1
|
Feng Y, Lin Z, Hu W, Wang X, Zhang Z, Lan X, Zhang X. Utilizing PET Imaging To Visualize Immune Activation and Tumor Apoptosis. Mol Pharm 2025; 22:2122-2130. [PMID: 40133236 DOI: 10.1021/acs.molpharmaceut.4c01314] [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] [Indexed: 03/27/2025]
Abstract
Immunotherapy-induced tumor apoptosis is one of the crucial pathways in tumor cell death. This study aimed to explore the potential of PET imaging for noninvasively visualizing pivotal processes in immunotherapy, specifically immune activation and tumor apoptosis, by targeting granzyme-B and caspase-3. Bioinformatic analyses validated granzyme-B and caspase-3 expression in cancer tissues and their associations with immune infiltration and patient prognosis using the GEPIA and TIMER databases. Two radiolabeled probes, [68Ga]Ga-GZP and [68Ga]Ga-AC3, were used to specifically target granzyme-B and caspase-3 for PET imaging, respectively. CT26 xenograft tumor models were assigned to PD-1 inhibitor or PBS control groups to receive treatment every 3 days, with imaging conducted at baseline and after each treatment. Imaging results showed significantly increased tumor uptake of both [68Ga]Ga-GZP and [68Ga]Ga-AC3 in the ICB-treated group compared to controls, indicating early molecular changes in immune activation and tumor apoptosis. Immunofluorescence analysis further supported these findings, revealing upregulated granzyme-B and caspase-3 expression in treated tumor tissues. Immunohistochemistry also confirmed increased T-cell infiltration and elevated levels of effector molecules, such as IFN-γ and TNF-α, in the ICB group. This study demonstrates that granzyme-B and caspase-3 PET/CT can noninvasively visualize early molecular changes in immunotherapy-induced CD8+ T cell activation and tumor apoptosis. These noninvasive diagnostic techniques hold significant promise for future clinical applications, particularly for a more accurate evaluation of immunotherapy efficacy.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| | - Zhaoguo Lin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| | - Wenzhu Hu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| | - Xingyi Wang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| | - Zihan Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| | - Xiao Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan 430022, Hubei Province, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, Wuhan 430022, Hubei Province, China
| |
Collapse
|
2
|
Zhang J, Pan T, Lee J, Goldberg S, King SA, Tang E, Hu Y, Chen L, Hoover A, Zhu L, Eng OS, Dekel B, Huang J, Wu X. Enabling tumor-specific drug delivery by targeting the Warburg effect of cancer. Cell Rep Med 2025; 6:101920. [PMID: 39809265 DOI: 10.1016/j.xcrm.2024.101920] [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: 08/31/2023] [Revised: 02/14/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
Metabolic reprogramming of tumor cells is an emerging hallmark of cancer. Among all the changes in cancer metabolism, increased glucose uptake and the accumulation of lactate under normoxic conditions (the "Warburg effect") is a common feature of cancer cells. In this study, we develop a lactate-responsive drug delivery platform by targeting the Warburg effect. We design and test a gold/mesoporous silica Janus nanoparticle system as a gated drug carrier, in which the gold particles are functionalized with lactate oxidase and the silica particles are capped with α-cyclodextrin through surface arylboronate modification. In the presence of lactate, the lactate oxidase generates hydrogen peroxide, which induces the self-immolation reaction of arylboronate, leading to uncapping and drug release. Our results demonstrate greatly improved drug delivery specificity and therapeutic efficacy with this platform for the treatment of different cancers. Our findings present an effective approach for drug delivery by metabolic targeting of tumors.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA; Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Jimmy Lee
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sanja Goldberg
- Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Aviv, Israel
| | - Sarah Ann King
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Yifei Hu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Lifeng Chen
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Alex Hoover
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Linyong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai 200237, China
| | - Oliver S Eng
- Department of Surgery, University of California, Irvine, Orange, CA 92868, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Aviv, Israel; Division of Pediatric Nephrology and Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Hasomer, Sago Center for Regenerative Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jun Huang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA.
