1
|
Fan P, Qiang H, Liu Z, Zhao Q, Wang Y, Liu T, Wang X, Chu T, Huang Y, Xu W, Qin S. Effective low-dose Anlotinib induces long-term tumor vascular normalization and improves anti-PD-1 therapy. Front Immunol 2022; 13:937924. [PMID: 35990640 PMCID: PMC9382125 DOI: 10.3389/fimmu.2022.937924] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022] Open
Abstract
Anlotinib is a new multitarget tyrosine kinase inhibitor for tumor angiogenesis, and its monotherapy exhibits a decent clinical efficacy. However, the process of combining Anlotinib and immune checkpoint therapy to achieve optimal antitumor effects while limiting side effects remains unclear. In this study, we found that effective low-dose Anlotinib was sufficient to inhibit tumor growth while reducing side effects compared with high doses. Effective low-dose Anlotinib treatments induced durable tumor vascular normalization and improved anti-PD-1 therapy in both short- and long-term treatment regimens. Mechanistically, the combination therapy increased the proportions of intratumoral CD4+ T, CD8+ T, and NK cells. Anlotinib-associated antitumor effects were independent of interferon γ; however, the combination therapy required CD8+ T cells to suppress tumor growth. Together, these results suggest that the combination of effective low-dose Anlotinib and PD-1 blockade induces durable antitumor effects with fewer side effects. Our findings indicate that antiangiogenic treatments combined with immune checkpoint therapy at an effective low-dose, rather than a tolerable high dose, would be more efficacious and safer.
Collapse
Affiliation(s)
- Peng Fan
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- Department of Experimental Medicine, University of Rome “Tor Vergata”, Rome, Italy
| | - Huiping Qiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenhua Liu
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Zhao
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Tingkun Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
| | - Xuan Wang
- Department of Immunology, Innovent Biologics, Inc., Suzhou, China
| | - Tianqing Chu
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhui Huang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou, China
- *Correspondence: Yuhui Huang, ; Wei Xu, ; Songbing Qin,
| | - Wei Xu
- Department of Immunology, Innovent Biologics, Inc., Suzhou, China
- *Correspondence: Yuhui Huang, ; Wei Xu, ; Songbing Qin,
| | - Songbing Qin
- Department of Radiotherapy, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Yuhui Huang, ; Wei Xu, ; Songbing Qin,
| |
Collapse
|
2
|
Seki T, Saida Y, Kishimoto S, Lee J, Otowa Y, Yamamoto K, Chandramouli GV, Devasahayam N, Mitchell JB, Krishna MC, Brender JR. PEGPH20, a PEGylated human hyaluronidase, induces radiosensitization by reoxygenation in pancreatic cancer xenografts. A molecular imaging study. Neoplasia 2022; 30:100793. [PMID: 35523073 PMCID: PMC9079680 DOI: 10.1016/j.neo.2022.100793] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/14/2022]
Abstract
PURPOSE PEGylated human hyaluronidase (PEGPH20) enzymatically depletes hyaluronan, an important component of the extracellular matrix, increasing the delivery of therapeutic molecules. Combinations of chemotherapy and PEGPH20, however, have been unsuccessful in Phase III clinical trials. We hypothesize that by increasing tumor oxygenation by improving vascular patency and perfusion, PEGPH20 will also act as a radiosensitization agent. EXPERIMENTAL DESIGN The effect of PEGPH20 on radiation treatment was analyzed with respect to tumor growth, survival time, p02, local blood volume, and the perfusion/permeability of blood vessels in a human pancreatic adenocarcinoma BxPC3 mouse model overexpressing hyaluronan synthase 3 (HAS3). RESULTS Mice overexpressing HAS3 developed fast growing, radiation resistant tumors that became rapidly more hypoxic as time progressed. Treatment with PEGPH20 increased survival times when used in combination with radiation therapy, significantly more than either radiation therapy or PEGPH20 alone. In mice that overexpressed HAS3, EPR imaging showed an increase in local pO2 that could be linked to increases in perfusion/permeability and local blood volume immediately after PEGPH20 treatment. Hyperpolarized [1-13C] pyruvate suggested PEGPH20 caused a metabolic shift towards decreased glycolytic flux. These effects were confined to the mice overexpressing HAS3 - no effect of PEGPH20 on survival, radiation treatment, or pO2 was seen in wild type BxPC3 tumors. CONCLUSIONS PEGPH20 may be useful for radiosensitization of pancreatic cancer but only in the subset of tumors with substantial hyaluronan accumulation. The response of the treatment may potentially be monitored by non-invasive imaging of the hemodynamic and metabolic changes in the tumor microenvironment.
Collapse
Affiliation(s)
- Tomohiro Seki
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States; Josai University, Faculty of Pharmaceutical Sciences, Sakado, Japan
| | - Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States; Department of Respiratory Medicine and Infectious Diseases, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - Jisook Lee
- Halozyme Therapeutics, San Diego, California, United States
| | - Yasunori Otowa
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - Gadisetti Vr Chandramouli
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States
| | - Jeffery R Brender
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, United States.
| |
Collapse
|
3
|
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
|
4
|
Kimura K, Iguchi N, Nakano H, Yasui H, Matsumoto S, Inanami O, Hirata H. Redox-Sensitive Mapping of a Mouse Tumor Model Using Sparse Projection Sampling of Electron Paramagnetic Resonance. Antioxid Redox Signal 2022; 36:57-69. [PMID: 33847172 PMCID: PMC8823265 DOI: 10.1089/ars.2021.0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/08/2021] [Accepted: 04/03/2021] [Indexed: 11/16/2022]
Abstract
Aims: This work aimed to establish an accelerated imaging system for redox-sensitive mapping in a mouse tumor model using electron paramagnetic resonance (EPR) and nitroxyl radicals. Results: Sparse sampling of EPR spectral projections was demonstrated for a solution phantom. The reconstructed three-dimensional (3D) images with filtered back-projection (FBP) and compressed sensing image reconstruction were quantitatively assessed for the solution phantom. Mouse xenograft models of a human-derived pancreatic ductal adenocarcinoma cell line, MIA PaCa-2, were also measured for redox-sensitive mapping with the sparse sampling technique. Innovation: A short-lifetime redox-sensitive nitroxyl radical (15N-labeled perdeuterated Tempone) could be measured to map the decay rates of the EPR signals for the mouse xenograft models. Acceleration of 3D EPR image acquisition broadened the choices of nitroxyl radical probes with various redox sensitivities to biological environments. Conclusion: Sparse sampling of EPR spectral projections accelerated image acquisition in the 3D redox-sensitive mapping of mouse tumor-bearing legs fourfold compared with conventional image acquisition with FBP. Antioxid. Redox Signal. 36, 57-69.
Collapse
Affiliation(s)
- Kota Kimura
- Division of Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Nami Iguchi
- Division of Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Hitomi Nakano
- Division of Bioengineering and Bioinformatics, Faculty of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Laboratory of Radiation Biology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shingo Matsumoto
- Division of Bioengineering and Bioinformatics, Faculty of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Osamu Inanami
- Laboratory of Radiation Biology, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Hirata
- Division of Bioengineering and Bioinformatics, Faculty of Information Science and Technology, Hokkaido University, Sapporo, Japan
| |
Collapse
|
5
|
Stewart NJ, Sato T, Takeda N, Hirata H, Matsumoto S. Hyperpolarized 13C Magnetic Resonance Imaging as a Tool for Imaging Tissue Redox State, Oxidative Stress, Inflammation, and Cellular Metabolism. Antioxid Redox Signal 2022; 36:81-94. [PMID: 34218688 PMCID: PMC8792501 DOI: 10.1089/ars.2021.0139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Magnetic resonance imaging (MRI) with hyperpolarized (HP) 13C-labeled redox-sensitive metabolic tracers can provide noninvasive functional imaging biomarkers, reflecting tissue redox state, oxidative stress, and inflammation, among others. The capability to use endogenous metabolites as 13C-enriched imaging tracers without structural modification makes HP 13C MRI a promising tool to evaluate redox state in patients with various diseases. Recent Advances: Recent studies have demonstrated the feasibility of in vivo metabolic imaging of 13C-labeled tracers polarized by parahydrogen-induced polarization techniques, which offer a cost-effective alternative to the more widely used dissolution dynamic nuclear polarization-based hyperpolarizers. Critical Issues: Although the fluxes of many metabolic pathways reflect the change in tissue redox state, they are not functionally specific. In the present review, we summarize recent challenges in the development of specific 13C metabolic tracers for biomarkers of redox state, including that for detecting reactive oxygen species. Future Directions: Applications of HP 13C metabolic MRI to evaluate redox state have only just begun to be investigated. The possibility to gain a comprehensive understanding of the correlations between tissue redox potential and metabolism under different pathological conditions by using HP 13C MRI is promoting its interest in the clinical arena, along with its noninvasive biomarkers to evaluate the extent of disease and treatment response.
