1
|
Chu T, Ning Y, Ma M, Zhao Z, Liu J, Wang W, Yu X, Wang Y, Zhang S. Phillygenin regulates the colorectal cancer tumor microenvironment by inhibiting hypoxia-inducible factor 1 alpha. Cytotechnology 2025; 77:17. [PMID: 39669690 PMCID: PMC11631830 DOI: 10.1007/s10616-024-00679-2] [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: 11/12/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
The tumor microenvironment (TME) is important in the recurrence and metastasis of colorectal cancer (CRC). Phillygenin is an effective component of Forsythiae fructus that has long been used in cancer therapy. The mechanism by which phillygenin regulates the TME remains unknown. Methods and Results: A co-culture system of CRC cells and Jurkat T cells was used to simulate the TME in vitro. Network pharmacology and Human XL cytokine arrays were used to preliminarily evaluate the role of phillygenin in the TME. The target of phillygenin was determined using transfection of plasmid-producing overexpression of hypoxia-inducible factor 1 alpha (HIF-1α) overexpression or abrogated HIF-1α expression via short hairpin RNA plasmid. The therapeutic effect of phillygenin in vivo was assessed in a subcutaneous tumor mouse model. In vitro, phillygenin enhanced the immune response of T cells and prevented the immune escape of cancer cells via the inhibition of HIF-1α. Phillygenin upregulated interleukin (IL)-2 and downregulates IL-10 and FOXP3 in Jurkat T cells co-cultured with CRC cells. Phillygenin inhibited expressions of HIF-1α, transforming growth factor-beta, vascular endothelial growth factor, and CD31 in CRC cells cultured alone or with Jurkat T cells. Phillygenin considerably suppressed tumor growth and improved the TME in vivo. Conclusions: Phillygenin can enhance the immune response and inhibit angiogenesis in the TME in CRC by inhibiting HIF-1α. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00679-2.
Collapse
Affiliation(s)
- Tianhao Chu
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, China
| | - Mingqian Ma
- Graduate School of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhenying Zhao
- Tianjin Institute of Coloproctology, Tianjin Union Medical Center, 190 Jieyuan Road, Hongqiao, Tianjin, 300121 China
| | - Jun Liu
- Department of Radiology, The Fourth Central Hospital Affiliated to Nankai University, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center of Nankai University, Tianjin, China
| | - Wei Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Xueer Yu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yijia Wang
- Tianjin Institute of Coloproctology, Tianjin Union Medical Center, 190 Jieyuan Road, Hongqiao, Tianjin, 300121 China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, 190 Jieyuan Road, Hongqiao, Tianjin, 300121 China
| |
Collapse
|
2
|
Bellaye PS, Dias AMM, Vrigneaud JM, Bouchard A, Moreau M, Petitot C, Bernhard C, Claron M, Froidurot L, Morgand V, Guillemin M, Monterrat M, Mirjolet C, Garrido C, Kohli E, Collin B. Targeted radionuclide therapy against GARP expressing T regulatory cells after tumour priming with external beam radiotherapy in a murine syngeneic model. Heliyon 2024; 10:e39543. [PMID: 39498075 PMCID: PMC11533616 DOI: 10.1016/j.heliyon.2024.e39543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 11/07/2024] Open
Abstract
Purpose Radiation therapy (RT) exerts its anti-tumour efficacy by inducing direct damage to cancer cells but also through modification of the tumour microenvironment (TME) by inducing immunogenic antitumor response. Conversely, RT also promotes an immunosuppressive TME notably through the recruitment of regulatory T cells (Tregs). Glycoprotein A repetitions predominant (GARP), a transmembrane protein highly expressed by activated Tregs, plays a key role in the activation of TGF-β and thus promotes the immunosuppressive action of Tregs. The development of a theranostic approach targeting GARP combining imaging and targeted radionuclide therapy (TRT) was carried out. Methods A preclinical model of 4T1 triple negative breast tumour-bearing BALB/c mice was used to show that GARP expression is increased after external beam radiation in the TME of our cancer model. We generated a theranostic probe through the bioconjugation of the chelating agent DOTAGA onto an anti-GARP monoclonal antibody. The bioconjugation with DOTAGA allows the radiolabelling of the DOTAGA-GARP conjugate with both Indium-111 for SPECT imaging and Lutetium-177 for TRT purposes. Results We demonstrate that GARP expression is increased following RT in vivo and can be specifically detected and quantified using in vivo SPECT imaging with [111In]In-DOTAGA-GARP. In addition, 177Lu-DOTAGA-GARP limits tumour growth in our cancer model. Conclusion This theranostic strategy may allow for the personalization of cancer treatments by early detection of activated Tregs infiltration following RT and identification of patients likely to respond to Tregs-targeted therapy via TRT.
