1
|
Li M, Guo X, Verma A, Rudkouskaya A, McKenna AM, Intes X, Wang G, Barroso M. Contrast-enhanced photon-counting micro-CT of tumor xenograft models. Phys Med Biol 2024; 69:155011. [PMID: 38670143 PMCID: PMC11258216 DOI: 10.1088/1361-6560/ad4447] [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: 01/12/2024] [Revised: 04/11/2024] [Accepted: 04/26/2024] [Indexed: 04/28/2024]
Abstract
Objective. Photon-counting micro-computed tomography (micro-CT) is a major advance in small animal preclinical imaging. Small molecule- and nanoparticle-based contrast agents have been widely used to enable the differentiation of liver tumors from surrounding tissues using photon-counting micro-CT. However, there is a notable gap in the application of these market-available agents to the imaging of breast and ovarian tumors using photon-counting micro-CT. Herein, we have used photon-counting micro-CT to determine the effectiveness of these contrast agents in differentiating ovarian and breast tumor xenografts in live, intact mice.Approach. Nude mice carrying different types of breast and ovarian tumor xenografts (AU565, MDA-MB-231 and SKOV-3 human cancer cells) were injected with ISOVUE-370 (a small molecule-based agent) or Exitron Nano 12000 (a nanoparticle-based agent) and subjected to photon-counting micro-CT. To improve tumor visualization using photon-counting micro-CT, we developed a novel color visualization method, which changes color tones to highlight contrast media distribution, offering a robust alternative to traditional material decomposition methods with less computational demand.Main results. Ourin vivoexperiments confirm the effectiveness of this color visualization approach, showing distinct enhancement characteristics for each contrast agent. Qualitative and quantitative analyses suggest that Exitron Nano 12000 provides superior vasculature enhancement and better quantitative consistency across scans, while ISOVUE-370 delivers a more comprehensive tumor enhancement but with significant variance between scans due to its short blood half-time. Further, a paired t-test on mean and standard deviation values within tumor volumes showed significant differences between the AU565 and SKOV-3 tumor models with the nanoparticle-based contrast agent (p-values < 0.02), attributable to their distinct vascularity, as confirmed by immunohistochemical analysis.Significance. These findings underscore the utility of photon-counting micro-CT in non-invasively assessing the morphology and anatomy of different tumor xenografts, which is crucial for tumor characterization and longitudinal monitoring of tumor progression and response to treatments.
Collapse
Affiliation(s)
- Mengzhou Li
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Xiaodong Guo
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Amit Verma
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| | - Antigone M McKenna
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Ge Wang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, United States of America
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, United States of America
| |
Collapse
|
2
|
Ozaki M, Kageyama K, Kimura K, Eguchi S, Yamamoto A, Tanaka R, Nota T, Yonezawa H, Nishiofuku H, Sakai Y, Tani N, Jogo A, Terai M, Sato T, Ishizawa T, Miki Y. A rat-based preclinical platform facilitating transcatheter hepatic arterial infusion in immunodeficient rats with liver xenografts of patient-derived pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:10529. [PMID: 38719893 PMCID: PMC11079078 DOI: 10.1038/s41598-024-61142-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
Liver metastases from pancreatic ductal adenocarcinoma (PDAC) are highly fatal. A rat-based patient-derived tumor xenograft (PDX) model is available for transcatheter therapy. This study aimed to create an immunodeficient rat model with liver xenografts of patient-derived primary PDAC and evaluate efficacy of hepatic arterial infusion chemotherapy with cisplatin in this model. Three patient-derived PDACs were transplanted into the livers of 21 rats each (totally, 63 rats), randomly assigned into hepatic arterial infusion, systemic venous infusion, and control groups (n = 7 each) four weeks post-implantation. Computed tomography evaluated tumor volumes before and four weeks after treatment. Post-euthanasia, resected tumor specimens underwent histopathological examination. A liver-implanted PDAC PDX rat model was established in all 63 rats, with first CT identifying all tumors. Four weeks post-treatment, arterial infusion groups exhibited significantly smaller tumor volumes than controls for all three tumors on second CT. Xenograft tumors histologically maintained adenocarcinoma features compared to original patient tumors. Ki67 expression was significantly lower in arterial infusion groups than in the other two for the three tumors, indicating reduced tumor growth in PDX rats. A liver-implanted PDAC PDX rat model was established as a rat-based preclinical platform. Arterial cisplatin infusion chemotherapy represents a potential therapy for PDAC liver metastasis.
Collapse
Affiliation(s)
- Masanori Ozaki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Ken Kageyama
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan.
| | - Kenjiro Kimura
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Shinpei Eguchi
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Akira Yamamoto
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Ryota Tanaka
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Takehito Nota
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Hiroki Yonezawa
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Hideyuki Nishiofuku
- Department of Diagnostic and Interventional Radiology, Nara Medical University, 840 Shijocho, Kashihara, Nara, 6348521, Japan
| | - Yuki Sakai
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Naoki Tani
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Atsushi Jogo
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Mizue Terai
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, 1024 Curtis Building, Philadelphia, PA, 19107, USA
| | - Takami Sato
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, 1015 Walnut Street, 1024 Curtis Building, Philadelphia, PA, 19107, USA
| | - Takeaki Ishizawa
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| | - Yukio Miki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3, Asahimachi, Abenoku, Osaka, 5458585, Japan
| |
Collapse
|
3
|
Kronfeld A, Rose P, Baumgart J, Brockmann C, Othman AE, Schweizer B, Brockmann MA. Quantitative multi-energy micro-CT: A simulation and phantom study for simultaneous imaging of four different contrast materials using an energy integrating detector. Heliyon 2024; 10:e23013. [PMID: 38148814 PMCID: PMC10750148 DOI: 10.1016/j.heliyon.2023.e23013] [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: 03/30/2023] [Revised: 11/23/2023] [Accepted: 11/23/2023] [Indexed: 12/28/2023] Open
Abstract
Emerging from the development of single-energy Computed Tomography (CT) and Dual-Energy Computed Tomography, Multi-Energy Computed Tomography (MECT) is a promising tool allowing advanced material and tissue decomposition and thereby enabling the use of multiple contrast materials in preclinical research. The scope of this work was to evaluate whether a usual preclinical micro-CT system is applicable for the decomposition of different materials using MECT together with a matrix-inversion method and how different changes of the measurement-environment affect the results. A matrix-inversion based algorithm to differentiate up to five materials (iodine, iron, barium, gadolinium, residual material) by applying four different acceleration voltages/energy levels was established. We carried out simulations using different ratios and concentrations (given in fractions of volume units, VU) of the four different materials (plus residual material) at different noise-levels for 30 keV, 40 keV, 50 keV, 60 keV, 80 keV and 100 keV (monochromatic). Our simulation results were then confirmed by using region of interest-based measurements in a phantom-study at corresponding acceleration voltages. Therefore, different mixtures of contrast materials were scanned using a micro-CT. Voxel wise evaluation of the phantom imaging data was conducted to confirm its usability for future imaging applications and to estimate the influence of varying noise-levels, scattering, artifacts and concentrations. The analysis of our simulations showed the smallest deviation of 0.01 (0.003-0.15) VU between given and calculated concentrations of the different contrast materials when using an energy-combination of 30 keV, 40 keV, 50 keV and 100 keV for MECT. Subsequent MECT phantom measurements, however, revealed a combination of acceleration voltages of 30 kV, 40 kV, 60 kV and 100 kV as most effective for performing material decomposition with a deviation of 0.28 (0-1.07) mg/ml. The feasibility of our voxelwise analyses using the proposed algorithm was then confirmed by the generation of phantom parameter-maps that matched the known contrast material concentrations. The results were mostly influenced by the noise-level and the concentrations used in the phantoms. MECT using a standard micro-CT combined with a matrix inversion method is feasible at four different imaging energies and allows the differentiation of mixtures of up to four contrast materials plus an additional residual material.
Collapse
Affiliation(s)
- Andrea Kronfeld
- University Medical Center of the Johannes Gutenberg University Mainz, Department of Neuroradiology, Langenbeck 1, 55131, Mainz, Germany
| | - Patrick Rose
- University Medical Center of the Johannes Gutenberg University Mainz, Department of Neuroradiology, Langenbeck 1, 55131, Mainz, Germany
- RheinMain University of Applied Sciences, Faculty of Engineering, Am Brückweg 26, 65428, Rüsselsheim am Main, Germany
| | - Jan Baumgart
- University Medical Center of the Johannes Gutenberg University Mainz, Translational Animal Research Center, Hanns-Dieter-Hüsch-Weg 19, 55128, Mainz, Germany
| | - Carolin Brockmann
- University Medical Center of the Johannes Gutenberg University Mainz, Department of Neuroradiology, Langenbeck 1, 55131, Mainz, Germany
| | - Ahmed E. Othman
- University Medical Center of the Johannes Gutenberg University Mainz, Department of Neuroradiology, Langenbeck 1, 55131, Mainz, Germany
| | - Bernd Schweizer
- RheinMain University of Applied Sciences, Faculty of Engineering, Am Brückweg 26, 65428, Rüsselsheim am Main, Germany
| | - Marc Alexander Brockmann
- University Medical Center of the Johannes Gutenberg University Mainz, Department of Neuroradiology, Langenbeck 1, 55131, Mainz, Germany
| |
Collapse
|
4
|
Li M, Guo X, Verma A, Rudkouskaya A, McKenna AM, Intes X, Wang G, Barroso M. Contrast-enhanced photon-counting micro-CT of tumor xenograft models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574097. [PMID: 38260707 PMCID: PMC10802390 DOI: 10.1101/2024.01.03.574097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Photon-counting micro computed tomography (micro-CT) offers new potential in preclinical imaging, particularly in distinguishing materials. It becomes especially helpful when combined with contrast agents, enabling the differentiation of tumors from surrounding tissues. There are mainly two types of contrast agents in the market for micro-CT: small molecule-based and nanoparticle-based. However, despite their widespread use in liver tumor studies, there is a notable gap in research on the application of these commercially available agents for photon-counting micro-CT in breast and ovarian tumors. Herein, we explored the effectiveness of these agents in differentiating tumor xenografts from various origins (AU565, MDA-MB-231, and SKOV-3) in nude mice, using photon-counting micro-CT. Specifically, ISOVUE-370 (a small molecule-based agent) and Exitrone Nano 12000 (a nanoparticle-based agent) were investigated in this context. To improve tumor visualization, we proposed a novel color visualization method for photon-counting micro-CT, which changes color tones to highlight contrast media distribution, offering a robust alternative to traditional material decomposition methods with less computational demand. Our in vivo experiments confirm its effectiveness, showing distinct enhancement characteristics for each contrast agent. Qualitative and quantitative analyses suggested that Exitrone Nano 12000 provides superior vasculature enhancement and better quantitative consistency across scans, while ISOVUE-370 gives more comprehensive tumor enhancement but with a significant variance between scans due to its short blood half-time. This variability leads to high sensitivity to timing and individual differences among mice. Further, a paired t-test on mean and standard deviation values within tumor volumes showed significant differences between the AU565 and SKOV-3 tumor models with the nanoparticle-based (p-values < 0.02), attributable to their distinct vascularity, as confirmed by immunohistochemistry. These findings underscore the utility of photon-counting micro-CT in non-invasively assessing the morphology and anatomy of different tumor xenografts, which is crucial for tumor characterization and longitudinal monitoring of tumor development and response to treatments.