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
3
|
Liu Q, Xu X, Yang Z, Zhang J, Li J, Qiao Y, Hu S, Liu X, Guo W, Song S. Granzyme B PET/CT Imaging Evaluates Early Response to Immunotherapy in Gastric Cancer. J Nucl Med 2024; 65:1695-1701. [PMID: 39389628 DOI: 10.2967/jnumed.124.267529] [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: 01/29/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
In several malignancies, only a limited number of patients respond to immune checkpoint inhibitors. Predicting and monitoring responses to these inhibitors represent an unmet clinical need. Here, we developed a PET/CT probe targeting granzyme B, [68Ga]Ga-NOTA-Gly-Gly-Gly-Ile-Glu-Pro-Asp-CHO (GSI), and aimed to investigate whether it can be used to monitor the effects of immune checkpoint inhibitors early in the course of therapy. Methods: Seventy-two patients with gastric cancer (stages III-IV) were recruited for [68Ga]Ga-NOTA-GSI PET/CT imaging after 2 or 3 cycles of the immunotherapy, and 40 patients were included in the final analysis. The SUVmax of primary tumors (SUVmax-t), SUVmax of metastatic lymph nodes (SUVmax-LN), and SUVmax of normal tissues (liver and blood pool) were measured, and their target-to-liver background ratio (TLR) and target-to-blood background ratio (TBR) were denoted for primary tumors as TLRtumor and TBRtumor and for metastatic lymph nodes as TLRLN and TBRLN, respectively. The treatment responses were assessed within 1 wk after full-course treatment according to RECIST version 1.1. Wilcoxon rank-sum tests were used to compare the PET/CT parameters between responders and nonresponders. Receiver operating characteristic curve analysis was used to assess the diagnostic efficacy of [68Ga]Ga-NOTA-GSI PET/CT parameters in identifying responders. Two-tailed P value of less than 0.05 was considered statistically significant. Results: We found that SUVmax-t, TLRtumor, TBRtumor, SUVmax-LN, and TBRLN were higher in responders than in nonresponders (2.49 ± 0.58 vs. 1.55 ± 0.48, P = 0.000; 2.24 ± 0.48 vs. 1.74 ± 0.67, P = 0.007; 1.38 ± 0.43 vs. 0.90 ± 0.23, P = 0.000; 2.24 ± 0.99 vs. 1.42 ± 0.55, P = 0.003; and 1.28 ± 0.68 vs. 0.83 ± 0.32, P = 0.012, respectively). According to receiver operating characteristic curve analysis, the area under the curve for SUVmax-t, TBRtumor, TLRtumor, SUVmax-LN, TLRLN, and TBRLN was 0.886, 0.866, 0.746, 0.772, 0.648, and 0.731, respectively. The threshold of SUVmax-t was 2.05, and its sensitivity and specificity were 81.0% and 84.2%, respectively. In addition, multivariate logistic regression indicated that TBRtumor was an independent predictor of treatment response (P = 0.03). Conclusion: Our results indicated that [68Ga]Ga-NOTA-GSI PET/CT is a promising tool for predicting early response to combined immunotherapy in gastric cancer patients.