Collapse
Affiliation(s)
- Pierre-Simon Bellaye
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Alexandre MM. Dias
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Jean-Marc Vrigneaud
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Alexanne Bouchard
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079, Dijon, France
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079, Dijon, France
| | - Camille Petitot
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Claire Bernhard
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079, Dijon, France
| | - Michael Claron
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079, Dijon, France
| | - Lisa Froidurot
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Véronique Morgand
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Mélanie Guillemin
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Marie Monterrat
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Céline Mirjolet
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
| | - Carmen Garrido
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079, Dijon, France
| | - Evelyne Kohli
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079, Dijon, France
- University Hospital Centre François Mitterrand, 21000, Dijon, France
| | - Bertrand Collin
- Centre George-François Leclerc, Service de Médecine Nucléaire, IMATHERA UMS INSERM BioSanD US58, 1 rue du Professeur Marion, 21079, Dijon, France
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079, Dijon, France
| |
Collapse
|
3
|
Yang N, Hellevik T, Berzaghi R, Martinez‐Zubiaurre I. Radiation-induced effects on TGF-β and PDGF receptor signaling in cancer-associated fibroblasts. Cancer Rep (Hoboken) 2024; 7:e2018. [PMID: 38488488 PMCID: PMC10941573 DOI: 10.1002/cnr2.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) consist of heterogeneous connective tissue cells and are often constituting the most abundant cell type in the tumor stroma. Radiation effects on tumor stromal components like CAFs in the context of radiation treatment is not well-described. AIM This study explores potential changes induced by ionizing radiation (IR) on platelet-derived growth factor (PDGF)/PDGFRs and transforming growth factor-beta (TGF-β)/TGFβRs signaling systems in CAFs. METHODS AND RESULTS Experiments were carried out by employing primary cultures of human CAFs isolated from freshly resected non-small cell lung carcinoma tumor tissues. CAF cultures from nine donors were treated with one high (1 × 18 Gy) or three fractionated (3 × 6 Gy) radiation doses. Alterations in expression levels of TGFβRII and PDGFRα/β induced by IR were analyzed by western blots and flow cytometry. In the presence or absence of cognate ligands, receptor activation was studied in nonirradiated and irradiated CAFs. Radiation exposure did not exert changes in expression of PDGF or TGF-β receptors in CAFs. Additionally, IR alone was unable to trigger activation of either receptor. The radiation regimens tested did not affect PDGFRβ signaling in the presence of PDGF-BB. In contrast, signaling via pSmad2/3 and pSmad1/5/8 appeared to be down-regulated in irradiated CAFs after stimulation with TGF-β, as compared with controls. CONCLUSION Our data demonstrate that IR by itself is insufficient to induce measurable changes in PDGF or TGF-β receptor expression levels or to induce receptor activation in CAFs. However, in the presence of their respective ligands, exposure to radiation at certain doses appear to interfere with TGF-β receptor signaling.