Collapse
Affiliation(s)
- Mengzhou Li
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Xiaodong Guo
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Amit Verma
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alena Rudkouskaya
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Antigone M. McKenna
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Xavier Intes
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Ge Wang
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
5
|
Peloso A, Lacotte S, Gex Q, Slits F, Moeckli B, Oldani G, Tihy M, Hautefort A, Kwak B, Rubbia-Brandt L, Toso C. Portosystemic shunting prevents hepatocellular carcinoma in non-alcoholic fatty liver disease mouse models. PLoS One 2023; 18:e0296265. [PMID: 38157359 PMCID: PMC10756526 DOI: 10.1371/journal.pone.0296265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/09/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver disease (NAFLD) is one of the leading cause of hepatocellular carcinoma (HCC). This association is supported by the translocation of bacteria products into the portal system, which acts on the liver through the gut-liver axis. We hypothesize that portosystemic shunting can disrupt this relationship, and prevent NAFLD-associated HCC. METHODS HCC carcinogenesis was tested in C57BL/6 mice fed a high-fat high-sucrose diet (HFD) and injected with diethylnitrosamine (DEN) at two weeks of age, and in double transgenic LAP-tTA and TRE-MYC (LAP-Myc) mice fed a methionine-choline-deficient diet. Portosystemic shunts were established by transposing the spleen to the sub-cutaneous tissue at eight weeks of age. RESULTS Spleen transposition led to a consistent deviation of part of the portal flow and a significant decrease in portal pressure. It was associated with a decrease in the number of HCC in both models. This effect was supported by the presence of less severe liver steatosis after 40 weeks, and lower expression levels of liver fatty acid synthase. Also, shunted mice exhibited lower liver oxygen levels, a key factor in preventing HCC as confirmed by the development of less HCCs in mice with hepatic artery ligation. CONCLUSIONS The present data show that portosystemic shunting prevents NAFLD-associated HCC, utilizing two independent mouse models. This effect is supported by the development of less steatosis, and a restored liver oxygen level. Portal pressure modulation and shunting deserve further exploration as potential prevention/treatment options for NAFLD and HCC.
Collapse
Affiliation(s)
- Andrea Peloso
- Division of Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| | - Stéphanie Lacotte
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| | - Quentin Gex
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| | - Florence Slits
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| | - Beat Moeckli
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| | - Graziano Oldani
- Division of Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| | - Matthieu Tihy
- Division of Clinical Pathology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Aurélie Hautefort
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Brenda Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Christian Toso
- Division of Abdominal Surgery, Department of Surgery, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
- Transplantation and Hepatology Laboratory, University of Geneva, Geneva, Switzerland
| |
Collapse
|
6
|
Zwiauer-Wolfbeisser V, Handschuh S, Tichy A, Nell B. Morphology and volume of Meibomian glands ex vivo pre and post partial tarsal plate excision, cryotherapy and laser therapy in the dog using microCT. Vet Ophthalmol 2023; 26 Suppl 1:98-108. [PMID: 36692053 DOI: 10.1111/vop.13057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To determine the morphology and volume of Meibomian glands (MG) of dogs with microCT before and after partial tarsal plate excision (PTPE), cryotherapy, and laser therapy. PROCEDURE MicroCT scans were made of 12 upper lids (ULs) and lower lids (LLs) of 12 dogs. After undergoing PTPE, 10 ULs and LLs were scanned again, and one UL and one LL was scanned after laser therapy and one UL and one LL after cryotherapy. RESULTS The length of the area containing MGs did not change pre- and post-PTPE, and cryo- or laser therapy. The mean number of MGs in the ULs and LLs was 30.50 and 29.42, respectively, and did not change during the procedures. The average length of one individual MG was 2.60 mm. The mean volume of MGs in the 10 ULs and LLs pre-PTPE was 21.45 and 17.2 mm3 , respectively, and 12.84 and 11.25 mm3 in the UL and LL after PTPE, respectively. The mean volume of MGs decreased from 29.78 mm3 precryotherapy to 28.91 mm3 post-treatment and in the lower eyelid from 22.87 to 22.4 mm3 after cryotherapy. The mean volume of MGs in the UL and LL before laser therapy was 8.95 and 6.78 mm3 , respectively, and after 9.25 and 6.38 mm3 , respectively. CONCLUSION MicroCT is a valuable tool to determine the morphology and the volume of MGs and to demonstrate changes that occur after PTPE, laser-, and cryotherapy. There is no need for additional preparation, such as staining, of the specimen prior to scanning.
Collapse
Affiliation(s)
| | - Stephan Handschuh
- VetCore Facility for Research, University of Veterinary Medicine, Vienna, Austria
| | - Alexander Tichy
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Barbara Nell
- Department of Companion Animals and Horses, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
7
|
Nota T, Kageyama K, Yamamoto A, Kakehashi A, Yonezawa H, Jogo A, Sohgawa E, Murai K, Ogawa S, Miki Y. Safety and Feasibility of Contrast-Enhanced Computed Tomography with a Nanoparticle Contrast Agent for Evaluation of Diethylnitrosamine-Induced Liver Tumors in a Rat Model. Acad Radiol 2023; 30:30-39. [PMID: 35680546 DOI: 10.1016/j.acra.2022.03.027] [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: 01/24/2022] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 11/17/2022]
Abstract
RATIONALE AND OBJECTIVES Safety and feasibility of contrast-enhanced computed tomography (CECT) with a nanoparticulate contrast agent, ExiTron nano 12000, was evaluated in a rat liver tumor model. MATERIALS AND METHODS This study employed eighteen 8-week-old male F344 rats. Six rats given tap water for 8 weeks further divided into two: Control group and Normal Liver with CECT group. Six rats each were given tap water containing diethylnitrosamine (DEN) at 100 ppm for 8 or 14 weeks; Adenoma group and Hepatocellular carcinoma (HCC) group, respectively. Biochemical marker values and adverse events were evaluated after CT imaging. ExiTron nano 12000 was evaluated for the hepatic contrast enhancement, and the detection and measurement of liver nodules by CECT after 8- and 14-weeks administration of DEN. Post-mortem liver specimens were evaluated by hematoxylin-eosin (HE) staining, and the number and size of liver nodules were measured. The HCC group was evaluated for diagnostic concordance between HE-stained and CECT-detected nodules. RESULTS The contrast agent enhanced liver and was tolerated after CECT in 15 rats. Biochemical parameter values did not differ significantly between the Control and Normal Liver groups. The numbers of CECT-detected nodules in the Adenoma and HCC groups were 14.8 ± 5.1, and 32.4 ± 8.1, respectively. The HCC group had 3.6 ± 2.7 of pathological HCCs, which were identified by CECT. The size of CECT-detected HCCs correlated significantly with that of pathological HCCs (r = 0.966, p < 0.0001). CONCLUSION CECT with ExiTron nano 12000 is a safe and feasible method to measure tumors in a rat liver tumor model.
Collapse
Affiliation(s)
- Takehito Nota
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Ken Kageyama
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan.
| | - Akira Yamamoto
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Anna Kakehashi
- Department of Molecular Pathology (A.K.), Graduate School of Medicine, Osaka City University (currently Osaka Metropolitan University), Abenoku, Osaka, Japan
| | - Hiroki Yonezawa
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Atsushi Jogo
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Etsuji Sohgawa
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Kazuki Murai
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Satoyuki Ogawa
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| | - Yukio Miki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine (T.N., K.K., A.Y., H.Y., A.J., E.S., K.M., S.O., Y.M.), Osaka City University (currently Osaka Metropolitan University), 1-4-3 Asahimachi, Abenoku, Osaka 545-8585, Japan
| |
Collapse
|
8
|
Thamm M, Rosenhain S, Leonardic K, Höfter A, Kiessling F, Osl F, Pöschinger T, Gremse F. Intrinsic Respiratory Gating for Simultaneous Multi-Mouse μCT Imaging to Assess Liver Tumors. Front Med (Lausanne) 2022; 9:878966. [PMID: 35872758 PMCID: PMC9299429 DOI: 10.3389/fmed.2022.878966] [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: 02/18/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Small animal micro computed tomography (μCT) is an important tool in cancer research and is used to quantify liver and lung tumors. A type of cancer that is intensively investigated with μCT is hepatocellular carcinoma (HCC). μCT scans acquire projections from different angles of the gantry which rotates X-ray source and detector around the animal. Motion of the animal causes inconsistencies between the projections which lead to artifacts in the resulting image. This is problematic in HCC research, where respiratory motion affects the image quality by causing hypodense intensity at the liver edge and smearing out small structures such as tumors. Dealing with respiratory motion is particularly difficult in a high throughput setting when multiple mice are scanned together and projection removal by retrospective respiratory gating may compromise image quality and dose efficiency. In mice, inhalation anesthesia leads to a regular respiration with short gasps and long phases of negligible motion. Using this effect and an iterative reconstruction which can cope with missing angles, we discard the relatively few projections in which the gasping motion occurs. Moreover, since gated acquisition, i.e., acquiring multiple projections from a single gantry angle is not a requirement, this method can be applied to existing scans. We applied our method in a high throughput setting in which four mice with HCC tumors were scanned simultaneously in a multi-mouse bed. To establish a ground truth, we manually selected projections with visible respiratory motion. Our automated intrinsic breathing projection selection achieved an accordance of 97% with manual selection. We reconstructed volumetric images and demonstrated that our intrinsic gating method significantly reduces the hypodense depiction at the cranial liver edge and improves the detectability of small tumors. Furthermore, we show that projection removal in a four mice scan discards only 7.5% more projections than in a single-mouse setting, i.e., four mouse scanning does not substantially compromise dose efficiency or image quality. To the best of our knowledge, no comparable method that combines multi-mouse scans for high throughput, intrinsic respiratory gating, and an available iterative reconstruction has been described for liver tumor imaging before.
Collapse
Affiliation(s)
- Mirko Thamm
- Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
- Gremse-IT GmbH, Aachen, Germany
| | | | | | | | - Fabian Kiessling
- Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Franz Osl
- Discovery Pharmacology, Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Thomas Pöschinger
- Discovery Pharmacology, Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Felix Gremse
- Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
- Gremse-IT GmbH, Aachen, Germany
- *Correspondence: Felix Gremse
| |
Collapse
|
9
|
Liu RL, Cai RQ. Recent advances in ultrasound-controlled fluorescence technology for deep tissue optical imaging. J Pharm Anal 2021; 12:530-540. [PMID: 36105171 PMCID: PMC9463483 DOI: 10.1016/j.jpha.2021.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/13/2021] [Accepted: 10/09/2021] [Indexed: 12/01/2022] Open
Abstract
Fluorescence imaging is a noninvasive and dynamic real-time imaging technique; however, it exhibits poor spatial resolution in centimeter-deep tissues because biological tissues are highly scattering media for optical radiation. The recently developed ultrasound-controlled fluorescence (UCF) imaging is a novel imaging technique that can overcome this bottleneck. Previous studies suggest that the effective contrast agent and sensitive imaging system are the two pivotal factors for generating high-resolution UCF images ex vivo and/or in vivo. Here, this review highlights the recent advances (2015–2020) in the design and synthesis of contrast agents and the improvement of imaging systems to realize high-resolution UCF imaging of deep tissues. The imaging performances of various UCF systems, including the signal-to-noise ratio, imaging resolution, and imaging depth, are specifically discussed. In addition, the challenges and prospects are highlighted. With continuously increasing research interest in this field and emerging multidisciplinary applications, UCF imaging with higher spatial resolution and larger imaging depth may be developed shortly, which is expected to have a far-reaching impact on disease surveillance and/or therapy.
