1
|
Koppensteiner L, Mathieson L, Neilson L, O'Connor RA, Akram AR. IFNγ and TNFα drive an inflammatory secretion profile in cancer-associated fibroblasts from human non-small cell lung cancer. FEBS Lett 2025. [PMID: 39743376 DOI: 10.1002/1873-3468.15083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are the dominant nonmalignant component of the tumour microenvironment (TME). CAFs demonstrate a high level of inter- and intra-tumour heterogeneity in solid tumours, though the drivers of CAF subpopulations are not fully understood. Here, we demonstrate that non-small cell lung cancer (NSCLC) patient-derived CAFs upregulate the secretion of inflammatory cytokines (IL6, LIF, IL33, GM-CSF, IL1ra) and chemokines (CCL2, CCL3, CCL4, CCL20, CXCL8, CXCL9, CXCL10, CXCL11) in response to in vitro co-culture with anti-CD3/anti-CD28-stimulated peripheral blood mononuclear cells (PBMCs) via IFNγ and TNFα. Furthermore, T-cell-derived IFNγ inhibits CXCL12 secretion by CAFs in vitro. Our results highlight the ability of T-cell effector cytokines to modulate the CAF secretome in NSCLC.
Collapse
Affiliation(s)
- Lilian Koppensteiner
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, UK
| | - Layla Mathieson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, UK
| | - Liam Neilson
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, UK
| | - Richard A O'Connor
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, UK
| | - Ahsan R Akram
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, UK
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, UK
| |
Collapse
|
2
|
Forsthuber A, Aschenbrenner B, Korosec A, Jacob T, Annusver K, Krajic N, Kholodniuk D, Frech S, Zhu S, Purkhauser K, Lipp K, Werner F, Nguyen V, Griss J, Bauer W, Soler Cardona A, Weber B, Weninger W, Gesslbauer B, Staud C, Nedomansky J, Radtke C, Wagner SN, Petzelbauer P, Kasper M, Lichtenberger BM. Cancer-associated fibroblast subtypes modulate the tumor-immune microenvironment and are associated with skin cancer malignancy. Nat Commun 2024; 15:9678. [PMID: 39516494 PMCID: PMC11549091 DOI: 10.1038/s41467-024-53908-9] [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: 04/10/2023] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in cancer progression and treatment outcome. This study dissects the intra-tumoral diversity of CAFs in basal cell carcinoma, squamous cell carcinoma, and melanoma using molecular and spatial single-cell analysis. We identify three distinct CAF subtypes: myofibroblast-like RGS5+ CAFs, matrix CAFs (mCAFs), and immunomodulatory CAFs (iCAFs). Large-cohort tissue analysis reveals significant shifts in CAF subtype patterns with increasing malignancy. Two CAF subtypes exhibit immunomodulatory properties via different mechanisms. mCAFs sythesize extracellular matrix and may restrict T cell invasion in low-grade tumors via ensheathing tumor nests, while iCAFs are enriched in late-stage tumors, and express high levels of cytokines and chemokines to aid immune cell recruitment and activation. This is supported by the induction of an iCAF-like phenotype with immunomodulatory functions in primary healthy fibroblasts exposed to skin cancer cell secretomes. Thus, targeting CAF variants holds promise to enhance immunotherapy efficacy in skin cancers.
Collapse
Affiliation(s)
- Agnes Forsthuber
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bertram Aschenbrenner
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ana Korosec
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Tina Jacob
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Annusver
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Krajic
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Daria Kholodniuk
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sophie Frech
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Shaohua Zhu
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Kim Purkhauser
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Lipp
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Franziska Werner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Vy Nguyen
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ana Soler Cardona
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Benedikt Weber
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Gesslbauer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Jakob Nedomansky
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan N Wagner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Beate M Lichtenberger
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
3
|
Dharani S, Cho H, Fernandez JP, Juillerat A, Valton J, Duchateau P, Poirot L, Das S. TALEN-edited allogeneic inducible dual CAR T cells enable effective targeting of solid tumors while mitigating off-tumor toxicity. Mol Ther 2024; 32:3915-3931. [PMID: 39169622 PMCID: PMC11573618 DOI: 10.1016/j.ymthe.2024.08.018] [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: 11/04/2023] [Revised: 04/29/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
Adoptive cell therapy using chimeric antigen receptor (CAR) T cells has proven to be lifesaving for many cancer patients. However, its therapeutic efficacy has been limited in solid tumors. One key factor for this is cancer-associated fibroblasts (CAFs) that modulate the tumor microenvironment (TME) to inhibit T cell infiltration and induce "T cell dysfunction." Additionally, the sparsity of tumor-specific antigens (TSA) and expression of CAR-directed tumor-associated antigens (TAA) on normal tissues often results in "on-target off-tumor" cytotoxicity, raising safety concerns. Using TALEN-mediated gene editing, we present here an innovative CAR T cell engineering strategy to overcome these challenges. Our allogeneic "Smart CAR T cells" are designed to express a constitutive CAR, targeting FAP+ CAFs in solid tumors. Additionally, a second CAR targeting a TAA such as mesothelin is specifically integrated at a TCR signaling-inducible locus like PDCD1. FAPCAR-mediated CAF targeting induces expression of the mesothelin CAR, establishing an IF/THEN-gated circuit sensitive to dual antigen sensing. Using this approach, we observe enhanced anti-tumor cytotoxicity, while limiting "on-target off-tumor" toxicity. Our study thus demonstrates TALEN-mediated gene editing capabilities for design of allogeneic IF/THEN-gated dual CAR T cells that efficiently target immunotherapy-recalcitrant solid tumors while mitigating potential safety risks, encouraging clinical development of this strategy.
