential mechanisms for radiosensitization by minimizing tumor hypoxia, operational/logistic complications and by overcoming tumor heterogeneity in radiotherapy/brachytherapy.
Collapse
Affiliation(s)
- Runze Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Di Zhao
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Narasimha M. Beeraka
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- Department of Pharmaceutical Chemistry, Jagadguru Sri Shivarathreeswara Academy of Higher Education and Research (JSS AHER), Jagadguru Sri Shivarathreeswara College of Pharmacy, Mysuru 570015, India
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
| | - Xiaoyan Wang
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Pengwei Lu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Ruixia Song
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Kuo Chen
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
8
|
Liu CH, Peng CM, Hwang JI, Liang PC, Chen PJ, Abi-Jaoudeh N, Giiang LH, Tyan YS. Phase I Dose-Escalation Study of Tirapazamine Chemoembolization for Unresectable Early- and Intermediate-Stage Hepatocellular Carcinoma. J Vasc Interv Radiol 2022; 33:926-933.e1. [PMID: 35504436 DOI: 10.1016/j.jvir.2022.04.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 04/03/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
PURPOSE To investigate the safety of replacing doxorubicin with tirapazamine in conventional transarterial chemoembolization (TACE) in an Asian population with hepatocellular carcinoma (HCC), and to determine the optimal tirapazamine dose for phase II studies. MATERIALS AND METHODS This was a phase I, 3 + 3 dose-escalation study for patients with unresectable early- and intermediate-stage HCC who received 5, 10, or 20 mg/m2 of intra-arterial (IA) tirapazamine followed by ethiodized oil/gelatin sponge-based embolization. Key eligibilities included HCCs no more than 10 cm in diameter, prior embolization allowed, Eastern Cooperative Oncology Group performance status of 0 or 1, Child-Pugh score of 5-7, and platelet count of ≥60,000 μL. Dose-limiting toxicity (DLT) was defined as any grade 3 nonhematological or grade 4 hematological toxicity, with the exception of transient elevation of aminotransferase levels after the procedure. RESULTS Seventeen patients were enrolled, 59% of whom had progression from a prior HCC therapy and 35% of whom had progression or recurrence after TACE. All patients tolerated the tirapazamine TACE well without any DLT or serious adverse event. Using the modified Response Evaluation Criteria in Solid Tumors, the complete response (CR) rate was 47%, and the CR + partial response rate was 65%. The median duration of response was not reached. The median time to progression was 12.6 months (95% confidence interval, 5.1-not reached). The median overall survival was 29.3 months. The selected phase II dose was set at a fixed dose of 35 mg of IA tirapazamine. CONCLUSIONS IA tirapazamine with transarterial embolization was well tolerated and showed promising efficacy signals in intermediate-stage HCC, justifying pursuit of a phase II study.
Collapse
Affiliation(s)
- Chang-Hsien Liu
- Department of Medical Imaging, China Medical University Hsinchu Hospital and China Medical University, Hsinchu, Taiwan, Republic of China; Department of Radiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan, Republic of China; Institute of Nuclear Engineering and Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China.
| | - Cheng-Ming Peng
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Jen-I Hwang
- Department of Radiology, Taichung Veteran General Hospital, and Department of Medical Imaging, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan, Republic of China
| | - Po-Chin Liang
- Department of Medical Imaging, National Taiwan University Hospital Hsin-Chu Branch, and National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Pei-Jer Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, Republic of China
| | - Nadine Abi-Jaoudeh
- Department of Radiology, University of California, Irvine Medical Center, Orange, California, USA
| | - Lung-Hui Giiang
- Department of Radiology, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Yu-Shen Tyan
- Department of Radiology, Chung Shan Medical University Hospital, Taichung, Taiwan, Republic of China
| |
Collapse
|
9
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
10
|
Liu Z, Zhang Y, Shen N, Sun J, Tang Z, Chen X. Destruction of tumor vasculature by vascular disrupting agents in overcoming the limitation of EPR effect. Adv Drug Deliv Rev 2022; 183:114138. [PMID: 35143895 DOI: 10.1016/j.addr.2022.114138] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/27/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023]
Abstract
Nanomedicine greatly improves the efficiency in the delivery of antitumor drugs into the tumor, but insufficient tumoral penetration impairs the therapeutic efficacy of most nanomedicines. Vascular disrupting agent (VDA) nanomedicines are distributed around the tumor vessels due to the low tissue penetration in solid tumors, and the released drugs can selectively destroy immature tumor vessels and block the supply of oxygen and nutrients, leading to the internal necrosis of the tumors. VDAs can also improve the vascular permeability of the tumor, further increasing the extravasation of VDA nanomedicines in the tumor site, markedly reducing the dependence of nanomedicines on the enhanced permeability and retention effect (EPR effect). This review highlights the progress of VDA nanomedicines in recent years and their application in cancer therapy. First, the mechanisms of different VDAs are introduced. Subsequently, different strategies of delivering VDAs are described. Finally, multiple combination strategies with VDA nanomedicines in cancer therapy are described in detail.
Collapse
|
11
|
Yuan Y, Tan L, Wang L, Zou D, Liu J, Lu X, Fu D, Wang G, Wang L, Wang Z. The Expression Pattern of Hypoxia-Related Genes Predicts the Prognosis and Mediates Drug Resistance in Colorectal Cancer. Front Cell Dev Biol 2022; 10:814621. [PMID: 35155430 PMCID: PMC8829070 DOI: 10.3389/fcell.2022.814621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths worldwide. However, due to the heterogeneity of CRC, the clinical therapy outcomes differ among patients. There is a need to identify predictive biomarkers to efficiently facilitate CRC treatment and prognosis. Methods: The expression profiles from Gene Expression Omnibus (GEO) database were used to identify cancer hallmarks associated with CRC outcomes. An accurate gene signature based on the prognosis related cancer hallmarks was further constructed. Results: Hypoxia was identified to be the primary factor that could influence CRC outcomes. Sixteen hypoxia-related genes were selected to construct a risk gene signature (HGS) associated with individuals’ prognosis, which was validated in three independent cohorts. Further, stromal and immune cells in tumor microenvironment (TME) were found to be associated with hypoxia. Finally, among the 16 hypoxia-related genes, six genes (DCBLD2, PLEC, S100A11, PLAT, PPAP2B and LAMC2) were identified as the most attributable ones to drug resistance. Conclusion: HGS can accurately predict CRC prognosis. The expression of the drug resistance-related genes is critical in CRC treatment decision-making.
Collapse
Affiliation(s)
- Ye Yuan
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lulu Tan
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danyi Zou
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Liu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohuan Lu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daan Fu
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guobin Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Guobin Wang, ; Lin Wang, ; Zheng Wang,
| | - Lin Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Guobin Wang, ; Lin Wang, ; Zheng Wang,
| | - Zheng Wang
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Guobin Wang, ; Lin Wang, ; Zheng Wang,
| |
Collapse
|
12
|
Spin Trapping Hydroxyl and Aryl Radicals of One-Electron Reduced Anticancer Benzotriazine 1,4-Dioxides. Molecules 2022; 27:molecules27030812. [PMID: 35164077 PMCID: PMC8840461 DOI: 10.3390/molecules27030812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Hypoxia in tumors results in resistance to both chemotherapy and radiotherapy treatments but affords an environment in which hypoxia-activated prodrugs (HAP) are activated upon bioreduction to release targeted cytotoxins. The benzotriazine 1,4-di-N-oxide (BTO) HAP, tirapazamine (TPZ, 1), has undergone extensive clinical evaluation in combination with radiotherapy to assist in the killing of hypoxic tumor cells. Although compound 1 did not gain approval for clinical use, it has spurred on the development of other BTOs, such as the 3-alkyl analogue, SN30000, 2. There is general agreement that the cytotoxin(s) from BTOs arise from the one-electron reduced form of the compounds. Identifying the cytotoxic radicals, and whether they play a role in the selective killing of hypoxic tumor cells, is important for continued development of the BTO class of anticancer prodrugs. In this study, nitrone spin-traps, combined with electron spin resonance, give evidence for the formation of aryl radicals from compounds 1, 2 and 3-phenyl analogues, compounds 3 and 4, which form carbon C-centered radicals. In addition, high concentrations of DEPMPO (5-(diethoxyphosphoryl)-5-methyl-1-pyrroline N-oxide) spin-trap the •OH radical. The combination of spin-traps with high concentrations of DMSO and methanol also give evidence for the involvement of strongly oxidizing radicals. The failure to spin-trap methyl radicals with PBN (N-tert-butylphenylnitrone) on the bioreduction of compound 2, in the presence of DMSO, implies that free •OH radicals are not released from the protonated radical anions of compound 2. The spin-trapping of •OH radicals by high concentrations of DEPMPO, and the radical species arising from DMSO and methanol give both direct and indirect evidence for the scavenging of •OH radicals that are involved in an intramolecular process. Hypoxia-selective cytotoxicity is not related to the formation of aryl radicals from the BTO compounds as they are associated with high aerobic cytotoxicity.