Collapse
Affiliation(s)
- Nannan Yang
- Department of Community Medicine, Faculty of Health SciencesUiT The Arctic University of NorwayTromsøNorway
| | - Turid Hellevik
- Department of Radiation OncologyUniversity Hospital of North NorwayTromsøNorway
| | - Rodrigo Berzaghi
- Department of Clinical Medicine, Faculty of Health SciencesUiT The Arctic University of NorwayTromsøNorway
| | - Inigo Martinez‐Zubiaurre
- Department of Clinical Medicine, Faculty of Health SciencesUiT The Arctic University of NorwayTromsøNorway
| |
Collapse
|
4
|
Wang X, Eichhorn PJA, Thiery JP. TGF-β, EMT, and resistance to anti-cancer treatment. Semin Cancer Biol 2023; 97:1-11. [PMID: 37944215 DOI: 10.1016/j.semcancer.2023.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 05/08/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Transforming growth factor-β (TGF-β) signaling regulates cell-specific programs involved in embryonic development, wound-healing, and immune homeostasis. Yet, during tumor progression, these TGF-β-mediated programs are altered, leading to epithelial cell plasticity and a reprogramming of epithelial cells into mesenchymal lineages through epithelial-to-mesenchymal transition (EMT), a critical developmental program in morphogenesis and organogenesis. These changes, in turn, lead to enhanced carcinoma cell invasion, metastasis, immune cell differentiation, immune evasion, and chemotherapy resistance. Here, we discuss EMT as one of the critical programs associated with carcinoma cell plasticity and the influence exerted by TGF-β on carcinoma status and function. We further explore the composition of carcinoma and other cell populations within the tumor microenvironment, and consider the relevant outcomes related to the programs associated with cancer treatment resistance.
Collapse
Affiliation(s)
- Xuecong Wang
- Guangzhou National Laboratory, Guangzhou, China; Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Pieter Johan Adam Eichhorn
- Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Curtin Medical School, Curtin University, GPO Box U1987, Perth, WA 6845, Australia; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore, Singapore
| | | |
Collapse
|
5
|
Xie L, Liang X, Xu J, Sun X, Liu K, Sun K, Li Y, Tang X, Li X, Zhan X, Niu X, Guo W. Exploratory study of an anti-PD-L1/TGF-β antibody, TQB2858, in patients with refractory or recurrent osteosarcoma and alveolar soft part sarcoma: a report from Chinese sarcoma study group (TQB2858-Ib-02). BMC Cancer 2023; 23:868. [PMID: 37715133 PMCID: PMC10503089 DOI: 10.1186/s12885-023-11390-4] [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: 06/27/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Novel and effective immunotherapies are required for refractory or recurrent sarcomas. Transforming growth factor-beta (TGF-β) is a diverse regulatory and fibrogenic protein expressed in multiple sarcoma tumors that promotes epithelial-mesenchymal transition and excessive deposition of extracellular matrix. This study evaluated the efficacy and safety of the anti-PD-L1/TGF-β antibody TQB2858 in patients with refractory osteosarcoma and alveolar soft part sarcoma (ASPS). METHODS This single-arm phase 1b exploratory study included patients with refractory osteosarcoma or ASPS who had previously undergone at least two lines of systemic therapy. Patients were administered 1200 mg of TQB2858 once every 3 weeks. The primary endpoint was objective response rate (ORR), with null and alternative hypotheses of ORR ≤5% and ≥20%, respectively. Exploratory biomarker analyses using immunohistochemistry (IHC) staining (for PD-L1 and TGF-β) were performed on pre-treatment tumor samples. RESULTS Eleven eligible patients were included in this study. TQB2858 did not demonstrate evidence of efficacy as 0/5 osteosarcomas had any objective response, while 2/6 ASPS showed a partial response. The median progression-free survivals were 1.51 (1.38, Not Evaluable) and 2.86 (1.38, Not Evaluable) months for the osteosarcoma and ASPS groups, respectively. None of the administered cycles met the criteria for unacceptable toxicity. Other Grade 3 toxicities included abnormal liver function and elevation of γ-glutamyl transferase. IHC analysis revealed that functional enrichment in the TGF-β pathway or PD-L1 was not associated with treatment outcomes. CONCLUSIONS The combination of PD-L1 and TQB2858 did not significantly improve the ORR in patients with recurrent osteosarcoma. However, it improved immunogenic responses in ASPS, even after progression upon anti-PD-1/PD-L1 therapy, with an acceptable safety profile. IHC profiling with pathway enrichment analysis may not have any predictive value for survival outcomes. TRIAL REGISTRATION Prospectively registered in the Ethical Review Committee of Peking University People's Hospital. The trial registration number is 2021PHA105-001 and 2021PHA140-001 and the registration date was March 2, 2022. CLINICALTRIALS gov Identifier CTR20213001 and CTR20220390.