Collapse
Affiliation(s)
- Neil J Stewart
- Division of Bioengineering & Bioinformatics, Graduate School of Information Science & Technology, Hokkaido University, Sapporo, Japan.,POLARIS, Imaging Sciences, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Tatsuyuki Sato
- Division of Cardiology and Metabolism Center for Molecular Medicine, Jichi Medical University, Shimotsuke-shi, Japan.,Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism Center for Molecular Medicine, Jichi Medical University, Shimotsuke-shi, Japan
| | - Hiroshi Hirata
- Division of Bioengineering & Bioinformatics, Graduate School of Information Science & Technology, Hokkaido University, Sapporo, Japan
| | - Shingo Matsumoto
- Division of Bioengineering & Bioinformatics, Graduate School of Information Science & Technology, Hokkaido University, Sapporo, Japan
| |
Collapse
|
6
|
Riggin CN, Rodriguez AB, Weiss SN, Raja HA, Chen M, Schultz SM, Sehgal CM, Soslowsky LJ. Modulation of vascular response after injury in the rat Achilles tendon alters healing capacity. J Orthop Res 2021; 39:2000-2016. [PMID: 32936495 PMCID: PMC7960560 DOI: 10.1002/jor.24861] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 02/04/2023]
Abstract
Tendons are relatively hypovascular but become hypervascular during both injury and degeneration. This is due to the angiogenic response, or the formation of new blood vessels, to tissue injury. The objective of this study was to evaluate the effect of vascular modulation in the rat Achilles tendons during healing. Fischer rats received a bilateral Achilles incisional injury followed by local injections of vascular endothelial growth factor (VEGF), anti-VEGF antibody (B20.4-1-1), or saline either early or late during the healing process. Vascular modulation and healing were evaluated using multiple in vivo ultrasound imaging modalities, in vivo functional assessment, and ex vivo measures of tendon compositional and mechanical properties. The late delivery of anti-VEGF antibody, B20, caused a temporary reduction in healing capacity during a time point where vascularity was also decreased, and then an improvement during a later time point where vascularity was increased relative to control. However, VEGF delivery had a minimal impact on healing and vascular changes in both early and late delivery times. This study was the first to evaluate vascular changes using both in vivo imaging methods and ex vivo histological methods, as well as functional and mechanical outcomes associated with these vascular changes. Clinical significance: this study demonstrates that the alteration of vascular response through the delivery of angiogenic growth factors has the ability to alter tendon healing properties.
Collapse
Affiliation(s)
- Corinne N Riggin
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Ashley B Rodriguez
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Stephanie N Weiss
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Harina A Raja
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Mengcun Chen
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104
| | - Susan M Schultz
- Department of Radiology, University of Pennsylvania, 1 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104
| | - Chandra M Sehgal
- Department of Radiology, University of Pennsylvania, 1 Silverstein, 3400 Spruce Street, Philadelphia, PA 19104
| | - Louis J Soslowsky
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104,Corresponding Author: Louis J Soslowsky, McKay Orthopaedic Research Laboratory, University of Pennsylvania, 307A Stemmler Hall, 36 Street & Hamilton Walk, Philadelphia, PA 19104, , Phone: 215-898-8653, Fax: 215-573-2133
| |
Collapse
|
7
|
Magnussen AL, Mills IG. Vascular normalisation as the stepping stone into tumour microenvironment transformation. Br J Cancer 2021; 125:324-336. [PMID: 33828258 PMCID: PMC8329166 DOI: 10.1038/s41416-021-01330-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/17/2021] [Accepted: 02/17/2021] [Indexed: 02/01/2023] Open
Abstract
A functional vascular system is indispensable for drug delivery and fundamental for responsiveness of the tumour microenvironment to such medication. At the same time, the progression of a tumour is defined by the interactions of the cancer cells with their surrounding environment, including neovessels, and the vascular network continues to be the major route for the dissemination of tumour cells in cancer, facilitating metastasis. So how can this apparent conflict be reconciled? Vessel normalisation-in which redundant structures are pruned and the abnormal vasculature is stabilised and remodelled-is generally considered to be beneficial in the course of anti-cancer treatments. A causality between normalised vasculature and improved response to medication and treatment is observed. For this reason, it is important to discern the consequence of vessel normalisation on the tumour microenvironment and to modulate the vasculature advantageously. This article will highlight the challenges of controlled neovascular remodelling and outline how vascular normalisation can shape disease management.
Collapse
Affiliation(s)
- Anette L Magnussen
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Ian G Mills
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
- Patrick G Johnston Centre for Cancer Research, Queen's University of Belfast, Belfast, UK.
- Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
8
|
Saida Y, Brender JR, Yamamoto K, Mitchell JB, Krishna MC, Kishimoto S. Multimodal Molecular Imaging Detects Early Responses to Immune Checkpoint Blockade. Cancer Res 2021; 81:3693-3705. [PMID: 33837042 DOI: 10.1158/0008-5472.can-20-3182] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/17/2021] [Accepted: 04/08/2021] [Indexed: 01/02/2023]
Abstract
Immune checkpoint blockade (ICB) has become a standard therapy for several cancers, however, the response to ICB is inconsistent and a method for noninvasive assessment has not been established to date. To investigate the capability of multimodal imaging to evaluate treatment response to ICB therapy, hyperpolarized 13C MRI using [1-13C] pyruvate and [1,4-13C2] fumarate and dynamic contrast enhanced (DCE) MRI was evaluated to detect early changes in tumor glycolysis, necrosis, and intratumor perfusion/permeability, respectively. Mouse tumor models served as platforms for high (MC38 colon adenocarcinoma) and low (B16-F10 melanoma) sensitivity to dual ICB of PD-L1 and CTLA4. Glycolytic flux significantly decreased following treatment only in the less sensitive B16-F10 tumors. Imaging [1,4-13C2] fumarate conversion to [1,4-13C2] malate showed a significant increase in necrotic cell death following treatment in the ICB-sensitive MC38 tumors, with essentially no change in B16-F10 tumors. DCE-MRI showed significantly increased perfusion/permeability in MC38-treated tumors, whereas a similar, but statistically nonsignificant, trend was observed in B16-F10 tumors. When tumor volume was also taken into consideration, each imaging biomarker was linearly correlated with future survival in both models. These results suggest that hyperpolarized 13C MRI and DCE MRI may serve as useful noninvasive imaging markers to detect early response to ICB therapy. SIGNIFICANCE: Hyperpolarized 13C MRI and dynamic contrast enhanced MRI in murine tumor models provide useful insight into evaluating early response to immune checkpoint blockade therapy.See related commentary by Cullen and Keshari, p. 3444.
