1
|
van der Haar Àvila I, Zhang T, Lorrain V, de Bruin F, Spreij T, Nakayama H, Iwabuchi K, García-Vallejo JJ, Wuhrer M, van Kooyk Y, van Vliet SJ. Limited impact of cancer-derived gangliosides on anti-tumor immunity in colorectal cancer. Glycobiology 2024; 34:cwae036. [PMID: 38785323 PMCID: PMC11137322 DOI: 10.1093/glycob/cwae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Aberrant glycosylation is a key mechanism employed by cancer cells to evade immune surveillance, induce angiogenesis and metastasis, among other hallmarks of cancer. Sialic acids, distinctive terminal glycan structures located on glycoproteins or glycolipids, are prominently upregulated across various tumor types, including colorectal cancer (CRC). Sialylated glycans modulate anti-tumor immune responses through their interactions with Siglecs, a family of glycan-binding receptors with specificity for sialic acid-containing glycoconjugates, often resulting in immunosuppression. In this paper, we investigated the immunomodulatory function of ST3Gal5, a sialyltransferase that catalyzes the addition of α2-3 sialic acids to glycosphingolipids, since lower expression of ST3Gal5 is associated with better survival of CRC patients. We employed CRISPR/Cas9 to knock out the ST3Gal5 gene in two murine CRC cell lines MC38 and CT26. Glycomics analysis confirmed the removal of sialic acids on glycolipids, with no discernible impact on glycoprotein sialylation. Although knocking out ST3Gal5 in both cell lines did not affect in vivo tumor growth, we observed enhanced levels of regulatory T cells in CT26 tumors lacking ST3Gal5. Moreover, we demonstrate that the absence of ST3Gal5 affected size and blood vessel density only in MC38 tumors. In summary, we ascertain that sialylation of glycosphingolipids has a limited influence on the anti-tumor immune response in CRC, despite detecting alterations in the tumor microenvironment, possibly due to a shift in ganglioside abundance.
Collapse
Affiliation(s)
- Irene van der Haar Àvila
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
- Cancer Immunology, Amterdam institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Victor Lorrain
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
| | - Florance de Bruin
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
| | - Tianne Spreij
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
| | - Hitoshi Nakayama
- Graduate School of Health Care and Nursing, Laboratory of Biochemistry, Juntendo University, 2-5-1 Takasu Urayasu-shi, Chiba, 279-0023, Japan
| | - Kazuhisa Iwabuchi
- Graduate School of Health Care and Nursing, Laboratory of Biochemistry, Juntendo University, 2-5-1 Takasu Urayasu-shi, Chiba, 279-0023, Japan
| | - Juan J García-Vallejo
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
- Cancer Immunology, Amterdam institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
- Cancer Immunology, Amterdam institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | - Sandra J van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, de Boelelaan 1117, 1081 HZ Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
- Cancer Immunology, Amterdam institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Guo J, Niu Z, Lv R, Yuan J, Zhang Z, Guan X, Li D, Zhang H, Zhao A, Feng J, Liu D, Zhou X, Gong J. A novel GARP humanized mouse model for efficacy assessment of GARP-targeting therapies. Int Immunopharmacol 2024; 130:111782. [PMID: 38442579 DOI: 10.1016/j.intimp.2024.111782] [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: 11/23/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/07/2024]
Abstract
Although breakthroughs have been achieved with immune checkpoint inhibitors (ICI) therapy, some tumors do not respond to those therapies due to primary or acquired resistance. GARP, a type I transmembrane cell surface docking receptor mediating latent transforming growth factor-β (TGF-β) and abundantly expressed on regulatory T lymphocytes and platelets, is a potential target to render these tumors responsive to ICI therapy, and enhancing anti-tumor response especially combined with ICI. To facilitate these research efforts, we developed humanized mouse models expressing humanized GARP (hGARP) instead of their mouse counterparts, enabling in vivo assessment of GARP-targeting agents. We created GARP-humanized mice by replacing the mouse Garp gene with its human homolog. Then, comprehensive experiments, including expression analysis, immunophenotyping, functional assessments, and pharmacologic assays, were performed to characterize the mouse model accurately. The Tregs and platelets in the B-hGARP mice (The letter B is the first letter of the company's English name, Biocytogen.) expressed human GARP, without expression of mouse GARP. Similar T, B, NK, DCs, monocytes and macrophages frequencies were identified in the spleen and blood of B-hGARP and WT mice, indicating that the humanization of GARP did not change the distribution of immune cell in these compartments. When combined with anti-PD-1, monoclonal antibodies (mAbs) against GARP/TGF-β1 complexes demonstrated enhanced in vivo anti-tumor activity compared to monotherapy with either agent. The novel hGARP model serves as a valuable tool for evaluating human GARP-targeting antibodies in immuno-oncology, which may enable preclinical studies to assess and validate new therapeutics targeting GARP. Furthermore, intercrosses of this model with ICI humanized models could facilitate the evaluation of combination therapies.