Collapse
MESH Headings
- Humans
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Mice
- Mesothelin
- Gene Editing
- Cell Line, Tumor
- Transcription Activator-Like Effector Nucleases
- Tumor Microenvironment/immunology
- Neoplasms/therapy
- Neoplasms/immunology
- Cancer-Associated Fibroblasts/metabolism
- Cancer-Associated Fibroblasts/immunology
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Xenograft Model Antitumor Assays
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- GPI-Linked Proteins/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Membrane Proteins
- Endopeptidases
Collapse
Affiliation(s)
| | - Hana Cho
- Cellectis Inc, New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Heimdörfer D, Artamonova N, Culig Z, Heidegger I. Unraveling molecular characteristics and tumor microenvironment dynamics of neuroendocrine prostate cancer. J Cancer Res Clin Oncol 2024; 150:462. [PMID: 39412660 PMCID: PMC11485041 DOI: 10.1007/s00432-024-05983-0] [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: 06/30/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024]
Abstract
Prostate cancer (PCa) is the most prevalent malignancy and the second leading cause of cancer-related deaths among men. While adenocarcinoma of the prostate (adeno-PCa) is well-characterized, neuroendocrine prostate cancer (NEPC) remains poorly understood. Generally, NEPC is a rare but highly aggressive histological variant, however its limited patho-physiological understanding leads to insufficient treatment options associated with low survival rates for NEPC patients. Current treatments for NEPC, including platinum-based therapies, offer some efficacy, but there is a significant need for more targeted approaches. This review summarizes the molecular characteristics of NEPC in contrast to adeno-PCa, providing a comprehensive comparison. A significant portion of the discussion is dedicated to the tumor microenvironment (TME), which has recently been identified as a key factor in tumor progression. The TME includes various cells, signaling molecules, and the extracellular matrix surrounding the tumor, all of which play critical roles in cancer development and response to treatment. Understanding the TME's influence on NEPC could uncover new avenues for innovative treatment strategies, potentially improving outcomes for patients with this challenging variant of PCa.
Collapse
Affiliation(s)
- David Heimdörfer
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria
| | - Nastasiia Artamonova
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria
| | - Zoran Culig
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria
| | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, Innsbruck, Anichstreet 35, Innsbruck, A-6020, Austria.
| |
Collapse
|
5
|
Guo W, Tan J, Wang L, Egelston CA, Simons DL, Ochoa A, Lim MH, Wang L, Solomon S, Waisman J, Wei CH, Hoffmann C, Song J, Schmolze D, Lee PP. Tumor draining lymph nodes connected to cold triple-negative breast cancers are characterized by Th2-associated microenvironment. Nat Commun 2024; 15:8592. [PMID: 39366933 PMCID: PMC11452381 DOI: 10.1038/s41467-024-52577-y] [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: 03/30/2023] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Tumor draining lymph nodes (TDLN) represent a key component of the tumor-immunity cycle. There are few studies describing how TDLNs impact lymphocyte infiltration into tumors. Here we directly compare tumor-free TDLNs draining "cold" and "hot" human triple negative breast cancers (TDLNCold and TDLNHot). Using machine-learning-based self-correlation analysis of immune gene expression, we find unbalanced intranodal regulations within TDLNCold. Two gene pairs (TBX21/GATA3-CXCR1) with opposite correlations suggest preferential priming of T helper 2 (Th2) cells by mature dendritic cells (DC) within TDLNCold. This is validated by multiplex immunofluorescent staining, identifying more mature-DC-Th2 spatial clusters within TDLNCold versus TDLNHot. Associated with this Th2 priming preference, more IL4 producing mast cells (MC) are found within sinus regions of TDLNCold. Downstream, Th2-associated fibrotic TME is found in paired cold tumors with increased Th2/T-helper-1-cell (Th1) ratio, upregulated fibrosis growth factors, and stromal enrichment of cancer associated fibroblasts. These findings are further confirmed in a validation cohort and public genomic data. Our results reveal a potential role of IL4+ MCs within TDLNs, associated with Th2 polarization and reduced immune infiltration into tumors.