Collapse
|
13
|
Brender JR, Saida Y, Devasahayam N, Krishna MC, Kishimoto S. Hypoxia Imaging As a Guide for Hypoxia-Modulated and Hypoxia-Activated Therapy. Antioxid Redox Signal 2022; 36:144-159. [PMID: 34428981 PMCID: PMC8856011 DOI: 10.1089/ars.2021.0176] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Significance: Oxygen imaging techniques, which can probe the spatiotemporal heterogeneity of tumor oxygenation, could be of significant clinical utility in radiation treatment planning and in evaluating the effectiveness of hypoxia-activated prodrugs. To fulfill these goals, oxygen imaging techniques should be noninvasive, quantitative, and capable of serial imaging, as well as having sufficient temporal resolution to detect the dynamics of tumor oxygenation to distinguish regions of chronic and acute hypoxia. Recent Advances: No current technique meets all these requirements, although all have strengths in certain areas. The current status of positron emission tomography (PET)-based hypoxia imaging, oxygen-enhanced magnetic resonance imaging (MRI), 19F MRI, and electron paramagnetic resonance (EPR) oximetry are reviewed along with their strengths and weaknesses for planning hypoxia-guided, intensity-modulated radiation therapy and detecting treatment response for hypoxia-targeted prodrugs. Critical Issues: Spatial and temporal resolution emerges as a major concern for these areas along with specificity and quantitative response. Although multiple oxygen imaging techniques have reached the investigative stage, clinical trials to test the therapeutic effectiveness of hypoxia imaging have been limited. Future Directions: Imaging elements of the redox environment besides oxygen by EPR and hyperpolarized MRI may have a significant impact on our understanding of the basic biology of the reactive oxygen species response and may extend treatment possibilities.
Collapse
Affiliation(s)
- Jeffrey R. Brender
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Li M, Zhang Y, Ren X, Niu W, Yuan Q, Cao K, Zhang J, Gao X, Su D. Activatable fluorogenic probe for accurate imaging of ulcerative colitis hypoxia in vivo. Chem Commun (Camb) 2021; 58:819-822. [PMID: 34928281 DOI: 10.1039/d1cc06577g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A simple but efficient fluorogenic probe is reported for accurate imaging of ulcerative colitis via hypoxia detection. The hypoxia produced by ulcerative colitis can lead to the upregulation of nitroreductase (NTR). NB-NO2 provides a unique response to NTR, enabling accurate imaging of Dextran sulphate sodium (DSS)-induced ulcerative colitis in vivo.
Collapse
Affiliation(s)
- Mingrui Li
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Yong Zhang
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Xiaojun Ren
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Wenchao Niu
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Qing Yuan
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Kai Cao
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Jinchao Zhang
- College of Chemistry & Environmental Science, Chemical Biology Key Laboratory of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding, 071002, P. R. China
| | - Xueyun Gao
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| | - Dongdong Su
- Department of Chemistry and Biology, Faculty of Environment and Life Science, Beijing University of Technology, Beijing, 100124, P. R. China.
| |
Collapse
|
15
|
Restoring Tumour Selectivity of the Bioreductive Prodrug PR-104 by Developing an Analogue Resistant to Aerobic Metabolism by Human Aldo-Keto Reductase 1C3. Pharmaceuticals (Basel) 2021; 14:ph14121231. [PMID: 34959631 PMCID: PMC8707548 DOI: 10.3390/ph14121231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
PR-104 is a phosphate ester pre-prodrug that is converted in vivo to its cognate alcohol, PR-104A, a latent alkylator which forms potent cytotoxins upon bioreduction. Hypoxia selectivity results from one-electron nitro reduction of PR-104A, in which cytochrome P450 oxidoreductase (POR) plays an important role. However, PR-104A also undergoes ‘off-target’ two-electron reduction by human aldo-keto reductase 1C3 (AKR1C3), resulting in activation in oxygenated tissues. AKR1C3 expression in human myeloid progenitor cells probably accounts for the dose-limiting myelotoxicity of PR-104 documented in clinical trials, resulting in human PR-104A plasma exposure levels 3.4- to 9.6-fold lower than can be achieved in murine models. Structure-based design to eliminate AKR1C3 activation thus represents a strategy for restoring the therapeutic window of this class of agent in humans. Here, we identified SN29176, a PR-104A analogue resistant to human AKR1C3 activation. SN29176 retains hypoxia selectivity in vitro with aerobic/hypoxic IC50 ratios of 9 to 145, remains a substrate for POR and triggers γH2AX induction and cell cycle arrest in a comparable manner to PR-104A. SN35141, the soluble phosphate pre-prodrug of SN29176, exhibited superior hypoxic tumour log cell kill (>4.0) to PR-104 (2.5–3.7) in vivo at doses predicted to be achievable in humans. Orthologues of human AKR1C3 from mouse, rat and dog were incapable of reducing PR-104A, thus identifying an underlying cause for the discrepancy in PR-104 tolerance in pre-clinical models versus humans. In contrast, the macaque AKR1C3 gene orthologue was able to metabolise PR-104A, indicating that this species may be suitable for evaluating the toxicokinetics of PR-104 analogues for clinical development. We confirmed that SN29176 was not a substrate for AKR1C3 orthologues across all four pre-clinical species, demonstrating that this prodrug analogue class is suitable for further development. Based on these findings, a prodrug candidate was subsequently identified for clinical trials.
Collapse
|
16
|
Datta A, West C, O'Connor JPB, Choudhury A, Hoskin P. Impact of hypoxia on cervical cancer outcomes. Int J Gynecol Cancer 2021; 31:1459-1470. [PMID: 34593564 DOI: 10.1136/ijgc-2021-002806] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/14/2021] [Indexed: 01/22/2023] Open
Abstract
The annual global incidence of cervical cancer is approximately 604 000 cases/342 000 deaths, making it the fourth most common cancer in women. Cervical cancer is a major healthcare problem in low and middle income countries where 85% of new cases and deaths occur. Secondary prevention measures have reduced incidence and mortality in developed countries over the past 30 years, but cervical cancer remains a major cause of cancer deaths in women. For women who present with Fédération Internationale de Gynécologie et d'Obstétrique (FIGO 2018) stages IB3 or upwards, chemoradiation is the established treatment. Despite high rates of local control, overall survival is less than 50%, largely due to distant relapse. Reducing the health burden of cervical cancer requires greater individualization of treatment, identifying those at risk of relapse and progression for modified or intensified treatment. Hypoxia is a well known feature of solid tumors and an established therapeutic target. Low tumorous oxygenation increases the risk of local invasion, metastasis and treatment failure. While meta-analyses show benefit, many individual trials targeting hypoxia failed in part due to not selecting patients most likely to benefit. This review summarizes the available hypoxia-targeted strategies and identifies further research and new treatment paradigms needed to improve patient outcomes. The applications and limitations of hypoxia biomarkers for treatment selection and response monitoring are discussed. Finally, areas of greatest unmet clinical need are identified to measure and target hypoxia and therefore improve cervical cancer outcomes.
Collapse
Affiliation(s)
- Anubhav Datta
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Radiology, The Christie NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
| | - James P B O'Connor
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, The Christie Hospital NHS Trust, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, The University of Manchester Faculty of Biology Medicine and Health, Manchester, UK
- Clinical Oncology, Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| |
Collapse
|
17
|
Hypoxic Jumbo Spheroids On-A-Chip (HOnAChip): Insights into Treatment Efficacy. Cancers (Basel) 2021; 13:cancers13164046. [PMID: 34439199 PMCID: PMC8394550 DOI: 10.3390/cancers13164046] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/25/2022] Open
Abstract
Hypoxia is a key characteristic of the tumor microenvironment, too rarely considered during drug development due to the lack of a user-friendly method to culture naturally hypoxic 3D tumor models. In this study, we used soft lithography to engineer a microfluidic platform allowing the culture of up to 240 naturally hypoxic tumor spheroids within an 80 mm by 82.5 mm chip. These jumbo spheroids on a chip are the largest to date (>750 µm), and express gold-standard hypoxic protein CAIX at their core only, a feature absent from smaller spheroids of the same cell lines. Using histopathology, we investigated response to combined radiotherapy (RT) and hypoxic prodrug Tirapazamine (TPZ) on our jumbo spheroids produced using two sarcoma cell lines (STS117 and SK-LMS-1). Our results demonstrate that TPZ preferentially targets the hypoxic core (STS117: p = 0.0009; SK-LMS-1: p = 0.0038), but the spheroids' hypoxic core harbored as much DNA damage 24 h after irradiation as normoxic spheroid cells. These results validate our microfluidic device and jumbo spheroids as potent fundamental and pre-clinical tools for the study of hypoxia and its effects on treatment response.