The unique UCF technique in high resolution imaging was described. The contrast agents and imaging systems of UCF imaging were discussed. Recent advances and prospects of NIR-UCF technique were summarized.
Collapse
Affiliation(s)
- Rui-Lin Liu
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
- Corresponding author. School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Ru-Qian Cai
- School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, China
| |
Collapse
|
10
|
PEG-modified gadolinium nanoparticles as contrast agents for in vivo micro-CT. Sci Rep 2021; 11:16603. [PMID: 34400681 PMCID: PMC8367985 DOI: 10.1038/s41598-021-95716-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/09/2021] [Indexed: 12/30/2022] Open
Abstract
Vascular research is largely performed in rodents with the goal of developing treatments for human disease. Micro-computed tomography (micro-CT) provides non-destructive three-dimensional imaging that can be used to study the vasculature of rodents. However, to distinguish vasculature from other soft tissues, long-circulating contrast agents are required. In this study, we demonstrated that poly(ethylene glycol) (PEG)-coated gadolinium nanoparticles can be used as a vascular contrast agent in micro-CT. The coated particles could be lyophilized and then redispersed in an aqueous solution to achieve 100 mg/mL of gadolinium. After an intravenous injection of the contrast agent into mice, micro-CT scans showed blood pool contrast enhancements of at least 200 HU for 30 min. Imaging and quantitative analysis of gadolinium in tissues showed the presence of contrast agent in clearance organs including the liver and spleen and very low amounts in other organs. In vitro cell culture experiments, subcutaneous injections, and analysis of mouse body weight suggested that the agents exhibited low toxicity. Histological analysis of tissues 5 days after injection of the contrast agent showed cytotoxicity in the spleen, but no abnormalities were observed in the liver, lungs, kidneys, and bladder.
Collapse
|
11
|
Sabel BO, Brand K, Rueckel J, Hoppe B, Fink N, Bartling S. Macrophage ablation significantly reduces uptake of imaging probe into organs of the reticuloendothelial system. Acta Radiol 2021; 62:882-889. [PMID: 32772706 DOI: 10.1177/0284185120943048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Macrophages engulf particulate contrast media, which is pivotal for biomedical imaging. PURPOSE To introduce a macrophage ablation animal model by showing its power to manipulate the kinetics of imaging probes. MATERIAL AND METHODS The kinetics of a particulate computed tomography (CT) contrast media was compared in macrophage ablative mice and normal mice. Liposomes (size 220 µg), loaded with clodronate, were injected into the peritoneum of three C57BL/6 mice. On the third day, 200 µL of the particulate agent ExiTron nano 6000 were injected into three macrophage-ablative mice and three control mice. CT scans were acquired before and 3 min, 1 h, 6 h, and 24 h after the ExiTron application. The animals were sacrificed, and their spleens and livers removed. Relative CT values (CTV) were measured and analyzed. RESULTS Liver and spleen enhancement of treated mice and controls were increasing over time. The median peak values were different with 225 CTV for treated mice and 582 CTV for controls in the liver (P = 0.032) and 431 CTV for treated and 974 CTV in controls in the spleen (P = 0.016). CONCLUSION Macrophage ablation leads to a decrease of enhancement in organs containing high numbers of macrophages, but only marginal changes in macrophage-poor organs. Macrophage ablation can influence the phagocytic activity and thus opens new potentials to investigate and manipulate the uptake of imaging probes.
Collapse
Affiliation(s)
- Bastian O Sabel
- Department of Radiology, German Cancer Research Center, Heidelberg, Germany
- Institute for Clinical Radiology, University Hospital Munich, Munich, Germany
| | | | - Johannes Rueckel
- Institute for Clinical Radiology, University Hospital Munich, Munich, Germany
| | - Boj Hoppe
- Institute for Clinical Radiology, University Hospital Munich, Munich, Germany
| | - Nicola Fink
- Institute for Clinical Radiology, University Hospital Munich, Munich, Germany
| | - Soenke Bartling
- Department of Radiology, German Cancer Research Center, Heidelberg, Germany
- Department of Clinical Radiology and Nuclear Medicine, Mannheim University Medical Center, Mannheim, Germany
| |
Collapse
|
12
|
Radiation-Induced Metabolic Shifts in the Hepatic Parenchyma: Findings from 18F-FDG PET Imaging and Tissue NMR Metabolomics in a Mouse Model for Hepatocellular Carcinoma. Molecules 2021; 26:molecules26092573. [PMID: 33925109 PMCID: PMC8125521 DOI: 10.3390/molecules26092573] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose: By taking advantage of 18F-FDG PET imaging and tissue nuclear magnetic resonance (NMR) metabolomics, we examined the dynamic metabolic alterations induced by liver irradiation in a mouse model for hepatocellular carcinoma (HCC). Methods: After orthotopic implantation with the mouse liver cancer BNL cells in the right hepatic lobe, animals were divided into two experimental groups. The first received irradiation (RT) at 15 Gy, while the second (no-RT) did not. Intergroup comparisons over time were performed, in terms of 18F-FDG PET findings, NMR metabolomics results, and the expression of genes involved in inflammation and glucose metabolism. Results: As of day one post-irradiation, mice in the RT group showed an increased 18F-FDG uptake in the right liver parenchyma compared with the no-RT group. However, the difference reached statistical significance only on the third post-irradiation day. NMR metabolomics revealed that glucose concentrations peaked on day one post-irradiation both, in the right and left lobes—the latter reflecting a bystander effect. Increased pyruvate and glutamate levels were also evident in the right liver on the third post-irradiation day. The expression levels of the glucose-6-phosphatase (G6PC) and fructose-1, 6-bisphosphatase 1 (FBP1) genes were down-regulated on the first and third post-irradiation days, respectively. Therefore, liver irradiation was associated with a metabolic shift from an impaired gluconeogenesis to an enhanced glycolysis from the first to the third post-irradiation day. Conclusion: Radiation-induced metabolic alterations in the liver parenchyma occur as early as the first post-irradiation day and show dynamic changes over time.
Collapse
|
13
|
Vøls KK, Kjelgaard‐Hansen M, Ley CD, Hansen AK, Petersen M. Initial joint bleed volume in a delayed on-demand treatment setup correlates with subsequent synovial changes in hemophilic mice. Animal Model Exp Med 2020; 3:160-168. [PMID: 32613175 PMCID: PMC7323705 DOI: 10.1002/ame2.12118] [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: 01/20/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Hemophilic arthropathy is a debilitating morbidity of hemophilia caused by recurrent joint bleeds. We investigated if the joint bleed volume, before initiation of treatment, was linked to the subsequent degree of histopathological changes and the development of bone pathology in a mouse model of hemophilic arthropathy. METHODS FVIII knock-out (F8-KO) mice were dosed with a micro-CT blood pool agent prior to induction of hemarthrosis. Eight hours after induction, the bleed volume was quantified with micro computed tomography (micro-CT) and recombinant FVIII treatment initiated. On Day 8, inflammation in the knees was characterized by fluorescence molecular tomography. On Day 14, knee pathology was characterized by micro-CT and histopathology. In a second study, contrast agent was injected into the knee of wild-type (WT) mice, followed by histopathological evaluation on Day 14. RESULTS The average joint bleed volume before treatment was 3.9 mm3. The inflammation-related fluorescent intensities in the injured knees were significantly increased on Day 8. The injured knees had significantly increased synovitis scores, vessel counts, and areas of hemosiderin compared to un-injured knees. However, no cartilage- or bone pathology was observed. The bleed volume before initiation of treatment correlated with the degree of synovitis and was associated with high fluorescent intensity on Day 8. In F8-KO and WT mice, persistence of contrast agent in the joint elicited morphological changes. CONCLUSION When applying a delayed on-demand treatment regimen to hemophilic mice subjected to an induced knee hemarthrosis, the degree of histopathological changes on Day 14 reflected the bleed volume prior to initiation of treatment.
Collapse
Affiliation(s)
- Kåre Kryger Vøls
- Global Drug DiscoveryNovo Nordisk A/SMaaloevDenmark
- Veterinary and Animal SciencesUniversity of CopenhagenFrederiksbergDenmark
| | | | | | | | - Maj Petersen
- Global Drug DiscoveryNovo Nordisk A/SMaaloevDenmark
| |
Collapse
|
14
|
El-Safty S, Shenashen M. Nanoscale dynamic chemical, biological sensor material designs for control monitoring and early detection of advanced diseases. Mater Today Bio 2020; 5:100044. [PMID: 32181446 PMCID: PMC7066237 DOI: 10.1016/j.mtbio.2020.100044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/25/2022] Open
Abstract
Early detection and easy continuous monitoring of emerging or re-emerging infectious, contagious or other diseases are of particular interest for controlling healthcare advances and developing effective medical treatments to reduce the high global cost burden of diseases in the backdrop of lack of awareness regarding advancing diseases. Under an ever-increasing demand for biosensor design reliability for early stage recognition of infectious agents or contagious diseases and potential proteins, nanoscale manufacturing designs had developed effective nanodynamic sensing assays and compact wearable devices. Dynamic developments of biosensor technology are also vital to detect and monitor advanced diseases, such as human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), diabetes, cancers, liver diseases, cardiovascular diseases (CVDs), tuberculosis, and central nervous system (CNS) disorders. In particular, nanoscale biosensor designs have indispensable contribution to improvement of health concerns by early detection of disease, monitoring ecological and therapeutic agents, and maintaining high safety level in food and cosmetics. This review reports an overview of biosensor designs and their feasibility for early investigation, detection, and quantitative determination of many advanced diseases. Biosensor strategies are highlighted to demonstrate the influence of nanocompact and lightweight designs on accurate analyses and inexpensive sensing assays. To date, the effective and foremost developments in various nanodynamic designs associated with simple analytical facilities and procedures remain challenging. Given the wide evolution of biosensor market requirements and the growing demand in the creation of early stage and real-time monitoring assays, precise output signals, and easy-to-wear and self-regulating analyses of diseases, innovations in biosensor designs based on novel fabrication of nanostructured platforms with active surface functionalities would produce remarkable biosensor devices. This review offers evidence for researchers and inventors to focus on biosensor challenge and improve fabrication of nanobiosensors to revolutionize consumer and healthcare markets.
Collapse
Affiliation(s)
- S.A. El-Safty
- National Institute for Materials Science (NIMS), 1-2-1 Sengen, Tsukubashi, Ibaraki-ken, 305-0047, Japan
| | | |
Collapse
|
15
|
Tracking Dynamics of Spontaneous Tumors in Mice Using Photon-Counting Computed Tomography. iScience 2019; 21:68-83. [PMID: 31655257 PMCID: PMC6820243 DOI: 10.1016/j.isci.2019.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Computed tomography is a powerful medical imaging modality for longitudinal studies in cancer to follow neoplasia progression and evaluate anticancer therapies. Here, we report the generation of a photon-counting micro-computed tomography (PC-CT) method based on hybrid pixel detectors with enhanced sensitivity and precision of tumor imaging. We then applied PC-CT for longitudinal imaging in a clinically relevant liver cancer model, the Alb-R26Met mice, and found a remarkable heterogeneity in the dynamics for tumors at the initiation phases. Instead, the growth curve of evolving tumors exhibited a comparable exponential growth, with a constant doubling time. Furthermore, longitudinal PC-CT imaging in mice treated with a combination of MEK and BCL-XL inhibitors revealed a drastic tumor regression accompanied by a striking remodeling of macrophages in the tumor microenvironment. Thus, PC-CT is a powerful system to detect cancer initiation and progression, and to monitor its evolution during treatment.