Collapse
Affiliation(s)
- Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Jeffrey R Brender
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
9
|
Matsumoto KI, Mitchell JB, Krishna MC. Multimodal Functional Imaging for Cancer/Tumor Microenvironments Based on MRI, EPRI, and PET. Molecules 2021; 26:1614. [PMID: 33799481 PMCID: PMC8002164 DOI: 10.3390/molecules26061614] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/23/2022] Open
Abstract
Radiation therapy is one of the main modalities to treat cancer/tumor. The response to radiation therapy, however, can be influenced by physiological and/or pathological conditions in the target tissues, especially by the low partial oxygen pressure and altered redox status in cancer/tumor tissues. Visualizing such cancer/tumor patho-physiological microenvironment would be a useful not only for planning radiotherapy but also to detect cancer/tumor in an earlier stage. Tumor hypoxia could be sensed by positron emission tomography (PET), electron paramagnetic resonance (EPR) oxygen mapping, and in vivo dynamic nuclear polarization (DNP) MRI. Tissue oxygenation could be visualized on a real-time basis by blood oxygen level dependent (BOLD) and/or tissue oxygen level dependent (TOLD) MRI signal. EPR imaging (EPRI) and/or T1-weighted MRI techniques can visualize tissue redox status non-invasively based on paramagnetic and diamagnetic conversions of nitroxyl radical contrast agent. 13C-DNP MRI can visualize glycometabolism of tumor/cancer tissues. Accurate co-registration of those multimodal images could make mechanisms of drug and/or relation of resulted biological effects clear. A multimodal instrument, such as PET-MRI, may have another possibility to link multiple functions. Functional imaging techniques individually developed to date have been converged on the concept of theranostics.
Collapse
Affiliation(s)
- Ken-ichiro Matsumoto
- Quantitative RedOx Sensing Group, Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Quantum Medical Science Directorate, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - James B. Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1002, USA;
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-1002, USA;
| |
Collapse
|
10
|
Economopoulou P, Kotsantis I, Psyrri A. Tumor Microenvironment and Immunotherapy Response in Head and Neck Cancer. Cancers (Basel) 2020; 12:E3377. [PMID: 33203092 PMCID: PMC7696050 DOI: 10.3390/cancers12113377] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/07/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment (TME) encompasses cellular and non-cellular components which play an important role in tumor evolution, invasion, and metastasis. A complicated interplay between tumor cells and adjacent TME cells, such as stromal cells, immune cells, inflammatory cells, and cytokines, leads to severe immunosuppression and the proliferation of cancer cells in several solid tumors. An immunosuppressive TME has a significant impact on treatment resistance and may guide response to immunotherapy. In head and neck cancer (HNC), immunotherapeutic drugs have been incorporated in everyday clinical practice. However, despite an exceptional rate of durable responses, only a low percentage of patients respond. In this review, we will focus on the complex interactions occurring in this dynamic system, the TME, which orchestrate key events that lead to tumor progression, immune escape, and resistance. Furthermore, we will summarize current clinical trials that depict the TME as a potential therapeutic target for improved patient selection.
Collapse
Affiliation(s)
| | | | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece; (P.E.); (I.K.)
| |
Collapse
|
11
|
Kishimoto S, Oshima N, Krishna MC, Gillies RJ. Direct and indirect assessment of cancer metabolism explored by MRI. NMR IN BIOMEDICINE 2019; 32:e3966. [PMID: 30169896 DOI: 10.1002/nbm.3966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/24/2018] [Accepted: 06/05/2018] [Indexed: 06/08/2023]
Abstract
Magnetic resonance-based approaches to obtain metabolic information on cancer have been explored for decades. Electron paramagnetic resonance (EPR) has been developed to pursue metabolic profiling and successfully used to monitor several physiologic parameters such as pO2 , pH, and redox status. All these parameters are associated with pathophysiology of various diseases. Especially in oncology, cancer hypoxia has been intensively studied because of its relationship with metabolic alterations, acquiring treatment resistance, or a malignant phenotype. Thus, pO2 imaging leads to an indirect metabolic assessment in this regard. Proton electron double-resonance imaging (PEDRI) is an imaging technique to visualize EPR by using the Overhauser effect. Most biological parameters assessed in EPR can be visualized using PEDRI. However, EPR and PEDRI have not been evaluated sufficiently for clinical application due to limitations such as toxicity of the probes or high specific absorption rate. Hyperpolarized (HP) 13 C MRI is a novel imaging technique that can directly visualize the metabolic profile. Production of metabolites of the HP 13 C probe delivered to target tissue are evaluated in this modality. Unlike EPR or PEDRI, which require the injection of radical probes, 13 C MRI requires a probe that can be physiologically metabolized and efficiently hyperpolarized. Among several methods for hyperpolarizing probes, dissolution dynamic nuclear hyperpolarization is a widely used technique for in vivo imaging. Pyruvate is the most suitable probe for HP 13 C MRI because it is part of the glycolytic pathway and the high efficiency of pyruvate-to-lactate conversion is a distinguishing feature of cancer. Its clinical applicability also makes it a promising metabolic imaging modality. Here, we summarize the applications of these indirect and direct MR-based metabolic assessments focusing on pO2 and pyruvate-to-lactate conversion. The two parameters are strongly associated with each other, hence the acquired information is potentially interchangeable when evaluating treatment response to oxygen-dependent cancer therapies.
Collapse
Affiliation(s)
- Shun Kishimoto
- Radiation Biology Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Nobu Oshima
- Urologic Oncology Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Murali C Krishna
- Radiation Biology Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Robert J Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
12
|
Gaustad JV, Simonsen TG, Wegner CS, Rofstad EK. Vascularization, Oxygenation, and the Effect of Sunitinib Treatment in Pancreatic Ductal Adenocarcinoma Xenografts. Front Oncol 2019; 9:845. [PMID: 31555596 PMCID: PMC6727195 DOI: 10.3389/fonc.2019.00845] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022] Open
Abstract
The purpose of the study was to investigate vascularization, oxygenation, and the effect of sunitinib treatment in pancreatic ductal adenocarcinoma (PDAC). BxPC-3 and Capan-2 xenografts grown in dorsal window chambers were used as preclinical models of human PDAC. Tumor angiogenesis and the morphology and function of tumor vascular networks were studied by intravital microscopy, and tumor hypoxia was assessed by immunohistochemistry. The PDAC models differed in vessel distribution and oxygenation, and the differences were induced by the initial tumor angiogenesis. In both models, sunitinib treatment reduced intratumor and peritumor vessel densities by selectively removing small-diameter vessels. Sunitinb treatment resulted in a general decrease in vessel density and scattered hypoxia in BxPC-3 tumors, and depleted most vessels and induced massive hypoxia in central parts of Capan-2 tumors. The study demonstrates that PDAC xenografts can differ in vascularization, and the differences can impact oxygenation and effects of treatment. Neoadjuvant sunitinib treatment is inappropriate in combination with conventional therapy for human PDACs resembling the PDAC xenografts used here, because sunitinib-induced hypoxia can impair the effect of most conventional therapies.
Collapse
Affiliation(s)
- Jon-Vidar Gaustad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Trude G Simonsen
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Catherine S Wegner
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Einar K Rofstad
- Group of Radiation Biology and Tumor Physiology, Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Kishimoto S, Brender JR, Crooks DR, Matsumoto S, Seki T, Oshima N, Merkle H, Lin P, Reed G, Chen AP, Ardenkjaer-Larsen JH, Munasinghe J, Saito K, Yamamoto K, Choyke PL, Mitchell J, Lane AN, Fan TWM, Linehan WM, Krishna MC. Imaging of glucose metabolism by 13C-MRI distinguishes pancreatic cancer subtypes in mice. eLife 2019; 8:e46312. [PMID: 31408004 PMCID: PMC6706239 DOI: 10.7554/elife.46312] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 08/08/2019] [Indexed: 12/13/2022] Open
Abstract
Metabolic differences among and within tumors can be an important determinant in cancer treatment outcome. However, methods for determining these differences non-invasively in vivo is lacking. Using pancreatic ductal adenocarcinoma as a model, we demonstrate that tumor xenografts with a similar genetic background can be distinguished by their differing rates of the metabolism of 13C labeled glucose tracers, which can be imaged without hyperpolarization by using newly developed techniques for noise suppression. Using this method, cancer subtypes that appeared to have similar metabolic profiles based on steady state metabolic measurement can be distinguished from each other. The metabolic maps from 13C-glucose imaging localized lactate production and overall glucose metabolism to different regions of some tumors. Such tumor heterogeneity would not be not detectable in FDG-PET.