Collapse
|
18
|
Romero J, Maihom T, Limão-Vieira P, Probst M. Electronic structure and reactivity of tirapazamine as a radiosensitizer. J Mol Model 2021; 27:177. [PMID: 34021836 PMCID: PMC8140980 DOI: 10.1007/s00894-021-04771-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/21/2021] [Indexed: 11/30/2022]
Abstract
Tirapazamine (TP) has been shown to enhance the cytotoxic effects of ionizing radiation in hypoxic cells, thus making it a candidate for a radiosensitizer. This selective behavior is often directly linked to the abundance of O2. In this paper, we study the electronic properties of TP in vacuum, micro-hydrated from one up to three molecules of water and embedded in a continuum of water. We discuss electron affinities, charge distribution, and bond dissociation energies of TP, and find that these properties do not change significantly upon hydration. In agreement with its large electron affinity, and bond breaking triggered by electron attachment requires energies higher than 2.5 eV, ruling out the direct formation of bioactive TP radicals. Our results suggest, therefore, that the selective behavior of TP cannot be explained by a one-electron reduction from a neighboring O2 molecule. Alternatively, we propose that TP's hypoxic selectivity could be a consequence of O2 scavenging hydrogen radicals.
Collapse
Affiliation(s)
- José Romero
- Institute of Ion Physics and Applied Physics, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
- Atomic and Molecular Collisions Laboratory, CEFITEC, Department of Physics, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| | - Thana Maihom
- School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology, Rayong, 21210, Thailand
| | - Paulo Limão-Vieira
- Atomic and Molecular Collisions Laboratory, CEFITEC, Department of Physics, Universidade NOVA de Lisboa, 2829-516, Caparica, Portugal.
| | - Michael Probst
- Institute of Ion Physics and Applied Physics, University of Innsbruck, Technikerstraße 25, 6020, Innsbruck, Austria.
- School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology, Rayong, 21210, Thailand.
| |
Collapse
|
19
|
Kang D, Cheung ST, Wong-Rolle A, Kim J. Enamine N-Oxides: Synthesis and Application to Hypoxia-Responsive Prodrugs and Imaging Agents. ACS CENTRAL SCIENCE 2021; 7:631-640. [PMID: 34056093 PMCID: PMC8155465 DOI: 10.1021/acscentsci.0c01586] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 05/10/2023]
Abstract
Tumor hypoxia induces the large-scale adaptive reprogramming of cancer cells, promoting their transformation into highly invasive and metastatic species that lead to highly negative prognoses for cancer patients. We describe the synthesis and application of a hypoxia-responsive trigger derived from previously inaccessible enamine N-oxide structures. Hypoxia-dependent reduction of this motif by hemeproteins results in the concomitant activation of a caged molecule and a latent electrophile. We exploit the former in a hypoxia-activated prodrug application using a caged staurosporine molecule as a proof-of-principle. We demonstrate the latter in in vivo tumor labeling applications with enamine-N-oxide-modified near-infrared probes. Hypoxia-activated prodrug development has long been complicated by the heterogeneity of tumor hypoxia in patients. The dual drug release and imaging modalities of the highly versatile enamine N-oxide motif present an attractive opportunity for theranostic development that can address the need not only for new therapeutics but paired methods for patient stratification.
Collapse
|
20
|
A rat toxicological study of intra-arterial injection of Tirapazamine, a hypoxia-activating Cancer therapeutic agent, followed by hepatic artery ligation. Invest New Drugs 2021; 39:747-755. [PMID: 33428079 DOI: 10.1007/s10637-020-01057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/20/2020] [Indexed: 11/27/2022]
Abstract
Background Tirapazamine's (TPZ) tolerability after an intra-arterial (IA) injection remains unclear. We investigated TPZ's safety and tolerability in rats by first injecting into the left hepatic artery and then performing a hepatic artery ligation, which recapitulates the transarterial embolization used clinically. Research design and methods: Forty-six rats in five groups were respectively injected with 0, 0.25, 0.50, 1.0, or more than 1.5 mL IA of TPZ (0.7 mg/mL) into the left hepatic artery and then subjected to hepatic artery ligation under laparotomy. Blood samples were collected four times daily up to day 15 after which the rats were euthanized and necropsied. The toxicity profile of IA injection of TPZ followed by hepatic artery ligation was then assessed. Results No significant changes to the rats' body weight and serum total bilirubin were observed. Serum alanine aminotransferase (ALT) levels increased slightly but remained below 100 U/L one day after treatment for most rats. Three rats in Groups 3 and 4 exhibited an over two-fold transient elevation of ALT. All ALT recovered to the baseline at day 14. Liver tissues were collected on day 15 using H&E staining. One rat in Group 3 showed ischemic coagulative necrosis in its liver tissue. Other sporadic pathological changes not related to TPZ doses were observed in Groups 2, 3, 4, and 5. Conclusion TPZ by IA injection followed by embolization is tolerated up to 7 mg/kg. This finding supports the strategy of administering an IA injection of TPZ followed by trans-arterial embolization to the liver.
Collapse
|
21
|
Arthur‐Baidoo E, Ameixa J, Ziegler P, Ferreira da Silva F, Ončák M, Denifl S. Reaktionen in Tirapazamin induziert durch die Anlagerung von niederenergetischen Elektronen: Dissoziation versus Roaming von OH. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Eugene Arthur‐Baidoo
- Institut für Ionenphysik und Angewandte Physik Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
- Center for Biomolecular Sciences Innsbruck (CMBI) Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
| | - João Ameixa
- Institut für Ionenphysik und Angewandte Physik Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
- Center for Biomolecular Sciences Innsbruck (CMBI) Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
- Atomic and Molecular Collisions Laboratory Department of Physics CEFITEC Universidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Patrick Ziegler
- Institut für Ionenphysik und Angewandte Physik Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
- Center for Biomolecular Sciences Innsbruck (CMBI) Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
| | - Filipe Ferreira da Silva
- Atomic and Molecular Collisions Laboratory Department of Physics CEFITEC Universidade NOVA de Lisboa 2829-516 Caparica Portugal
| | - Milan Ončák
- Institut für Ionenphysik und Angewandte Physik Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
| | - Stephan Denifl
- Institut für Ionenphysik und Angewandte Physik Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
- Center for Biomolecular Sciences Innsbruck (CMBI) Leopold-Franzens-Universität Innsbruck Technikerstrasse 25 A-6020 Innsbruck Österreich
| |
Collapse
|
22
|
Arthur‐Baidoo E, Ameixa J, Ziegler P, Ferreira da Silva F, Ončák M, Denifl S. Reactions in Tirapazamine Induced by the Attachment of Low-Energy Electrons: Dissociation Versus Roaming of OH. Angew Chem Int Ed Engl 2020; 59:17177-17181. [PMID: 32543771 PMCID: PMC7540495 DOI: 10.1002/anie.202006675] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/09/2020] [Indexed: 11/27/2022]
Abstract
Tirapazamine (TPZ) has been tested in clinical trials on radio-chemotherapy due to its potential highly selective toxicity towards hypoxic tumor cells. It was suggested that either the hydroxyl radical or benzotriazinyl radical may form as bioactive radical after the initial reduction of TPZ in solution. In the present work, we studied low-energy electron attachment to TPZ in the gas phase and investigated the decomposition of the formed TPZ- anion by mass spectrometry. We observed the formation of the (TPZ-OH)- anion accompanied by the dissociation of the hydroxyl radical as by far the most abundant reaction pathway upon attachment of a low-energy electron. Quantum chemical calculations suggest that NH2 pyramidalization is the key reaction coordinate for the reaction dynamics upon electron attachment. We propose an OH roaming mechanism for other reaction channels observed, in competition with the OH dissociation.