Development of photon-counting micro-computed tomography (PC-CT) with hybrid pixels PC-CT allows longitudinal imaging of tumor dynamics in mouse cancer models RTK-driven tumors are heterogeneous at onset, but grow steadily during progression MEK + BCL-XL targeting leads to tumor regression and microenvironment remodeling
Collapse
|
16
|
Wang Z, Kuang G, Yu Z, Li A, Zhou D, Huang Y. Light-activatable dual prodrug polymer nanoparticle for precise synergistic chemotherapy guided by drug-mediated computed tomography imaging. Acta Biomater 2019; 94:459-468. [PMID: 31128323 DOI: 10.1016/j.actbio.2019.05.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/09/2019] [Accepted: 05/19/2019] [Indexed: 12/22/2022]
Abstract
The synergistic efficacy and clinical application of light-responsive polymeric co-delivery systems are severely restricted by uncontrollable/imprecise drug loading, release, and adverse effects caused by the introduction of additional light-responsive molecules or contrast agents when diagnostic imaging is applied to guide therapy. Here, we report the design of a light-activatable dual prodrug polymer nanoparticle (DPP NP) for precise synergistic chemotherapy guided by drug-mediated computed tomography (DMCT) imaging without the introduction of any additional diagnostic imaging agent. DPP NP enables visible light-triggered prodrug polymer backbone cleavage and bioactive Pt(II) release in cancer cell/tumor site; the light-cleaved polymer fragments are further hydrolyzed to produce demethyl cantharidin (DMC). Notably, the drug loading ratio of Pt(IV) and DMC in DPP NP was fixed at an optimal value to achieve maximum synergistic cancer cell killing, which was kept even after cellular uptake, thereby resulting in enhanced synergistic antitumor efficacy both in vitro and in vivo. Because of the high content of the heavy metal Pt in the polymer chain, the spatial/temporal dynamic biodistribution as well as metabolism of DPP NP in vivo can be monitored by Pt DMCT imaging to guide the light irradiation parameters for optimized light-activatable synergistic chemotherapy. Guided by Pt DMCT imaging, DPP NP was able to achieve an improved light-activatable antitumor efficacy, with 75% tumors fully cured and low toxicity. The light-activatable DDP NP system exhibits tremendous potential as precise theranostic nanomedicine. STATEMENT OF SIGNIFICANCE: The synergistic efficacy and clinical application of light-responsive polymeric co-delivery systems are severely restricted by uncontrollable/imprecise drug loading, delivery, and release, as well as adverse effects caused by the introduction of additional light-responsive molecules or contrast agents when diagnostic imaging is applied to guide therapy. Herein, we report the design of a light-activatable dual prodrug polymer nanoparticle (DPP NP) for precise synergistic chemotherapy guided by drug-mediated computed tomography imaging without the introduction of any additional diagnostic imaging agents. Notably, the drug loading ratio of Pt(II) and DMC in DPP NP was fixed at an optimal value to achieve maximum synergistic cancer cell killing, which was kept even after cellular uptake, thereby resulting in enhanced synergistic antitumor efficacy both in vitro and in vivo. The light-activatable DDP NP system exhibits tremendous potential as precise theranostic nanomedicine.
Collapse
Affiliation(s)
- Zigui Wang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Gaizhen Kuang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, PR China
| | - Zhiqiang Yu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Aimin Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, PR China.
| | - Dongfang Zhou
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Yubin Huang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China.
| |
Collapse
|
17
|
Abstract
The conventional fluorescence imaging has limited spatial resolution in centimeter-deep tissue because of the tissue's high scattering property. Ultrasound-switchable fluorescence (USF) imaging, a new imaging technique, was recently proposed to realize high-resolution fluorescence imaging in centimeter-deep tissue. However, in vivo USF imaging has not been achieved so far because of the lack of stable near-infrared contrast agents in a biological environment and the lack of data about their biodistributions. In this study, for the first time, we achieved in vivo USF imaging successfully in mice with high resolution. USF imaging in porcine heart tissue and mouse breast tumor via local injections were studied and demonstrated. In vivo and ex vivo USF imaging of the mouse spleen via intravenous injections was also successfully achieved. The results showed that the USF contrast agent adopted in this study was very stable in a biological environment, and it was mainly accumulated into the spleen of the mice. By comparing the results of CT imaging and the results of USF imaging, the accuracy of USF imaging was proved.
Collapse
Affiliation(s)
- Tingfeng Yao
- Ultrasound and Optical Imaging Laboratory, Department of Bioengineering, The University of Texas at Arlington, Arlington, TX, 76019, USA
- Joint Biomedical Engineering Program, The University of Texas at Arlington and The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shuai Yu
- Ultrasound and Optical Imaging Laboratory, Department of Bioengineering, The University of Texas at Arlington, Arlington, TX, 76019, USA
- Joint Biomedical Engineering Program, The University of Texas at Arlington and The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yang Liu
- Ultrasound and Optical Imaging Laboratory, Department of Bioengineering, The University of Texas at Arlington, Arlington, TX, 76019, USA
- Joint Biomedical Engineering Program, The University of Texas at Arlington and The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Baohong Yuan
- Ultrasound and Optical Imaging Laboratory, Department of Bioengineering, The University of Texas at Arlington, Arlington, TX, 76019, USA.
- Joint Biomedical Engineering Program, The University of Texas at Arlington and The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
18
|
In Vivo Detection and Measurement of Aortic Aneurysm and Dissection in Mouse Models Using Microcomputed Tomography with Contrast Agent. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:5940301. [PMID: 30956627 PMCID: PMC6431409 DOI: 10.1155/2019/5940301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/11/2019] [Indexed: 02/03/2023]
Abstract
Objectives The aim of this study was to evaluate the potential of microcomputed tomography (micro-CT) using the intravascular contrast agent ExiTron nano 12000 for aorta imaging and monitoring the dynamic changing process of the aorta in mouse models with aortic aneurysm and dissection. Materials and Methods Experiments were performed on healthy mice and mice with aortic dissection. Mice that were developing aortic dissection and healthy mice underwent micro-CT imaging after injection of ExiTron nano 12000. Time-dependent signal enhancement (at 1, 2, 3, 6, and 12 hours after intravenous injection of the contrast agent, respectively) in the aorta of healthy mice was measured to confirm the optimal imaging time of aorta. Various contrast agent doses (70, 100, and 150 μl per 25 g mouse, respectively) were investigated to determine the optimal required dose for imaging of the aorta. The mice were scanned with micro-CT at 1, 14, and 28 days after onset of aneurysm and dissection to monitor the dynamic changing process of the aorta. Mouse aortas were stained with hematoxylin and eosin staining, and the diameter of the aorta was measured and compared with those obtained by micro-CT. Results Time-dependent signal enhancement in the aorta shows that the contrast agent has a long blood half-life of 6 hours, with a peak enhancement at 2 hours after injection. Injection of 100 μl ExiTron nano 12000 per 25 g mouse allows for effective visualization of the aorta. Micro-CT combined with contrast agent can monitor the changing process of the aorta in the mouse model of aortic aneurysm and dissection dynamically. The values of the diameter of the aortas obtained from the in vivo micro-CT imaging were compared with those obtained from histology and showed a significant correlation (R2 = 0.96). Conclusions These data demonstrate that in vivo micro-CT is an accurate and feasible technique to detect aortic aneurysm or dissection in a mouse model, and the micro-CT technique using the innovative contrast agent ExiTron nano 12000 allows for monitoring various processes dynamically such as aortic remodeling in longitudinal studies.
Collapse
|
19
|
Liu CN, Morin J, Dokmanovich M, Bluette CT, Goldstein R, Manickam B, Bagi CM. Nanoparticle contrast-enhanced micro-CT: A preclinical tool for the 3D imaging of liver and spleen in longitudinal mouse studies. J Pharmacol Toxicol Methods 2019; 96:67-77. [PMID: 30738209 DOI: 10.1016/j.vascn.2019.02.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 01/04/2023]
Abstract
In drug discovery and development, X-ray micro-computed tomography (micro-CT) has gained increasing importance over the past decades. In recent years, micro-CT imaging of soft tissues has become popular due to the introduction of a variety of radiopaque contrast agents. More recently, nanoparticle-based ExiTron nano 12,000 has become commercially available for the nonclinical micro-CT imaging of soft tissues in rodents. Phagocytosis and accumulation of the contrast agent by Kupffer cells in the liver, as well as macrophages in the spleen, increase the soft tissue X-ray attenuation for up to 6 months. Therefore, it is essential to understand the potential toxicity of this nanomaterial in micro-CT imaging prior to its application in pharmacology and/or toxicology studies. Herein, we describe the time-course and distribution of the contrast in the liver, spleen and blood after a single intravenous injection (IV) of this nanoparticle contrast agent at 0.1 ml/mouse. Thoracic images of male adult C57BL/6 mice were acquired using a Bruker SkyScan 1276 micro-CT over a period of 29 days. The stability of X-ray attenuation enhancement in the above tissues was also tested after a single dose of Kupffer cell toxicant gadolinium chloride (GdCl3) at 15 mg/kg on day 2. The liver, spleen and kidney were examined microscopically on days 15 and 29 post treatment. Serum and liver cytokines (IL-1β, IL-2, IL-6, IL-10, IL-12p70, IFN-γ, IP-10, MIP1-α, MIP1-β and TNF-α) were quantified on days 15 and 29 as indicators of a pro-inflammatory response to treatment. This study determined that there was an accumulation of amphophilic granular material in the cells of the mononuclear phagocyte system in the liver and spleen following a single dose of ExiTron nano 12,000 and a second dose of GdCl3 or its vehicle. However, ExiTron nano12000 contrast administration did not cause any hepatotoxicity in the liver, nor did pro-inflammatory cytokines release in the liver or serum. Similarly, there were no adverse pathologies in the spleen or kidneys. In summary, ExiTron nano12000 contrast agent-enhanced micro-CT could be used as a safe method in up to 29-day longitudinal efficacy and toxicology mouse studies for the non-invasive assessment of the liver and spleen.