Collapse
Affiliation(s)
- Shun Kishimoto
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - Jeffrey R Brender
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIHBethesdaUnited States
| | - Shingo Matsumoto
- Graduate School of Information Science and Technology, Division of Bioengineering and BioinformaticsHokkaido UniversitySapporoJapan
- JST, PRESTSaitamaJapan
| | - Tomohiro Seki
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - Nobu Oshima
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | | | - Penghui Lin
- Center for Environmental and Systems BiochemistryUniversity of KentuckyLexingtonUnited States
| | | | | | - Jan Henrik Ardenkjaer-Larsen
- GE HealthCareChicagoUnited States
- Department of Electrical EngineeringTechnical University of DenmarkKongens LyngbyDenmark
| | | | - Keita Saito
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - Kazutoshi Yamamoto
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - James Mitchell
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| | - Andrew N Lane
- Center for Environmental and Systems BiochemistryUniversity of KentuckyLexingtonUnited States
- Markey Cancer CenterUniversity of KentuckyLexingtonUnited States
| | - Teresa WM Fan
- Center for Environmental and Systems BiochemistryUniversity of KentuckyLexingtonUnited States
- Markey Cancer CenterUniversity of KentuckyLexingtonUnited States
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIHBethesdaUnited States
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer ResearchNCI, NIHBethesdaUnited States
| |
Collapse
|
14
|
Alpha lipoic acid attenuates hypoxia-induced apoptosis, inflammation and mitochondrial oxidative stress via inhibition of TRPA1 channel in human glioblastoma cell line. Biomed Pharmacother 2018; 111:292-304. [PMID: 30590317 DOI: 10.1016/j.biopha.2018.12.077] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Apoptosis, overload Ca2+ entry and oxidative stress are induced in neurons by hypoxia. Drug-resistant cancer cells are killed by hypoxic conditions. α-Lipoic acid (ALA) has antioxidant and pro-oxidant functions. The TRPA1 channel is activated by oxidative stress and pro-oxidant ALA may have a regulator role in the TRPA1 activity in the human glioblastoma (DBTRG) cells. The aim of this study was to evaluate if a combination therapy of ALA with a hypoxia can alter the effect of this hypoxia through TRPA1 activation in the DBTRG cells. The DBTRG cells were divided into four treatment groups as control, ALA (50 μM), and hypoxia and hypoxia + ALA. Hypoxia in the cells was induced by CoCl2 (200 μM). Apoptosis, Annexin V, mitochondrial membrane depolarization (JC-1), reactive oxygen species (ROS) production, IL-1β, IL-18, caspase 3 and 9 values were increased through activation of TRPA1 (cinnamaldehyde) in the cells by the hypoxia induction, although cell viability, reduced glutathione and glutathione peroxidase values were decreased by the treatments. The values were modulated in the cells by TRPA1 blocker (AP18) and ALA treatments. Involvements of TRPA1 activity on values in the cells were also confirmed by patch-clamp and laser confocal microscopy analyses. In conclusion, apoptotic, inflammatory and oxidant effects of hypoxia were increased by activation of TRPA1, but its action on the values was decreased by the ALA treatment. ALA treatment could be used as an effective strategy in the treatment of hypoxia-induced oxidative stress, apoptosis and inflammation in the neurons.
Collapse
|
15
|
Takakusagi Y, Naz S, Takakusagi K, Ishima M, Murata H, Ohta K, Miura M, Sugawara F, Sakaguchi K, Kishimoto S, Munasinghe JP, Mitchell JB, Krishna MC. A Multimodal Molecular Imaging Study Evaluates Pharmacological Alteration of the Tumor Microenvironment to Improve Radiation Response. Cancer Res 2018; 78:6828-6837. [PMID: 30301838 DOI: 10.1158/0008-5472.can-18-1654] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/03/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022]
Abstract
: Hypoxic zones in solid tumors contribute to radioresistance, and pharmacologic agents that increase tumor oxygenation prior to radiation, including antiangiogenic drugs, can enhance treatment response to radiotherapy. Although such strategies have been applied, imaging assessments of tumor oxygenation to identify an optimum time window for radiotherapy have not been fully explored. In this study, we investigated the effects of α-sulfoquinovosylacyl-1,3-propanediol (SQAP or CG-0321; a synthetic derivative of an antiangiogenic agent) on the tumor microenvironment in terms of oxygen partial pressure (pO2), oxyhemoglobin saturation (sO2), blood perfusion, and microvessel density using electron paramagnetic resonance imaging, photoacoustic imaging, dynamic contrast-enhanced MRI with Gd-DTPA injection, and T2*-weighted imaging with ultrasmall superparamagnetic iron oxide (USPIO) contrast. SCCVII and A549 tumors were grown by injecting tumor cells into the hind legs of mice. Five days of daily radiation (2 Gy) combined with intravenous injection of SQAP (2 mg/kg) 30 minutes prior to irradiation significantly delayed growth of tumor xenografts. Three days of daily treatment improved tumor oxygenation and decreased tumor microvascular density on T2*-weighted images with USPIO, suggesting vascular normalization. Acute effects of SQAP on tumor oxygenation were examined by pO2, sO2, and Gd-DTPA contrast-enhanced imaging. SQAP treatment improved perfusion and tumor pO2 (ΔpO2: 3.1 ± 1.0 mmHg) and was accompanied by decreased sO2 (20%-30% decrease) in SCCVII implants 20-30 minutes after SQAP administration. These results provide evidence that SQAP transiently enhanced tumor oxygenation by facilitating oxygen dissociation from oxyhemoglobin and improving tumor perfusion. Therefore, SQAP-mediated sensitization to radiation in vivo can be attributed to increased tumor oxygenation. SIGNIFICANCE: A multimodal molecular imaging study evaluates pharmacological alteration of the tumor microenvironment to improve radiation response.
Collapse
Affiliation(s)
- Yoichi Takakusagi
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Sarwat Naz
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Kaori Takakusagi
- Center for Biologics Evaluation and Research, Food and Drug Administration, Rockville, Maryland
| | | | | | | | - Masahiko Miura
- Department of Oral Radiation Oncology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fumio Sugawara
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Kengo Sakaguchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Tokyo, Japan
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jeeva P Munasinghe
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorder and Stroke, NIH, Bethesda, Maryland
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
16
|
Matsumoto S, Kishimoto S, Saito K, Takakusagi Y, Munasinghe JP, Devasahayam N, Hart CP, Gillies RJ, Mitchell JB, Krishna MC. Metabolic and Physiologic Imaging Biomarkers of the Tumor Microenvironment Predict Treatment Outcome with Radiation or a Hypoxia-Activated Prodrug in Mice. Cancer Res 2018; 78:3783-3792. [PMID: 29792309 PMCID: PMC8092078 DOI: 10.1158/0008-5472.can-18-0491] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 04/05/2018] [Accepted: 05/18/2018] [Indexed: 01/11/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by hypoxic niches that lead to treatment resistance. Therefore, studies of tumor oxygenation and metabolic profiling should contribute to improved treatment strategies. Here, we define two imaging biomarkers that predict differences in tumor response to therapy: (i) partial oxygen pressure (pO2), measured by EPR imaging; and (ii) [1-13C] pyruvate metabolism rate, measured by hyperpolarized 13C MRI. Three human PDAC xenografts with varying treatment sensitivity (Hs766t, MiaPaCa2, and Su.86.86) were grown in mice. The median pO2 of the mature Hs766t, MiaPaCa2, and Su.86.86 tumors was 9.1 ± 1.7, 11.1 ± 2.2, and 17.6 ± 2.6 mm Hg, and the rate of pyruvate-to-lactate conversion was 2.72 ± 0.48, 2.28 ± 0.26, and 1.98 ± 0.51 per minute, respectively (n = 6, each). These results are in agreement with steady-state data of matabolites quantified by mass spectroscopy and histologic analysis, indicating glycolytic and hypoxia profile in Hs766t, MiaPaca2, and Su.86.86 tumors. Fractionated radiotherapy (5 Gy × 5) resulted in a tumor growth delay of 16.7 ± 1.6 and 18.0 ± 2.7 days in MiaPaca2 and Su.86.86 tumors, respectively, compared with 6.3 ± 2.7 days in hypoxic Hs766t tumors. Treatment with gemcitabine, a first-line chemotherapeutic agent, or the hypoxia-activated prodrug TH-302 was more effective against Hs766t tumors (20.0 ± 3.5 and 25.0 ± 7.7 days increase in survival time, respectively) than MiaPaCa2 (2.7 ± 0.4 and 6.7 ± 0.7 days) and Su.86.86 (4.7 ± 0.6 and 0.7 ± 0.6 days) tumors. Collectively, these results demonstrate the ability of molecular imaging biomarkers to predict the response of PDAC to treatment with radiotherapy and TH-302.Significance: pO2 imaging data and clinically available metabolic imaging data provide useful insight into predicting the treatment efficacy of chemotherapy, radiation, and a hypoxia-activated prodrug as monotherapies and combination therapies in PDAC tumor xenograft models. Cancer Res; 78(14); 3783-92. ©2018 AACR.