Collapse
Affiliation(s)
- Eugene Arthur‐Baidoo
- Institut für Ionenphysik und Angewandte PhysikLeopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
- Center for Biomolecular Sciences Innsbruck (CMBI)Leopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
| | - João Ameixa
- Institut für Ionenphysik und Angewandte PhysikLeopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
- Center for Biomolecular Sciences Innsbruck (CMBI)Leopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
- Atomic and Molecular Collisions LaboratoryDepartment of PhysicsCEFITECUniversidade NOVA de Lisboa2829-516CaparicaPortugal
| | - Patrick Ziegler
- Institut für Ionenphysik und Angewandte PhysikLeopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
- Center for Biomolecular Sciences Innsbruck (CMBI)Leopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
| | - Filipe Ferreira da Silva
- Atomic and Molecular Collisions LaboratoryDepartment of PhysicsCEFITECUniversidade NOVA de Lisboa2829-516CaparicaPortugal
| | - Milan Ončák
- Institut für Ionenphysik und Angewandte PhysikLeopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
| | - Stephan Denifl
- Institut für Ionenphysik und Angewandte PhysikLeopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
- Center for Biomolecular Sciences Innsbruck (CMBI)Leopold-Franzens-Universität InnsbruckTechnikerstrasse 25A-6020InnsbruckAustria
| |
Collapse
|
23
|
Editor's Pick: Tumour-Associated Hypoxia: Can We Give Chimeric Antigen Receptor T Cells More Breathing Space? EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR)-engineered T cells has encountered important limitations in the transition of their use from liquid to solid tumours. Success is dependent upon T-cell trafficking to, and functional persistence within, tumours that often present a metabolically and immunologically hostile microenvironment. Moreover, CAR targets that are tumour specific are extremely scarce. To address these issues, several strategies have been proposed to improve both tumour selectivity and safety. One approach involves the engineering of CAR-T cells that only deploy their effector function at tumour sites. Conceptually, a solution for this exploits the oxygen-limited nature of advanced tumour deposits through the engineering of CAR that are exclusively expressed or activated under conditions of profound hypoxia. T cells have a complex inter-relationship with oxygen, which also needs to be factored into the refinement of these technologies. Ideally, oxygen-sensing CAR should only function when oxygen tension is below 2%, as is commonly the case in solid tumours but rare in healthy tissue. Successful advancement of such technologies presents opportunities for solid tumour immunotherapy because it should broaden the target repertoire that may safely be exploited in this context.
Collapse
|
24
|
Wang M, Chang M, Chen Q, Wang D, Li C, Hou Z, Lin J, Jin D, Xing B. Au 2Pt-PEG-Ce6 nanoformulation with dual nanozyme activities for synergistic chemodynamic therapy / phototherapy. Biomaterials 2020; 252:120093. [PMID: 32422490 DOI: 10.1016/j.biomaterials.2020.120093] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
Although synergistic therapy for tumors has displayed significant promise for effective treatment of cancer, developing a simple and effective strategy to build a multi-functional nanoplatform is still a huge challenge. By virtue of the characteristics of tumor microenvironment, such as hypoxia, slight acidity and H2O2 overexpression, Au2Pt-PEG-Ce6 nanoformulation is constructed for collaborative chemodynamic/phototherapy of tumors. Specifically, the Au2Pt nanozymes with multiple functions are synthesized in one step at room temperature. The photosensitizer chlorin e6 (Ce6) is covalently linked to Au2Pt nanozymes for photodynamic therapy (PDT). Interestingly, the Au2Pt nanozymes possess catalase- and peroxidase-like activities simultaneously, which not only can generate O2 for relaxation of tumor hypoxia and enhancement of PDT efficiency but also can produce ∙OH for chemodynamic therapy (CDT). In addition, the high photothermal conversion efficiency (η = 31.5%) of Au2Pt-PEG-Ce6 nanoformulation provides the possibility for photoacoustic (PA) and photothermal (PT) imaging guided photothermal therapy (PTT). Moreover, the presence of high-Z elements (Au and Pt) in Au2Pt-PEG-Ce6 nanoformulation endows it with the ability to act as an X-ray computed tomography (CT) imaging contrast agent. All in all, the Au2Pt-PEG-Ce6 exhibits great potential in multimodal imaging-guided synergistic PTT/PDT/CDT with remarkably tumor specificity and enhanced therapy.
Collapse
Affiliation(s)
- Man Wang
- Institute of Frontier and Interdisciplinarity Science and Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, PR China; Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Mengyu Chang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Qing Chen
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China
| | - Chunxia Li
- Institute of Frontier and Interdisciplinarity Science and Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, PR China; Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, Zhejiang, PR China.
| | - Zhiyao Hou
- Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou, Guangdong, 511436, PR China.
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Dayong Jin
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW, 2007, Australia
| | - Bengang Xing
- School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
25
|
Ni K, Lan G, Song Y, Hao Z, Lin W. Biomimetic nanoscale metal-organic framework harnesses hypoxia for effective cancer radiotherapy and immunotherapy. Chem Sci 2020; 11:7641-7653. [PMID: 34094142 PMCID: PMC8159451 DOI: 10.1039/d0sc01949f] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tumor hypoxia presents a major impediment to effective cancer therapy with ionizing radiation and immune checkpoint inhibitors. Here we report the design of a biomimetic nanoscale metal–organic-framework (nMOF), Hf-DBP-Fe, with catalase-like activity to decompose elevated levels of H2O2 in hypoxic tumors to generate oxygen and hydroxyl radical. The generated oxygen attenuates hypoxia to enable radiodynamic therapy upon X-ray irradiation and fixes DNA damage while hydroxyl radical inflicts direct damage to tumor cells to afford chemodynamic therapy. Hf-DBP-Fe thus mediates effective local therapy of hypoxic cancer with low-dose X-ray irradiation, leading to highly immunogenic tumor microenvironments for synergistic combination with anti-PD-L1 immune checkpoint blockade. This combination treatment not only eradicates primary tumors but also rejects distant tumors through systemic anti-tumor immunity. We have thus advanced an nMOF-based strategy to harness hypoxic tumor microenvironments for highly effective cancer therapy using a synergistic combination of low dose radiation and immune checkpoint blockade. Biomimetic Hf-DBP-Fe harnesses tumor hypoxia for cancer treatment via RT-RDT and CDT as well as synergistic combination with immune checkpoint blockade.![]()
Collapse
Affiliation(s)
- Kaiyuan Ni
- Department of Chemistry, The University of Chicago Chicago IL 60637 USA
| | - Guangxu Lan
- Department of Chemistry, The University of Chicago Chicago IL 60637 USA
| | - Yang Song
- Department of Chemistry, The University of Chicago Chicago IL 60637 USA
| | - Ziyang Hao
- Department of Chemistry, The University of Chicago Chicago IL 60637 USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago Chicago IL 60637 USA .,Department of Radiation and Cellular Oncology, Ludwig Center for Metastasis Research, The University of Chicago Chicago IL 60637 USA
| |
Collapse
|
26
|
Silva VL, Kaassis A, Dehsorkhi A, Koffi CR, Severic M, Abdelhamid M, Nyimanu D, Morris CJ, Al-Jamal WT. Enhanced selectivity, cellular uptake, and in vitro activity of an intrinsically fluorescent copper-tirapazamine nanocomplex for hypoxia targeted therapy in prostate cancer. Biomater Sci 2020; 8:2420-2433. [PMID: 32236169 DOI: 10.1039/c9bm01905g] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In the present work, a copper-tirapazamine (TPZ) nanocomplex [Cu(TPZ)2] was synthesized for selective hypoxia-targeted therapy. The nanocomplex revealed a crystalline form, and exhibited higher lipophilicity, compared to TPZ. Furthermore, its stability was confirmed in different media, with minimum dissociation in serum (∼20% up to 72 h). In contrast to other hypoxia-targeted agents, our intrinsically fluorescent nanocomplex offered an invaluable tool to monitor its cellular uptake and intracellular distribution under both normoxia and hypoxia. The conferred higher cellular uptake of the nanocomplex, especially under hypoxia, and its biocompatible reductive potential resulted in superior hypoxia selectivity in two prostate cancer (PC) cell lines. More promisingly, the nanocomplex showed higher potency in three-dimensional tumor spheroids, compared to TPZ, due to its slower metabolism, and probably deeper penetration in tumor spheroids. Interestingly, the nuclear localization of the intact nanocomplex, combined with its higher DNA binding affinity, as evidenced by the DNA binding assay, resulted in significant S-phase cell-cycle arrest, followed by apoptosis in the three-dimensional spheroid model. In conclusion, the presented findings suggested that the Cu(TPZ)2 nanocomplex can be a promising hypoxia-targeted therapeutic, which could potentiate the efficacy of the existing chemo- and radiotherapy in PC.