Collapse
Affiliation(s)
- Weihua Guo
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jiayi Tan
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Lei Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- International Cancer Center, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Colt A Egelston
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Diana L Simons
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Aaron Ochoa
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Min Hui Lim
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Genomics Core, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Lu Wang
- Mork Family Department of Chemical Engineering & Material Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Shawn Solomon
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - James Waisman
- Department of Medical Oncology, City of Hope, Duarte, CA, 91010, USA
| | - Christina H Wei
- Department of Pathology, City of Hope, Duarte, CA, 91010, USA
- Pathology Laboratory Administration, Los Angeles General Medical Center, Los Angeles, CA, 90033, USA
| | - Caroline Hoffmann
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Owkin, Inc., New York, NY, 10003, USA
| | - Joo Song
- Department of Pathology, City of Hope, Duarte, CA, 91010, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope, Duarte, CA, 91010, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
6
|
Takei J, Maeda M, Fukasawa N, Kawashima M, Miyake M, Tomoto K, Nawate S, Teshigawara A, Suzuki T, Yamamoto Y, Nagashima H, Mori R, Fukushima R, Matsushima S, Kino H, Muroi A, Tsurubuchi T, Sakamoto N, Nishiwaki K, Yano S, Hasegawa Y, Murayama Y, Akasaki Y, Shimoda M, Ishikawa E, Tanaka T. Comparative analyses of immune cells and alpha-smooth muscle actin-positive cells under the immunological microenvironment between with and without dense fibrosis in primary central nervous system lymphoma. Brain Tumor Pathol 2024; 41:97-108. [PMID: 39186169 PMCID: PMC11499374 DOI: 10.1007/s10014-024-00488-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Histopathologic examinations of primary central nervous system lymphoma (PCNSL) reveal concentric accumulation of lymphocytes in the perivascular area with fibrosis. However, the nature of this fibrosis in "stiff" PCNSL remains unclear. We have encountered some PCNSLs with hard masses as surgical findings. This study investigated the dense fibrous status and tumor microenvironment of PCNSLs with or without stiffness. We evaluated by silver-impregnation nine PCNSLs with stiffness and 26 PCNSLs without stiffness. Six of the nine stiff PCNSLs showed pathological features of prominent fibrosis characterized by aggregation of reticulin fibers, and collagen accumulations. Alpha-smooth muscle actin (αSMA)-positive spindle cells as a cancer-associated fibroblast, the populations of T lymphocytes, and macrophages were compared between fibrous and control PCNSLs. Fibrous PCNSLs included abundant αSMA-positive cells in both intra- and extra-tumor environments (5/6, 87% and 3/6, 50%, respectively). Conversely, only one out of the seven control PCNSL contained αSMA-positive cells in the intra-tumoral area. Furthermore, the presence of extra-tumoral αSMA-positive cells was associated with infiltration of T lymphocytes and macrophages. In conclusion, recognizing the presence of dense fibrosis in PCNSL can provide insights into the tumor microenvironment. These results may help stratify patients with PCNSL and improve immunotherapies for these patients.
Collapse
Affiliation(s)
- Jun Takei
- Department of Neurosurgery, The Jikei University Katsushika Medical Center, 6-41-2 Aoto, Katsushika-ku, Tokyo, 125-8506, Japan
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Miku Maeda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Nei Fukasawa
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Masaharu Kawashima
- Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Misayo Miyake
- Department of Pathology, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Kyoichi Tomoto
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Shohei Nawate
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Akihiko Teshigawara
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Tomoya Suzuki
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yohei Yamamoto
- Department of Neurosurgery, The Jikei University Daisan Hospital, 4-11-1 Izumi-honcho, Komae-shi, Tokyo, 201-8601, Japan
| | - Hiroyasu Nagashima
- Department of Neurosurgery, The Jikei University Katsushika Medical Center, 6-41-2 Aoto, Katsushika-ku, Tokyo, 125-8506, Japan
| | - Ryosuke Mori
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Ryoko Fukushima
- Division of Clinical Oncology and Hematology, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Satoshi Matsushima
- Department of Radiology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hiroyoshi Kino
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Ai Muroi
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takao Tsurubuchi
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Noriaki Sakamoto
- Department of Clinical Pathology, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kaichi Nishiwaki
- Division of Clinical Oncology and Hematology, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yuzuru Hasegawa
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan
| | - Yuichi Murayama
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yasuharu Akasaki
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Toshihide Tanaka
- Department of Neurosurgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan.
- Department of Neurosurgery, The Jikei University Kashiwa Hospital, 163-1 Kashiwa-shita, Kashiwa, Chiba, 277-8567, Japan.