Collapse
Affiliation(s)
- Jing Guo
- School of Life Science, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, China; Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Zhenlan Niu
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Ruili Lv
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Jiangfeng Yuan
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Zhi Zhang
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Xuewa Guan
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Dirui Li
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Haichao Zhang
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Ang Zhao
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Jia Feng
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China
| | - Dong Liu
- School of Life Science, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, China.
| | - Xiaofei Zhou
- Biocytogen Pharmaceuticals (Beijing), Beijing 102600, China.
| | - Jie Gong
- School of Life Science, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, China.
| |
Collapse
|
3
|
Qian C, Liu C, Liu W, Zhou R, Zhao L. Targeting vascular normalization: a promising strategy to improve immune-vascular crosstalk in cancer immunotherapy. Front Immunol 2023; 14:1291530. [PMID: 38193080 PMCID: PMC10773740 DOI: 10.3389/fimmu.2023.1291530] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/01/2023] [Indexed: 01/10/2024] Open
Abstract
Blood vessels are a key target for cancer therapy. Compared with the healthy vasculature, tumor blood vessels are extremely immature, highly permeable, and deficient in pericytes. The aberrantly vascularized tumor microenvironment is characterized by hypoxia, low pH, high interstitial pressure, and immunosuppression. The efficacy of chemotherapy, radiotherapy, and immunotherapy is affected by abnormal blood vessels. Some anti-angiogenic drugs show vascular normalization effects in addition to targeting angiogenesis. Reversing the abnormal state of blood vessels creates a normal microenvironment, essential for various cancer treatments, specifically immunotherapy. In addition, immune cells and molecules are involved in the regulation of angiogenesis. Therefore, combining vascular normalization with immunotherapy may increase the efficacy of immunotherapy and reduce the risk of adverse reactions. In this review, we discussed the structure, function, and formation of abnormal vessels. In addition, we elaborated on the role of the immunosuppressive microenvironment in the formation of abnormal vessels. Finally, we described the clinical challenges associated with the combination of immunotherapy with vascular normalization, and highlighted future research directions in this therapeutic area.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Chaoqun Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weiwei Liu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Rui Zhou
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Liang Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Zimmer N, Trzeciak ER, Müller A, Licht P, Sprang B, Leukel P, Mailänder V, Sommer C, Ringel F, Tuettenberg J, Kim E, Tuettenberg A. Nuclear Glycoprotein A Repetitions Predominant (GARP) Is a Common Trait of Glioblastoma Stem-like Cells and Correlates with Poor Survival in Glioblastoma Patients. Cancers (Basel) 2023; 15:5711. [PMID: 38136258 PMCID: PMC10741777 DOI: 10.3390/cancers15245711] [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: 10/20/2023] [Revised: 11/17/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Glioblastoma (GB) is notoriously resistant to therapy. GB genesis and progression are driven by glioblastoma stem-like cells (GSCs). One goal for improving treatment efficacy and patient outcomes is targeting GSCs. Currently, there are no universal markers for GSCs. Glycoprotein A repetitions predominant (GARP), an anti-inflammatory protein expressed by activated regulatory T cells, was identified as a possible marker for GSCs. This study evaluated GARP for the detection of human GSCs utilizing a multidimensional experimental design that replicated several features of GB: (1) intratumoral heterogeneity, (2) cellular hierarchy (GSCs with varied degrees of self-renewal and differentiation), and (3) longitudinal GSC evolution during GB recurrence (GSCs from patient-matched newly diagnosed and recurrent GB). Our results indicate that GARP is expressed by GSCs across various cellular states and disease stages. GSCs with an increased GARP expression had reduced self-renewal but no alterations in proliferative capacity or differentiation commitment. Rather, GARP correlated inversely with the expression of GFAP and PDGFR-α, markers of astrocyte or oligodendrocyte differentiation. GARP had an abnormal nuclear localization (GARPNU+) in GSCs and was negatively associated with patient survival. The uniformity of GARP/GARPNU+ expression across different types of GSCs suggests a potential use of GARP as a marker to identify GSCs.