Collapse
Affiliation(s)
- Shingo Matsumoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- Division of Bioengineering and Bioinformatics, Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
- JST, PREST, Saitama, Japan
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Keita Saito
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Yoichi Takakusagi
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
- National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Jeeva P Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorder and Stroke, NIH, Bethesda, Maryland
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Robert J Gillies
- Department of Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
17
|
Yasui H, Kawai T, Matsumoto S, Saito K, Devasahayam N, Mitchell JB, Camphausen K, Inanami O, Krishna MC. Quantitative imaging of pO 2 in orthotopic murine gliomas: hypoxia correlates with resistance to radiation. Free Radic Res 2018; 51:861-871. [PMID: 29076398 DOI: 10.1080/10715762.2017.1388506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia is considered one of the microenvironmental factors associated with the malignant nature of glioblastoma. Thus, evaluating intratumoural distribution of hypoxia would be useful for therapeutic planning as well as assessment of its effectiveness during the therapy. Electron paramagnetic resonance imaging (EPRI) is an imaging technique which can generate quantitative maps of oxygen in vivo using the exogenous paramagnetic compound, triarylmethyl and monitoring its line broadening caused by oxygen. In this study, the feasibility of EPRI for assessment of oxygen distribution in the glioblastoma using orthotopic U87 and U251 xenograft model is examined. Heterogeneous distribution of pO2 between 0 and 50 mmHg was observed throughout the tumours except for the normal brain tissue. U251 glioblastoma was more likely to exhibit hypoxia than U87 for comparable tumour size (median pO2; 29.7 and 18.2 mmHg, p = .028, in U87 and U251, respectively). The area with pO2 under 10 mmHg on the EPR oximetry (HF10) showed a good correlation with pimonidazole staining among tumours with evaluated size. In subcutaneous xenograft model, irradiation was relatively less effective for U251 compared with U87. In conclusion, EPRI is a feasible method to evaluate oxygen distribution in the brain tumour.
Collapse
Affiliation(s)
- Hironobu Yasui
- a Central Institute of Isotope Science, Hokkaido University , Sapporo , Japan
| | - Tatsuya Kawai
- b Radiation Oncology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Shingo Matsumoto
- c Division of Bioengineering and Bioinformatics , Graduate School of Information Science and Technology, Hokkaido University , Sapporo , Japan
| | - Keita Saito
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Nallathamby Devasahayam
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - James B Mitchell
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Kevin Camphausen
- b Radiation Oncology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| | - Osamu Inanami
- e Laboratory of Radiation Biology, Department of Environmental Veterinary Sciences, Graduate School of Veterinary Medicine , Hokkaido University , Sapporo , Japan
| | - Murali C Krishna
- d Radiation Biology Branch , Center for Cancer Research, National Cancer Institute, National Health Institutes , Bethesda , MD , USA
| |
Collapse
|
18
|
El Alaoui-Lasmaili K, Faivre B. Antiangiogenic therapy: Markers of response, "normalization" and resistance. Crit Rev Oncol Hematol 2018; 128:118-129. [PMID: 29958627 DOI: 10.1016/j.critrevonc.2018.06.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 02/08/2023] Open
Abstract
Currently in cancer treatment, one premise is to use antiangiogenic therapies in association with chemotherapy or radiotherapy to augment their efficacy by benefiting from the vascular "normalization" induced by antiangiogenic therapy. This concept defines the time during which the tumor blood vessels adopt normal-like morphology and functionality, i.e. the blood vessels become more mature, the perfusion augments and hypoxia decreases. To date, there is such a diversity of treatment protocols where the type of antiangiogenic to adopt, its dose and duration of administration are different, that knowing when and how to treat is problematic. In this review, we analyzed thoroughly preclinical and clinical studies that use antiangiogenic treatments to benefit from the "normalization" and showed that the effects depend on the type of antiangiogenic administrated (anti-VEGF, anti-VEGFR, Multi-Kinase Inhibitor) and on the duration of treatment. Finally, biomarkers of "normalization" and resistance that could be used in the clinic are presented.
Collapse
Affiliation(s)
| | - Béatrice Faivre
- Université de Lorraine, CNRS, CRAN, F-54000 Nancy, France; Université de Lorraine, Faculté de Pharmacie, Nancy, France.
| |
Collapse
|
19
|
Shan Y, Wang B, Zhang J. New strategies in achieving antiangiogenic effect: Multiplex inhibitors suppressing compensatory activations of RTKs. Med Res Rev 2018; 38:1674-1705. [DOI: 10.1002/med.21517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/19/2018] [Accepted: 05/19/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Yuanyuan Shan
- Department of Pharmacy; The First Affiliated Hospital of Xi'an Jiaotong University; Xi'an China
| | - Binghe Wang
- Department of Chemistry; Center for Diagnostics and Therapeutics; Georgia State University; Atlanta GA USA
| | - Jie Zhang
- School of Pharmacy, Health Science Center; Xi'an Jiaotong University; Xi'an China
| |
Collapse
|
20
|
Kishimoto S, Matsumoto KI, Saito K, Enomoto A, Matsumoto S, Mitchell JB, Devasahayam N, Krishna MC. Pulsed Electron Paramagnetic Resonance Imaging: Applications in the Studies of Tumor Physiology. Antioxid Redox Signal 2018; 28:1378-1393. [PMID: 29130334 PMCID: PMC5910045 DOI: 10.1089/ars.2017.7391] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Electron paramagnetic resonance imaging (EPRI) is capable of generating images of tissue oxygenation using exogenous paramagnetic probes such as trityl radicals or nitroxyl radicals. The spatial distribution of the paramagnetic probe can be generated using magnetic field gradients as in magnetic resonance imaging and, from its spectral features, spatial maps of oxygen can be obtained from live objects. In this review, two methods of signal acquisition and image formation/reconstruction are described. The probes used and its application to study tumor physiology and monitor treatment response with chemotherapy drugs in mouse models of human cancer are summarized. Recent Advances: By implementing phase encoding/Fourier reconstruction in EPRI in time domain mode, the frequency contribution to the spatial resolution was avoided and images with improved spatial resolution were obtained. The EPRI-generated pO2 maps in tumor were useful to detect and evaluate the effects of various antitumor therapies on tumor physiology. Coregistration with other imaging modalities provided a better understanding of hypoxia-related alteration in physiology. CRITICAL ISSUES The high radiofrequency (RF) power of EPR irradiation and toxicity profile of radical probes are the main obstacles for clinical application. The improvement of RF low power pulse sequences may allow for clinical translation. FUTURE DIRECTIONS Pulsed EPR oximetry can be a powerful tool to research various diseases involving hypoxia such as cancer, ischemic heart diseases, stroke, and diabetes. With appropriate paramagnetic probes, it can also be applied for various other purposes such as detecting local acid-base balance or oxidative stress. Antioxid. Redox Signal. 28, 1378-1393.
