1
|
Honda M, Sigmund EE, Le Bihan D, Pinker K, Clauser P, Karampinos D, Partridge SC, Fallenberg E, Martincich L, Baltzer P, Mann RM, Camps-Herrero J, Iima M. Advanced breast diffusion-weighted imaging: what are the next steps? A proposal from the EUSOBI International Breast Diffusion-weighted Imaging working group. Eur Radiol 2024:10.1007/s00330-024-11010-0. [PMID: 39379708 DOI: 10.1007/s00330-024-11010-0] [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: 01/19/2024] [Revised: 05/25/2024] [Accepted: 07/23/2024] [Indexed: 10/10/2024]
Abstract
OBJECTIVES This study by the EUSOBI International Breast Diffusion-weighted Imaging (DWI) working group aimed to evaluate the current and future applications of advanced DWI in breast imaging. METHODS A literature search and a comprehensive survey of EUSOBI members to explore the clinical use and potential of advanced DWI techniques and a literature search were involved. Advanced DWI approaches such as intravoxel incoherent motion (IVIM), diffusion kurtosis imaging (DKI), and diffusion tensor imaging (DTI) were assessed for their current status and challenges in clinical implementation. RESULTS Although a literature search revealed an increasing number of publications and growing academic interest in advanced DWI, the survey revealed limited adoption of advanced DWI techniques among EUSOBI members, with 32% using IVIM models, 17% using non-Gaussian diffusion techniques for kurtosis analysis, and only 8% using DTI. A variety of DWI techniques are used, with IVIM being the most popular, but less than half use it, suggesting that the study identified a gap between the potential benefits of advanced DWI and its actual use in clinical practice. CONCLUSION The findings highlight the need for further research, standardization and simplification to transition advanced DWI from a research tool to regular practice in breast imaging. The study concludes with guidelines and recommendations for future research directions and clinical implementation, emphasizing the importance of interdisciplinary collaboration in this field to improve breast cancer diagnosis and treatment. CLINICAL RELEVANCE STATEMENT Advanced DWI in breast imaging, while currently in limited clinical use, offers promising improvements in diagnosis, staging, and treatment monitoring, highlighting the need for standardized protocols, accessible software, and collaborative approaches to promote its broader integration into routine clinical practice. KEY POINTS Increasing number of publications on advanced DWI over the last decade indicates growing research interest. EUSOBI survey shows that advanced DWI is used primarily in research, not extensively in clinical practice. More research and standardization are needed to integrate advanced DWI into routine breast imaging practice.
Collapse
Affiliation(s)
- Maya Honda
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Diagnostic Radiology, Kansai Electric Power Hospital, Osaka, Japan
| | - Eric E Sigmund
- Department of Radiology, NYU Langone Health, 6, 60 1st Avenue, New York, NY, 10016, USA
| | - Denis Le Bihan
- NeuroSpin/Joliot, CEA-Saclay Center, Paris-Saclay University, Gif-sur-Yvette, France
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- National Institute for Physiological Sciences, Okazaki, Japan
| | - Katja Pinker
- Department of Radiology, Breast Imaging Division, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Paola Clauser
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Structural Preclinical Imaging, Medical University of Vienna/Vienna General Hospital, Wien, Austria
| | - Dimitrios Karampinos
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Savannah C Partridge
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Eva Fallenberg
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Laura Martincich
- Unit of Radiodiagnostics, Ospedale Cardinal G. Massaia -ASL AT, Via Conte Verde 125, 14100, Asti, Italy
| | - Pascal Baltzer
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Ritse M Mann
- Department of Diagnostic Imaging, Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - Mami Iima
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Department of Fundamental Development for Advanced Low Invasive Diagnostic Imaging, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
2
|
Schraven S, Brück R, Rosenhain S, Lemainque T, Heines D, Noormohammadian H, Pabst O, Lederle W, Gremse F, Kiessling F. CT- and MRI-Aided Fluorescence Tomography Reconstructions for Biodistribution Analysis. Invest Radiol 2024; 59:504-512. [PMID: 38038691 DOI: 10.1097/rli.0000000000001052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