Collapse
Affiliation(s)
- Lu Xie
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Xin Liang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Jie Xu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Xin Sun
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Kuisheng Liu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China
| | - Kunkun Sun
- Pathology Department, Peking University People's Hospital, Beijing, 100044, China
| | - Yuan Li
- Radiology Department and Nuclear Medicine Department, Peking University People's Hospital, Beijing, 100044, China
| | - Xiaodong Tang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China.
| | - Xianan Li
- Orthopedic Oncology Department, Hunan Cancer Hospital, Changsha, 410013, China
| | - Xing Zhan
- Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xiaohui Niu
- Orthopedic Oncology Department, Beijing Jishuitan Hospital, Beijing, 100035, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, 100044, China.
| |
Collapse
|
6
|
Wang D, Sun Z, Zhu X, Zheng X, Zhou Y, Lu Y, Yan P, Wang H, Liu H, Jin J, Zhu H, Sun R, Wang Y, Fu B, Tian Z, Wei H. GARP-mediated active TGF-β1 induces bone marrow NK cell dysfunction in AML patients with early relapse post-allo-HSCT. Blood 2022; 140:2788-2804. [PMID: 35981475 PMCID: PMC10653097 DOI: 10.1182/blood.2022015474] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 07/22/2022] [Accepted: 08/04/2022] [Indexed: 01/05/2023] Open
Abstract
Relapse is a leading cause of death after allogeneic hematopoietic stem cell transplantation (allo-HSCT) for acute myeloid leukemia (AML). However, the underlying mechanisms remain poorly understood. Natural killer (NK) cells play a crucial role in tumor surveillance and cancer immunotherapy, and NK cell dysfunction has been observed in various tumors. Here, we performed ex vivo experiments to systematically characterize the mechanisms underlying the dysfunction of bone marrow-derived NK (BMNK) cells isolated from AML patients experiencing early relapse after allo-HSCT. We demonstrated that higher levels of active transforming growth factor β1 (TGF-β1) were associated with impaired effector function of BMNK cells in these AML patients. TGF-β1 activation was induced by the overexpression of glycoprotein A repetitions predominant on the surface of CD4+ T cells. Active TGF-β1 significantly suppressed mTORC1 activity, mitochondrial oxidative phosphorylation, the proliferation, and cytotoxicity of BMNK cells. Furthermore, pretreatment with the clinical stage TGF-β1 pathway inhibitor, galunisertib, significantly restored mTORC1 activity, mitochondrial homeostasis, and cytotoxicity. Importantly, the blockade of the TGF-β1 signaling improved the antitumor activity of NK cells in a leukemia xenograft mouse model. Thus, our findings reveal a mechanism explaining BMNK cell dysfunction and suggest that targeted inhibition of TGF-β1 signaling may represent a potential therapeutic intervention to improve outcomes in AML patients undergoing allo-HSCT or NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Dongyao Wang
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Zimin Sun
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoyu Zhu
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaohu Zheng
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yonggang Zhou
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yichen Lu
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Peidong Yan
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Huiru Wang
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Huilan Liu
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Jing Jin
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Huaiping Zhu
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Wang
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Binqing Fu
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
| | - Haiming Wei
- Division of Life Sciences and Medicine, Department of Hematology, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Institute of Immunology, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical Center, University of Science and Technology of China, Hefei, Anhui, China
- Blood and Cell Therapy Institute, Anhui Provincial Key Laboratory of Blood Research and Applications, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
7
|
Lahimchi MR, Eslami M, Yousefi B. New insight into GARP striking role in cancer progression: application for cancer therapy. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:33. [PMID: 36460874 DOI: 10.1007/s12032-022-01881-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 10/31/2022] [Indexed: 12/04/2022]
Abstract