Collapse
Affiliation(s)
- Chang-Ning Liu
- Comparative Medicine, Pfizer Worldwide R&D, Groton, CT 06340, USA.
| | | | | | | | | | | | - Cedo M Bagi
- Comparative Medicine, Pfizer Worldwide R&D, Groton, CT 06340, USA
| |
Collapse
|
20
|
Akimoto J, Nakayama M, Takagi S, Okano T. Efficient intrahepatic tumor generation by cell sheet transplantation to fabricate orthotopic hepatocarcinoma-bearing model mice for drug testing. J Biomed Mater Res A 2019; 107:1071-1079. [PMID: 30706672 DOI: 10.1002/jbm.a.36641] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 01/24/2019] [Indexed: 11/10/2022]
Abstract
Subcutaneous tumor-bearing mice are commonly used to evaluate antitumor activity in preclinical studies of anticancer drugs. However, these models often exhibit excessive antitumor responses to anticancer drug candidates. In this study, intrahepatic tumor-bearing mice as orthotopic tumor models were fabricated by transplanting hepatocarcinoma cell monolayers (sheets) to investigate differences in ectopic versus orthotopic antitumor response. Cell sheets, harvested from temperature-responsive cell culture dishes using thin gelatin gel supporters, were transferred onto mouse liver surfaces. Cell sheet transplantation drastically improved intrahepatic tumor formation compared with direct intrahepatic injection of dispersed cells. In particular, all cell sheet-transplanted mice formed well-developed tumors inside the liver following removal of the mesothelial membrane at the liver surface. Notably, these mice exhibited comparable life spans, indicating similar intrahepatic tumor development rates. Antitumor activity of doxorubicin (DOX) was examined using both subcutaneous and intrahepatic tumor-bearing mice. Although DOX administration yielded decreased subcutaneous tumor volumes, intrahepatic tumors exhibited no significant antitumor response. The results were considered to represent pharmacokinetic and histological structure differences between ectopic and orthotopic tumors, and partially supported the clinical uses of DOX. Therefore, cancer cell sheet transplantation constitutes a promising method to fabricate intrahepatic tumor-bearing mice for drug screening test in preclinical studies. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1071-1079, 2019.
Collapse
Affiliation(s)
- Jun Akimoto
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Masamichi Nakayama
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Soichi Takagi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), Kawada-cho 8-1, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
21
|
Vøls KK, Kjelgaard-Hansen M, Ley CD, Hansen AK, Petersen M. Bleed volume of experimental knee haemarthrosis correlates with the subsequent degree of haemophilic arthropathy. Haemophilia 2019; 25:324-333. [PMID: 30648774 DOI: 10.1111/hae.13672] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Haemophilic arthropathy is the main morbidity of haemophilia. The individual pathological response to the same number of clinically evident joint bleeds is highly variable; thus, it remains unknown if certain joint bleeding characteristics are critical for the development of arthropathy. AIM To study the relation between bleed volume and subsequent development of arthropathy, we aimed to develop quantitative in vivo imaging of active joint bleeds in a mouse model of haemophilia. METHODS Haemophilia A (F8-KO) and wild-type (WT) mice were IV-dosed with a micro-CT blood pool contrast agent prior to an induced knee haemarthrosis or sham procedure. The mice were micro-CT scanned five times the following 2 days to characterise and quantify the induced haemarthrosis in vivo. On Day 14, the mice were euthanized and pathological changes evaluated by histology and micro-CT. Additionally, bleeding characteristics in vehicle-treated F8-KO mice were compared with those of recombinant FVIII (rFVIII)-treated F8-KO mice. RESULTS F8-KO mice had a significantly larger bleed volume than WT mice at all scan time points. The bleed volume 12 hours after induction of haemarthrosis correlated with the subsequent degree of arthropathy. Presence of µCT-detectable bone pathology was associated with a significantly increased bleed volume among F8-KO mice. rFVIII treatment significantly reduced bleed volume in F8-KO mice. CONCLUSION Quantitative in vivo contrast-enhanced micro-CT imaging can be used to characterize and quantify joint bleeds in a mouse model of haemophilic arthropathy. The bleed volume correlates with the subsequent degree of arthropathy.
Collapse
Affiliation(s)
- Kåre Kryger Vøls
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark.,Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | | | - Axel Kornerup Hansen
- Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maj Petersen
- Global Drug Discovery, Novo Nordisk A/S, Maaloev, Denmark
| |
Collapse
|
22
|
A preclinical micro-computed tomography database including 3D whole body organ segmentations. Sci Data 2018; 5:180294. [PMID: 30561432 PMCID: PMC6298256 DOI: 10.1038/sdata.2018.294] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022] Open
Abstract
The gold-standard of preclinical micro-computed tomography (μCT) data processing is still manual delineation of complete organs or regions by specialists. However, this method is time-consuming, error-prone, has limited reproducibility, and therefore is not suitable for large-scale data analysis. Unfortunately, robust and accurate automated whole body segmentation algorithms are still missing. In this publication, we introduce a database containing 225 murine 3D whole body μCT scans along with manual organ segmentation of most important organs including heart, liver, lung, trachea, spleen, kidneys, stomach, intestine, bladder, thigh muscle, bone, as well as subcutaneous tumors. The database includes native and contrast-enhanced, regarding spleen and liver, μCT data. All scans along with organ segmentation are freely accessible at the online repository Figshare. We encourage researchers to reuse the provided data to evaluate and improve methods and algorithms for accurate automated organ segmentation which may reduce manual segmentation effort, increase reproducibility, and even reduce the number of required laboratory animals by reducing a source of variability and having access to a reliable reference group.
Collapse
|
23
|
Groza D, Gehrig S, Kudela P, Holcmann M, Pirker C, Dinhof C, Schueffl HH, Sramko M, Hoebart J, Alioglu F, Grusch M, Ogris M, Lubitz W, Keppler BK, Pashkunova-Martic I, Kowol CR, Sibilia M, Berger W, Heffeter P. Bacterial ghosts as adjuvant to oxaliplatin chemotherapy in colorectal carcinomatosis. Oncoimmunology 2018; 7:e1424676. [PMID: 29721389 DOI: 10.1080/2162402x.2018.1424676] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/29/2017] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and a major cause of cancer mortality worldwide. At late stage of the disease CRC often shows (multiple) metastatic lesions in the peritoneal cavity which cannot be efficiently targeted by systemic chemotherapy. This is one major factor contributing to poor prognosis. Oxaliplatin is one of the most commonly used systemic treatment options for advanced CRC. However, drug resistance - often due to insufficient drug delivery - is still hampering successful treatment. The anticancer activity of oxaliplatin includes besides DNA damage also a strong immunogenic component. Consequently, the aim of this study was to investigate the effect of bacterial ghosts (BGs) as adjuvant immunostimulant on oxaliplatin efficacy. BGs are empty envelopes of gram-negative bacteria with a distinct immune-stimulatory potential. Indeed, we were able to show that the combination of BGs with oxaliplatin treatment had strong synergistic anticancer activity against the CT26 allograft, resulting in prolonged survival and even a complete remission in this murine model of CRC carcinomatosis. This synergistic effect was based on an enhanced induction of immunogenic cell death and activation of an efficient T-cell response leading to long-term anti-tumor memory effects. Taken together, co-application of BGs strengthens the immunogenic component of the oxaliplatin anticancer response and thus represents a promising natural immune-adjuvant to chemotherapy in advanced CRC.
Collapse
Affiliation(s)
- Diana Groza
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Austria
| | - Sebastian Gehrig
- Laboratory of MacroMolecular Cancer Therapeutics ( MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | | | - Martin Holcmann
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Carina Dinhof
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Hemma H Schueffl
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Austria
| | | | - Julia Hoebart
- Laboratory of MacroMolecular Cancer Therapeutics ( MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Fatih Alioglu
- Laboratory of MacroMolecular Cancer Therapeutics ( MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Manfred Ogris
- Laboratory of MacroMolecular Cancer Therapeutics ( MMCT), Center of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | | | - Bernhard K Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Austria
| | - Irena Pashkunova-Martic
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.,Research Cluster "Translational Cancer Therapy Research", University of Vienna and Medical University of Vienna, Austria
| |
Collapse
|
24
|
In Vivo Quantification of Myocardial Infarction in Mice Using Micro-CT and a Novel Blood Pool Agent. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:2617047. [PMID: 29114173 PMCID: PMC5662822 DOI: 10.1155/2017/2617047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 09/12/2017] [Indexed: 11/25/2022]
Abstract
We herein developed a micro-CT method using the innovative contrast agent ExiTron™ MyoC 8000 to longitudinally monitor cardiac processes in vivo in small animals. Experiments were performed on healthy mice and mice with myocardial infarction inflicted by ligation of the left anterior descending artery. Time-dependent signal enhancement in different tissues of healthy mice was measured and various contrast agent doses were investigated so as to determine the minimum required dose for imaging of the myocardium. Due to its ability to be taken up by healthy myocardium but not by infarct tissue, ExiTron MyoC 8000 enables detection of myocardial infarction even at a very low dose. The signal enhancement in the myocardium of infarcted mice after contrast agent injection was exploited for quantification of infarct size. The values of infarct size obtained from the imaging method were compared with those obtained from histology and showed a significant correlation (R2 = 0.98). Thus, the developed micro-CT method allows for monitoring of a variety of processes such as cardiac remodeling in longitudinal studies.
Collapse
|
25
|
Lattin CR, Pechenenko AV, Carson RE. Experimentally reducing corticosterone mitigates rapid captivity effects on behavior, but not body composition, in a wild bird. Horm Behav 2017; 89:121-129. [PMID: 28065712 PMCID: PMC5359069 DOI: 10.1016/j.yhbeh.2016.12.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/23/2016] [Accepted: 12/29/2016] [Indexed: 01/02/2023]
Abstract
Wild animals and captives display physiological and behavioral differences, and it has been hypothesized, but rarely tested, that these differences are caused by sustained elevation of the hormone corticosterone. We used repeated computed tomography (CT) imaging to examine body composition changes in breeding male and female wild house sparrows (Passer domesticus; n=20) in response to two weeks of captivity, and assessed behavioral changes using video recordings. Half of the birds received the drug mitotane, which significantly decreased stress-induced corticosterone titers compared to controls. Based on the CT images, fat volumes increased, and pectoralis muscle density and heart and testes volumes decreased, over the two weeks of captivity in both groups of birds. However, beak-wiping, a behavior that can indicate anxiety and aggression, showed increased occurrence in controls compared to mitotane-treated birds. While our results do not support the hypothesis that these body composition changes were primarily driven by stress-induced corticosterone, our data suggest that experimentally reducing stress-induced corticosterone may mitigate some captivity-induced behavioral changes. Broadly, our results emphasize that researchers should take behavioral and physiological differences between free-living animals and captives into consideration when designing studies and interpreting results. Further, time in captivity should be minimized when birds will be reintroduced back to the wild.
Collapse
Affiliation(s)
- Christine R Lattin
- Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT 06520-8048, United States.
| | - Anita V Pechenenko
- Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT 06520-8048, United States
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, 801 Howard Avenue, PO Box 208048, New Haven, CT 06520-8048, United States
| |
Collapse
|
26
|
Xing R, De Wilde D, McCann G, Ridwan Y, Schrauwen JTC, van der Steen AFW, Gijsen FJH, Van der Heiden K. Contrast-enhanced micro-CT imaging in murine carotid arteries: a new protocol for computing wall shear stress. Biomed Eng Online 2016; 15:156. [PMID: 28155699 PMCID: PMC5259814 DOI: 10.1186/s12938-016-0270-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Wall shear stress (WSS) is involved in the pathophysiology of atherosclerosis. The correlation between WSS and atherosclerosis can be investigated over time using a WSS-manipulated atherosclerotic mouse model. To determine WSS in vivo, detailed 3D geometry of the vessel network is required. However, a protocol to reconstruct 3D murine vasculature using this animal model is lacking. In this project, we evaluated the adequacy of eXIA 160, a small animal contrast agent, for assessing murine vascular network on micro-CT. Also, a protocol was established for vessel geometry segmentation and WSS analysis. METHODS A tapering cast was placed around the right common carotid artery (RCCA) of ApoE-/- mice (n = 8). Contrast-enhanced micro-CT was performed using eXIA 160. An innovative local threshold-based segmentation procedure was implemented to reconstruct 3D geometry of the RCCA. The reconstructed RCCA was compared to the vessel geometry using a global threshold-based segmentation method. Computational fluid dynamics was applied to compute the velocity field and WSS distribution along the RCCA. RESULTS eXIA 160-enhanced micro-CT allowed clear visualization and assessment of the RCCA in all eight animals. No adverse biological effects were observed from the use of eXIA 160. Segmentation using local threshold values generated more accurate RCCA geometry than the global threshold-based approach. Mouse-specific velocity data and the RCCA geometry generated 3D WSS maps with high resolution, enabling quantitative analysis of WSS. In all animals, we observed low WSS upstream of the cast. Downstream of the cast, asymmetric WSS patterns were revealed with variation in size and location between animals. CONCLUSIONS eXIA 160 provided good contrast to reconstruct 3D vessel geometry and determine WSS patterns in the RCCA of the atherosclerotic mouse model. We established a novel local threshold-based segmentation protocol for RCCA reconstruction and WSS computation. The observed differences between animals indicate the necessity to use mouse-specific data for WSS analysis. For our future work, our protocol makes it possible to study in vivo WSS longitudinally over a growing plaque.