OBJECTIVES Optical fluorescence imaging can track the biodistribution of fluorophore-labeled drugs, nanoparticles, and antibodies longitudinally. In hybrid computed tomography-fluorescence tomography (CT-FLT), CT provides the anatomical information to generate scattering and absorption maps supporting a 3-dimensional reconstruction from the raw optical data. However, given the CT's limited soft tissue contrast, fluorescence reconstruction and quantification can be inaccurate and not sufficiently detailed. Magnetic resonance imaging (MRI) can overcome these limitations and extend the options for tissue characterization. Thus, we aimed to establish a hybrid CT-MRI-FLT approach for whole-body imaging and compared it with CT-FLT. MATERIALS AND METHODS The MRI-based hybrid imaging approaches were established first by scanning a water and coconut oil-filled phantom, second by quantifying Cy7 concentrations of inserts in dead mice, and finally by analyzing the biodistribution of AF750-labeled immunoglobulins (IgG, IgA) in living SKH1 mice. Magnetic resonance imaging, acquired with a fat-water-separated mDixon sequence, CT, and FLT were co-registered using markers in the mouse holder frame filled with white petrolatum, which was solid, stable, and visible in both modalities. RESULTS Computed tomography-MRI fusion was confirmed by comparing the segmentation agreement using Dice scores. Phantom segmentations showed good agreement, after correction for gradient linearity distortion and chemical shift. Organ segmentations in dead and living mice revealed adequate agreement for fusion. Marking the mouse holder frame and the successful CT-MRI fusion enabled MRI-FLT as well as CT-MRI-FLT reconstructions. Fluorescence tomography reconstructions supported by CT, MRI, or CT-MRI were comparable in dead mice with 60 pmol fluorescence inserts at different locations. Although standard CT-FLT reconstruction only considered general values for soft tissue, skin, lung, fat, and bone scattering, MRI's more versatile soft tissue contrast enabled the additional consideration of liver, kidneys, and brain. However, this did not change FLT reconstructions and quantifications significantly, whereas for extending scattering maps, it was important to accurately segment the organs and the entire mouse body. The various FLT reconstructions also provided comparable results for the in vivo biodistribution analyses with fluorescent immunoglobulins. However, MRI additionally enabled the visualization of gallbladder, thyroid, and brain. Furthermore, segmentations of liver, spleen, and kidney were more reliable due to better-defined contours than in CT. Therefore, the improved segmentations enabled better assignment of fluorescence signals and more differentiated conclusions with MRI-FLT. CONCLUSIONS Whole-body CT-MRI-FLT was implemented as a novel trimodal imaging approach, which allowed to more accurately assign fluorescence signals, thereby significantly improving pharmacokinetic analyses.
Collapse
Affiliation(s)
- Sarah Schraven
- From the Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany (S.S., R.B., S.R., T.L., D.H., W.L., F.G., F.K.); Institute of Molecular Medicine, RWTH Aachen University, Aachen, Germany (H.N., O.P.); Gremse-IT GmbH, Aachen, Germany (S.R., F.G.); Department for Diagnostic and Interventional Radiology, RWTH Aachen University, Aachen, Germany (T.L.); Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany (F.K.); and Fraunhofer MEVIS, Institute for Medical Image Computing, Aachen, Germany (F.K.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Wang D, Liu S, Fu J, Zhang P, Zheng S, Qiu B, Liu H, Ye Y, Guo J, Zhou Y, Jiang H, Yin S, He H, Xie C, Liu H. Correlation of K trans derived from dynamic contrast-enhanced MRI with treatment response and survival in locally advanced NSCLC patients undergoing induction immunochemotherapy and concurrent chemoradiotherapy. J Immunother Cancer 2024; 12:e008574. [PMID: 38910009 PMCID: PMC11328668 DOI: 10.1136/jitc-2023-008574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