T regulatory cells play a crucial role in antitumor immunity suppression. Glycoprotein-A repetitions predominant (GARP), transmembrane cell surface marker, is mostly expressed on Tregs and mediates intracellular organization of transforming growth factor-beta (TGF-β). The physiological role of GARP is immune system homeostasis, while it may cause tumor development by upregulating TGF-β secretion. Despite the vast application of anti- programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte Antigen-4 (CTLA-4) antibodies in immunotherapy, anti-GARP antibodies have the advantage of better response in patients who has resistance to anti-PD-1/PD-L1. Furthermore, simultaneous administration of anti-GARP antibody and anti-PD-1/PD-L1 antibody is much more effective than anti-PD-1/PD-L1 alone. It is worth mentioning that the GARP-mTGF-β complex is more potent than secretory TGF-β to induce T helper 17 cells differentiation in HIV + patients. On the other hand, TGF-β is an effective cytokine in cancer development, and some microRNAs could control its secretion by regulating GARP. In the present review, some information is provided about the undeniable role of GARP in cancer progression and its probable importance as a novel prognostic biomarker. Anti-GARP antibodies are also suggested for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Majid Eslami
- Food Safety Research Center (Salt), Semnan University of Medical Sciences, Semnan, Iran.,Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran. .,Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
8
|
McRitchie BR, Akkaya B. Exhaust the exhausters: Targeting regulatory T cells in the tumor microenvironment. Front Immunol 2022; 13:940052. [PMID: 36248808 PMCID: PMC9562032 DOI: 10.3389/fimmu.2022.940052] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022] Open
Abstract
The concept of cancer immunotherapy has gained immense momentum over the recent years. The advancements in checkpoint blockade have led to a notable progress in treating a plethora of cancer types. However, these approaches also appear to have stalled due to factors such as individuals' genetic make-up, resistant tumor sub-types and immune related adverse events (irAE). While the major focus of immunotherapies has largely been alleviating the cell-intrinsic defects of CD8+ T cells in the tumor microenvironment (TME), amending the relationship between tumor specific CD4+ T cells and CD8+ T cells has started driving attention as well. A major roadblock to improve the cross-talk between CD4+ T cells and CD8+ T cells is the immune suppressive action of tumor infiltrating T regulatory (Treg) cells. Despite their indispensable in protecting tissues against autoimmune threats, Tregs have also been under scrutiny for helping tumors thrive. This review addresses how Tregs establish themselves at the TME and suppress anti-tumor immunity. Particularly, we delve into factors that promote Treg migration into tumor tissue and discuss the unique cellular and humoral composition of TME that aids survival, differentiation and function of intratumoral Tregs. Furthermore, we summarize the potential suppression mechanisms used by intratumoral Tregs and discuss ways to target those to ultimately guide new immunotherapies.
Collapse
Affiliation(s)
- Bayley R. McRitchie
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Billur Akkaya
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
Chan MKK, Chung JYF, Tang PCT, Chan ASW, Ho JYY, Lin TPT, Chen J, Leung KT, To KF, Lan HY, Tang PMK. TGF-β signaling networks in the tumor microenvironment. Cancer Lett 2022; 550:215925. [DOI: 10.1016/j.canlet.2022.215925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/05/2022] [Accepted: 09/17/2022] [Indexed: 11/02/2022]
|
10
|
Zimmer N, Trzeciak ER, Graefen B, Satoh K, Tuettenberg A. GARP as a Therapeutic Target for the Modulation of Regulatory T Cells in Cancer and Autoimmunity. Front Immunol 2022; 13:928450. [PMID: 35898500 PMCID: PMC9309211 DOI: 10.3389/fimmu.2022.928450] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Regulatory T cells (Treg) play a critical role in immune homeostasis by suppressing several aspects of the immune response. Herein, Glycoprotein A repetitions predominant (GARP), the docking receptor for latent transforming growth factor (LTGF-β), which promotes its activation, plays a crucial role in maintaining Treg mediated immune tolerance. After activation, Treg uniquely express GARP on their surfaces. Due to its location and function, GARP may represent an important target for immunotherapeutic approaches, including the inhibition of Treg suppression in cancer or the enhancement of suppression in autoimmunity. In the present review, we will clarify the cellular and molecular regulation of GARP expression not only in human Treg but also in other cells present in the tumor microenvironment. We will also examine the overall roles of GARP in the regulation of the immune system. Furthermore, we will explore potential applications of GARP as a predictive and therapeutic biomarker as well as the targeting of GARP itself in immunotherapeutic approaches.
