1
|
Lin P, Xie W, Li Y, Zhang C, Wu H, Wan H, Gao M, Liang F, Han P, Chen R, Cheng G, Liu X, Fan S, Huang X. Intratumoral and peritumoral radiomics of MRIs predicts pathologic complete response to neoadjuvant chemoimmunotherapy in patients with head and neck squamous cell carcinoma. J Immunother Cancer 2024; 12:e009616. [PMID: 39500529 PMCID: PMC11552555 DOI: 10.1136/jitc-2024-009616] [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] [Accepted: 10/01/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND For patients with locally advanced head and neck squamous cell carcinoma (HNSCC), combined programmed death receptor-1 inhibitor and chemotherapy improved response rate to neoadjuvant therapy. However, treatment response varies among patients. There is no tool to predict pathologic complete response (pCR) with high accuracy for now. To develop a tool based on radiomics features of MRI to predict pCR to neoadjuvant chemoimmunotherapy (NACI) may provide valuable assistance in treatment regimen determination for HNSCC. METHODS From January 2021 to April 2024, a total of 172 patients with HNSCC from three medical center, who received NACI followed by surgery, were included and allocated into a training set (n=84), an internal validation set (n=37) and an external validation set (n=51). Radiomics features were extracted from intratumoral and different peritumoral areas, and radiomics signature (Rad-score) for each area was constructed. A radiomics-clinical nomogram was developed based on Rad-scores and clinicopathological characteristics, tested in the validation sets, and compared with clinical nomogram and combined positive score (CPS) in predicting pCR. RESULTS The radiomics-clinical nomogram, incorporating peritumoral Rad-score, intratumoral Rad-score and CPS, achieved the highest accuracy with areas under the receiver operating characteristic curve of 0.904 (95% CI, 0.835 to 0.972) in the training cohort, 0.860 (95% CI, 0.722 to 0.998) in the internal validation cohort, and 0.849 (95% CI, 0.739 to 0.959) in the external validation cohort, respectively, which outperformed the clinical nomogram and CPS in predict pCR to NACI for HNSCC. CONCLUSION A nomogram developed based on intratumoral and peritumoral MRI radiomics features outperformed CPS, a widely employed biomarker, in predict pCR to NACI for HNSCC, which would provide incremental value in treatment regimen determination.
Collapse
Affiliation(s)
- Peiliang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Wenqian Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Yong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Chenjia Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Huiqian Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Pathology Department, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Huan Wan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Ming Gao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Faya Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Ping Han
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Renhui Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Gui Cheng
- Department of Otolaryngology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-sen University, Shanwei, China
| | - Xuekui Liu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Xiaoming Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
XU SULING, LI XIAO, MA WENXUE. Redefining the tumor microenvironment with emerging therapeutic strategies. Oncol Res 2024; 32:1701-1708. [PMID: 39449800 PMCID: PMC11497178 DOI: 10.32604/or.2024.055161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 07/23/2024] [Indexed: 10/26/2024] Open
Abstract
The environment surrounding a tumor, known as the tumor microenvironment (TME), plays a role in how cancer progresses and responds to treatment. It poses both challenges and opportunities for improving cancer therapy. Recent progress in understanding the TME complexity and diversity has led to approaches for treating cancer. This perspective discusses the strategies for targeting the TME, such as adjusting networks using extracellular vesicles to deliver drugs and enhancing immune checkpoint inhibitors (ICIS) through combined treatments. Furthermore, it highlights adoptive cell transfer (ACT) therapies as an option for tumors. By studying how components of the TME interact and utilizing technologies like single-cell RNA sequencing and spatial transcriptomics, we can develop more precise and efficient treatments for cancer. This article emphasizes the need to reshape the TME to boost antitumor immunity and overcome resistance to therapy, providing guidance for research and clinical practices in precision oncology.