PURPOSE This study aimed to investigate the prognostic significance of pretreatment dynamic contrast-enhanced (DCE)-MRI parameters concerning tumor response following induction immunochemotherapy and survival outcomes in patients with locally advanced non-small cell lung cancer (NSCLC) who underwent immunotherapy-based multimodal treatments. MATERIAL AND METHODS Unresectable stage III NSCLC patients treated by induction immunochemotherapy, concurrent chemoradiotherapy (CCRT) with or without consolidative immunotherapy from two prospective clinical trials were screened. Using the two-compartment Extend Tofts model, the parameters including Ktrans, Kep, Ve, and Vp were calculated from DCE-MRI data. The apparent diffusion coefficient was calculated from diffusion-weighted-MRI data. The receiver operating characteristic (ROC) curve and the area under the curve (AUC) were used to assess the predictive performance of MRI parameters. The Cox regression model was used for univariate and multivariate analysis. RESULTS 111 unresectable stage III NSCLC patients were enrolled. Patients received two cycles of induction immunochemotherapy and CCRT, with or without consolidative immunotherapy. With the median follow-up of 22.3 months, the median progression-free survival (PFS) and overall survival (OS) were 16.3 and 23.8 months. The multivariate analysis suggested that Eastern Cooperative Oncology Group score, TNM stage and the response to induction immunochemotherapy were significantly related to both PFS and OS. After induction immunochemotherapy, 67 patients (59.8%) achieved complete response or partial response and 44 patients (40.2%) had stable disease or progressive disease. The Ktrans of primary lung tumor before induction immunochemotherapy yielded the best performance in predicting the treatment response, with an AUC of 0.800. Patients were categorized into two groups: high-Ktrans group (n=67, Ktrans>164.3×10-3/min) and low-Ktrans group (n=44, Ktrans≤164.3×10-3/min) based on the ROC analysis. The high-Ktrans group had a significantly higher objective response rate than the low-Ktrans group (85.1% (57/67) vs 22.7% (10/44), p<0.001). The high-Ktrans group also presented better PFS (median: 21.1 vs 11.3 months, p=0.002) and OS (median: 34.3 vs 15.6 months, p=0.035) than the low-Ktrans group. CONCLUSIONS Pretreatment Ktrans value emerged as a significant predictor of the early response to induction immunochemotherapy and survival outcomes in unresectable stage III NSCLC patients who underwent immunotherapy-based multimodal treatments. Elevated Ktrans values correlated positively with enhanced treatment response, leading to extended PFS and OS durations.
Collapse
Affiliation(s)
- DaQuan Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - SongRan Liu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Jia Fu
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - PengXin Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - ShiYang Zheng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Bo Qiu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- United Imaging Healthcare, ShangHai, China
| | - YongQuan Ye
- United Imaging of Healthcare America, Houston, Texas, USA
| | - JinYu Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Yin Zhou
- SuZhou TongDiao Company, Suzhou, China
| | | | - ShaoHan Yin
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - HaoQiang He
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - ChuanMiao Xie
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center of Cancer Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Hoffmann E, Masthoff M, Kunz WG, Seidensticker M, Bobe S, Gerwing M, Berdel WE, Schliemann C, Faber C, Wildgruber M. Multiparametric MRI for characterization of the tumour microenvironment. Nat Rev Clin Oncol 2024; 21:428-448. [PMID: 38641651 DOI: 10.1038/s41571-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 04/21/2024]
Abstract
Our understanding of tumour biology has evolved over the past decades and cancer is now viewed as a complex ecosystem with interactions between various cellular and non-cellular components within the tumour microenvironment (TME) at multiple scales. However, morphological imaging remains the mainstay of tumour staging and assessment of response to therapy, and the characterization of the TME with non-invasive imaging has not yet entered routine clinical practice. By combining multiple MRI sequences, each providing different but complementary information about the TME, multiparametric MRI (mpMRI) enables non-invasive assessment of molecular and cellular features within the TME, including their spatial and temporal heterogeneity. With an increasing number of advanced MRI techniques bridging the gap between preclinical and clinical applications, mpMRI could ultimately guide the selection of treatment approaches, precisely tailored to each individual patient, tumour and therapeutic modality. In this Review, we describe the evolving role of mpMRI in the non-invasive characterization of the TME, outline its applications for cancer detection, staging and assessment of response to therapy, and discuss considerations and challenges for its use in future medical applications, including personalized integrated diagnostics.