Collapse
Affiliation(s)
- Shun Kishimoto
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | - Ken-Ichiro Matsumoto
- 2 Quantitative RedOx Sensing Team, Department of Basic Medical Sciences for Radiation Damages, Chiba, Japan
| | - Keita Saito
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | - Ayano Enomoto
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | - Shingo Matsumoto
- 3 Division of Bioengineering and Bioinformatics, Hokkaido University , Sapporo, Japan
| | - James B Mitchell
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | - Nallathamby Devasahayam
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| | - Murali C Krishna
- 1 Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, Maryland
| |
Collapse
|
21
|
Matsuo M, Kawai T, Kishimoto S, Saito K, Munasinghe J, Devasahayam N, Mitchell JB, Krishna MC. Co-imaging of the tumor oxygenation and metabolism using electron paramagnetic resonance imaging and 13-C hyperpolarized magnetic resonance imaging before and after irradiation. Oncotarget 2018; 9:25089-25100. [PMID: 29861855 PMCID: PMC5982751 DOI: 10.18632/oncotarget.25317] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/02/2018] [Indexed: 01/18/2023] Open
Abstract
To examine the relationship between local oxygen partial pressure and energy metabolism in the tumor, electron paramagnetic resonance imaging (EPRI) and magnetic resonance imaging (MRI) with hyperpolarized [1-13C] pyruvate were performed. SCCVII and HT29 solid tumors implanted in the mouse leg were imaged by EPRI using OX063, a paramagnetic probe and 13C-MRI using hyperpolarized [1-13C] pyruvate. Local partial oxygen pressure and pyruvate metabolism in the two tumor implants were examined. The effect of a single dose of 5-Gy irradiation on the pO2 and metabolism was also investigated by sequential imaging of EPRI and 13C-MRI in HT29 tumors. A phantom study using tubes filled with different concentration of [1-13C] pyruvate, [1-13C] lactate, and OX063 at different levels of oxygen confirmed the validity of this sequential imaging of EPRI and hyperpolarized 13C-MRI. In vivo studies revealed SCCVII tumor had a significantly larger hypoxic fraction (pO2 < 8 mmHg) compared to HT29 tumor. The flux of pyruvate-to-lactate conversion was also higher in SCCVII than HT29. The lactate-to-pyruvate ratio in hypoxic regions (pO2 < 8 mmHg) 24 hours after 5-Gy irradiation was significantly higher than those without irradiation (0.76 vs. 0.36) in HT29 tumor. The in vitro study showed an increase in extracellular acidification rate after irradiation. In conclusion, co-imaging of pO2 and pyruvate-to-lactate conversion kinetics successfully showed the local metabolic changes especially in hypoxic area induced by radiation therapy.
Collapse
Affiliation(s)
- Masayuki Matsuo
- Radiation Biology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Department of Radiology, Gifu University Graduate School of Medicine, Gifu City, Japan
| | - Tatsuya Kawai
- Radiation Oncology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Keita Saito
- Radiation Biology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeeva Munasinghe
- MRI Research Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
22
|
Nitta N, Takakusagi Y, Kokuryo D, Shibata S, Tomita A, Higashi T, Aoki I, Harada M. Intratumoral evaluation of 3D microvasculature and nanoparticle distribution using a gadolinium-dendron modified nano-liposomal contrast agent with magnetic resonance micro-imaging. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1315-1324. [PMID: 29626524 DOI: 10.1016/j.nano.2018.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 03/23/2018] [Accepted: 03/25/2018] [Indexed: 12/22/2022]
Abstract
The enhanced permeability and retention (EPR) effect is variable depending on nanoparticle properties and tumor/vessel conditions. Thus, intratumoral evaluations of the vasculature and nanoparticle distribution are important for predicting the therapeutic efficacy and the intractability of tumors. We aimed to develop a tumor vasculature evaluation method and high-resolution nanoparticle delivery imaging using magnetic resonance (MR) micro-imaging technology with a gadolinium (Gd)-dendron assembled liposomal contrast agent. Using the Gd-liposome and a cryogenic receiving coil, we achieved 50-μm isotropic MR angiography with clear visualization of tumor micro-vessel structure. The Gd-liposome-enhanced MR micro-imaging revealed differences in the vascular structures between Colon26- and SU-DHL6-grafted mice models. The vessel volumes and diameters measured for both tumors were significantly correlated with histological observations. The MR micro-imaging methods facilitate the evaluation of intratumoral vascularization patterns, the quantitative assessment of vascular-properties that alter tumor malignancy, particle retentivity, and the effects of treatment.
Collapse
Affiliation(s)
- Nobuhiro Nitta
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan; Group of Quantum-state Controlled MRI, QST, Chiba, Japan; Graduate School of Medicine, Tokushima University, Tokushima, Japan
| | - Yoichi Takakusagi
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan; Group of Quantum-state Controlled MRI, QST, Chiba, Japan
| | - Daisuke Kokuryo
- Graduate School of System Informatics, Kobe University, Kobe, Hyogo, Japan
| | - Sayaka Shibata
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan; Group of Quantum-state Controlled MRI, QST, Chiba, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Tatsuya Higashi
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Ichio Aoki
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan; Group of Quantum-state Controlled MRI, QST, Chiba, Japan.
| | - Masafumi Harada
- Graduate School of Medicine, Tokushima University, Tokushima, Japan
| |
Collapse
|
23
|
Dizeux A, Payen T, Le Guillou-Buffello D, Comperat E, Gennisson JL, Tanter M, Oelze M, Bridal SL. In Vivo Multiparametric Ultrasound Imaging of Structural and Functional Tumor Modifications during Therapy. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:2000-2012. [PMID: 28554540 DOI: 10.1016/j.ultrasmedbio.2017.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 05/26/2023]
Abstract
Longitudinal imaging techniques are needed that can meaningfully probe the tumor microenvironment and its spatial heterogeneity. Contrast-enhanced ultrasound, shear wave elastography and quantitative ultrasound are ultrasound-based techniques that provide information on the vascular function and micro-/macroscopic tissue structure. Modifications of the tumor microenvironment induced by cytotoxic and anti-angiogenic molecules in ectopic murine Lewis lung carcinoma tumors were monitored. The most heterogenous structures were found in tumors treated with anti-angiogenic drug that simultaneously accumulated the highest levels of necrosis and fibrosis. The anti-angiogenic group presented the highest number of correlations between parameters related to vascular function and those related to the micro-/macrostructure of the tumor microenvironment. Results suggest how patterns of multiparametric ultrasound modifications can be related to provide a more insightful marker of changes occurring within tumors during therapy.