Collapse
Affiliation(s)
- Niklas Zimmer
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Emily R. Trzeciak
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Barbara Graefen
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kazuki Satoh
- Early Clinical Development Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, Johannes Gutenberg University Mainz, Mainz, Germany
- *Correspondence: Andrea Tuettenberg,
| |
Collapse
|
11
|
Chelerythrine Chloride Inhibits Stemness of Melanoma Cancer Stem-Like Cells (CSCs) Potentially via Inducing Reactive Oxygen Species and Causing Mitochondria Dysfunction. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4000733. [PMID: 35761835 PMCID: PMC9233603 DOI: 10.1155/2022/4000733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/27/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023]
Abstract
Growing evidence has demonstrated that high heterogeneity contributes to poor prognosis and malignancies. The existence of melanoma cancer stem-like cells (CSCs), which are a small subpopulation of melanoma cells, is responsible for tumour resistance to therapies. Recently, plant secondary metabolites have attracted attention because they are considered promising compounds that are isolated from herbs that could help to target different subpopulations of tumours. In the present study, we aimed to identify the antitumourigenic activities of the medicinal compound chelerythrine chloride (CHE) on melanoma CSCs. CHE (30-40 μmol/L) induced apoptosis in A375 and A2058 CSCs. A relatively low dose of CHE (1-5 μmol/L) inhibited the stemness of melanoma CSCs without inducing apoptosis. Coculture of CHE with A375 and A2058 cells also inhibited sphere formation and decreased stemness factors, including Nanog, Oct4, and Sox2. In functional characterizations, we observed that CHE treatment increased both cellular reactive oxygen species (ROS) and mitochondrial ROS, which resulted in a decrease in mitochondrial energy production and sphere formation. Abolishing CHE-induced ROS by N-acetyl-L-cysteine (NAC), a ROS scavenger, reversed the inhibitory effects of CHE on sphere formation, suggesting that CHE-induced ROS are the potential cause of the inhibition of sphere formation. In conclusion, CHE may exert its antitumour effect as an antistem cell natural compound, suggesting that selection of the antistem cell effects of natural compounds might be a promising strategy to overcome the poor prognosis of melanoma due to the presence of CSCs.
Collapse
|
12
|
Ortal A, Rodríguez A, Solis-Hernández MP, de Prado M, Rey V, Tornín J, Estupiñán Ó, Gallego B, Murillo D, Huergo C, García-Llano JL, Costilla S, Rodríguez R. Proof of concept for the use of trained sniffer dogs to detect osteosarcoma. Sci Rep 2022; 12:6911. [PMID: 35484295 PMCID: PMC9051207 DOI: 10.1038/s41598-022-11013-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/07/2022] [Indexed: 12/19/2022] Open
Abstract
Sarcomas are mesenchymal cancers which often show an aggressive behavior and patient survival largely depends on an early detection. In last years, much attention has been given to the fact that cancer patients release specific odorous volatile organic compounds (VOCs) that can be efficiently detected by properly trained sniffer dogs. Here, we have evaluated for the first time the ability of sniffer dogs (n = 2) to detect osteosarcoma cell cultures and patient samples. One of the two dogs was successfully trained to discriminate osteosarcoma patient-derived primary cells from mesenchymal stem/stromal cells (MSCs) obtained from healthy individuals. After the training phase, the dog was able to detect osteosarcoma specific odor cues in a different panel of 6 osteosarcoma cell lines with sensitivity and specificity rates between 95 and 100%. Moreover, the same VOCs were also detected by the sniffer dog in saliva samples from osteosarcoma patients (n = 2) and discriminated from samples from healthy individuals with a similar efficacy. Altogether, these results indicate that there are common odor profiles shared by cultures of osteosarcoma cells and body fluid samples from patients and provide a first proof of concept about the potential of canine odor detection as a non-invasive screening method to detect osteosarcomas.