Collapse
Affiliation(s)
- SULING XU
- Department of Dermatology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - XIAO LI
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - WENXUE MA
- Department of Medicine, Sanford Stem Cell Institute and Moores Cancer Center, University of California San Diego, La Jolla, CA92093, USA
| |
Collapse
|
3
|
Yosofvand M, Edmiston SN, Smithy JW, Peng X, Kostrzewa CE, Lin B, Ehrich F, Reiner A, Miedema J, Moy AP, Orlow I, Postow MA, Panageas K, Seshan VE, Callahan MK, Thomas NE, Shen R. Spatial Immunophenotyping from Whole-Slide Multiplexed Tissue Imaging Using Convolutional Neural Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608247. [PMID: 39229153 PMCID: PMC11370407 DOI: 10.1101/2024.08.16.608247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The multiplexed immunofluorescence (mIF) platform enables biomarker discovery through the simultaneous detection of multiple markers on a single tissue slide, offering detailed insights into intratumor heterogeneity and the tumor-immune microenvironment at spatially resolved single cell resolution. However, current mIF image analyses are labor-intensive, requiring specialized pathology expertise which limits their scalability and clinical application. To address this challenge, we developed CellGate, a deep-learning (DL) computational pipeline that provides streamlined, end-to-end whole-slide mIF image analysis including nuclei detection, cell segmentation, cell classification, and combined immuno-phenotyping across stacked images. The model was trained on over 750,000 single cell images from 34 melanomas in a retrospective cohort of patients using whole tissue sections stained for CD3, CD8, CD68, CK-SOX10, PD-1, PD-L1, and FOXP3 with manual gating and extensive pathology review. When tested on new whole mIF slides, the model demonstrated high precision-recall AUC. Further validation on whole-slide mIF images of 9 primary melanomas from an independent cohort confirmed that CellGate can reproduce expert pathology analysis with high accuracy. We show that spatial immuno-phenotyping results using CellGate provide deep insights into the immune cell topography and differences in T cell functional states and interactions with tumor cells in patients with distinct histopathology and clinical characteristics. This pipeline offers a fully automated and parallelizable computing process with substantially improved consistency for cell type classification across images, potentially enabling high throughput whole-slide mIF tissue image analysis for large-scale clinical and research applications.
Collapse
|
4
|
Fu M, Li Z, Ma J, Shen F, Zhang X. Study on the predictive value of pretreatment peripheral blood inflammatory markers regarding immunotherapy in patients with inoperable advanced or locally advanced oesophageal squamous cell carcinoma. Scand J Gastroenterol 2024; 59:722-729. [PMID: 38362884 DOI: 10.1080/00365521.2024.2319319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVE To explore the effects of pretreatment peripheral blood panimmune-inflammation value (PIV), systemic immune-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) on the efficacy and prognostic value of immunotherapy in patients with inoperable advanced or locally advanced oesophageal squamous cell carcinoma (ESCC). METHODS Clinical data of 107 inoperable advanced or locally advanced ESCC patients were retrospectively analysed between May 2019 and August 2023, the receiver operating characteristic curves (ROCs) of PIV, SII, NLR, and PLR in patients prior to immunotherapy were plotted, and their optimal cutoff values were determined. The risk factors were determined by univariate and multivariate analyses in groups based on the optimal cut-off values. RESULTS Peripheral blood PIV, SII and PLR before immunotherapy had predictive value for the optimal efficacy of immunotherapy in patients with inoperable advanced or locally advanced ESCC; patients with PIV ≥415.885, SII ≥834.295 and NLR ≥3.740 had a low objective response rate (ORR), disease control rate (DCR), a short progression-free survival (PFS) and overall survival (OS) after immunotherapy (p < 0.05). Patient tumour stage, distant lymph node metastasis, lung metastasis, liver metastasis, PIV, SII, and NLR were risk factors affecting PFS and OS (p < 0.05). Tumour stage and SII were independent risk factors affecting PFS and OS (p < 0.05). CONCLUSION In patients with inoperable advanced or locally advanced ESCC, peripheral blood PIV, SII, and NLR have predictive value for immunotherapy outcome, SII is an independent risk factor affecting the survival prognosis, and SII ≥834.295 suggests a poor prognosis from immunotherapy.