Collapse
Affiliation(s)
- Ruoyu Xing
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Wytemaweg 80, Ee2338, 3015CN, Rotterdam, The Netherlands
| | - David De Wilde
- IBiTech-bioMMeda, iMinds Medical IT, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - Gayle McCann
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Wytemaweg 80, Ee2338, 3015CN, Rotterdam, The Netherlands
| | - Yanto Ridwan
- Department of Genetics, Erasmus MC, Wytemaweg 80, Ee720, 3015CN, Rotterdam, The Netherlands
| | - Jelle T. C. Schrauwen
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Wytemaweg 80, Ee2338, 3015CN, Rotterdam, The Netherlands
| | - Anton F. W. van der Steen
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Wytemaweg 80, Ee2338, 3015CN, Rotterdam, The Netherlands
| | - Frank J. H. Gijsen
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Wytemaweg 80, Ee2338, 3015CN, Rotterdam, The Netherlands
| | - Kim Van der Heiden
- Department of Biomedical Engineering, Thorax Center, Erasmus MC, Wytemaweg 80, Ee2338, 3015CN, Rotterdam, The Netherlands
| |
Collapse
|
27
|
Kirschner S, Mürle B, Felix M, Arns A, Groden C, Wenz F, Hug A, Glatting G, Kramer M, Giordano FA, Brockmann MA. Imaging of Orthotopic Glioblastoma Xenografts in Mice Using a Clinical CT Scanner: Comparison with Micro-CT and Histology. PLoS One 2016; 11:e0165994. [PMID: 27829015 PMCID: PMC5102379 DOI: 10.1371/journal.pone.0165994] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/23/2016] [Indexed: 01/24/2023] Open
Abstract
Purpose There is an increasing need for small animal in vivo imaging in murine orthotopic glioma models. Because dedicated small animal scanners are not available ubiquitously, the applicability of a clinical CT scanner for visualization and measurement of intracerebrally growing glioma xenografts in living mice was validated. Materials and Methods 2.5x106 U87MG cells were orthotopically implanted in NOD/SCID/ᵞc-/- mice (n = 9). Mice underwent contrast-enhanced (300 μl Iomeprol i.v.) imaging using a micro-CT (80 kV, 75 μAs, 360° rotation, 1,000 projections, scan time 33 s, resolution 40 x 40 x 53 μm) and a clinical CT scanner (4-row multislice detector; 120 kV, 150 mAs, slice thickness 0.5 mm, feed rotation 0.5 mm, resolution 98 x 98 x 500 μm). Mice were sacrificed and the brain was worked up histologically. In all modalities tumor volume was measured by two independent readers. Contrast-to-noise ratio (CNR) and Signal-to-noise ratio (SNR) were measured from reconstructed CT-scans (0.5 mm slice thickness; n = 18). Results Tumor volumes (mean±SD mm3) were similar between both CT-modalities (micro-CT: 19.8±19.0, clinical CT: 19.8±18.8; Wilcoxon signed-rank test p = 0.813). Moreover, between reader analyses for each modality showed excellent agreement as demonstrated by correlation analysis (Spearman-Rho >0.9; p<0.01 for all correlations). Histologically measured tumor volumes (11.0±11.2) were significantly smaller due to shrinkage artifacts (p<0.05). CNR and SNR were 2.1±1.0 and 1.1±0.04 for micro-CT and 23.1±24.0 and 1.9±0.7 for the clinical CTscanner, respectively. Conclusion Clinical CT scanners may reliably be used for in vivo imaging and volumetric analysis of brain tumor growth in mice.
Collapse
Affiliation(s)
- Stefanie Kirschner
- Department of Neuroradiology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Bettina Mürle
- Department of Neuroradiology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Manuela Felix
- Medical Radiation Physics/Radiation Protection, Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Anna Arns
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Christoph Groden
- Department of Neuroradiology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Frederik Wenz
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Andreas Hug
- Spinal Cord Injury Center, University Hospital Heidelberg, Schlierbacher Landstr. 200a, 69118, Heidelberg, Germany
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Martin Kramer
- Department of Veterinary Clinical Sciences, Small Animal Clinic, Justus-Liebig-University, 35392, Giessen, Germany
| | - Frank A. Giordano
- Department of Radiation Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Marc A. Brockmann
- Department of Neuroradiology, University Medical Center of the Johannes Gutenberg University Mainz, 55131, Mainz, Germany
- * E-mail:
| |
Collapse
|
28
|
Ignat M, Akladios CY, Lindner V, Khetchoumian K, Teletin M, Muttter D, Aprahamian PM, Marescaux J. Development of a methodology for in vivo follow-up of hepatocellular carcinoma in hepatocyte specific Trim24-null mice treated with myo-inositol trispyrophosphate. J Exp Clin Cancer Res 2016; 35:155. [PMID: 27686696 PMCID: PMC5041534 DOI: 10.1186/s13046-016-0434-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 09/21/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Genetically induced hepatocellular carcinoma (HCC) models are generally used to investigate carcinogenesis pathways, but very few attempts were made to valorize them for pharmacological testing. This study describes a micro-computed tomography (micro-CT) - based methodology for the diagnostic and lifelong follow-up of HCC in the hepatocyte-specific Trim24-null mouse line. Myo-inositol trispyrophosphate (ITPP) was tested as anti-cancer drug. METHODS Partial hepatectomy was performed in 2 months-old Trim24-null mice, in order to accelerate the carcinogenesis process. HCC diagnosis was obtained by micro-CT scan with double contrast agent: 10 μl/g Fenestra™ LC was injected intraperitoneally 6 h prior to imaging and 10 μl/g Fenestra™ VC was injected intravenously 15 min prior to imaging. Twenty three hepatocyte-specific Trim24-null mice were considered for ITPP testing (3 mg/g/week intraperitoneally during 10 months in 12 mice, versus 11 controls). Lifelong follow-up was performed using micro-CT. Comparative analysis was performed using unpaired t test with Welch correction and survival curves were compared by log-rank test. Gene expression analysis was performed using the RT q-PCR technique. RESULTS Double contrast micro-CT scan allowed HCC diagnosis as hypodense, isodense or hyperdense nodules. Positive predictive value was 81.3 %. Negative predictive value was 83.3 %. Tumor growth could be objectified by micro-CT scan before the ITPP treatment was started, and at 3 and 9 months follow-up. Significant progression of tumor volume was demonstrated in the both groups, with no difference between groups (p > 0.05). In the ITPP group, a mild decrease in tumor doubling time was first observed (31.9 +/- 12 days, p > 0.05) followed by a significant increase (59.8 +/- 18.3 days, p = 0.008). However, tumor doubling time was not different between groups (p > 0.05). Median survival after treatment initiation was 223 days (controls) versus 296 days (ITPP group, p = 0.0027). HIF1α, VEGF, glutamine synthase, osteopontin expression levels were not significantly modified at the end of follow-up. In the ITPP group, the p53 expression profile was inversed as compared to the control group, higher in non-tumor livers than in tumors. CONCLUSION ITPP treatment allowed for a two-month survival improvement, with better tolerance of tumor burden and apoptosis increase in non-tumor, pathological livers.
Collapse
Affiliation(s)
- Mihaela Ignat
- IRCAD, 1 place de l’hôpital, 67091 Strasbourg, France
- Department of Digestive and Endocrine Surgery, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg, France
| | - Cherif Youssef Akladios
- Department of Digestive and Endocrine Surgery, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg, France
| | - Véronique Lindner
- Department of Digestive and Endocrine Surgery, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg, France
| | - Konstantin Khetchoumian
- Institute of Genetics and Molecular and Cellular Biology, F-67404 Illkirch, France
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), Montréal, QC H2W 1R7 Canada
| | - Marius Teletin
- Department of Digestive and Endocrine Surgery, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg, France
- Institute of Genetics and Molecular and Cellular Biology, F-67404 Illkirch, France
| | - Didier Muttter
- IRCAD, 1 place de l’hôpital, 67091 Strasbourg, France
- Department of Digestive and Endocrine Surgery, University Hospital of Strasbourg, 1 place de l’Hôpital, 67091 Strasbourg, France
| | | | | |
Collapse
|
29
|
Ghaghada KB, Sato AF, Starosolski ZA, Berg J, Vail DM. Computed Tomography Imaging of Solid Tumors Using a Liposomal-Iodine Contrast Agent in Companion Dogs with Naturally Occurring Cancer. PLoS One 2016; 11:e0152718. [PMID: 27031614 PMCID: PMC4816501 DOI: 10.1371/journal.pone.0152718] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/17/2016] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVES Companion dogs with naturally occurring cancer serve as an important large animal model in translational research because they share strong similarities with human cancers. In this study, we investigated a long circulating liposomal-iodine contrast agent (Liposomal-I) for computed tomography (CT) imaging of solid tumors in companion dogs with naturally occurring cancer. MATERIALS AND METHODS The institutional animal ethics committees approved the study and written informed consent was obtained from all owners. Thirteen dogs (mean age 10.1 years) with a variety of masses including primary and metastatic liver tumors, sarcomas, mammary carcinoma and lung tumors, were enrolled in the study. CT imaging was performed pre-contrast and at 15 minutes and 24 hours after intravenous administration of Liposomal-I (275 mg/kg iodine dose). Conventional contrast-enhanced CT imaging was performed in a subset of dogs, 90 minutes prior to administration of Liposomal-I. Histologic or cytologic diagnosis was obtained for each dog prior to admission into the study. RESULTS Liposomal-I resulted in significant (p < 0.05) enhancement and uniform opacification of the vascular compartment. Non-renal, reticulo-endothelial systemic clearance of the contrast agent was demonstrated. Liposomal-I enabled visualization of primary and metastatic liver tumors. Sub-cm sized liver lesions grossly appeared as hypo-enhanced compared to the surrounding normal parenchyma with improved lesion conspicuity in the post-24 hour scan. Large liver tumors (> 1 cm) demonstrated a heterogeneous pattern of intra-tumoral signal with visibly higher signal enhancement at the post-24 hour time point. Extra-hepatic, extra-splenic tumors, including histiocytic sarcoma, anaplastic sarcoma, mammary carcinoma and lung tumors, were visualized with a heterogeneous enhancement pattern in the post-24 hour scan. CONCLUSIONS The long circulating liposomal-iodine contrast agent enabled prolonged visualization of small and large tumors in companion dogs with naturally occurring cancer. The study warrants future work to assess the sensitivity and specificity of the Liposomal-I agent in various types of naturally occurring canine tumors.