| |
Collapse
|
7
|
Yuan Y, Mishra F, Li B, Peng G, Chan P, Yang J, Liu Z. Modulating Tumor Immunity by Targeting Tumor Fibrotic Stroma and Angiogenic Vessels for Lung Cancer Treatment. Cancers (Basel) 2024; 16:2483. [PMID: 39001545 PMCID: PMC11240634 DOI: 10.3390/cancers16132483] [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: 06/02/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Fibrotic stroma and angiogenic tumor vessels play an important role in modulating tumor immunity. We previously reported a rationally designed protein (ProAgio) that targets integrin αvβ3 at a novel site. ProAgio induces the apoptosis of cells that express high levels of the integrin. Both activated cancer-associated fibroblasts (CAFs) and angiogenic endothelial cells (aECs) in tumors express high levels of integrin αvβ3. ProAgio simultaneously and specifically induces apoptosis in CAFs and aECs in tumors. We provide evidence here that the depletion of CAFs and the elimination of leaky tumor angiogenic vessels by ProAgio alter tumor immunity. ProAgio reduces CD4+ Treg and Myeloid-derived suppressor cells (MDSCs), increases CD8+ T-cells, and increases the M1/M2 macrophage ratio in the tumor. The depletion of dense fibrotic stroma (CAFs) by ProAgio decreases the Programmed Death Ligand 1 (PDL-1) levels in the stroma areas surrounding the tumors, and thus strongly increases the delivery of anti-PDL-1 antibody to the target cancer cells. The impact of ProAgio on tumor immunity provides strong synergistical effects of checkpoint inhibitors on lung cancer treatment.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Falguni Mishra
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Bin Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Guangda Peng
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Payton Chan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Jenny Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA;
| | - Zhiren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| |
Collapse
|
8
|
Li X, González-Maroto C, Tavassoli M. Crosstalk between CAFs and tumour cells in head and neck cancer. Cell Death Discov 2024; 10:303. [PMID: 38926351 PMCID: PMC11208506 DOI: 10.1038/s41420-024-02053-9] [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: 02/02/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are amongst the most aggressive, complex, and heterogeneous malignancies. The standard of care treatments for HNC patients include surgery, radiotherapy, chemotherapy, or their combination. However, around 50% do not benefit while suffering severe toxic side effects, costing the individuals and society. Decades have been spent to improve HNSCC treatment outcomes with only limited success. Much of the research in HNSCC treatment has focused on understanding the genetics of the HNSCC malignant cells, but it has become clear that tumour microenvironment (TME) plays an important role in the progression as well as treatment response in HNSCC. Understanding the crosstalk between cancer cells and TME is crucial for inhibiting progression and treatment resistance. Cancer-associated fibroblasts (CAFs), the predominant component of stroma in HNSCC, serve as the primary source of extra-cellular matrix (ECM) and various pro-tumoral composites in TME. The activation of CAFs in HNSCC is primarily driven by cancer cell-secreted molecules, which in turn induce phenotypic changes, elevated secretive status, and altered ECM production profile. Concurrently, CAFs play a pivotal role in modulating the cell cycle, stemness, epithelial-mesenchymal transition (EMT), and resistance to targeted and chemoradiotherapy in HNSCC cells. This modulation occurs through interactions with secreted molecules or direct contact with the ECM or CAF. Co-culture and 3D models of tumour cells and other TME cell types allows to mimic the HNSCC tumour milieu and enable modulating tumour hypoxia and reprograming cancer stem cells (CSC). This review aims to provide an update on the development of HNSCC tumour models comprising CAFs to obtain better understanding of the interaction between CAFs and tumour cells, and for providing preclinical testing platforms of current and combination with emerging therapeutics.
Collapse
Affiliation(s)
- Xinyang Li
- Head and Neck Oncology Group, Centre for Host Microbiome Interaction, King's College London, Hodgkin Building, London, SE1 1UL, UK
| | - Celia González-Maroto
- Head and Neck Oncology Group, Centre for Host Microbiome Interaction, King's College London, Hodgkin Building, London, SE1 1UL, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mahvash Tavassoli
- Head and Neck Oncology Group, Centre for Host Microbiome Interaction, King's College London, Hodgkin Building, London, SE1 1UL, UK.
| |
Collapse
|
9
|
Acharjee A, Okyere D, Nath D, Nagar S, Gkoutos GV. Network dynamics and therapeutic aspects of mRNA and protein markers with the recurrence sites of pancreatic cancer. Heliyon 2024; 10:e31437. [PMID: 38803850 PMCID: PMC11128524 DOI: 10.1016/j.heliyon.2024.e31437] [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: 11/03/2023] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/29/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease that typically manifests late patient presentation and poor outcomes. Furthermore, PDAC recurrence is a common challenge. Distinct patterns of PDAC recurrence have been associated with differential activation of immune pathway-related genes and specific inflammatory responses in their tumour microenvironment. However, the molecular associations between and within cellular components that underpin PDAC recurrence require further development, especially from a multi-omics integration perspective. In this study, we identified stable molecular associations across multiple PDAC recurrences and utilised integrative analytics to identify stable and novel associations via simultaneous feature selection. Spatial transcriptome and proteome datasets were used to perform univariate analysis, Spearman partial correlation analysis, and univariate analyses by Machine Learning methods, including regularised canonical correlation analysis and sparse partial least squares. Furthermore, networks were constructed for reported and new stable associations. Our findings revealed gene and protein associations across multiple PDAC recurrence groups, which can provide a better understanding of the multi-layer disease mechanisms that contribute to PDAC recurrence. These findings may help to provide novel association targets for clinical studies for constructing precision medicine and personalised surveillance tools for patients with PDAC recurrence.