Collapse
Affiliation(s)
- Vera L Silva
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Song C, Xu W, Wei Z, Ou C, Wu J, Tong J, Cai Y, Dong X, Han W. Anti-LDLR modified TPZ@Ce6-PEG complexes for tumor hypoxia-targeting chemo-/radio-/photodynamic/photothermal therapy. J Mater Chem B 2020; 8:648-654. [PMID: 31898718 DOI: 10.1039/c9tb02248a] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hypoxia, a state of low oxygen tension in solid tumors, is not only closely correlated with resistance to both radiotherapy and chemotherapy, but also associated with poor prognosis of tumors and regional lymph node status. Herein, based on the analysis of cell samples from tumor patients, low-density lipoprotein receptor (LDLR) was found to be overexpressed on the surface of hypoxic tumor cell membranes, and confirmed to be an effective hypoxia marker through specific binding with anti-LDLR antibody in solid tumors. In addition, using the special therapeutic microenvironment of hypoxia, tirapazamine (TPZ, which can be used as both a hypoxia-activated chemotherapy prodrug and radiotherapy sensitizer) was integrated with PEGylated photosensitizer chlorin e6 (Ce6-PEG) by self-assembly, and anti-LDLR was then modified on the surface to form tumor hypoxia-targeting multifunctional nanoparticles (CPTA). CPTA possesses a multimodal antitumor effect via a simultaneous photothermal therapy (PTT)/photodynamic therapy (PDT) effect generated by Ce6, and chemotherapy/radiotherapy actions sensitized by TPZ. It is noteworthy that tumor oxygen was consumed in the process of PDT and the hypoxia was subsequently exacerbated, which can greatly increase the TPZ-sensitized chemotherapy and lead to a synergistic antitumor effect. Both in vitro and in vivo experiments demonstrated that CPTA possesses an excellent therapeutic effect through PTT, PDT, and TPZ sensitized radiotherapy and chemotherapy. This hypoxic tumor targeting synergetic therapeutic strategy has great potential for future clinical transformation.
Collapse
Affiliation(s)
- Chuanhui Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China. and Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China.
| | - Wenguang Xu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China. and Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China.
| | - Zheng Wei
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China. and Pediatric Dentistry, Nanjing Stomatology Hospital, Medical school of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China
| | - Changjin Ou
- School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, China.
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Jinlong Tong
- Department of Radiotherapy, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, No. 1 Zhongfu Road, Nanjing, 210003, China
| | - Yu Cai
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China.
| | - Xiaochen Dong
- School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing 210044, China.
| | - Wei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, No. 30 Zhongyang Road, Nanjing, 210008, China.
| |
Collapse
|
28
|
Evans MA, Shields CW, Krishnan V, Wang LL, Zhao Z, Ukidve A, Lewandowski M, Gao Y, Mitragotri S. Macrophage‐Mediated Delivery of Hypoxia‐Activated Prodrug Nanoparticles. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Michael A. Evans
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - C. Wyatt Shields
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Vinu Krishnan
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Lily Li‐Wen Wang
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of Technology Cambridge MA 02139 USA
| | - Zhongmin Zhao
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Anvay Ukidve
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Michael Lewandowski
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Yongsheng Gao
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| |
Collapse
|
29
|
Wang M, Wang D, Chen Q, Li C, Li Z, Lin J. Recent Advances in Glucose-Oxidase-Based Nanocomposites for Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1903895. [PMID: 31747128 DOI: 10.1002/smll.201903895] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/18/2019] [Indexed: 06/10/2023]
Abstract
Glucose oxidase (GOx) can react with intracellular glucose and oxygen (O2 ) to produce hydrogen peroxide (H2 O2 ) and gluconic acid, which can cut off the nutrition source of cancer cells and consequently inhibit their proliferation. Therefore, GOx is recognised as an ideal endogenous oxido-reductase for cancer starvation therapy. This process can further regulate the tumor microenvironment by increasing the hypoxia and the acidity. Thus, GOx offers new possibilities for the elaborate design of multifunctional nanocomposites for tumor therapy. However, natural GOx is expensive to prepare and purify and exhibits immunogenicity, short in vivo half-life, and systemic toxicity. Furthermore, GOx is highly prone to degrade after exposure to biological conditions. These intrinsic shortcomings will undoubtedly limit its biomedical applications. Accordingly, some nanocarriers can be used to protect GOx from the surrounding environment, thus controlling or preserving the activity. A variety of nanocarriers including hollow mesoporous silica nanoparticles, metal-organic frameworks, organic polymers, and magnetic nanoparticles are summarized for the construction of GOx-based nanocomposites for multimodal synergistic cancer therapy. In addition, current challenges and promising developments in this area are highlighted.
Collapse
Affiliation(s)
- Man Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Dongmei Wang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Qing Chen
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Chunxia Li
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Zhengquan Li
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Zhejiang Normal University, Jinhua, 321004, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| |
Collapse
|
30
|
Perfluorocarbon regulates the intratumoural environment to enhance hypoxia-based agent efficacy. Nat Commun 2019; 10:1580. [PMID: 30952842 PMCID: PMC6450981 DOI: 10.1038/s41467-019-09389-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/06/2019] [Indexed: 12/18/2022] Open
Abstract
Hypoxia-based agents (HBAs), such as anaerobic bacteria and bioreductive prodrugs, require both a permeable and hypoxic intratumoural environment to be fully effective. To solve this problem, herein, we report that perfluorocarbon nanoparticles (PNPs) can be used to create a long-lasting, penetrable and hypoxic tumour microenvironment for ensuring both the delivery and activation of subsequently administered HBAs. In addition to the increased permeability and enhanced hypoxia caused by the PNPs, the PNPs can be retained to further achieve the long-term inhibition of intratumoural O2 reperfusion while enhancing HBA accumulation for over 24 h. Therefore, perfluorocarbon materials may have great potential for reigniting clinical research on hypoxia-based drugs. Hypoxia-based agents need permeable and hypoxic intratumour environment to be effective. Here, the authors show that perfluorocarbon nanoparticles promote increased permeability and sustained hypoxia to improve accumulation of hypoxia-based agents, and inhibit intratumour oxygen reperfusion.
Collapse
|
31
|
Marx S, Van Gysel M, Breuer A, Dal Maso T, Michiels C, Wouters J, Le Calvé B. Potentialization of anticancer agents by identification of new chemosensitizers active under hypoxia. Biochem Pharmacol 2019; 162:224-236. [DOI: 10.1016/j.bcp.2019.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022]
|
32
|
Chen C, Su W, Liu Y, Zhang J, Zuo C, Yao Z, Bu W. Artificial anaerobic cell dormancy for tumor gaseous microenvironment regulation therapy. Biomaterials 2019; 200:48-55. [DOI: 10.1016/j.biomaterials.2019.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/21/2019] [Accepted: 02/07/2019] [Indexed: 01/15/2023]
|
33
|
Conway JRW, Herrmann D, Evans TRJ, Morton JP, Timpson P. Combating pancreatic cancer with PI3K pathway inhibitors in the era of personalised medicine. Gut 2019; 68:742-758. [PMID: 30396902 PMCID: PMC6580874 DOI: 10.1136/gutjnl-2018-316822] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 10/02/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the most deadly solid tumours. This is due to a generally late-stage diagnosis of a primarily treatment-refractory disease. Several large-scale sequencing and mass spectrometry approaches have identified key drivers of this disease and in doing so highlighted the vast heterogeneity of lower frequency mutations that make clinical trials of targeted agents in unselected patients increasingly futile. There is a clear need for improved biomarkers to guide effective targeted therapies, with biomarker-driven clinical trials for personalised medicine becoming increasingly common in several cancers. Interestingly, many of the aberrant signalling pathways in PDAC rely on downstream signal transduction through the mitogen-activated protein kinase and phosphoinositide 3-kinase (PI3K) pathways, which has led to the development of several approaches to target these key regulators, primarily as combination therapies. The following review discusses the trend of PDAC therapy towards molecular subtyping for biomarker-driven personalised therapies, highlighting the key pathways under investigation and their relationship to the PI3K pathway.