Collapse
Affiliation(s)
- Qiufang Liu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Xiaoping Xu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Ziyi Yang
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Jianping Zhang
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Jindian Li
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Ying Qiao
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Silong Hu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Xiaosheng Liu
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| | - Weijian Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China;
- Department of Gastrointestinal Medical Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Shaoli Song
- Department of Nuclear Medicine, Shanghai Cancer Center, Fudan University, Shanghai, China;
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Engineering Research Center of Molecular Imaging Probes, Shanghai, China
- Key Laboratory of Nuclear Physics and Ion-Beam Application, MOE, Fudan University, Shanghai, China; and
| |
Collapse
|
4
|
Pandey A, Chopra S, Cleary SJ, López-Álvarez M, Quimby FM, Alanizi AAA, Sakhamuri S, Zhang N, Looney MR, Craik CS, Wilson DM, Evans MJ. Imaging the Granzyme Mediated Host Immune Response to Viral and Bacterial Pathogens In Vivo Using Positron Emission Tomography. ACS Infect Dis 2024; 10:2108-2117. [PMID: 38819300 DOI: 10.1021/acsinfecdis.4c00114] [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] [Indexed: 06/01/2024]
Abstract
Understanding how the host immune system engages complex pathogens is essential to developing therapeutic strategies to overcome their virulence. While granzymes are well understood to trigger apoptosis in infected host cells or bacteria, less is known about how the immune system mobilizes individual granzyme species in vivo to combat diverse pathogens. Toward the goal of studying individual granzyme function directly in vivo, we previously developed a new class of radiopharmaceuticals termed "restricted interaction peptides (RIPs)" that detect biochemically active endoproteases using positron emission tomography (PET). In this study, we showed that secreted granzyme B proteolysis in response to diverse viral and bacterial pathogens could be imaged with [64Cu]Cu-GRIP B, a RIP that specifically targets granzyme B. Wild-type or germline granzyme B knockout mice were instilled intranasally with the A/PR/8/34 H1N1 influenza A strain to generate pneumonia, and granzyme B production within the lungs was measured using [64Cu]Cu-GRIP B PET/CT. Murine myositis models of acute bacterial (E. coli, P. aeruginosa, K. pneumoniae, and L. monocytogenes) infection were also developed and imaged using [64Cu]Cu-GRIP B. In all cases, the mice were studied in vivo using mPET/CT and ex vivo via tissue-harvesting, gamma counting, and immunohistochemistry. [64Cu]Cu-GRIP B uptake was significantly higher in the lungs of wild-type mice that received A/PR/8/34 H1N1 influenza A strain compared to mice that received sham or granzyme B knockout mice that received either treatment. In wild-type mice, [64Cu]Cu-GRIP B uptake was significantly higher in the infected triceps muscle versus normal muscle and the contralateral triceps inoculated with heat killed bacteria. In granzyme B knockout mice, [64Cu]Cu-GRIP B uptake above the background was not observed in the infected triceps muscle. Interestingly, live L. monocytogenes did not induce detectable granzyme B on PET, despite prior in vitro data, suggesting a role for granzyme B in suppressing their pathogenicity. In summary, these data show that the granzyme response elicited by diverse human pathogens can be imaged using PET. These results and data generated via additional RIPs specific for other granzyme proteases will allow for a deeper mechanistic study analysis of their complex in vivo biology.
Collapse
Affiliation(s)
- Apurva Pandey
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Shalini Chopra
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Simon J Cleary
- Department of Medicine, University of California, San Francisco, San Francisco, California 94158, United States
| | - Marina López-Álvarez
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Fiona M Quimby
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Aryn A A Alanizi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Sasank Sakhamuri
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Ningjing Zhang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Mark R Looney
- Department of Medicine, University of California, San Francisco, San Francisco, California 94158, United States
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158, United States
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California 94158, United States
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California 94158, United States
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
5
|
Zhao D, Li Z, Ji DK, Xia Q. Recent Progress of Multifunctional Molecular Probes for Triple-Negative Breast Cancer Theranostics. Pharmaceutics 2024; 16:803. [PMID: 38931924 PMCID: PMC11207493 DOI: 10.3390/pharmaceutics16060803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Breast cancer (BC) poses a significant threat to women's health, with triple-negative breast cancer (TNBC) representing one of the most challenging and aggressive subtypes due to the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Traditional TNBC treatments often encounter issues such as low drug efficiency, limited tumor enrichment, and substantial side effects. Therefore, it is crucial to explore novel diagnostic and treatment systems for TNBC. Multifunctional molecular probes (MMPs), which integrate target recognition as well as diagnostic and therapeutic functions, introduce advanced molecular tools for TNBC theranostics. Using an MMP system, molecular drugs can be precisely delivered to the tumor site through a targeted ligand. Real-time dynamic monitoring of drug release achieved using imaging technology allows for the evaluation of drug enrichment at the tumor site. This approach enables accurate drug release, thereby improving the therapeutic effect. Therefore, this review summarizes the recent advancements in MMPs for TNBC theranostics, encompassing the design and synthesis of MMPs as well as their applications in the field of TNBC theranostics.