Collapse
Affiliation(s)
- Niklas Zimmer
- Department of Dermatology, University Medical Center Mainz, 55131 Mainz, Germany (P.L.)
| | - Emily R. Trzeciak
- Department of Dermatology, University Medical Center Mainz, 55131 Mainz, Germany (P.L.)
| | - Andreas Müller
- Department of Neurosurgery, University Medical Center Mainz, 55131 Mainz, Germany
- Laboratory of Experimental Neurooncology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Philipp Licht
- Department of Dermatology, University Medical Center Mainz, 55131 Mainz, Germany (P.L.)
| | - Bettina Sprang
- Department of Neurosurgery, University Medical Center Mainz, 55131 Mainz, Germany
- Laboratory of Experimental Neurooncology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Petra Leukel
- Institute of Neuropathology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Volker Mailänder
- Department of Dermatology, University Medical Center Mainz, 55131 Mainz, Germany (P.L.)
- Research Center for Immunotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Florian Ringel
- Department of Neurosurgery, University Medical Center Mainz, 55131 Mainz, Germany
| | - Jochen Tuettenberg
- Department of Neurosurgery, SHG-Klinikum Idar-Oberstein, 55743 Idar-Oberstein, Germany;
| | - Ella Kim
- Department of Neurosurgery, University Medical Center Mainz, 55131 Mainz, Germany
- Laboratory of Experimental Neurooncology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center Mainz, 55131 Mainz, Germany (P.L.)
- Research Center for Immunotherapy, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
5
|
Zhang J, Liu S, Chen X, Xu X, Xu F. Non-immune cell components in tumor microenvironment influencing lung cancer Immunotherapy. Biomed Pharmacother 2023; 166:115336. [PMID: 37591126 DOI: 10.1016/j.biopha.2023.115336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023] Open
Abstract
Lung cancer (LC) is one of the leading causes of cancer-related deaths worldwide, with a significant morbidity and mortality rate, endangering human life and health. The introduction of immunotherapies has significantly altered existing cancer treatment strategies and is expected to improve immune responses, objective response rates, and survival rates. However, a better understanding of the complex immunological networks of LC is required to improve immunotherapy efficacy further. Tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs) are significantly expressed by LC cells, which activate dendritic cells, initiate antigen presentation, and activate lymphocytes to exert antitumor activity. However, as tumor cells combat the immune system, an immunosuppressive microenvironment forms, enabling the enactment of a series of immunological escape mechanisms, including the recruitment of immunosuppressive cells and induction of T cell exhaustion to decrease the antitumor immune response. In addition to the direct effect of LC cells on immune cell function, the secreting various cytokines, chemokines, and exosomes, changes in the intratumoral microbiome and the function of cancer-associated fibroblasts and endothelial cells contribute to LC cell immune escape. Accordingly, combining various immunotherapies with other therapies can elicit synergistic effects based on the complex immune network, improving immunotherapy efficacy through multi-target action on the tumor microenvironment (TME). Hence, this review provides guidance for understanding the complex immune network in the TME and designing novel and effective immunotherapy strategies for LC.