Collapse
Affiliation(s)
- Alexandre Dizeux
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France.
| | - Thomas Payen
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France
| | | | - Eva Comperat
- Academic Department of Pathology, Pitie-Salpetriere Hospital, AP-HP, UPMC Univ Paris 06, Paris, France
| | - Jean-Luc Gennisson
- Institut Langevin-Ondes et Images, ESPCI ParisTech, PSL Research University, CNRS UMR7587, INSERM U979, Paris, France
| | - Mickael Tanter
- Institut Langevin-Ondes et Images, ESPCI ParisTech, PSL Research University, CNRS UMR7587, INSERM U979, Paris, France
| | - Michael Oelze
- Bioacoustics Research Laboratory, Department of Electrical and Computer Engineering, University of Illinois, Urbana, Illinois, USA
| | - S Lori Bridal
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France
| |
Collapse
|
24
|
Abstract
OPINION STATEMENT The survival rate for patients with advanced stages of squamous cell carcinoma of the head and neck (SCCHN) remains poor despite multimodal treatment options. Cetuximab, an anti-EGFR inhibitor, is the only FDA-approved targeted agent for this disease. Recent findings have implicated modifications of the microenvironment and, consequently, phenotypical modifications of the cancer cell, in treatment resistance mechanisms. For many years, cancer research has focused mainly on targetable sites on or inside the cancer cell. Nowadays, in preclinical and clinical studies, a greater emphasis is being placed on drugs that target the tumor microenvironment. Potential targets relate to tumor vascularization, immunology, extracellular matrix components, or cancer-associated fibroblasts. The combination of these new agents with standard treatment options is of particular interest to overcome resistance mechanisms and/or to increase treatment efficacy. Whereas antiangiogenic agents show poor clinical activity, immunotherapy seems to be a more promising tool with an objective response rate (ORR) of 20 % in patients with recurrent and/or metastatic squamous cell carcinoma (R/M SCC). Other targets, located inside the extracellular matrix or on cancer associated fibroblasts, are under preclinical investigation. These new agents all need to be tested in clinical trials alone, or in combination with standard treatment modalities, based on preclinical data. To increase our knowledge of the complex network between the cancer cell and its environment, preclinical studies should consider co-culture models, and clinical studies should incorporate a translational research objective.
Collapse
|
25
|
Gallez B, Neveu MA, Danhier P, Jordan BF. Manipulation of tumor oxygenation and radiosensitivity through modification of cell respiration. A critical review of approaches and imaging biomarkers for therapeutic guidance. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2017; 1858:700-711. [DOI: 10.1016/j.bbabio.2017.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/05/2017] [Accepted: 01/06/2017] [Indexed: 11/17/2022]
|
26
|
Colliez F, Gallez B, Jordan BF. Assessing Tumor Oxygenation for Predicting Outcome in Radiation Oncology: A Review of Studies Correlating Tumor Hypoxic Status and Outcome in the Preclinical and Clinical Settings. Front Oncol 2017; 7:10. [PMID: 28180110 PMCID: PMC5263142 DOI: 10.3389/fonc.2017.00010] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/10/2017] [Indexed: 12/30/2022] Open
Abstract
Tumor hypoxia is recognized as a limiting factor for the efficacy of radiotherapy, because it enhances tumor radioresistance. It is strongly suggested that assessing tumor oxygenation could help to predict the outcome of cancer patients undergoing radiation therapy. Strategies have also been developed to alleviate tumor hypoxia in order to radiosensitize tumors. In addition, oxygen mapping is critically needed for intensity modulated radiation therapy (IMRT), in which the most hypoxic regions require higher radiation doses and the most oxygenated regions require lower radiation doses. However, the assessment of tumor oxygenation is not yet included in day-to-day clinical practice. This is due to the lack of a method for the quantitative and non-invasive mapping of tumor oxygenation. To fully integrate tumor hypoxia parameters into effective improvements of the individually tailored radiation therapy protocols in cancer patients, methods allowing non-invasively repeated, safe, and robust mapping of changes in tissue oxygenation are required. In this review, non-invasive methods dedicated to assessing tumor oxygenation with the ultimate goal of predicting outcome in radiation oncology are presented, including positron emission tomography used with nitroimidazole tracers, magnetic resonance methods using endogenous contrasts (R1 and R2*-based methods), and electron paramagnetic resonance oximetry; the goal is to highlight results of studies establishing correlations between tumor hypoxic status and patients’ outcome in the preclinical and clinical settings.
Collapse
Affiliation(s)
- Florence Colliez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Bénédicte F Jordan
- Biomedical Magnetic Resonance Group, Louvain Drug Research Institute, Université Catholique de Louvain , Brussels , Belgium
| |
Collapse
|
27
|
Noninvasive mapping of the redox status of dimethylnitrosamine-induced hepatic fibrosis using in vivo dynamic nuclear polarization-magnetic resonance imaging. Sci Rep 2016; 6:32604. [PMID: 27587186 PMCID: PMC5009327 DOI: 10.1038/srep32604] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/09/2016] [Indexed: 12/13/2022] Open
Abstract
Hepatic fibrosis is a chronic disorder caused by viral infection and/or metabolic, genetic and cholestatic disorders. A noninvasive procedure that enables the detection of liver fibrosis based on redox status would be useful for disease identification and monitoring, and the development of treatments. However, an appropriate technique has not been reported. This study describes a novel method for assessing the redox status of the liver using in vivo dynamic nuclear polarization-magnetic resonance imaging (DNP-MRI) with the nitroxyl radical carbamoyl-PROXYL as a molecular imaging probe, which was tested in dimethylnitrosamine-treated mice as a model of liver fibrosis. Based on the pharmacokinetics of carbamoyl-PROXYL in control livers, reduction rate mapping was performed in fibrotic livers. Reduction rate maps demonstrated a clear difference between the redox status of control and fibrotic livers according to the expression of antioxidants. These findings indicate that in vivo DNP-MRI with a nitroxyl radical probe enables noninvasive detection of changes in liver redox status.
Collapse
|
28
|
Matsumoto S. Prediction of Cancer Treatment Response by Physiologic and Metabolic Imaging. YAKUGAKU ZASSHI 2016; 136:1101-5. [DOI: 10.1248/yakushi.15-00234-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Shingo Matsumoto
- Graduate School of Information Science and Technology, Hokkaido University
| |
Collapse
|
29
|
Matsumoto KI. Development of Magnetic Resonance-based Functional Imaging: The Past, the Present, and the Future. YAKUGAKU ZASSHI 2016; 136:1075-80. [DOI: 10.1248/yakushi.15-00234-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Ken-ichiro Matsumoto
- Quantitative RedOx Sensing Team, Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology
| |
Collapse
|
30
|
El Kaffas A, Sadeghi-Naini A, Falou O, Tran WT, Zhou S, Hashim A, Fernandes J, Giles A, Czarnota GJ. Assessment of tumor response to radiation and vascular targeting therapy in mice using quantitative ultrasound spectroscopy. Med Phys 2016; 42:4965-73. [PMID: 26233222 DOI: 10.1118/1.4926554] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE It is now recognized that the tumor vasculature is in part responsible for regulating tumor responses to radiation therapy. However, the extent to which radiation-based vascular damage contributes to tumor cell death remains unknown. In this work, quantitative ultrasound spectroscopy (QUS) methods were used to investigate the acute responses of tumors to radiation-based vascular treatments. METHODS Tumor xenografts (MDA-MB-231) were treated with single radiation doses of 2 or 8 Gy alone, or in combination with pharmacological agents that modulate vascular radiosensitivity. The midband fit, the slope, and the 0-MHz intercept QUS parameters were obtained from a linear-regression fit to the averaged power spectrum of frequency-dependent ultrasound backscatter and were used to quantify acute tumor responses following treatment administration. Power spectrums were extracted from raw volumetric radio-frequency ultrasound data obtained before and 24 h following treatment administration. These parameters have previously been correlated to tumor cell death. Staining using in situ end labeling, carbonic anhydrase 9 and cluster of differentiation 31 of tumor sections were used to assess cell death, oxygenation, and vasculature distributions, respectively. RESULTS Results indicate a significant midband fit QUS parameter increases of 3.2 ± 0.3 dBr and 5.4 ± 0.5 dBr for tumors treated with 2 and 8 Gy radiation combined with the antiangiogenic agent Sunitinib, respectively. In contrast, tumors treated with radiation alone demonstrated a significant midband fit increase of 4.4 ± 0.3 dBr at 8 Gy only. Preadministration of basic fibroblast growth factor, an endothelial radioprotector, acted to minimize tumor response following single large doses of radiation. Immunohistochemical analysis was in general agreement with QUS findings; an R(2) of 0.9 was observed when quantified cell death was correlated with changes in midband fit. CONCLUSIONS Results from QUS analysis presented in this study confirm that acute tumor response is linked to a vascular effect following high doses of radiation therapy. Overall, this is in agreement with previous reports suggesting that acute tumor radiation response is regulated by a vascular-driven response. Data also suggest that Sunitinib may enhance tumor radiosensitivity through a vascular remodeling process, and that QUS may be sensitive to changes in tissue properties associated with vascular remodeling. Finally, the work also demonstrates the ability of QUS methods to monitor response to radiation-based vascular strategies.