Collapse
Affiliation(s)
- Deyi Zhao
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (D.Z.); (Z.L.)
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhe Li
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (D.Z.); (Z.L.)
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ding-Kun Ji
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Qian Xia
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
6
|
Zhang Y, Deshane JS, Yang ES, Larimer B. A Novel Translational PET Imaging Approach to Quantifying Distal Tumor Immune Activation After Targeted Radiation Therapy and Checkpoint Blockade. Int J Radiat Oncol Biol Phys 2024; 118:1217-1227. [PMID: 38199384 PMCID: PMC11907763 DOI: 10.1016/j.ijrobp.2023.12.047] [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: 05/17/2023] [Revised: 12/20/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024]
Abstract
PURPOSE This study aimed to provide a novel noninvasive method to quantify abscopal immune activation and predict combinational treatment response using [68Ga]-NOTA-GZP positron emission tomography (PET) imaging. METHODS AND MATERIALS 4T1 breast cancer cells were implanted bilaterally in the mammary fat pad of Balb/c mice and Lewis's lung cancer cells (LLC) were implanted bilaterally on the shoulders of C57/Bl6 mice. One of the tumors received a single fraction of 12 Gy irradiation followed by combination of concurrent PD-1 and CTLA-4 inhibitors or controls. Tumor growth of the irradiated and nonirradiated tumors was measured and compared with 12 Gy irradiation only, checkpoint inhibitor only, and no treatment control group. Changes in granzyme B activity were assessed with [68Ga]-NOTA-GZP PET imaging from baseline and every 3 days until day 9. RESULTS In the 4T1 model, concurrent treatment with dual checkpoint inhibitors and radiation resulted in reduction of the irradiated tumor volume at day 30. At this same time point, the nonirradiated tumor volume for combination treatment decreased significantly, consistent with abscopal immune activation. Similarly, in the LLC model, concurrent treatment inhibited tumor growth on the nonirradiated tumor at day 15. On day 9, granzyme B PET signal in both 4T1 and LLC models was significantly higher in the nonirradiated tumors that responded to concurrent treatment compared with subsequent nonresponding tumors. A similar lack of granzyme B signal was observed in the nonirradiated tumors from mice that received radiation or checkpoint inhibitors only and control tumors. Receiver operating characteristic analysis identified a PET threshold of 1.505 and 1.233 on day 9 that predicted treatment response in 4T1 and LLC models, respectively. CONCLUSIONS [68Ga]-NOTA-GZP PET imaging was able to noninvasively predict abscopal immune activation before subsequent tumor volume changes after combination treatment. It provides a potential translational paradigm for investigating distal immune activation postradiation in a clinical setting.
Collapse
Affiliation(s)
- Yujun Zhang
- Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Jessy S Deshane
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Eddy S Yang
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Benjamin Larimer
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
7
|
Guz W, Podgórski R, Bober Z, Aebisher D, Truszkiewicz A, Olek M, Machorowska Pieniążek A, Kawczyk-Krupka A, Bartusik-Aebisher D. In Vitro MRS of Cells Treated with Trastuzumab at 1.5 Tesla. Int J Mol Sci 2024; 25:1719. [PMID: 38338997 PMCID: PMC10855746 DOI: 10.3390/ijms25031719] [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: 12/30/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The aim of the study was to investigate the effect of Trastuzumab on the MCF-7 and CRL-2314 breast cancer cell lines. Additionally, an attempt was made to optimize magnetic resonance spectroscopy (MRS) for cell culture studies, with particular emphasis on the impact of treatment with Trastuzumab. The research materials included MCF-7 and CRL-2314 breast cancer cell lines. The study examined the response of these cell lines to treatment with Trastuzumab. The clinical magnetic resonance imaging (MRI) system, OPTIMA MR360 manufactured by GEMS, with a magnetic field induction of 1.5 T, was used. Due to the nature of the tested objects, their size and shape, it was necessary to design and manufacture additional receiving coils. They were used to image the tested cell cultures and record the spectroscopic signal. The spectra obtained by MRS were confirmed by NMR using a 300 MHz NMR Fourier 300 with the TopSpin 3.1 system from Bruker. The designed receiving coils allowed for conducting experiments with the cell lines in a satisfactory manner. These tests would not be possible using factory-delivered coils due to their parameters and the size of the test objects, whose volume did not exceed 1 mL. MRS studies revealed an increase in the metabolite at 1.9 ppm, which indicates the induction of histone acetylation. Changes in histone acetylation play a very important role in both cell development and differentiation processes. The use of Trastuzumab therapy in breast cancer cells increases the levels of acetylated histones. MRS studies and spectra obtained from the 300 MHz NMR system are consistent with the specificity inherent in both systems.