Collapse
Affiliation(s)
- James RW Conway
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Division, Sydney, New South Wales, Australia
| | - David Herrmann
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Division, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - TR Jeffry Evans
- Cancer Department, Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jennifer P Morton
- Cancer Department, Cancer Research UK Beatson Institute, Glasgow, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Paul Timpson
- Garvan Institute of Medical Research & The Kinghorn Cancer Centre, Cancer Division, Sydney, New South Wales, Australia
- St Vincent’s Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
34
|
Elsaidi HR, Yang XH, Ahmadi F, Weinfeld M, Wiebe LI, Kumar P. Putative electron-affinic radiosensitizers and markers of hypoxic tissue: Synthesis and preliminary in vitro biological characterization of C3-amino-substituted benzotriazine dioxides (BTDOs). Eur J Med Chem 2019; 165:216-224. [PMID: 30684798 DOI: 10.1016/j.ejmech.2019.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 01/04/2019] [Accepted: 01/05/2019] [Indexed: 01/16/2023]
Abstract
INTRODUCTION The redox characteristics of 1,2,4-benzotriazine-1,4-dioxides (BTDOs) make them potential radiosensitizing agents for hypoxic cells in solid human cancers. Tirapazamine (TPZ) is the most clinically tested BTDO radiosensitizer, despite its toxicity at effective doses. To date, no BTDOs have been developed as diagnostic markers of tissue hypoxia. HYPOTHESIS TPZ analogues with appropriate reporting groups can act as potential radiosensitizers and hypoxia selective diagnostics. EXPERIMENTAL AND RESULTS 3-Chloro-1,2,4-benzotriazine 1-oxide was substituted at the C3 position to afford 3-(2-hydroxyethoxyethyl)-amino-1,2,4-benzotriazine-1-oxide, which was oxidized to 3-(2-hydroxyethoxyethyl)-amino-1,2,4-benzotriazine-1,4-dioxide (HO-EOE-TPZ) or converted to 3-(2-tosyloxyethoxyethyl)-amino-1,2,4-benzotriazine-1,4-dioxide (Tos-EOE-TPZ). Tos-EOE-TPZ was intended for use as a synthon for preparing 3-(2-azidoethoxyethyl)-amino-1,2,4-benzotriazine-1,4-dioxide (N3-EOE-TPZ) and 3-(2-iodoethoxyethyl)-amino-1,2,4-benzotriazine-1,4-dioxide (I-EOE-TPZ). The logP values (-0.69 to 0.61) for these molecules bracketed that of TPZ (-0.34). Cell line dependent cytotoxicities (IC50) in air were in the 10-100 μM range, with Hypoxia Cytotoxicity Ratios (HCR; IC50-air/IC50-hypoxia) of 5-10. LUMO calculations indicated that these molecules are in the optimal redox range for radiosensitization, offering cell-line-specific Relative Radiosensitization Ratios (RRSR; SER/OER) of 0.58-0.88, compared to TPZ (0.67-0.76). CONCLUSION The LUMO, IC50, HCR and RRSR values of 3-(2-substituted ethoxyethyl)-amino-1,2,4-benzotriazine-1,4-dioxides are similar to the corresponding values for TPZ, supporting the conclusion that these TPZ analogues are potentially useful as hypoxia-activated radiosensitizers. Further studies into their biodistributions in animal models are being pursued to determine the in vivo potential in hypoxia management.
Collapse
Affiliation(s)
- Hassan Rh Elsaidi
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue Edmonton, Alberta, T6G 1Z2, Canada
| | - Xiao-Hong Yang
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue Edmonton, Alberta, T6G 1Z2, Canada
| | - Fatemeh Ahmadi
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue Edmonton, Alberta, T6G 1Z2, Canada
| | - Michael Weinfeld
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue Edmonton, Alberta, T6G 1Z2, Canada
| | - Leonard I Wiebe
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue Edmonton, Alberta, T6G 1Z2, Canada; Joint Appointment to Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Piyush Kumar
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, 11560 University Avenue Edmonton, Alberta, T6G 1Z2, Canada.
| |
Collapse
|
35
|
Tang Z, Liu Y, He M, Bu W. Chemodynamic Therapy: Tumour Microenvironment‐Mediated Fenton and Fenton‐like Reactions. Angew Chem Int Ed Engl 2019; 58:946-956. [DOI: 10.1002/anie.201805664] [Citation(s) in RCA: 920] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/22/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Zhongmin Tang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P.R. China
- University of Chinese Academy of Sciences Beijing 100049 P.R. China
| | - Yanyan Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical ProcessesSchool of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 P.R. China
| | - Mingyuan He
- Shanghai Key Laboratory of Green Chemistry and Chemical ProcessesSchool of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 P.R. China
| | - Wenbo Bu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P.R. China
- Shanghai Key Laboratory of Green Chemistry and Chemical ProcessesSchool of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 P.R. China
| |
Collapse
|
36
|
Development of [ 131I]I-EOE-TPZ and [ 131I]I-EOE-TPZMO: Novel Tirapazamine (TPZ)-Based Radioiodinated Pharmaceuticals for Application in Theranostic Management of Hypoxia. Pharmaceuticals (Basel) 2019; 12:ph12010003. [PMID: 30609671 PMCID: PMC6469288 DOI: 10.3390/ph12010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/18/2018] [Accepted: 12/20/2018] [Indexed: 01/14/2023] Open
Abstract
Introduction: Benzotriazine-1,4-dioxides (BTDOs) such as tirapazamine (TPZ) and its derivatives act as radiosensitizers of hypoxic tissues. The benzotriazine-1-monoxide (BTMO) metabolite (SR 4317, TPZMO) of TPZ also has radiosensitizing properties, and via unknown mechanisms, is a potent enhancer of the radiosensitizing effects of TPZ. Unlike their 2-nitroimidazole radiosensitizer counterparts, radiolabeled benzotriazine oxides have not been used as radiopharmaceuticals for diagnostic imaging or molecular radiotherapy (MRT) of hypoxia. The radioiodination chemistry for preparing model radioiodinated BTDOs and BTMOs is now reported. Hypothesis: Radioiodinated 3-(2-iodoethoxyethyl)-amino-1,2,4-benzotriazine-1,4-dioxide (I-EOE-TPZ), a novel bioisosteric analogue of TPZ, and 3-(2-iodoethoxyethyl)-amino-1,2,4-benzotriazine-1-oxide (I-EOE-TPZMO), its monoxide analogue, are candidates for in vivo and in vitro investigations of biochemical mechanisms in pathologies that develop hypoxic microenvironments. In theory, both radiotracers can be prepared from the same precursors. Methods: Radioiodination procedures were based on classical nucleophilic [131I]iodide substitution on Tos-EOE-TPZ (P1) and by [131I]iodide exchange on I-EOE-TPZ (P2). Reaction parameters, including temperature, reaction time, solvent and the influence of pivalic acid on products’ formation and the corresponding radiochemical yields (RCY) were investigated. Results: The [131I]iodide labeling reactions invariably led to the synthesis of both products, but with careful manipulation of conditions the preferred product could be recovered as the major product. Radioiodide exchange on P2 in ACN at 80 ± 5 °C for 30 min afforded the highest RCY, 89%, of [131I]I-EOE-TPZ, which upon solid phase purification on an alumina cartridge gave 60% yield of the product with over 97% of radiochemical purity. Similarly, radioiodide exchange on P2 in ACN at 50 ± 5 °C for 30 min with pivalic acid afforded the highest yield, 92%, of [131I]I-EOE-TPZMO exclusively with no trace of [131I]I-EOE-TPZ. In both cases, extended reaction times and/or elevated temperatures resulted in the formation of at least two additional radioactive reaction products. Conclusions: Radioiodination of P1 and P2 with [131I]iodide leads to the facile formation of [131I]I-EOE-TPZMO. At 80 °C and short reaction times, the facile reduction of the N-4-oxide moiety was minimized to afford acceptable radiochemical yields of [131I]I-EOE-TPZ from either precursor. Regeneration of [131I]I-EOE-TPZ from [131I]I-EOE-TPZMO is impractical after reaction work-up.