Collapse
Affiliation(s)
- Maodong Fu
- Department of Integrative Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, People's Republic of China
| | - Zhiyong Li
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, People's Republic of China
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, People's Republic of China
| | - Jun Ma
- Department of Integrative Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, People's Republic of China
| | - Feng Shen
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, People's Republic of China
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, People's Republic of China
| | - Xiuping Zhang
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, People's Republic of China
- Department of Medical Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, People's Republic of China
| |
Collapse
|
5
|
Fu M, Zhang X, Shen F, Ma J, Li Z. Prognostic value of peripheral blood neutrophil/lymphocyte ratio, platelet/lymphocyte ratio, pan-immune-inflammation value and systemic immune-inflammation index for the efficacy of immunotherapy in patients with advanced gastric cancer. Immunotherapy 2024. [PMID: 38578121 DOI: 10.2217/imt-2024-0031] [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] [Indexed: 04/06/2024] Open
Abstract
Aim: The study aimed to assess the value of pretreatment peripheral blood neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio (PLR), pan-immune-inflammation value (PIV) and systemic immune-inflammation index (SII) for predicting immunotherapy prognosis and efficacy in advanced gastric cancer (GC). Methods: A total of 84 advanced GC patients received immunotherapy were retrospectively collected. The optimal cut-off values were determined by receiver operating characteristic curves. The univariate and multivariate analysis investigated the effects of NLR, PLR, PIV and SII on patients prognosis. Results: NLR, PLR, PIV and SII had predictive value of efficacy. NLR ≥3.65 was an independent risk factor for worse outcomes. Conclusion: NLR, PLR, PIV and SII have predictive value of efficacy and NLR ≥3.65 suggests a poor prognosis following immunotherapy in advanced GC.
Collapse
Affiliation(s)
- Maodong Fu
- Department of Integrated Traditional Chinese & Western Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Xiuping Zhang
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Feng Shen
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Jun Ma
- Department of Integrated Traditional Chinese & Western Medicine, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| | - Zhiyong Li
- Department of Oncology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, Fujian, 361015, People's Republic of China
- Xiamen Clinical Research Center for Cancer Therapy, Xiamen, Fujian, 361015, People's Republic of China
| |
Collapse
|
6
|
Gao S, Tang X, Gao C, Gao X, Guo X, Luo Y, Li S, Gong G, Zhang Y, Lin S. CXCL family-related classification predicts prognosis and response to immunotherapy in patients with head and neck squamous cell carcinoma based on TCGA and GEO databases. Transl Cancer Res 2024; 13:999-1015. [PMID: 38482440 PMCID: PMC10928609 DOI: 10.21037/tcr-23-1299] [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: 07/23/2023] [Accepted: 12/07/2023] [Indexed: 11/01/2024]
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is the sixth most prevalent malignant cancer worldwide. The cysteine X cysteine (CXC) chemokine family contains 17 members, which are reportedly crucial for the growth, invasion, metastasis, and microenvironment of tumor cells. Although the precise functions of CXC ligands (CXCLs) in HNSCC are unclear, these proteins may play important roles in controlling tumor growth and forming the tumor immune environment. Methods We downloaded the RNA sequencing and matched clinicopathological data of 379 patients with HNSCC as the training set from The Cancer Genome Atlas and two datasets from the Gene Expression Omnibus for use as validation sets. Results Through consensus clustering, we identified two subtypes of HNSCC associated with the CXCL family, named cluster1 and cluster2. Patients with the cluster1 subtype showed favourable clinical outcomes, significant immune cell infiltration, and improved immune response signalling pathway modulation. We also developed a nomogram of CXCL family scores for therapeutic use and for predicting the overall survival (OS) of patients with HNSCC. Patients with lower scores showed longer OS and higher immune cell infiltration in their tissues. Conclusions We developed a new classification method for HNSCC using the CXCL gene family, which can be used clinically to evaluate the prognosis and response to immunotherapy in patients with HNSCC.