Collapse
Affiliation(s)
- Wiesław Guz
- Department of Diagnostic Imaging and Nuclear Medicine, Medical College of Rzeszów University, 35-959 Rzeszów, Poland;
| | - Rafal Podgórski
- Department of Biochemistry and General Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (R.P.); (D.B.-A.)
| | - Zuzanna Bober
- Department of Photomedicine and Physical Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (Z.B.); (A.T.)
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (Z.B.); (A.T.)
| | - Adrian Truszkiewicz
- Department of Photomedicine and Physical Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (Z.B.); (A.T.)
| | - Marcin Olek
- Department of Densitry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland (A.M.P.)
| | - Agnieszka Machorowska Pieniążek
- Department of Densitry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland (A.M.P.)
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of Rzeszów University, 35-959 Rzeszów, Poland; (R.P.); (D.B.-A.)
| |
Collapse
|
8
|
Jarrett AM, Song PN, Reeves K, Lima EABF, Larimer B, Yankeelov TE, Sorace AG. Investigating tumor-host response dynamics in preclinical immunotherapy experiments using a stepwise mathematical modeling strategy. Math Biosci 2023; 366:109106. [PMID: 37931781 PMCID: PMC10841996 DOI: 10.1016/j.mbs.2023.109106] [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: 02/09/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023]
Abstract
Immunotherapies such as checkpoint blockade to PD1 and CTLA4 can have varied effects on individual tumors. To quantify the successes and failures of these therapeutics, we developed a stepwise mathematical modeling strategy and applied it to mouse models of colorectal and breast cancer that displayed a range of therapeutic responses. Using longitudinal tumor volume data, an exponential growth model was utilized to designate response groups for each tumor type. The exponential growth model was then extended to describe the dynamics of the quality of vasculature in the tumors via [18F] fluoromisonidazole (FMISO)-positron emission tomography (PET) data estimating tumor hypoxia over time. By calibrating the mathematical system to the PET data, several biological drivers of the observed deterioration of the vasculature were quantified. The mathematical model was then further expanded to explicitly include both the immune response and drug dosing, so that model simulations are able to systematically investigate biological hypotheses about immunotherapy failure and to generate experimentally testable predictions of immune response. The modeling results suggest elevated immune response fractions (> 30 %) in tumors unresponsive to immunotherapy is due to a functional immune response that wanes over time. This experimental-mathematical approach provides a means to evaluate dynamics of the system that could not have been explored using the data alone, including tumor aggressiveness, immune exhaustion, and immune cell functionality.
Collapse
Affiliation(s)
- Angela M Jarrett
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, USA; Livestrong Cancer Institutes, The University of Texas at Austin, USA
| | - Patrick N Song
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Kirsten Reeves
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama USA; Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Ernesto A B F Lima
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, USA; Livestrong Cancer Institutes, The University of Texas at Austin, USA
| | - Benjamin Larimer
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, USA; Livestrong Cancer Institutes, The University of Texas at Austin, USA; Departments of Biomedical Engineering, The University of Texas at Austin, USA; Diagnostic Medicine, The University of Texas at Austin, USA; Oncology, The University of Texas at Austin, USA; Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | - Anna G Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama USA; Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama USA.