Collapse
Affiliation(s)
- Jingtao Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Shuai Liu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiubao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xiangdong Xu
- Central Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Fei Xu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
6
|
Nixon BG, Gao S, Wang X, Li MO. TGFβ control of immune responses in cancer: a holistic immuno-oncology perspective. Nat Rev Immunol 2023; 23:346-362. [PMID: 36380023 PMCID: PMC10634249 DOI: 10.1038/s41577-022-00796-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2022] [Indexed: 11/16/2022]
Abstract
The immune system responds to cancer in two main ways. First, there are prewired responses involving myeloid cells, innate lymphocytes and innate-like adaptive lymphocytes that either reside in premalignant tissues or migrate directly to tumours, and second, there are antigen priming-dependent responses, in which adaptive lymphocytes are primed in secondary lymphoid organs before homing to tumours. Transforming growth factor-β (TGFβ) - one of the most potent and pleiotropic regulatory cytokines - controls almost every stage of the tumour-elicited immune response, from leukocyte development in primary lymphoid organs to their priming in secondary lymphoid organs and their effector functions in the tumour itself. The complexity of TGFβ-regulated immune cell circuitries, as well as the contextual roles of TGFβ signalling in cancer cells and tumour stromal cells, necessitates the use of rigorous experimental systems that closely recapitulate human cancer, such as autochthonous tumour models, to uncover the underlying immunobiology. The diverse functions of TGFβ in healthy tissues further complicate the search for effective and safe cancer therapeutics targeting the TGFβ pathway. Here we discuss the contextual complexity of TGFβ signalling in tumour-elicited immune responses and explain how understanding this may guide the development of mechanism-based cancer immunotherapy.
Collapse
Affiliation(s)
- Briana G Nixon
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA
| | - Shengyu Gao
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xinxin Wang
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School of Biomedical Sciences, Cornell University, New York, NY, USA.
- Louis V. Gerstner, Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
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
|
Quach HT, Hou Z, Bellis RY, Saini JK, Amador-Molina A, Adusumilli PS, Xiong Y. Next-generation immunotherapy for solid tumors: combination immunotherapy with crosstalk blockade of TGFβ and PD-1/PD-L1. Expert Opin Investig Drugs 2022; 31:1187-1202. [PMID: 36448335 PMCID: PMC10085570 DOI: 10.1080/13543784.2022.2152323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022]
Abstract
INTRODUCTION In solid tumor immunotherapy, less than 20% of patients respond to anti-programmed cell death 1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) agents. The role of transforming growth factor β (TGFβ) in diverse immunity is well-established; however, systemic blockade of TGFβ is associated with toxicity. Accumulating evidence suggests the role of crosstalk between TGFβ and PD-1/PD-L1 pathways. AREAS COVERED We focus on TGFβ and PD-1/PD-L1 signaling pathway crosstalk and the determinant role of TGFβ in the resistance of immune checkpoint blockade. We provide the rationale for combination anti-TGFβ and anti-PD-1/PD-L1 therapies for solid tumors and discuss the current status of dual blockade therapy in preclinical and clinical studies. EXPERT OPINION The heterogeneity of tumor microenvironment across solid tumors complicates patient selection, treatment regimens, and response and toxicity assessment for investigation of dual blockade agents. However, clinical knowledge from single-agent studies provides infrastructure to translate dual blockade therapies. Dual TGFβ and PD-1/PD-L1 blockade results in enhanced T-cell infiltration into tumors, a primary requisite for successful immunotherapy. A bifunctional fusion protein specifically targets TGFβ in the tumor microenvironment, avoiding systemic toxicity, and prevents interaction of PD-1+ cytotoxic cells with PD-L1+ tumor cells.
Collapse
Affiliation(s)
- Hue Tu Quach
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Rebecca Y. Bellis
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jasmeen K. Saini
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Alfredo Amador-Molina
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Prasad S. Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Director, Mesothelioma Program; Head, Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Yuquan Xiong
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
9
|