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Wolfgang G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Seidensticker
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Stefanie Bobe
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | | | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
5
|
Jiang W, Wu R, Yang T, Yu S, Xing W. Profiling regulatory T lymphocytes within the tumor microenvironment of breast cancer via radiomics. Cancer Med 2023; 12:21861-21872. [PMID: 38083903 PMCID: PMC10757114 DOI: 10.1002/cam4.6757] [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: 06/18/2023] [Revised: 10/13/2023] [Accepted: 11/16/2023] [Indexed: 12/31/2023] Open
Abstract
OBJECTIVE To generate an image-driven biomarker (Rad_score) to predict tumor-infiltrating regulatory T lymphocytes (Treg) in breast cancer (BC). METHODS Overall, 928 BC patients were enrolled from the Cancer Genome Atlas (TCGA) for survival analysis; MRI (n = 71 and n = 30 in the training and validation sets, respectively) from the Cancer Imaging Archive (TCIA) were retrieved and subjected to repeat least absolute shrinkage and selection operator for feature reduction. The radiomic scores (rad_score) for Treg infiltration estimation were calculated via support vector machine (SVM) and logistic regression (LR) algorithms, and validated on the remaining patients. RESULTS Landmark analysis indicated Treg infiltration was a risk factor for BC patients in the first 5 years and after 10 years of diagnosis (p = 0.007 and 0.018, respectively). Altogether, 108 radiomic features were extracted from MRI images, 4 of which remained for model construction. Areas under curves (AUCs) of the SVM model were 0.744 (95% CI 0.622-0.867) and 0.733 (95% CI 0.535-0.931) for training and validation sets, respectively, while for the LR model, AUCs were 0.771 (95% CI 0.657-0.885) and 0.724 (95% CI 0.522-0.926). The calibration curves indicated good agreement between prediction and true value (p > 0.05), and DCA shows the high clinical utility of the radiomic model. Rad_score was significantly correlated with immune inhibitory genes like CTLA4 and PDCD1. CONCLUSIONS High Treg infiltration is a risk factor for patients with BC. The Rad_score formulated on radiomic features is a novel tool to predict Treg abundance in the tumor microenvironment.
Collapse
Affiliation(s)
- Wenying Jiang
- Department of RadiologyThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
- Department of Breast SurgeryThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Ruoxi Wu
- Department of RadiologyThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Tao Yang
- Department of Breast SurgeryGansu Provincial Maternity and Child Care HospitalLanzhouChina
| | - Shengnan Yu
- Department of RadiologyThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| | - Wei Xing
- Department of RadiologyThe Third Affiliated Hospital of Soochow UniversityChangzhouChina
| |
Collapse
|
6
|
Chen M, Shen Y, Pu Y, Zhou B, Bing J, Ge M, Zhu Y, Gao S, Wu W, Zhou M, Shi J. Biomimetic inducer enabled dual ferroptosis of tumor and M2-type macrophages for enhanced tumor immunotherapy. Biomaterials 2023; 303:122386. [PMID: 37977008 DOI: 10.1016/j.biomaterials.2023.122386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/29/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
Tumor-associated macrophages (TAMs) are abundant in the tumor microenvironment which promotes the formation of the immunosuppressive tumor microenvironment (ITME) through multiple mechanisms, severely counteracting the therapeutic efficacy of immunotherapy. In this study, a novel biomimetic ferroptosis inducer (D@FMN-M) capable of ITME regulation for enhanced cancer ferroptosis immunotherapy is reported. Upon tumor accumulation of D@FMN-M, the intratumoral mild acidity triggers the biodegradation of Fe-enriched nanocarriers and the concurrent co-releases of dihydroartemisinin (DHA) and Fe3+. The released Fe3+ is reduced to Fe2+ by consuming intratumoral glutathione (GSH), which promotes abundant free radical generation via triggering Fenton and Fe2+-DHA reactions, thus inducing ferroptosis of both cancer cells and M2-type TAMs. Resultantly, the anticancer immune response is strongly activated by the massive tumor-associated antigens released by ferroptositic cancer cells. Also importantly, the ferroptosis-sensitive M2-type TAMs will be either damaged or gradually domesticated to ferroptosis-resistant M1 TAMs under the ferroptosis stress, favoring the normalization of ITME and finally amplifying cancer ferroptosis immunotherapeutic efficacy. This work provides a novel strategy for ferroptosis immunotherapy of solid tumors featuring TAMs infiltration and immunosuppression by inducing dual ferroptosis of tumor cells and M2-type TAMs.