| |
Collapse
|
9
|
Zhang H, Mi J, Xin Q, Cao W, Song C, Zhang N, Yuan C. Recent research and clinical progress of CTLA-4-based immunotherapy for breast cancer. Front Oncol 2023; 13:1256360. [PMID: 37860188 PMCID: PMC10582933 DOI: 10.3389/fonc.2023.1256360] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023] Open
Abstract
Breast cancer is characterized by a high incidence rate and its treatment challenges, particularly in certain subtypes. Consequently, there is an urgent need for the development of novel therapeutic approaches. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) is currently gaining momentum for the treatment of breast cancer. Substantial progress has been made in clinical studies employing cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) inhibitors for breast cancer, but the cure rates are relatively low. To improve the efficacy of CTLA-4-based therapy for breast cancer, further research is imperative to explore more effective immune-based treatment strategies. In addition to monotherapy, CTLA-4 inhibitors are also being investigated in combination with other ICIs or alternative medications. However, it should be noted that immune-based treatments may cause adverse events. This review focuses on the mechanisms of CTLA-4 inhibitor monotherapy or combination therapy in breast cancer. We systematically summarize the latest research and clinical advances in CTLA-4-based immunotherapy for breast cancer, providing new perspectives on the treatment of breast cancer. In addition, this review highlights the immune-related adverse events (irAEs) associated with CTLA-4 inhibitors, providing insights into the development of appropriate clinical tumor immunotherapy regimens and intervention strategies.
Collapse
Affiliation(s)
- Hongsheng Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jintao Mi
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qi Xin
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weiwei Cao
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| | - Chunjiao Song
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| | - Naidan Zhang
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| | - Chengliang Yuan
- Department of Clinical Laboratory, People’s Hospital of Deyang City, Deyang, China
| |
Collapse
|
10
|
Mizoguchi K, Kawaji H, Kai M, Morisaki T, Hayashi S, Takao Y, Yamada M, Shimazaki A, Osako T, Arima N, Okido M, Oda Y, Nakamura M, Kubo M. Granzyme B Expression in the Tumor Microenvironment as a Prognostic Biomarker for Patients with Triple-Negative Breast Cancer. Cancers (Basel) 2023; 15:4456. [PMID: 37760424 PMCID: PMC10526301 DOI: 10.3390/cancers15184456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Tumor-infiltrating lymphocytes in the tumor microenvironment are important in the treatment of triple-negative breast cancer (TNBC). Cytotoxic T cells produce cytokines and cytotoxic factors, such as perforin and granzyme, which induce apoptosis by damaging target cells. To identify biomarkers of these cells, we investigated granzyme B (GZMB) in the tumor microenvironment as a biomarker of treatment response and prognosis in 230 patients with primary TNBC who underwent surgery without preoperative chemotherapy between January 2004 and December 2014. Programmed cell death ligand 1 (PD-L1) positivity was defined as a composite positive score ≥10 based on the PD-L1 immunostaining of tumor cells and immune cells. GZMB-high was defined as positivity in ≥1% of tumor-infiltrating lymphocytes (TILs). Among the 230 TNBC patients, 117 (50.9%) had CD8-positive infiltrating tumors. In the PD-L1-positive group, a Kaplan-Meier analysis showed that GZMB-high TNBC patients had better recurrence-free survival (RFS) and overall survival (OS) than GZMB-low patients and that OS was significantly longer (RFS: p = 0.0220, OS: p = 0.0254). A multivariate analysis also showed significantly better OS in PD-L1- and GZMB-high patients (hazard ratio: 0.25 (95% IC: 0.07-0.88), p = 0.03). Our findings indicate that GZMB is a useful prognostic biomarker in PD-L1-positive TNBC patients.
Collapse
Affiliation(s)
- Kimihisa Mizoguchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Hitomi Kawaji
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Masaya Kai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Takafumi Morisaki
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Saori Hayashi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Yuka Takao
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Mai Yamada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Akiko Shimazaki
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Tomofumi Osako
- Breast Center, Kumamoto Shinto General Hospital, 3-2-65 Oe, Chuo-ku, Kumamoto 862-8655, Japan
| | - Nobuyuki Arima
- Department of Pathology, Kumamoto Shinto General Hospital, 3-2-65 Oe, Chuo-ku, Kumamoto 862-8655, Japan
| | - Masayuki Okido
- Department of Surgery, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka 810-8539, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| | - Makoto Kubo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.M.); (H.K.); (M.K.); (T.M.); (S.H.); (Y.T.); (M.Y.)