Collapse
Affiliation(s)
- Ketan B. Ghaghada
- The Singleton Department of Pediatric Radiology, Texas Children’s Hospital, Houston, Texas, United States of America
- * E-mail:
| | - Amy F. Sato
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - Zbigniew A. Starosolski
- The Singleton Department of Pediatric Radiology, Texas Children’s Hospital, Houston, Texas, United States of America
| | - John Berg
- Department of Clinical Sciences, Tufts Cummings School of Veterinary Medicine, North Grafton, Massachusetts, United States of America
| | - David M. Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
30
|
Mannheim JG, Schlichthaerle T, Kuebler L, Quintanilla-Martinez L, Kohlhofer U, Kneilling M, Pichler BJ. Comparison of small animal CT contrast agents. CONTRAST MEDIA & MOLECULAR IMAGING 2016; 11:272-84. [PMID: 26991457 DOI: 10.1002/cmmi.1689] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 12/21/2015] [Accepted: 01/16/2016] [Indexed: 11/09/2022]
Abstract
Non-invasive in vivo small animal computed tomography (CT) imaging provides high resolution bone scans but cannot differentiate between soft tissues. For most applications injections of contrast agents (CAs) are necessary. Aim of this study was to uncover the advantages and disadvantages of commercially available CT CAs (ExiTron nano 12 000 and 6000, eXIA 160 and 160XL, Fenestra VC and LC) regarding their pharmacokinetics, toxicological side-effects and the influence of anesthesia on the biodistribution, based on an injection volume of 100 μL/25 g body weight. The pharmacokinetics of the CAs were determined for up to five days. The CA-induced toxicological/physiological side-effects were evaluated by determining blood counts, liver enzymes, thyroxine and total protein values, pro-inflammatory mediators (messenger ribonucleic acid (mRNA)), histology and immunohistochemistry. ExiTron nano 12 000 and 6000 yielded a long-term contrast enhancement (CE) in the liver and spleen for up to five days. Some of the evaluated CAs did not show any CE at all. Anesthesia did not impair the CAs' biodistribution. The CAs differentially affected the body weight, blood counts, liver enzymes, thyroxine and total protein values. ExiTron nano 12 000 and 6000 induced histiocytes in the liver and spleen. Moreover, ExiTron nano 12 000 and eXIA 160 enhanced tumor necrosis factor (TNF) mRNA expression levels in the kidneys. Thus, we recommend ExiTron nano 12 000 and 6000 when multiple injections should be avoided. We recommend careful selection of the employed CA in order to achieve an acceptable CE in the organs of interest and to avoid influences on the animal physiology. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Julia G Mannheim
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Thomas Schlichthaerle
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Laura Kuebler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | | | - Ursula Kohlhofer
- Institute of Pathology, University Hospital Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Manfred Kneilling
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| |
Collapse
|
31
|
Das NM, Hatsell S, Nannuru K, Huang L, Wen X, Wang L, Wang LH, Idone V, Meganck JA, Murphy A, Economides A, Xie L. In Vivo Quantitative Microcomputed Tomographic Analysis of Vasculature and Organs in a Normal and Diseased Mouse Model. PLoS One 2016; 11:e0150085. [PMID: 26910759 PMCID: PMC4765930 DOI: 10.1371/journal.pone.0150085] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/09/2016] [Indexed: 02/07/2023] Open
Abstract
Non-bone in vivo micro-CT imaging has many potential applications for preclinical evaluation. Specifically, the in vivo quantification of changes in the vascular network and organ morphology in small animals, associated with the emergence and progression of diseases like bone fracture, inflammation and cancer, would be critical to the development and evaluation of new therapies for the same. However, there are few published papers describing the in vivo vascular imaging in small animals, due to technical challenges, such as low image quality and low vessel contrast in surrounding tissues. These studies have primarily focused on lung, cardiovascular and brain imaging. In vivo vascular imaging of mouse hind limbs has not been reported. We have developed an in vivo CT imaging technique to visualize and quantify vasculature and organ structure in disease models, with the goal of improved quality images. With 1–2 minutes scanning by a high speed in vivo micro-CT scanner (Quantum CT), and injection of a highly efficient contrast agent (Exitron nano 12000), vasculature and organ structure were semi-automatically segmented and quantified via image analysis software (Analyze). Vessels of the head and hind limbs, and organs like the heart, liver, kidneys and spleen were visualized and segmented from density maps. In a mouse model of bone metastasis, neoangiogenesis was observed, and associated changes to vessel morphology were computed, along with associated enlargement of the spleen. The in vivo CT image quality, voxel size down to 20 μm, is sufficient to visualize and quantify mouse vascular morphology. With this technique, in vivo vascular monitoring becomes feasible for the preclinical evaluation of small animal disease models.
Collapse
Affiliation(s)
- Nanditha Mohan Das
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Sarah Hatsell
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Kalyan Nannuru
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Lily Huang
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Xialing Wen
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Lili Wang
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Li-Hsien Wang
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Vincent Idone
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Jeffrey A. Meganck
- Research and Development, PerkinElmer, Hopkinton, Massachusetts, United States of America
| | - Andrew Murphy
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - Aris Economides
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
| | - LiQin Xie
- Department of Skeletal Diseases – Therapeutic Focus Areas, Regeneron Pharmaceuticals Inc., Tarrytown, New York, United States of America
- * E-mail:
| |
Collapse
|
32
|
van Deel E, Ridwan Y, van Vliet JN, Belenkov S, Essers J. In Vivo Quantitative Assessment of Myocardial Structure, Function, Perfusion and Viability Using Cardiac Micro-computed Tomography. J Vis Exp 2016:53603. [PMID: 26967592 PMCID: PMC4828165 DOI: 10.3791/53603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The use of Micro-Computed Tomography (MicroCT) for in vivo studies of small animals as models of human disease has risen tremendously due to the fact that MicroCT provides quantitative high-resolution three-dimensional (3D) anatomical data non-destructively and longitudinally. Most importantly, with the development of a novel preclinical iodinated contrast agent called eXIA160, functional and metabolic assessment of the heart became possible. However, prior to the advent of commercial MicroCT scanners equipped with X-ray flat-panel detector technology and easy-to-use cardio-respiratory gating, preclinical studies of cardiovascular disease (CVD) in small animals required a MicroCT technologist with advanced skills, and thus were impractical for widespread implementation. The goal of this work is to provide a practical guide to the use of the high-speed Quantum FX MicroCT system for comprehensive determination of myocardial global and regional function along with assessment of myocardial perfusion, metabolism and viability in healthy mice and in a cardiac ischemia mouse model induced by permanent occlusion of the left anterior descending coronary artery (LAD).
Collapse
Affiliation(s)
- Elza van Deel
- Department of Genetics, Erasmus MC, Rotterdam; Department of Experimental Cardiology, Erasmus MC, Rotterdam
| | | | | | | | - Jeroen Essers
- Department of Genetics, Erasmus MC, Rotterdam; Department of Vascular Surgery, Erasmus MC, Rotterdam; Department of Radiation Oncology, Erasmus MC, Rotterdam;
| |
Collapse
|
33
|
Flechsig P, Kratochwil C, Warth A, Rath D, Eichwald V, Huber PE, Kauczor HU, Haberkorn U, Giesel FL. A Comparison of microCT and microPET for Evaluating Lymph Node Metastasis in a Rat Model. Mol Imaging Biol 2015; 18:243-8. [DOI: 10.1007/s11307-015-0890-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
34
|
Hua XW, Lu TF, Li DW, Wang WG, Li J, Liu ZZ, Lin WW, Zhang JJ, Xia Q. Contrast-enhanced micro-computed tomography using ExiTron nano6000 for assessment of liver injury. World J Gastroenterol 2015; 21:8043-8051. [PMID: 26185375 PMCID: PMC4499346 DOI: 10.3748/wjg.v21.i26.8043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/13/2015] [Accepted: 03/31/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the potential of contrast-enhanced computed tomography (CECT) using ExiTron nano6000 for assessment of liver lesions in mouse models.
METHODS: Three mouse models of liver lesions were used: bile duct ligation (BDL), lipopolysaccharide (LPS)/D-galactosamine (D-GalN), and alcohol. After injection with the contrast agent ExiTron nano6000, the mice were scanned with micro-CT. Liver lesions were evaluated using CECT images, hematoxylin and eosin staining, and serum aminotransferase levels. Macrophage distribution in the injury models was shown by immunohistochemical staining of CD68. The in vitro studies measured the densities of RAW264.7 under different conditions by CECT.
RESULTS: In the in vitro studies, CECT provided specific and strong contrast enhancement of liver in mice. CECT could present heterogeneous images and densities of injured livers induced by BDL, LPS/D-GalN, and alcohol. The liver histology and immunochemistry of CD68 demonstrated that both dilated biliary tracts and necrosis in the injured livers could lead to the heterogeneous distribution of macrophages. The in vitro study showed that the RAW264.7 cell masses had higher densities after LPS activation.
CONCLUSION: Micro-CT with the contrast agent ExiTron nano6000 is feasible for detecting various liver lesions by emphasizing the heterogeneous textures and densities of CECT images.
Collapse
|
35
|
Felix MC, Fleckenstein J, Kirschner S, Hartmann L, Wenz F, Brockmann MA, Glatting G, Giordano FA. Image-Guided Radiotherapy Using a Modified Industrial Micro-CT for Preclinical Applications. PLoS One 2015; 10:e0126246. [PMID: 25993010 PMCID: PMC4438006 DOI: 10.1371/journal.pone.0126246] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/30/2015] [Indexed: 11/30/2022] Open
Abstract
Purpose/Objective Although radiotherapy is a key component of cancer treatment, its implementation into pre-clinical in vivo models with relatively small target volumes is frequently omitted either due to technical complexity or expected side effects hampering long-term observational studies. We here demonstrate how an affordable industrial micro-CT can be converted into a small animal IGRT device at very low costs. We also demonstrate the proof of principle for the case of partial brain irradiation of mice carrying orthotopic glioblastoma implants. Methods/Materials A commercially available micro-CT originally designed for non-destructive material analysis was used. It consists of a CNC manipulator, a transmission X-ray tube (10–160 kV) and a flat-panel detector, which was used together with custom-made steel collimators (1–5 mm aperture size). For radiation field characterization, an ionization chamber, water-equivalent slab phantoms and radiochromic films were used. A treatment planning tool was implemented using a C++ application. For proof of principle, NOD/SCID/γc−/− mice were orthotopically implanted with U87MG high-grade glioma cells and irradiated using the novel setup. Results The overall symmetry of the radiation field at 150 kV was 1.04±0.02%. The flatness was 4.99±0.63% and the penumbra widths were between 0.14 mm and 0.51 mm. The full width at half maximum (FWHM) ranged from 1.97 to 9.99 mm depending on the collimator aperture size. The dose depth curve along the central axis followed a typical shape of keV photons. Dose rates measured were 10.7 mGy/s in 1 mm and 7.6 mGy/s in 5 mm depth (5 mm collimator aperture size). Treatment of mice with a single dose of 10 Gy was tolerated well and resulted in central tumor necrosis consistent with therapeutic efficacy. Conclusion A conventional industrial micro-CT can be easily modified to allow effective small animal IGRT even of critical target volumes such as the brain.