Collapse
Affiliation(s)
- Animesh Acharjee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Health Data Research UK (HDR UK), Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, B15 2TT, United Kingdom
- Centre for Health Data Research, University of Birmingham, B15 2TT, United Kingdom
| | - Daniella Okyere
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Dipanwita Nath
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shruti Nagar
- Eureka Tutorials, Muzaffarnagar, U.P., 251201, India
| | - Georgios V. Gkoutos
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
- MRC Health Data Research UK (HDR UK), Birmingham, United Kingdom
- Institute of Translational Medicine, University Hospitals Birmingham NHS, Foundation Trust, B15 2TT, United Kingdom
- Centre for Health Data Research, University of Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
10
|
Milosevic V, Östman A. Interactions between cancer-associated fibroblasts and T-cells: functional crosstalk with targeting and biomarker potential. Ups J Med Sci 2024; 129:10710. [PMID: 38863724 PMCID: PMC11165253 DOI: 10.48101/ujms.v129.10710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 06/13/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population recognized as a key component of the tumour microenvironment (TME). Cancer-associated fibroblasts are known to play an important role in maintaining and remodelling the extracellular matrix (ECM) in the tumour stroma, supporting cancer progression and inhibiting the immune system's response against cancer cells. This review aims to summarize the immunomodulatory roles of CAFs, particularly focussing on their T-cell suppressive effects. Cancer-associated fibroblasts have several ways by which they can affect the tumour's immune microenvironment (TIME). For example, their interactions with macrophages and dendritic cells (DCs) create an immunosuppressive milieu that can indirectly affect T-cell anticancer immunity and enable immune evasion. In addition, a number of recent studies have confirmed CAF-mediated direct suppressive effects on T-cell anticancer capacity through ECM remodelling, promoting the expression of immune checkpoints, cytokine secretion and the release of extracellular vesicles. The consequential impact of CAFs on T-cell function is then reflected in affecting T-cell proliferation and apoptosis, migration and infiltration, differentiation and exhaustion. Emerging evidence highlights the existence of specific CAF subsets with distinct capabilities to modulate the immune landscape of TME in various cancers, suggesting the possibility of their exploitation as possible prognostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Vladan Milosevic
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Arne Östman
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
11
|
Buruiană A, Gheban BA, Gheban-Roșca IA, Georgiu C, Crișan D, Crișan M. The Tumor Stroma of Squamous Cell Carcinoma: A Complex Environment That Fuels Cancer Progression. Cancers (Basel) 2024; 16:1727. [PMID: 38730679 PMCID: PMC11083853 DOI: 10.3390/cancers16091727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/25/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
The tumor microenvironment (TME), a complex assembly of cellular and extracellular matrix (ECM) components, plays a crucial role in driving tumor progression, shaping treatment responses, and influencing metastasis. This narrative review focuses on the cutaneous squamous cell carcinoma (cSCC) tumor stroma, highlighting its key constituents and their dynamic contributions. We examine how significant changes within the cSCC ECM-specifically, alterations in fibronectin, hyaluronic acid, laminins, proteoglycans, and collagens-promote cancer progression, metastasis, and drug resistance. The cellular composition of the cSCC TME is also explored, detailing the intricate interplay of cancer-associated fibroblasts (CAFs), mesenchymal stem cells (MSCs), endothelial cells, pericytes, adipocytes, and various immune cell populations. These diverse players modulate tumor development, angiogenesis, and immune responses. Finally, we emphasize the TME's potential as a therapeutic target. Emerging strategies discussed in this review include harnessing the immune system (adoptive cell transfer, checkpoint blockade), hindering tumor angiogenesis, disrupting CAF activity, and manipulating ECM components. These approaches underscore the vital role that deciphering TME interactions plays in advancing cSCC therapy. Further research illuminating these complex relationships will uncover new avenues for developing more effective treatments for cSCC.
Collapse
Affiliation(s)
- Alexandra Buruiană
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Bogdan-Alexandru Gheban
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
- Emergency Clinical County Hospital, 400347 Cluj-Napoca, Romania
| | - Ioana-Andreea Gheban-Roșca
- Department of Medical Informatics and Biostatistics, Iuliu Hațieganu University of Medicine and Pharmacy, 400129 Cluj-Napoca, Romania;
| | - Carmen Georgiu
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Doința Crișan
- Department of Pathology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.B.); (C.G.); (D.C.)
| | - Maria Crișan
- Department of Histology, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
12
|
Tang H, Chen L, Liu X, Zeng S, Tan H, Chen G. Pan-cancer dissection of vasculogenic mimicry characteristic to provide potential therapeutic targets. Front Pharmacol 2024; 15:1346719. [PMID: 38694917 PMCID: PMC11061449 DOI: 10.3389/fphar.2024.1346719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/30/2024] [Indexed: 05/04/2024] Open
Abstract