| |
Collapse
|
11
|
Massicano AVF, Song PN, Mansur A, White SL, Sorace AG, Lapi SE. [ 89Zr]-Atezolizumab-PET Imaging Reveals Longitudinal Alterations in PDL1 during Therapy in TNBC Preclinical Models. Cancers (Basel) 2023; 15:2708. [PMID: 37345044 PMCID: PMC10216761 DOI: 10.3390/cancers15102708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 06/23/2023] Open
Abstract
Triple-negative breast cancers (TNBCs) currently have limited treatment options; however, PD-L1 is an indicator of susceptibility to immunotherapy. Currently, assessment of PD-L1 is limited to biopsy samples. These limitations may be overcome with molecular imaging. In this work, we describe chemistry development and optimization, in vitro, in vivo, and dosimetry of [89Zr]-Atezolizumab for PD-L1 imaging. Atezolizumab was conjugated to DFO and radiolabeled with 89Zr. Tumor uptake and heterogeneity in TNBC xenograft and patient-derived xenograft (PDX) mouse models were quantified following [89Zr]-Atezolizumab-PET imaging. PD-L1 expression in TNBC PDX models undergoing therapy and immunohistochemistry (IHC) was used to validate imaging. SUV from PET imaging was quantified and used to identify heterogeneity. PET/CT imaging using [89Zr]-Atezolizumab identified a significant increase in tumor:muscle SUVmean 1 and 4 days after niraparib therapy and revealed an increased trend in PD-L1 expression following other cytotoxic therapies. A preliminary dosimetry study indicated the organs that will receive a higher dose are the spleen, adrenals, kidneys, and liver. [89Zr]-Atezolizumab PET/CT imaging reveals potential for the noninvasive detection of PD-L1-positive TNBC tumors and allows for quantitative and longitudinal assessment. This has potential significance for understanding tumor heterogeneity and monitoring early expression changes in PD-L1 induced by therapy.
Collapse
Affiliation(s)
| | - Patrick N. Song
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Graduate Biomedical Sciences, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ameer Mansur
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sharon L. White
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Anna G. Sorace
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Suzanne E. Lapi
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- O’Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Department of Chemistry, The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
12
|
Vaz SC, Graff SL, Ferreira AR, Debiasi M, de Geus-Oei LF. PET/CT in Patients with Breast Cancer Treated with Immunotherapy. Cancers (Basel) 2023; 15:cancers15092620. [PMID: 37174086 PMCID: PMC10177398 DOI: 10.3390/cancers15092620] [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: 03/29/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Significant advances in breast cancer (BC) treatment have been made in the last decade, including the use of immunotherapy and, in particular, immune checkpoint inhibitors that have been shown to improve the survival of patients with triple negative BC. This narrative review summarizes the studies supporting the use of immunotherapy in BC. Furthermore, the usefulness of 2-deoxy-2-[18F]fluoro-D-glucose (2-[18F]FDG) positron emission/computerized tomography (PET/CT) to image the tumor heterogeneity and to assess treatment response is explored, including the different criteria to interpret 2-[18F]FDG PET/CT imaging. The concept of immuno-PET is also described, by explaining the advantages of mapping treatment targets with a non-invasive and whole-body tool. Several radiopharmaceuticals in the preclinical phase are referred too, and, considering their promising results, translation to human studies is needed to support their use in clinical practice. Overall, this is an evolving field in BC treatment, despite PET imaging developments, the future trends also include expanding immunotherapy to early-stage BC and using other biomarkers.