Collapse
|
37
|
Xie J, Gong L, Zhu S, Yong Y, Gu Z, Zhao Y. Emerging Strategies of Nanomaterial-Mediated Tumor Radiosensitization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1802244. [PMID: 30156333 DOI: 10.1002/adma.201802244] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Indexed: 05/23/2023]
Abstract
Nano-radiosensitization has been a hot concept for the past ten years, and the nanomaterial-mediated tumor radiosensitization method is mainly focused on increasing intracellular radiation deposition by high atomic number (high Z) nanomaterials, particularly gold (Au)-mediated radiation enhancement. Recently, various new nanomaterial-mediated radiosensitive approaches have been successively reported, such as catalyzing reactive oxygen species (ROS) generation, consuming intracellular reduced glutathione (GSH), overcoming tumor hypoxia, and various synergistic radiotherapy ways. These strategies may open a new avenue for enhancing the radiotherapeutic effect and avoiding its side effects. Nevertheless, reviews systematically summarizing these newly emerging methods and their radiosensitive mechanisms are still rare. Therefore, the general strategies of nanomaterial-mediated tumor radiosensitization are comprehensively summarized, particularly aiming at introducing the emerging radiosensitive methods. The strategies are divided into three general parts. First, methods on account of the intrinsic radiosensitive properties of nanoradiosensitizers for radiosensitization are highlighted. Then, newly developed synergistic strategies based on multifunctional nanomaterials for enhancing radiotherapy efficacy are emphasized. Third, nanomaterial-mediated radioprotection approaches for increasing the radiotherapeutic ratio are discussed. Importantly, the clinical translation of nanomaterial-mediated tumor radiosensitization is also covered. Finally, further challenges and outlooks in this field are discussed.
Collapse
Affiliation(s)
- Jiani Xie
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Linji Gong
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Zhu
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuan Yong
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanjun Gu
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuliang Zhao
- Prof. Z. Gu, Prof. Y. Zhao, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Chinese Academy of Sciences, Beijing, 100190, China
| |
Collapse
|
38
|
Tang Z, Liu Y, He M, Bu W. Chemodynamic Therapy: Tumour Microenvironment‐Mediated Fenton and Fenton‐like Reactions. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201805664] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Zhongmin Tang
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P.R. China
- University of Chinese Academy of Sciences Beijing 100049 P.R. China
| | - Yanyan Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical ProcessesSchool of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 P.R. China
| | - Mingyuan He
- Shanghai Key Laboratory of Green Chemistry and Chemical ProcessesSchool of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 P.R. China
| | - Wenbo Bu
- State Key Laboratory of High Performance Ceramics and Superfine MicrostructureShanghai Institute of CeramicsChinese Academy of Sciences Shanghai 200050 P.R. China
- Shanghai Key Laboratory of Green Chemistry and Chemical ProcessesSchool of Chemistry and Molecular EngineeringEast China Normal University Shanghai 200062 P.R. China
| |
Collapse
|
39
|
Nunes AS, Barros AS, Costa EC, Moreira AF, Correia IJ. 3D tumor spheroids as in vitro models to mimic in vivo human solid tumors resistance to therapeutic drugs. Biotechnol Bioeng 2018; 116:206-226. [DOI: 10.1002/bit.26845] [Citation(s) in RCA: 309] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/30/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Ana S. Nunes
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Andreia S. Barros
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Elisabete C. Costa
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - André F. Moreira
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
| | - Ilídio J. Correia
- Health Sciences Research Centre, Universidade da Beira Interior (CICS-UBI); Covilhã Portugal
- Departamento de Engenharia Química; Universidade de Coimbra, (CIEPQF); Coimbra Portugal
| |
Collapse
|
40
|
Xie H, Jiao Y, Fan Q, Hai M, Yang J, Hu Z, Yang Y, Shuai J, Chen G, Liu R, Liu L. Modeling three-dimensional invasive solid tumor growth in heterogeneous microenvironment under chemotherapy. PLoS One 2018; 13:e0206292. [PMID: 30365511 PMCID: PMC6203364 DOI: 10.1371/journal.pone.0206292] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 10/10/2018] [Indexed: 01/08/2023] Open
Abstract
A systematic understanding of the evolution and growth dynamics of invasive solid tumors in response to different chemotherapy strategies is crucial for the development of individually optimized oncotherapy. Here, we develop a hybrid three-dimensional (3D) computational model that integrates pharmacokinetic model, continuum diffusion-reaction model and discrete cell automaton model to investigate 3D invasive solid tumor growth in heterogeneous microenvironment under chemotherapy. Specifically, we consider the effects of heterogeneous environment on drug diffusion, tumor growth, invasion and the drug-tumor interaction on individual cell level. We employ the hybrid model to investigate the evolution and growth dynamics of avascular invasive solid tumors under different chemotherapy strategies. Our simulations indicate that constant dosing is generally more effective in suppressing primary tumor growth than periodic dosing, due to the resulting continuous high drug concentration. In highly heterogeneous microenvironment, the malignancy of the tumor is significantly enhanced, leading to inefficiency of chemotherapies. The effects of geometrically-confined microenvironment and non-uniform drug dosing are also investigated. Our computational model, when supplemented with sufficient clinical data, could eventually lead to the development of efficient in silico tools for prognosis and treatment strategy optimization.
Collapse
Affiliation(s)
- Hang Xie
- College of Physics, Chongqing University, Chongqing, China
| | - Yang Jiao
- Materials Science and Engineering, Arizona State University, Tempe, AZ, United States of America
| | - Qihui Fan
- Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Science, Beijing, China
| | - Miaomiao Hai
- College of Physics, Chongqing University, Chongqing, China
| | - Jiaen Yang
- College of Physics, Chongqing University, Chongqing, China
| | - Zhijian Hu
- College of Physics, Chongqing University, Chongqing, China
| | - Yue Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Haidian District, Beijing, China
| | - Jianwei Shuai
- Department of Physics, Xiamen University, Xiamen, China
| | - Guo Chen
- College of Physics, Chongqing University, Chongqing, China
| | - Ruchuan Liu
- College of Physics, Chongqing University, Chongqing, China
| | - Liyu Liu
- College of Physics, Chongqing University, Chongqing, China
| |
Collapse
|
41
|
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: 3.0] [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
|
42
|
Sahu A, Choi WI, Tae G. Recent Progress in the Design of Hypoxia-Specific Nano Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| | - Won Il Choi
- Center for Convergence Bioceramic Materials; Convergence R&D Division; Korea Institute of Ceramic Engineering and Technology; 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu Cheongju Chungbuk 28160 Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| |
Collapse
|
43
|
Zhu H, Wang DD, Yuan T, Yan FJ, Zeng CM, Dai XY, Chen ZB, Chen Y, Zhou T, Fan GH, Ying M, Cao J, Luo P, Liu XJ, Hu Y, Peng Y, He Q, Yang B. Multikinase Inhibitor CT-707 Targets Liver Cancer by Interrupting the Hypoxia-Activated IGF-1R-YAP Axis. Cancer Res 2018; 78:3995-4006. [PMID: 29669759 DOI: 10.1158/0008-5472.can-17-1548] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 01/22/2018] [Accepted: 04/12/2018] [Indexed: 11/16/2022]
Abstract
Given that Yes-associated protein (YAP) signaling acts as a critical survival input for hypoxic cancer cells in hepatocellular carcinoma (HCC), disruption of YAP function and the maintenance of hypoxia is an attractive way to treat HCC. Utilizing a cell-based YAP-TEAD luciferase reporter assay and functional analyses, we identified CT-707, a China-FDA approved multi-kinase inhibitor under clinical trial with remarkable inhibitory activity against YAP function. CT-707 exhibited prominent cytotoxicity under hypoxia on HCC cells, which was attributable to the inhibition of YAP signaling. CT-707 arrested tumor growth in HepG2, Bel-7402, and HCC patient-derived xenografts. Mechanistically, the inhibitory activity of CT-707 on YAP signaling was due to the interruption of hypoxia-activated IGF1R. Overall, these findings not only identify CT-707 as a promising hypoxia-targeting agent against HCC, but they also unveil IGF1R as a new modulator specifically regulating hypoxia-activated YAP signaling.Significance: CT-707 may represent a novel clinical approach for patients with HCC suffering poor drug response due to intratumor hypoxia. Cancer Res; 78(14); 3995-4006. ©2018 AACR.
Collapse
Affiliation(s)
- Hong Zhu
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Dan-Dan Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tao Yuan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fang-Jie Yan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chen-Ming Zeng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiao-Yang Dai
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
| | - Zi-Bo Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Tianyi Zhou
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Guang-Han Fan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meidan Ying
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ji Cao
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Peihua Luo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xi-Jie Liu
- Medicinal Chemistry Department, Centaurus BioPharma Co., Ltd., Beijing, China
| | - Yuandong Hu
- Medicinal Chemistry Department, Centaurus BioPharma Co., Ltd., Beijing, China
| | - Yong Peng
- Discovery Biology Department, Centaurus BioPharma Co., Ltd., Beijing, China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
44
|
Abstract
In 2012, cancer affected 14.1 million people worldwide and was responsible for 8.2 million deaths. The disease predominantly affects aged populations and is one of the leading causes of death in most western countries. In tumors, the aggressive growth of the neoplastic cell population and associated overexpression of pro-angiogenic factors lead to the development of disorganized blood vessel networks that are structurally and functionally different from normal vasculature. A disorganized labyrinth of vessels that are immature, tortuous and hyperpermeable typifies tumor vasculature. Functionally, the ability of the tumor vasculature to deliver nutrients and remove waste products is severely diminished. A critical consequence of the inadequate vascular networks in solid tumors is the development of regions of hypoxia [low oxygen tensions typically defined as oxygen tensions (pO2 values) < 10 mm Hg]. Tumor cells existing in such hypoxic environments have long been known to be resistant to anticancer therapy, display an aggressive phenotype, and promote tumor progression and dissemination. This review discusses the physiological basis of hypoxia, methods of detection, and strategies to overcome the resulting therapy resistance.