Spourquet C, Delcorte O, Lemoine P, Dauguet N, Loriot A, Achouri Y, Hollmén M, Jalkanen S, Huaux F, Lucas S, Meerkeeck PV, Knauf JA, Fagin JA, Dessy C, Mourad M, Henriet P, Tyteca D, Marbaix E, Pierreux CE. BRAFV600E Expression in Thyrocytes Causes Recruitment of Immunosuppressive STABILIN-1 Macrophages. Cancers (Basel) 2022; 14:cancers14194687. [PMID: 36230610 PMCID: PMC9563029 DOI: 10.3390/cancers14194687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Incidence of thyroid cancer, including papillary thyroid cancer, is rapidly increasing. Oncogenes, such as the BRAFV600E, have been identified, and their effect on thyroid cancer cells have been studied in vitro and in mouse models. What is less understood is the impact of these mutations on thyroid cancer microenvironment and, in turn, the effect of changes in the microenvironment on tumor progression. We investigated the modifications in the cellular composition of thyroid cancer microenvironment using an inducible mouse model. We focused on a subpopulation of macrophages, expressing the STABILIN-1 protein, recruited in the thyroid tumor microenvironment following BRAFV600E expression. CRISPR/Cas9 genetic inactivation of Stablin-1 did not change macrophage recruitment but highlighted the immunosuppressive role of STABILIN-1-expressing macrophages. The identification of a similar subpopulation of STABILIN-1 macrophages in human thyroid diseases supports a conserved role for these macrophages and offers an opportunity for intervention. Abstract Papillary thyroid carcinoma (PTC) is the most frequent histological subtype of thyroid cancers (TC), and BRAFV600E genetic alteration is found in 60% of this endocrine cancer. This oncogene is associated with poor prognosis, resistance to radioiodine therapy, and tumor progression. Histological follow-up by anatomo-pathologists revealed that two-thirds of surgically-removed thyroids do not present malignant lesions. Thus, continued fundamental research into the molecular mechanisms of TC downstream of BRAFV600E remains central to better understanding the clinical behavior of these tumors. To study PTC, we used a mouse model in which expression of BRAFV600E was specifically switched on in thyrocytes by doxycycline administration. Upon daily intraperitoneal doxycycline injection, thyroid tissue rapidly acquired histological features mimicking human PTC. Transcriptomic analysis revealed major changes in immune signaling pathways upon BRAFV600E induction. Multiplex immunofluorescence confirmed the abundant recruitment of macrophages, among which a population of LYVE-1+/CD206+/STABILIN-1+ was dramatically increased. By genetically inactivating the gene coding for the scavenger receptor STABILIN-1, we showed an increase of CD8+ T cells in this in situ BRAFV600E-dependent TC. Lastly, we demonstrated the presence of CD206+/STABILIN-1+ macrophages in human thyroid pathologies. Altogether, we revealed the recruitment of immunosuppressive STABILIN-1 macrophages in a PTC mouse model and the interest to further study this macrophage subpopulation in human thyroid tissues.
Collapse
Affiliation(s)
- Catherine Spourquet
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Ophélie Delcorte
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Pascale Lemoine
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Nicolas Dauguet
- CYTF Platform, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Axelle Loriot
- CBIO Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Younes Achouri
- Transgenesis Platform, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Maija Hollmén
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, 20500 Turku, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, 20500 Turku, Finland
| | - François Huaux
- LTAP Unit, IREC, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Sophie Lucas
- GECE Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), 1300 Wavre, Belgium
| | - Pierre Van Meerkeeck
- GECE Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Jeffrey A. Knauf
- Department of Otolaryngology Head & Neck Surgery in the Cleveland Clinic Lerner, College of Medicine of Case Western Reserve University, Cleveland, OH 44106, USA
| | - James A. Fagin
- Department of Medicine and Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chantal Dessy
- FATH & MORF Unit, IREC, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Michel Mourad
- Surgery and Abdominal Transplantation Division, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Patrick Henriet
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Donatienne Tyteca
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Etienne Marbaix
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christophe E. Pierreux
- CELL Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
- Correspondence: ; Tel.:+32-2-764-65-22
| |
Collapse
|
10
|
Zhang M, Zhang YY, Chen Y, Wang J, Wang Q, Lu H. TGF-β Signaling and Resistance to Cancer Therapy. Front Cell Dev Biol 2021; 9:786728. [PMID: 34917620 PMCID: PMC8669610 DOI: 10.3389/fcell.2021.786728] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor β (TGF-β) pathway, which is well studied for its ability to inhibit cell proliferation in early stages of tumorigenesis while promoting epithelial-mesenchymal transition and invasion in advanced cancer, is considered to act as a double-edged sword in cancer. Multiple inhibitors have been developed to target TGF-β signaling, but results from clinical trials were inconsistent, suggesting that the functions of TGF-β in human cancers are not yet fully explored. Multiple drug resistance is a major challenge in cancer therapy; emerging evidence indicates that TGF-β signaling may be a key factor in cancer resistance to chemotherapy, targeted therapy and immunotherapy. Finally, combining anti-TGF-β therapy with other cancer therapy is an attractive venue to be explored for the treatment of therapy-resistant cancer.
Collapse
Affiliation(s)
- Maoduo Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ying Yi Zhang
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Yongze Chen
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jia Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hezhe Lu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|