Collapse
Affiliation(s)
- Sofia C Vaz
- Nuclear Medicine-Radiopharmacology, Champalimaud Center for the Unkown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
- Department of Radiology, Leiden University Medical Center, P.O. Box 9600-2300 RC Leiden, The Netherlands
| | - Stephanie L Graff
- Division of Hematology/Oncology, Lifespan Cancer Institute, Providence, RI 02903, USA
- Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Arlindo R Ferreira
- Católica Medical School, Universidade Católica Portuguesa, 2635-631 Lisbon, Portugal
| | - Márcio Debiasi
- Breast Cancer Unit, Champalimaud Center for the Unkown, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, P.O. Box 9600-2300 RC Leiden, The Netherlands
- Biomedical Photonic Imaging Group, University of Twente, P.O. Box 217-7500 AE Enschede, The Netherlands
- Department of radiation Science & Technology, Delft University of Technology, P.O. Postbus 5 2600 AA Delft, The Netherlands
| |
Collapse
|
13
|
Napier TS, Lynch SE, Lu Y, Song PN, Burns AC, Sorace AG. Molecular Imaging of Oxygenation Changes during Immunotherapy in Combination with Paclitaxel in Triple Negative Breast Cancer. Biomedicines 2023; 11:125. [PMID: 36672633 PMCID: PMC9856084 DOI: 10.3390/biomedicines11010125] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/16/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is a common feature of the tumor microenvironment, including that of triple-negative breast cancer (TNBC), an aggressive breast cancer subtype with a high five-year mortality rate. Using [18F]-fluoromisonidazole (FMISO) positron emission tomography (PET) imaging, we aimed to monitor changes in response to immunotherapy (IMT) with chemotherapy in TNBC. TNBC-tumor-bearing mice received paclitaxel (PTX) ± immune checkpoint inhibitors anti-programmed death 1 and anti-cytotoxic T-lymphocyte 4. FMISO-PET imaging was performed on treatment days 0, 6, and 12. Max and mean standard uptake values (SUVmax and SUVmean, respectively), histological analyses, and flow cytometry results were compared. FMISO-PET imaging revealed differences in tumor biology between treatment groups prior to tumor volume changes. 4T1 responders showed SUVmean 1.6-fold lower (p = 0.02) and 1.8-fold lower (p = 0.02) than non-responders on days 6 and 12, respectively. E0771 responders showed SUVmean 3.6-fold lower (p = 0.001) and 2.7-fold lower (p = 0.03) than non-responders on days 6 and 12, respectively. Immunohistochemical analyses revealed IMT plus PTX decreased hypoxia and proliferation and increased vascularity compared to control. Combination IMT/PTX recovered the loss of CD4+ T-cells observed with single-agent therapies. PET imaging can provide timely, longitudinal data on the TNBC tumor microenvironment, specifically intratumoral hypoxia, predicting therapeutic response to IMT plus chemotherapy.
Collapse
Affiliation(s)
- Tiara S. Napier
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Shannon E. Lynch
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yun Lu
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Patrick N. Song
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Andrew C. Burns
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anna G. Sorace
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
14
|
Ma X, Zhang MJ, Wang J, Zhang T, Xue P, Kang Y, Sun ZJ, Xu Z. Emerging Biomaterials Imaging Antitumor Immune Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2204034. [PMID: 35728795 DOI: 10.1002/adma.202204034] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/19/2022] [Indexed: 06/15/2023]
Abstract
Immunotherapy is one of the most promising clinical modalities for the treatment of malignant tumors and has shown excellent therapeutic outcomes in clinical settings. However, it continues to face several challenges, including long treatment cycles, high costs, immune-related adverse events, and low response rates. Thus, it is critical to predict the response rate to immunotherapy by using imaging technology in the preoperative and intraoperative. Here, the latest advances in nanosystem-based biomaterials used for predicting responses to immunotherapy via the imaging of immune cells and signaling molecules in the immune microenvironment are comprehensively summarized. Several imaging methods, such as fluorescence imaging, magnetic resonance imaging, positron emission tomography imaging, ultrasound imaging, and photoacoustic imaging, used in immune predictive imaging, are discussed to show the potential of nanosystems for distinguishing immunotherapy responders from nonresponders. Nanosystem-based biomaterials aided by various imaging technologies are expected to enable the effective prediction and diagnosis in cases of tumors, inflammation, and other public diseases.
Collapse
Affiliation(s)
- Xianbin Ma
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
- Institute of Engineering Medicine, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Meng-Jie Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Jingting Wang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Tian Zhang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Peng Xue
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Yuejun Kang
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Zhigang Xu
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, School of Materials and Energy and Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, P. R. China
| |
Collapse
|