Collapse
Affiliation(s)
- Mingqi Chen
- Endoscopy Center, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Yucui Shen
- Endoscopy Center, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Yinying Pu
- Department of Medical Ultrasound, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, PR China
| | - Bangguo Zhou
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, PR China
| | - Jinhong Bing
- Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Min Ge
- Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Yaxuan Zhu
- Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China
| | - Shuang Gao
- Endoscopy Center, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China
| | - Wencheng Wu
- Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China.
| | - Min Zhou
- Endoscopy Center, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, PR China.
| | - Jianlin Shi
- Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, PR China.
| |
Collapse
|
7
|
Gerwing M, Hoffmann E, Geyer C, Helfen A, Maus B, Schinner R, Wachsmuth L, Heindel W, Eisenblaetter M, Faber C, Wildgruber M. Intratumoral heterogeneity after targeted therapy in murine cancer models with differing degrees of malignancy. Transl Oncol 2023; 37:101773. [PMID: 37666208 PMCID: PMC10483060 DOI: 10.1016/j.tranon.2023.101773] [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: 06/30/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/06/2023] Open
Abstract
INTRODUCTION Conventional morphologic and volumetric assessment of treatment response is not suitable for adequately assessing responses to targeted cancer therapy. The aim of this study was to evaluate changes in tumor composition after targeted therapy in murine models of breast cancer with differing degrees of malignancy via non-invasive magnetic resonance imaging (MRI). MATERIALS AND METHODS Mice bearing highly malignant 4T1 tumors or low malignant 67NR tumors were treated with either a combination of two immune checkpoint inhibitors (ICI, anti-PD1 and anti-CTLA-4) or the multi-tyrosine kinase inhibitor sorafenib, following experiments with macrophage-depleting clodronate-loaded liposomes and vessel-stabilizing angiopoietin-1. Mice were imaged on a 9.4 T small animal MRI system with a multiparametric (mp) protocol, comprising T1 and T2 mapping and diffusion-weighted imaging. Tumors were analyzed ex vivo with histology. RESULTS AND DISCUSSIONS All treatments led to an increase in non-viable areas, but therapy-induced intratumoral changes differed between the two tumor models and the different targeted treatments. While ICI treatment led to intratumoral hemorrhage, sorafenib treatment mainly induced intratumoral necrosis. Treated 4T1 tumors showed increasing and extensive areas of necrosis, in comparison to 67NR tumors with only small, but also increasing, necrotic areas. After either of the applied treatments, intratumoral heterogeneity, was increased in both tumor models, and confirmed ex vivo by histology. Apparent diffusion coefficient with subsequent histogram analysis proved to be the most sensitive MRI sequence. In conclusion, mp MRI enables to assess dedicated therapy-related intratumoral changes and may serve as a biomarker for treatment response assessment.
Collapse
Affiliation(s)
- M Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany.
| | - E Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany
| | - C Geyer
- Clinic of Radiology, University of Münster, Münster, Germany
| | - A Helfen
- Clinic of Radiology, University of Münster, Münster, Germany
| | - B Maus
- Clinic of Radiology, University of Münster, Münster, Germany
| | - R Schinner
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - L Wachsmuth
- Clinic of Radiology, University of Münster, Münster, Germany
| | - W Heindel
- Clinic of Radiology, University of Münster, Münster, Germany
| | - M Eisenblaetter
- Department of Diagnostic and Interventional Radiology, Medical Faculty OWL, University of Bielefeld, Bielefeld, Germany
| | - C Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - M Wildgruber
- Clinic of Radiology, University of Münster, Münster, Germany; Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|