Collapse
Affiliation(s)
- Manuela C. Felix
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jens Fleckenstein
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefanie Kirschner
- Department of Neuroradiology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Linda Hartmann
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Frederik Wenz
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marc A. Brockmann
- Department of Neuroradiology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Diagnostic and Interventional Neuroradiology, University Hospital Aachen, Aachen, Germany
| | - Gerhard Glatting
- Medical Radiation Physics/Radiation Protection, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- * E-mail:
| | - Frank A. Giordano
- Department of Radiation Oncology, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
36
|
Rothe JH, Rudolph I, Rohwer N, Kupitz D, Gregor-Mamoudou B, Derlin T, Furth C, Amthauer H, Brenner W, Buchert R, Cramer T, Apostolova I. Time course of contrast enhancement by micro-CT with dedicated contrast agents in normal mice and mice with hepatocellular carcinoma: comparison of one iodinated and two nanoparticle-based agents. Acad Radiol 2015; 22:169-78. [PMID: 25282584 DOI: 10.1016/j.acra.2014.07.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/27/2014] [Accepted: 07/28/2014] [Indexed: 11/16/2022]
Abstract
RATIONALE AND OBJECTIVES The aim of the present study was to characterize the kinetics of two nanoparticle-based contrast agents for preclinical imaging, Exitron nano 6000 and Exitron nano 12000, and the iodinated agent eXIA 160 in both healthy mice and in a mouse model of hepatocellular carcinoma (HCC). Semiautomatic segmentation of liver lesions for estimation of total tumor load of the liver was evaluated in HCC mice. MATERIALS AND METHODS The normal time course of contrast enhancement was assessed in 15 healthy C57BL/6 mice. Imaging of tumor spread in the liver was evaluated in 15 mice harboring a transgenic HCC model (ASV-B mice). Automatic segmentation of liver lesions for determination of total tumor burden of the liver was tested in three additional ASV-B mice before and after an experimental therapy. RESULTS In healthy mice, clearance of the contrast agent from blood was completed within 3-4 hours for eXIA 160 and Exitron nano 6000, whereas complete blood clearance of Exitron nano 12000 required about 24 hours. eXIA 160 provided maximum liver contrast at 1 hour post injection (p.i.) followed by a continuous decline. Enhancement of liver contrast with Exitron nano 6000 and Exitron nano 12000 reached a plateau at about 4 hours p.i., which lasted until the end of the measurements at 96 hours p.i. Maximum contrast enhancement of the liver was not statistically different between Exitron nano 6000 and Exitron nano 12000, but was about three times lower for eXIA 160 (P < .05). Visually Exitron nano 12000 provided the best liver-to-tumor contrast. Semiautomatic liver and tumor segmentation was feasible after the administration of Exitron nano 12000 but did not work properly for the other two contrast agents. CONCLUSIONS Both nanoparticle-based contrast agents provided stronger and longer lasting contrast enhancement of healthy liver parenchyma. Exitron nano 12000 allowed automatic segmentation of tumor lesions for estimation of the total tumor load in the liver.
Collapse
Affiliation(s)
- Jan H Rothe
- Clinic of Nuclear Medicine, University Medicine Charité, Berlin, Germany
| | - Ines Rudolph
- Clinic of Hepatology and Gastroenterology, University Medicine Charité, Berlin, Germany; German Cancer Consortium, Deutsches Krebsforschungzentrum (DKFZ), Heidelberg, Germany
| | - Nadine Rohwer
- Clinic of Hepatology and Gastroenterology, University Medicine Charité, Berlin, Germany
| | - Dennis Kupitz
- Department of Radiology and Nuclear Medicine, Medical School, Otto-von-Guericke University, Magdeburg A.ö.R., Magdeburg, Germany
| | | | - Thorsten Derlin
- Clinic of Radiology, University Medical Center, Hamburg, Germany
| | - Christian Furth
- Department of Radiology and Nuclear Medicine, Medical School, Otto-von-Guericke University, Magdeburg A.ö.R., Magdeburg, Germany
| | - Holger Amthauer
- Department of Radiology and Nuclear Medicine, Medical School, Otto-von-Guericke University, Magdeburg A.ö.R., Magdeburg, Germany
| | - Winfried Brenner
- Clinic of Nuclear Medicine, University Medicine Charité, Berlin, Germany
| | - Ralph Buchert
- Clinic of Nuclear Medicine, University Medicine Charité, Berlin, Germany
| | - Thorsten Cramer
- Clinic of Hepatology and Gastroenterology, University Medicine Charité, Berlin, Germany
| | - Ivayla Apostolova
- Department of Radiology and Nuclear Medicine, Medical School, Otto-von-Guericke University, Magdeburg A.ö.R., Magdeburg, Germany.
| |
Collapse
|
37
|
Kirschner S, Felix MC, Hartmann L, Bierbaum M, Maros ME, Kerl HU, Wenz F, Glatting G, Kramer M, Giordano FA, Brockmann MA. In vivo micro-CT imaging of untreated and irradiated orthotopic glioblastoma xenografts in mice: capabilities, limitations and a comparison with bioluminescence imaging. J Neurooncol 2015; 122:245-54. [PMID: 25605299 DOI: 10.1007/s11060-014-1708-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/24/2014] [Indexed: 11/28/2022]
Abstract
Small animal imaging is of increasing relevance in biomedical research. Studies systematically assessing the diagnostic accuracy of contrast-enhanced in vivo micro-CT of orthotopic glioma xenografts in mice do not exist. NOD/SCID/γc(-/-) mice (n = 27) underwent intracerebral implantation of 2.5 × 10(6) GFP-Luciferase-transduced U87MG cells. Mice underwent bioluminescence imaging (BLI) to detect tumor growth and afterwards repeated contrast-enhanced (300 µl Iomeprol i.v.) micro-CT imaging (80 kV, 75 µAs, 360° rotation, 1,000 projections, 33 s scan time, resolution 40 × 40 × 53 µm, 0.5 Gy/scan). Presence of tumors, tumor diameter and tumor volume in micro-CT were rated by two independent readers. Results were compared with histological analyses. Six mice with tumors confirmed by micro-CT received fractionated irradiation (3 × 5 Gy every other day) using the micro-CT (5 mm pencil beam geometry). Repeated micro-CT scans were tolerated well. Tumor engraftment rate was 74 % (n = 20). In micro-CT, mean tumor volume was 30 ± 33 mm(3), and the smallest detectable tumor measured 360 × 620 µm. The inter-rater agreement (n = 51 micro-CT scans) for the item tumor yes/no was excellent (Spearman-Rho = 0.862, p < 0.001). Sensitivity and specificity of micro-CT were 0.95 and 0.71, respectively (PPV = 0.91, NPV = 0.83). BLI on day 21 after tumor implantation had a sensitivity and specificity of 0.90 and 1.0, respectively (PPV = 1.0, NPV = 0.5). Maximum tumor diameter and volume in micro-CT and histology correlated excellently (tumor diameter: 0.929, p < 0.001; tumor volume: 0.969, p < 0.001, n = 17). Irradiated animals showed a large central tumor necrosis. Longitudinal contrast enhanced micro-CT imaging of brain tumor growth in live mice is feasible at high sensitivity levels and with excellent inter-rater agreement and allows visualization of radiation effects.
Collapse
Affiliation(s)
- Stefanie Kirschner
- Department of Neuroradiology, Medical Faculty Mannheim, University, Medical Center Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang H, Thorling CA, Liang X, Bridle KR, Grice JE, Zhu Y, Crawford DHG, Xu ZP, Liu X, Roberts MS. Diagnostic imaging and therapeutic application of nanoparticles targeting the liver. J Mater Chem B 2015; 3:939-958. [PMID: 32261972 DOI: 10.1039/c4tb01611d] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liver diseases, particularly viral hepatitis, cirrhosis and hepatocellular carcinoma, are common in clinical practice with high morbidity and mortality worldwide. Many substances for diagnostic imaging and therapy of liver diseases may have either severe adverse effects or insufficient effectiveness in vivo because of their nonspecific uptake. Therefore, by targeting the delivery of drugs into the liver or specific liver cells, drug efficiency may be largely improved. This review summarizes the up-to-date research progress focusing on nanoparticles targeting the liver for both diagnostic and therapeutic purposes. Targeting strategies, mechanisms of enhanced effects, and clinical applications of nanoparticles are discussed specifically. We believe that new targeting nanotechnology such as nanoprobes for multi-modality imaging and multifunctional nanoparticles would facilitate significant advancements in this active research area in the near future.
Collapse
Affiliation(s)
- Haolu Wang
- Therapeutics Research Centre, School of Medicine, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, QLD 4102, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Wathen CA, Caldwell C, Chanda N, Upendran A, Zambre A, Afrasiabi Z, Chapaman SE, Foje N, Leevy WM, Kannan R. Selective X-ray contrast enhancement of the spleen of living mice mediated by gold nanorods. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 10:188-93. [PMID: 25169942 DOI: 10.1002/cmmi.1617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 04/14/2014] [Accepted: 06/25/2014] [Indexed: 11/10/2022]
Abstract
Gold nanomaterials (AuNPs) represent a promising new class of contrast agents for X-ray computed tomographic (CT) imaging in both research and clinical settings. These materials exhibit superior X-ray absorption properties compared with other iodinated agents, and thus require lower injection doses. Gold is nonimmunogenic and therefore contributes to safety profile in living specimens. Unfortunately, most reports on the use of AuNPs as X-ray CT enhancers only demonstrate marginal enhancement of the intended anatomical structure. In this study, we demonstrate the dramatic properties of gold nanorods (GNR) to serve as robust X-ray CT contrast-enhancing agent for selective imaging of the spleen. These organ-specific uptake properties were delineated by performing longitudinal CT imaging of living mice that were dosed with GNR at 2 day intervals. Rapid uptake in spleen was noted within 12 h of first systemic administration with a change in contrast enhancement of 90 Hounsfield units (ΔHU = 90) and with two subsequent injections a total contrast enhancement of over 200 HU was observed. The resulting images provide excellent contrast that will enable the detailed anatomical visualization and study of a range of pre-clinical models of spleen disease including infection and cancer.
Collapse
Affiliation(s)
- Connor A Wathen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Chuck Caldwell
- Department of Biological Engineering, University of Missouri-Columbia, Columbia, MO, 65212, USA
| | - Nripen Chanda
- Department of Radiology, University of Missouri-Columbia, Columbia, MO, 65212, USA
| | - Anandhi Upendran
- Department of Physics, University of Missouri-Columbia, Columbia, MO, 65212, USA
| | - Ajit Zambre
- Department of Radiology, University of Missouri-Columbia, Columbia, MO, 65212, USA
| | - Zahra Afrasiabi
- Department of Chemistry, Lincoln University, Jefferson City, Missouri, USA
| | - Sarah E Chapaman
- Department of Notre Dame Integrated Imaging Facility, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Nathan Foje
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - W Matthew Leevy
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA.,Department of Notre Dame Integrated Imaging Facility, University of Notre Dame, Notre Dame, IN, 46556, USA.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Raghuraman Kannan
- Department of Biological Engineering, University of Missouri-Columbia, Columbia, MO, 65212, USA.,Department of Radiology, University of Missouri-Columbia, Columbia, MO, 65212, USA.,Center for Micro/Nano Systems and Nanotechnology, University of Missouri-Columbia, Columbia, MO, 65212, USA
| |
Collapse
|