Collapse
Affiliation(s)
- Shun Gao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xuemei Tang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chao Gao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xinrui Gao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xuanzhu Guo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuyao Luo
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Sijie Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guotao Gong
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Zhang
- Department of Oncology, Luzhou Municipal People’s Hospital, Luzhou, China
| | - Sheng Lin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou, China
| |
Collapse
|
7
|
Yang Y, Wang S, Wang XX, Guo S, Wang H, Shi Q, Tian Y, Wang H, Zhao T, Zhang H, Zhang B, Gao T, Li C, Yi X, Guo W. Tumorous IRE1α facilitates CD8 +T cells-dependent anti-tumor immunity and improves immunotherapy efficacy in melanoma. Cell Commun Signal 2024; 22:83. [PMID: 38291473 PMCID: PMC10826282 DOI: 10.1186/s12964-024-01470-8] [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: 06/24/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Tumor cells frequently suffer from endoplasmic reticulum (ER) stress. Previous studies have extensively elucidated the role of tumorous unfolded protein response in melanoma cells, whereas the effect on tumor immunology and the underlying mechanism remain elusive. METHODS Bioinformatics, biochemical assays and pre-clinical mice model were employed to demonstrate the role of tumorous inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) in anti-tumor immunity and the underlying mechanism. RESULTS We firstly found that IRE1α signaling activation was positively associated with the feature of tumor-infiltrating lymphocytes. Then, pharmacological ER stress induction by HA15 exerted prominent anti-tumor effect in immunocompetent mice and was highly dependent on CD8+T cells, paralleled with the reshape of immune cells in tumor microenvironment via tumorous IRE1α-XBP1 signal. Subsequently, tumorous IRE1α facilitated the expression and secretion of multiple chemokines and cytokines via XBP1-NF-κB axis, leading to increased infiltration and anti-tumor capacity of CD8+T cells. Ultimately, pharmacological induction of tumorous ER stress by HA15 brought potentiated therapeutic effect along with anti-PD-1 antibody on melanoma in vivo. CONCLUSIONS Tumorous IRE1α facilitates CD8+T cells-dependent anti-tumor immunity and improves immunotherapy efficacy by regulating chemokines and cytokines via XBP1-NF-κB axis. The combination of ER stress inducer and anti-PD-1 antibody could be promising for increasing the efficacy of melanoma immunotherapy.
Collapse
Affiliation(s)
- Yuqi Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sijia Wang
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiang-Xu Wang
- Department of Clinical Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sen Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qiong Shi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yangzi Tian
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hao Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tao Zhao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hengxiang Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Baolu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianwen Gao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Xiuli Yi
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
8
|
Gordon PB, So WY, Azubuike UF, Johnson B, Cicala J, Sturgess V, Wong C, Bishop K, Bresciani E, Sood R, Ganesan S, Tanner K. Organ specific microenvironmental MR1 expression in cutaneous melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.28.573554. [PMID: 38313277 PMCID: PMC10836068 DOI: 10.1101/2023.12.28.573554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The microenvironment is an important regulator of intertumoral trafficking and activity of immune cells. Understanding how the immune system can be tailored to maintain anti-tumor killing responses in metastatic disease remains an important goal. Thus, immune mediated eradication of metastasis requires the consideration of organ specific microenvironmental cues. Using a xenograft model of melanoma metastasis in adult zebrafish, we perturbed the dynamic balance between the infiltrating immune cells in the metastatic setting using a suite of different transgenic zebrafish. We employed intravital imaging coupled with metabolism imaging (FLIM) to visualize and map the organ specific metabolism with near simultaneity in multiple metastatic lesions. Of all the MHC complexes examined for brain and skeletal metastases, we determined that there is an organ specific expression of mhc1uba (human ortholog, MR1) for both the melanoma cells and the resident and infiltrating immune cells. Specifically, immune clusters did not express mhc1uba in brain metastatic lesions in immune competent fish. Finally, the differential immune response drove organ specific metabolism where tumor glycolysis was increased in brain metastases compared to skeletal and parental lines as measured using fluorescence lifetime imaging microscopy (FLIM). As MR1 belongs to the MHC class I molecules and is a target of immunotherapeutic drugs, we believe that our data presents an opportunity to understand the relationship between organ specific tumor metabolism and drug efficacy in the metastatic setting.
Collapse
Affiliation(s)
- Patricia B. Gordon
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Woong Young So
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Udochi F Azubuike
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bailey Johnson
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James Cicala
- Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD
| | - Victoria Sturgess
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Claudia Wong
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kevin Bishop
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Erica Bresciani
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Raman Sood
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Sundar Ganesan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Kandice Tanner
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|