Introduction Vasculogenic mimicry (VM) represents a novel form of tumor angiogenesis that is associated with tumor invasiveness and drug resistance. However, the VM landscape across cancer types remains poorly understood. In this study, we elucidate the characterizations of VM across cancers based on multi-omics data and provide potential targeted therapeutic strategies. Methods Multi-omics data from The Cancer Genome Atlas was used to conduct comprehensive analyses of the characteristics of VM related genes (VRGs) across cancer types. Pan-cancer vasculogenic mimicry score was established to provide a depiction of the VM landscape across cancer types. The correlation between VM and cancer phenotypes was conducted to explore potential regulatory mechanisms of VM. We further systematically examined the relationship between VM and both tumor immunity and tumor microenvironment (TME). In addition, cell communication analysis based on single-cell transcriptome data was used to investigate the interactions between VM cells and TME. Finally, transcriptional and drug response data from the Genomics of Drug Sensitivity in Cancer database were utilized to identify potential therapeutic targets and drugs. The impact of VM on immunotherapy was also further clarified. Results Our study revealed that VRGs were dysregulated in tumor and regulated by multiple mechanisms. Then, VM level was found to be heterogeneous among different tumors and correlated with tumor invasiveness, metastatic potential, malignancy, and prognosis. VM was found to be strongly associated with epithelial-mesenchymal transition (EMT). Further analyses revealed cancer-associated fibroblasts can promote EMT and VM formation. Furthermore, the immune-suppressive state is associated with a microenvironment characterized by high levels of VM. VM score can be used as an indicator to predict the effect of immunotherapy. Finally, seven potential drugs targeting VM were identified. Conclusion In conclusion, we elucidate the characteristics and key regulatory mechanisms of VM across various cancer types, underscoring the pivotal role of CAFs in VM. VM was further found to be associated with the immunosuppressive TME. We also provide clues for the research of drugs targeting VM. Our study provides an initial overview and reference point for future research on VM, opening up new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Haibin Tang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liuxun Chen
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xvdong Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengjie Zeng
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Tan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Chen
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Agioti S, Zaravinos A. Immune Cytolytic Activity and Strategies for Therapeutic Treatment. Int J Mol Sci 2024; 25:3624. [PMID: 38612436 PMCID: PMC11011457 DOI: 10.3390/ijms25073624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/14/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Intratumoral immune cytolytic activity (CYT), calculated as the geometric mean of granzyme-A (GZMA) and perforin-1 (PRF1) expression, has emerged as a critical factor in cancer immunotherapy, with significant implications for patient prognosis and treatment outcomes. Immune checkpoint pathways, the composition of the tumor microenvironment (TME), antigen presentation, and metabolic pathways regulate CYT. Here, we describe the various methods with which we can assess CYT. The detection and analysis of tumor-infiltrating lymphocytes (TILs) using flow cytometry or immunohistochemistry provide important information about immune cell populations within the TME. Gene expression profiling and spatial analysis techniques, such as multiplex immunofluorescence and imaging mass cytometry allow the study of CYT in the context of the TME. We discuss the significant clinical implications that CYT has, as its increased levels are associated with positive clinical outcomes and a favorable prognosis. Moreover, CYT can be used as a prognostic biomarker and aid in patient stratification. Altering CYT through the different methods targeting it, offers promising paths for improving treatment responses. Overall, understanding and modulating CYT is critical for improving cancer immunotherapy. Research into CYT and the factors that influence it has the potential to transform cancer treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Stephanie Agioti
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus;
| | - Apostolos Zaravinos
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), 1516 Nicosia, Cyprus;
- Department of Life Sciences, School of Sciences, European University Cyprus, 1516 Nicosia, Cyprus
| |
Collapse
|
14
|
Ozmen E, Demir TD, Ozcan G. Cancer-associated fibroblasts: protagonists of the tumor microenvironment in gastric cancer. Front Mol Biosci 2024; 11:1340124. [PMID: 38562556 PMCID: PMC10982390 DOI: 10.3389/fmolb.2024.1340124] [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: 11/17/2023] [Accepted: 01/31/2024] [Indexed: 04/04/2024] Open
Abstract
Enhanced knowledge of the interaction of cancer cells with their environment elucidated the critical role of tumor microenvironment in tumor progression and chemoresistance. Cancer-associated fibroblasts act as the protagonists of the tumor microenvironment, fostering the metastasis, stemness, and chemoresistance of cancer cells and attenuating the anti-cancer immune responses. Gastric cancer is one of the most aggressive cancers in the clinic, refractory to anti-cancer therapies. Growing evidence indicates that cancer-associated fibroblasts are the most prominent risk factors for a poor tumor immune microenvironment and dismal prognosis in gastric cancer. Therefore, targeting cancer-associated fibroblasts may be central to surpassing resistance to conventional chemotherapeutics, molecular-targeted agents, and immunotherapies, improving survival in gastric cancer. However, the heterogeneity in cancer-associated fibroblasts may complicate the development of cancer-associated fibroblast targeting approaches. Although single-cell sequencing studies started dissecting the heterogeneity of cancer-associated fibroblasts, the research community should still answer these questions: "What makes a cancer-associated fibroblast protumorigenic?"; "How do the intracellular signaling and the secretome of different cancer-associated fibroblast subpopulations differ from each other?"; and "Which cancer-associated fibroblast subtypes predominate specific cancer types?". Unveiling these questions can pave the way for discovering efficient cancer-associated fibroblast targeting strategies. Here, we review current knowledge and perspectives on these questions, focusing on how CAFs induce aggressiveness and therapy resistance in gastric cancer. We also review potential therapeutic approaches to prevent the development and activation of cancer-associated fibroblasts via inhibition of CAF inducers and CAF markers in cancer.