Collapse
Affiliation(s)
- Agustín Ortal
- Canvida Detection Organization, CP 33212, Gijon, Spain
| | - Aida Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain
| | - María Pilar Solis-Hernández
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.,Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Verónica Rey
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain.,CIBER en Oncología (CIBERONC), 28029, Madrid, Spain
| | - Juan Tornín
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain
| | - Óscar Estupiñán
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain
| | - Borja Gallego
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain
| | - Dzohara Murillo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain
| | - Carmen Huergo
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain
| | - Juan Luis García-Llano
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Serafín Costilla
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain.,Department of Radiology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - René Rodríguez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Av. de Roma s/n, 33011, Oviedo, Spain. .,Instituto Universitario de Oncología del Principado de Asturias, 33011, Oviedo, Spain. .,CIBER en Oncología (CIBERONC), 28029, Madrid, Spain.
| |
Collapse
|
13
|
Bouchard A, Collin B, Garrido C, Bellaye PS, Kohli E. GARP: A Key Target to Evaluate Tumor Immunosuppressive Microenvironment. BIOLOGY 2021; 10:biology10090836. [PMID: 34571713 PMCID: PMC8470583 DOI: 10.3390/biology10090836] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/12/2021] [Indexed: 01/16/2023]
Abstract
Simple Summary Tumors are not only composed of cancer cells but also of various infiltrating cells constituting the tumor microenvironment (TME); all these cells produce growth factors which contribute to tumor progression and invasiveness. Among them, transforming growth factor-β1 (TGF-β1) has been shown to be a potent immunosuppressive cytokine favoring cell proliferation and invasion and to be associated with resistance to anticancer treatments. Glycoprotein-A repetition predominant (GARP) plays a critical role in the activation of TGF-β1 and has been shown to be expressed at the membrane of cancer cells and also of regulatory T cells and platelets in the TME. An increased GARP expression has been shown in a variety of cancers. The objective of this review is to highlight GARP’s expression and function in cancer and to evaluate its potential as a predictive and therapeutic follow-up biomarker that could be assessed, in real time, by molecular imaging. Abstract Glycoprotein-A repetitions predominant (GARP) is the docking receptor for latent transforming growth factor (LTGF-β) and promotes its activation. In cancer, increased GARP expression has been found in many types of cancer. GARP is expressed by regulatory T cells and platelets in the tumor microenvironment (TME) and can be also expressed by tumor cells themselves. Thus, GARP can be widely present in tumors in which it plays a major role in the production of active TGF-β, contributing to immune evasion and cancer progression via the GARP-TGF-β pathway. The objective of this review is to highlight GARP expression and function in cancer and to evaluate the potential of membrane GARP as a predictive and therapeutic follow-up biomarker that could be assessed, in real time, by molecular imaging. Moreover, as GARP can be secreted, a focus will also be made on soluble GARP as a circulating biomarker.
Collapse
Affiliation(s)
- Alexanne Bouchard
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France; (A.B.); (B.C.); (C.G.)
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Bertrand Collin
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France; (A.B.); (B.C.); (C.G.)
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR CNRS/uB 6302, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Carmen Garrido
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France; (A.B.); (B.C.); (C.G.)
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
| | - Pierre-Simon Bellaye
- Centre George-François Leclerc, Service de Médecine Nucléaire, Plateforme d’Imagerie et de Radiothérapie Précliniques, 1 rue du Professeur Marion, 21079 Dijon, France; (A.B.); (B.C.); (C.G.)
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
- Correspondence: (P.-S.B.); (E.K.)
| | - Evelyne Kohli
- UMR INSERM/uB/AGROSUP 1231, Labex LipSTIC, Faculty of Health Sciences, Université de Bourgogne Franche-Comté, 21079 Dijon, France
- CHU Dijon, 21079 Dijon, France
- Correspondence: (P.-S.B.); (E.K.)
| |
Collapse
|