Collapse
Affiliation(s)
- Veronica S Hughes
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| | - Jennifer M Wiggins
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| | - Dietmar W Siemann
- 1 Department of Radiation Oncology, University of Florida, Cancer Genetic Research Complex , Gainesville, FL , USA
| |
Collapse
|
45
|
Shen X, Laber CH, Sarkar U, Galazzi F, Johnson KM, Mahieu NG, Hillebrand R, Fuchs-Knotts T, Barnes CL, Baker GA, Gates KS. Exploiting the Inherent Photophysical Properties of the Major Tirapazamine Metabolite in the Development of Profluorescent Substrates for Enzymes That Catalyze the Bioreductive Activation of Hypoxia-Selective Anticancer Prodrugs. J Org Chem 2018; 83:3126-3131. [DOI: 10.1021/acs.joc.7b03035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Fan W, Yung B, Huang P, Chen X. Nanotechnology for Multimodal Synergistic Cancer Therapy. Chem Rev 2017; 117:13566-13638. [DOI: 10.1021/acs.chemrev.7b00258] [Citation(s) in RCA: 1059] [Impact Index Per Article: 151.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Wenpei Fan
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
- Key
Laboratory of Optoelectronic Devices and Systems of Ministry of Education
and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Bryant Yung
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peng Huang
- Guangdong
Key Laboratory for Biomedical Measurements and Ultrasound Imaging,
School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Xiaoyuan Chen
- Laboratory
of Molecular Imaging and Nanomedicine, National Institute of Biomedical
Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
47
|
Shi Y, Oeh J, Hitz A, Hedehus M, Eastham-Anderson J, Peale FV, Hamilton P, O'Brien T, Sampath D, Carano RAD. Monitoring and Targeting Anti-VEGF Induced Hypoxia within the Viable Tumor by 19F-MRI and Multispectral Analysis. Neoplasia 2017; 19:950-959. [PMID: 28987998 PMCID: PMC5635323 DOI: 10.1016/j.neo.2017.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 01/21/2023] Open
Abstract
The effect of anti-angiogenic agents on tumor oxygenation has been in question for a number of years, where both increases and decreases in tumor pO2 have been observed. This dichotomy in results may be explained by the role of vessel normalization in the response of tumors to anti-angiogenic therapy, where anti-angiogenic therapies may initially improve both the structure and the function of tumor vessels, but more sustained or potent anti-angiogenic treatments will produce an anti-vascular response, producing a more hypoxic environment. The first goal of this study was to employ multispectral (MS) 19F–MRI to noninvasively quantify viable tumor pO2 and evaluate the ability of a high dose of an antibody to vascular endothelial growth factor (VEGF) to produce a strong and prolonged anti-vascular response that results in significant tumor hypoxia. The second goal of this study was to target the anti-VEGF induced hypoxic tumor micro-environment with an agent, tirapazamine (TPZ), which has been designed to target hypoxic regions of tumors. These goals have been successfully met, where an antibody that blocks both murine and human VEGF-A (B20.4.1.1) was found by MS 19F–MRI to produce a strong anti-vascular response and reduce viable tumor pO2 in an HM-7 xenograft model. TPZ was then employed to target the anti-VEGF-induced hypoxic region. The combination of anti-VEGF and TPZ strongly suppressed HM-7 tumor growth and was superior to control and both monotherapies. This study provides evidence that clinical trials combining anti-vascular agents with hypoxia-activated prodrugs should be considered to improved efficacy in cancer patients.
Collapse
Affiliation(s)
- Yunzhou Shi
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, CA
| | - Jason Oeh
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Anna Hitz
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Maj Hedehus
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, CA
| | | | - Franklin V Peale
- Department of Pathology, Genentech Inc., South San Francisco, CA
| | - Patricia Hamilton
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Thomas O'Brien
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Deepak Sampath
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA
| | - Richard A D Carano
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, CA.
| |
Collapse
|
48
|
Stornetta A, Zimmermann M, Cimino GD, Henderson PT, Sturla SJ. DNA Adducts from Anticancer Drugs as Candidate Predictive Markers for Precision Medicine. Chem Res Toxicol 2017; 30:388-409. [PMID: 27936622 PMCID: PMC5379252 DOI: 10.1021/acs.chemrestox.6b00380] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Indexed: 01/23/2023]
Abstract
Biomarker-driven drug selection plays a central role in cancer drug discovery and development, and in diagnostic strategies to improve the use of traditional chemotherapeutic drugs. DNA-modifying anticancer drugs are still used as first line medication, but drawbacks such as resistance and side effects remain an issue. Monitoring the formation and level of DNA modifications induced by anticancer drugs is a potential strategy for stratifying patients and predicting drug efficacy. In this perspective, preclinical and clinical data concerning the relationship between drug-induced DNA adducts and biological response for platinum drugs and combination therapies, nitrogen mustards and half-mustards, hypoxia-activated drugs, reductase-activated drugs, and minor groove binding agents are presented and discussed. Aspects including measurement strategies, identification of adducts, and biological factors that influence the predictive relationship between DNA modification and biological response are addressed. A positive correlation between DNA adduct levels and response was observed for the majority of the studies, demonstrating the high potential of using DNA adducts from anticancer drugs as mechanism-based biomarkers of susceptibility, especially as bioanalysis approaches with higher sensitivity and throughput emerge.
Collapse
Affiliation(s)
- Alessia Stornetta
- Department
of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| | - Maike Zimmermann
- Department
of Internal Medicine, Division of Hematology and Oncology and the
UC Davis Comprehensive Cancer Center, University
of California Davis, 4501 X Street, Sacramento, California 95655, United States
- Accelerated
Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, California 95618, United States
| | - George D. Cimino
- Accelerated
Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, California 95618, United States
| | - Paul T. Henderson
- Department
of Internal Medicine, Division of Hematology and Oncology and the
UC Davis Comprehensive Cancer Center, University
of California Davis, 4501 X Street, Sacramento, California 95655, United States
- Accelerated
Medical Diagnostics, Inc., 2121 Second Street, B101, Davis, California 95618, United States
| | - Shana J. Sturla
- Department
of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, 8092 Zurich, Switzerland
| |
Collapse
|
49
|
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: 17.0] [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
|
50
|
Hypoxia-activated cytotoxic agent tirapazamine enhances hepatic artery ligation-induced killing of liver tumor in HBx transgenic mice. Proc Natl Acad Sci U S A 2016; 113:11937-11942. [PMID: 27702890 DOI: 10.1073/pnas.1613466113] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Transarterial chemoembolization (TACE) is the main treatment for intermediate stage hepatocellular carcinoma (HCC) with Barcelona Clinic Liver Cancer classification because of its exclusive arterial blood supply. Although TACE achieves substantial necrosis of the tumor, complete tumor necrosis is uncommon, and the residual tumor generally rapidly recurs. We combined tirapazamine (TPZ), a hypoxia-activated cytotoxic agent, with hepatic artery ligation (HAL), which recapitulates transarterial embolization in mouse models, to enhance the efficacy of TACE. The effectiveness of this combination treatment was examined in HCC that spontaneously developed in hepatitis B virus X protein (HBx) transgenic mice. We proved that the tumor blood flow in this model was exclusively supplied by the hepatic artery, in contrast to conventional orthotopic HCC xenografts that receive both arterial and venous blood supplies. At levels below the threshold oxygen levels created by HAL, TPZ was activated and killed the hypoxic cells, but spared the normoxic cells. This combination treatment clearly limited the toxicity of TPZ to HCC, which caused the rapid and near-complete necrosis of HCC. In conclusion, the combination of TPZ and HAL showed a synergistic tumor killing activity that was specific for HCC in HBx transgenic mice. This preclinical study forms the basis for the ongoing clinical program for the TPZ-TACE regimen in HCC treatment.
Collapse
|