Collapse
Affiliation(s)
- Ece Ozmen
- Koç University Graduate School of Health Sciences, Istanbul, Türkiye
| | - Tevriz Dilan Demir
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
| | - Gulnihal Ozcan
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Türkiye
- Department of Medical Pharmacology, Koç University School of Medicine, Istanbul, Türkiye
| |
Collapse
|
15
|
Gu Y, Chen Q, Yin H, Zeng M, Gao S, Wang X. Cancer-associated fibroblasts in neoadjuvant setting for solid cancers. Crit Rev Oncol Hematol 2024; 193:104226. [PMID: 38056580 DOI: 10.1016/j.critrevonc.2023.104226] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/15/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
Therapeutic approaches for cancer have become increasingly diverse in recent times. A comprehensive understanding of the tumor microenvironment (TME) holds great potential for enhancing the precision of tumor therapies. Neoadjuvant therapy offers the possibility of alleviating patient symptoms and improving overall quality of life. Additionally, it may facilitate the reduction of inoperable tumors and prevent potential preoperative micrometastases. Within the TME, cancer-associated fibroblasts (CAFs) play a prominent role as they generate various elements that contribute to tumor progression. Particularly, extracellular matrix (ECM) produced by CAFs prevents immune cell infiltration into the TME, hampers drug penetration, and diminishes therapeutic efficacy. Therefore, this review provides a summary of the heterogeneity and interactions of CAFs within the TME, with a specific focus on the influence of neoadjuvant therapy on the microenvironment, particularly CAFs. Finally, we propose several potential and promising therapeutic strategies targeting CAFs, which may efficiently eliminate CAFs to decrease stroma density and impair their functions.
Collapse
Affiliation(s)
- Yanan Gu
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China; Department of Interventional Radiology, Zhongshan Hospital Fudan University Shanghai, 200032, China
| | - Qiangda Chen
- Department of Pancreatic Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hanlin Yin
- Department of Pancreatic Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China
| | - Shanshan Gao
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China.
| | - Xiaolin Wang
- Department of Radiology, Zhongshan Hospital and Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, China; Department of Interventional Radiology, Zhongshan Hospital Fudan University Shanghai, 200032, China.
| |
Collapse
|
16
|
Wang WN, Koguchi-Yoshioka H, Nimura K, Watanabe R, Tanemura A, Fujimoto M, Wataya-Kaneda M. Distinct Transcriptional Profiles in the Different Phenotypes of Neurofibroma from the Same Subject with Neurofibromatosis 1. J Invest Dermatol 2024; 144:133-141.e4. [PMID: 37301319 DOI: 10.1016/j.jid.2023.03.1688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 06/12/2023]
Abstract
Neurofibromatosis 1 is a prevalent hereditary neurocutaneous disorder. Among the clinical phenotypes of neurofibromatosis 1, cutaneous neurofibroma (cNF) and plexiform neurofibroma (pNF) have distinct clinical manifestations, and pNF should be closely monitored owing to its malignant potential. However, the detailed distinct features of neurofibromatosis 1 phenotypes remain unknown. To determine whether the transcriptional features and microenvironment of cNF and pNF differ, single-cell RNA sequencing was performed on isolated cNF and pNF cells from the same patient. Six cNF and five pNF specimens from different subjects were also immunohistochemically analyzed. Our findings revealed that cNF and pNF had distinct transcriptional profiles even within the same subject. pNF is enriched in Schwann cells with characteristics similar to those of their malignant counterpart, fibroblasts, with a cancer-associated fibroblast-like phenotype, angiogenic endothelial cells, and M2-like macrophages, whereas cNF is enriched in CD8 T cells with tissue residency markers. The results of immunohistochemical analyses performed on different subjects agreed with those of single-cell RNA sequencing. This study found that cNF and pNF, the different neurofibromatosis phenotypes in neurofibromatosis 1, from the same subject are transcriptionally distinct in terms of the cell types involved, including T cells.
Collapse
Affiliation(s)
- Wei-Ning Wang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan; Division of Health Science, Department of Neurocutaneous Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rei Watanabe
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan; Department of Integrative Medicine for Allergic and Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan; Division of Health Science, Department of Neurocutaneous Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
17
|
DuCote TJ, Naughton KJ, Skaggs EM, Bocklage TJ, Allison DB, Brainson CF. Using Artificial Intelligence to Identify Tumor Microenvironment Heterogeneity in Non-Small Cell Lung Cancers. J Transl Med 2023; 103:100176. [PMID: 37182840 PMCID: PMC10527157 DOI: 10.1016/j.labinv.2023.100176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023] Open
Abstract
Lung cancer heterogeneity is a major barrier to effective treatments and encompasses not only the malignant epithelial cell phenotypes and genetics but also the diverse tumor-associated cell types. Current techniques used to investigate the tumor microenvironment can be time-consuming, expensive, complicated to interpret, and often involves destruction of the sample. Here we use standard hematoxylin and eosin-stained tumor sections and the HALO AI nuclear phenotyping software to characterize 6 distinct cell types (epithelial, mesenchymal, macrophage, neutrophil, lymphocyte, and plasma cells) in both murine lung cancer models and human lung cancer samples. CD3 immunohistochemistry and lymph node sections were used to validate lymphocyte calls, while F4/80 immunohistochemistry was used for macrophage validation. Consistent with numerous prior studies, we demonstrated that macrophages predominate the adenocarcinomas, whereas neutrophils predominate the squamous cell carcinomas in murine samples. In human samples, we showed a strong negative correlation between neutrophils and lymphocytes as well as between mesenchymal cells and lymphocytes and that higher percentages of mesenchymal cells correlate with poor prognosis. Taken together, we demonstrate the utility of this AI software to identify, quantify, and compare distributions of cell types on standard hematoxylin and eosin-stained slides. Given the simplicity and cost-effectiveness of this technique, it may be widely beneficial for researchers designing new therapies and clinicians working to select favorable treatments for their patients.