Collapse
Affiliation(s)
- Ahmed El Kaffas
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Departments of Medical Biophysics and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Ali Sadeghi-Naini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Departments of Medical Biophysics and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Omar Falou
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Departments of Medical Biophysics and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - William Tyler Tran
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Departments of Medical Biophysics and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Stephanie Zhou
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - Amr Hashim
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada and Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - Jason Fernandes
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - Anoja Giles
- Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada
| | - Gregory J Czarnota
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; Imaging Research and Physical Sciences, Sunnybrook Health Sciences Centre, Toronto, Ontario M4N 3M5, Canada; and Departments of Medical Biophysics and Radiation Oncology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
31
|
Hutchinson LG, Gaffney EA, Maini PK, Wagg J, Phipps A, Byrne HM. Vascular phenotype identification and anti-angiogenic treatment recommendation: A pseudo-multiscale mathematical model of angiogenesis. J Theor Biol 2016; 398:162-80. [PMID: 26987523 DOI: 10.1016/j.jtbi.2016.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/29/2016] [Accepted: 03/03/2016] [Indexed: 12/23/2022]
Abstract
The development of anti-angiogenic drugs for cancer therapy has yielded some promising candidates, but novel approaches for interventions to angiogenesis have led to disappointing results. In addition, there is a shortage of biomarkers that are predictive of response to anti-angiogenic treatments. Consequently, the complex biochemical and physiological basis for tumour angiogenesis remains incompletely understood. We have adopted a mathematical approach to address these issues, formulating a spatially averaged multiscale model that couples the dynamics of VEGF, Ang1, Ang2 and PDGF, with those of mature and immature endothelial cells and pericyte cells. The model reproduces qualitative experimental results regarding pericyte coverage of vessels after treatment by anti-Ang2, anti-VEGF and combination anti-VEGF/anti-Ang2 antibodies. We used the steady state behaviours of the model to characterise angiogenic and non-angiogenic vascular phenotypes, and used mechanistic perturbations representing hypothetical anti-angiogenic treatments to generate testable hypotheses regarding transitions to non-angiogenic phenotypes that depend on the pre-treatment vascular phenotype. Additionally, we predicted a synergistic effect between anti-VEGF and anti-Ang2 treatments when applied to an immature pre-treatment vascular phenotype, but not when applied to a normalised angiogenic pre-treatment phenotype. Based on these findings, we conclude that changes in vascular phenotype are predicted to be useful as an experimental biomarker of response to treatment. Further, our analysis illustrates the potential value of non-spatial mathematical models for generating tractable predictions regarding the action of anti-angiogenic therapies.
Collapse
Affiliation(s)
- L G Hutchinson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK.
| | - E A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| | - P K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| | - J Wagg
- Roche Pharmaceutical Research and Early Development, Clinical Pharmacology, Roche Innovation Centre Basel, Switzerland
| | - A Phipps
- Pharma Research and Early Development, Roche Innovation Centre Welwyn, 6 Falcon Way, Shire Park, Welwyn Garden City, AL7 1TW, UK
| | - H M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Radcliffe Observatory Quarter, Woodstock Road, Oxford OX2 6GG, UK
| |
Collapse
|
32
|
De Souza R, Spence T, Huang H, Allen C. Preclinical imaging and translational animal models of cancer for accelerated clinical implementation of nanotechnologies and macromolecular agents. J Control Release 2015; 219:313-330. [PMID: 26409122 DOI: 10.1016/j.jconrel.2015.09.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/22/2015] [Accepted: 09/22/2015] [Indexed: 01/08/2023]
Abstract
The majority of animal models of cancer have performed poorly in terms of predicting clinical performance of new therapeutics, which are most often first evaluated in patients with advanced, metastatic disease. The development and use of metastatic models of cancer may enhance clinical translatability of preclinical studies focused on the development of nanotechnology-based drug delivery systems and macromolecular therapeutics, potentially accelerating their clinical implementation. It is recognized that the development and use of such models are not without challenge. Preclinical imaging tools offer a solution by allowing temporal and spatial characterization of metastatic lesions. This paper provides a review of imaging methods applicable for evaluation of novel therapeutics in clinically relevant models of advanced cancer. An overview of currently utilized models of oncology in small animals is followed by image-based development and characterization of visceral metastatic cancer models. Examples of imaging tools employed for metastatic lesion detection, evaluation of anti-tumor and anti-metastatic potential and biodistribution of novel therapies, as well as the co-development and/or use of imageable surrogates of response, are also discussed. While the focus is on development of macromolecular and nanotechnology-based therapeutics, examples with small molecules are included in some cases to illustrate concepts and approaches that can be applied in the assessment of nanotechnologies or macromolecules.
Collapse
Affiliation(s)
- Raquel De Souza
- Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, Ontario M5S 3M2, Canada.
| | - Tara Spence
- Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, Ontario M5S 3M2, Canada
| | - Huang Huang
- DLVR Therapeutics, 661 University Avenue, Toronto, Ontario M5G 0A3, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, 144 College Street, Toronto, Ontario M5S 3M2, Canada.
| |
Collapse
|
33
|
Bailey KM, Cornnell HH, Ibrahim-Hashim A, Wojtkowiak JW, Hart CP, Zhang X, Leos R, Martinez GV, Baker AF, Gillies RJ. Evaluation of the "steal" phenomenon on the efficacy of hypoxia activated prodrug TH-302 in pancreatic cancer. PLoS One 2014; 9:e113586. [PMID: 25532146 PMCID: PMC4273999 DOI: 10.1371/journal.pone.0113586] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/26/2014] [Indexed: 11/19/2022] Open
Abstract
Pancreatic ductal adenocarcinomas are desmoplastic and hypoxic, both of which are associated with poor prognosis. Hypoxia-activated prodrugs (HAPs) are specifically activated in hypoxic environments to release cytotoxic or cytostatic effectors. TH-302 is a HAP that is currently being evaluated in a Phase III clinical trial in pancreatic cancer. Using animal models, we show that tumor hypoxia can be exacerbated using a vasodilator, hydralazine, improving TH-302 efficacy. Hydralazine reduces tumor blood flow through the "steal" phenomenon, in which atonal immature tumor vasculature fails to dilate in coordination with normal vasculature. We show that MIA PaCa-2 tumors exhibit a "steal" effect in response to hydralazine, resulting in decreased tumor blood flow and subsequent tumor pH reduction. The effect is not observed in SU.86.86 tumors with mature tumor vasculature, as measured by CD31 and smooth muscle actin (SMA) immunohistochemistry staining. Combination therapy of hydralazine and TH-302 resulted in a reduction in MIA PaCa-2 tumor volume growth after 18 days of treatment. These studies support a combination mechanism of action for TH-302 with a vasodilator that transiently increases tumor hypoxia.
Collapse
Affiliation(s)
- Kate M. Bailey
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida 33612, United States of America
| | - Heather H. Cornnell
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Arig Ibrahim-Hashim
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Jonathan W. Wojtkowiak
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Charles P. Hart
- Threshold Pharmaceuticals, South San Francisco, California 94080, United States of America
| | - Xiaomeng Zhang
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Rafael Leos
- Arizona Cancer Center, Hematology/Oncology Section, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States of America
| | - Gary V. Martinez
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
| | - Amanda F. Baker
- Arizona Cancer Center, Hematology/Oncology Section, College of Medicine, University of Arizona, Tucson, Arizona 85724, United States of America
| | - Robert J. Gillies
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, United States of America
- * E-mail:
| |
Collapse
|