Collapse
Affiliation(s)
- Tanner J DuCote
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Kassandra J Naughton
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Erika M Skaggs
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Therese J Bocklage
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Derek B Allison
- Markey Cancer Center, University of Kentucky, Lexington, Kentucky; Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, Kentucky
| | - Christine F Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky; Markey Cancer Center, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
18
|
Koppensteiner L, Mathieson L, Pattle S, Dorward DA, O'Connor R, Akram AR. Location of CD39 + T cell subpopulations within tumors predict differential outcomes in non-small cell lung cancer. J Immunother Cancer 2023; 11:e006770. [PMID: 37648263 PMCID: PMC10471883 DOI: 10.1136/jitc-2023-006770] [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: 06/25/2023] [Indexed: 09/01/2023] Open
Abstract
PURPOSE An improved mechanistic understanding of immunosuppressive pathways in non-small cell lung cancer (NSCLC) is important to develop novel diagnostic and therapeutic approaches. Here, we investigate the prognostic significance of the ectonucleotidases CD39 and CD73 in NSCLC. EXPERIMENTAL DESIGN The expression and localization of CD39, CD73 and CD103 was digitally quantified in a cohort of 162 early treatment naïve NSCLC patients using multiplex-immunofluorescence and related to patient outcome. Expression among different cell-populations was assessed via flow cytometry. Targeted RNA-Seq was performed on CD4+ and CD8+ T cells from digested NSCLC tumor tissue and single-cell RNA-Seq data was analyzed to investigate the functional significance of CD39+ T cell populations. RESULTS We demonstrate that flow cytometry of early untreated NSCLC patients shows an upregulation of CD39 expression in the tumor tissue among natural killer (NK) cells, fibroblasts and T cells. CD73 expression is mainly found among fibroblasts and Epcam+cells in the tumor tissue. Multiplex Immunofluorescence in a cohort of 162 early untreated NSCLC patients demonstrates that CD39 expression is mainly localized in the tumor stroma while CD73 expression is equally distributed between tumor nest and stroma, and high expression of CD39 and CD73 in the tumor stroma is associated with poor recurrence-free survival (RFS) at 5 years. Additionally, we find that CD8+T cells located in the tumor nest express CD103 and the density of CD39+CD103+CD8+ T cells in the tumor nest predicts improved RFS at 5 years. Targeted RNA-Seq shows that the tumor microenvironment of NSCLC upregulates regulatory pathways in CD4+ T cells and exhaustion in CD8+ T cells, and analysis of a single cell RNA sequencing dataset shows that CD39+CD4+ cells are enriched in Treg signature gene-sets, and CD39+CD103+ cytotoxic T lymphocyte show gene signatures indicative of an exhausted cytotoxic phenotype with upregulated expression of CXCL13. CONCLUSIONS Knowledge of patterns of distribution and location are required to understand the prognostic impact of CD39+ T cell populations in NSCLC. This study provides an improved understanding of spatial and functional characteristics of CD39+ T cells and their significance to patient outcome.
Collapse
Affiliation(s)
| | - Layla Mathieson
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Samuel Pattle
- Department of Pathology, Royal Infirmary, Edinburgh, UK
| | | | - Richard O'Connor
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
| | - Ahsan R Akram
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, UK
- Cancer Research UK Edinburgh Centre, Institute of Genetics & Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
19
|
O’Connor RA, Martinez BR, Koppensteiner L, Mathieson L, Akram AR. Cancer-associated fibroblasts drive CXCL13 production in activated T cells via TGF-beta. Front Immunol 2023; 14:1221532. [PMID: 37520560 PMCID: PMC10373066 DOI: 10.3389/fimmu.2023.1221532] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Tumour-reactive T cells producing the B-cell attractant chemokine CXCL13, in solid tumours, promote development of tertiary lymphoid structures (TLS) and are associated with improved prognosis and responsiveness to checkpoint immunotherapy. Cancer associated fibroblasts are the dominant stromal cell type in non-small cell lung cancer (NSCLC) where they co-localise with T cells and can influence T cell activation and exhaustion. We questioned whether CAF directly promote CXCL13-production during T cell activation. Methods We characterised surface markers, cytokine production and transcription factor expression in CXCL13-producing T cells in NSCLC tumours and paired non-cancerous lung samples using flow cytometry. We then assessed the influence of human NSCLC-derived primary CAF lines on T cells from healthy donors and NSCLC patients during activation in vitro measuring CXCL13 production and expression of cell-surface markers and transcription factors by flow cytometry. Results CAFs significantly increased the production of CXCL13 by both CD4+ and CD8+ T cells. CAF-induced CXCL13-producing cells lacked expression of CXCR5 and BCL6 and displayed a T peripheral helper cell phenotype. Furthermore, we demonstrate CXCL13 production by T cells is induced by TGF-β and limited by IL-2. CAF provide TGF-β during T cell activation and reduce availability of IL-2 both directly (by reducing the capacity for IL-2 production) and indirectly, by expanding a population of activated Treg. Inhibition of TGF-β signalling prevented both CAF-driven upregulation of CXCL13 and Treg expansion. Discussion Promoting CXCL13 production represents a newly described immune-regulatory function of CAF with the potential to shape the immune infiltrate of the tumour microenvironment both by altering the effector-function of tumour infiltrating T-cells and their capacity to attract B cells and promote TLS formation.
Collapse
Affiliation(s)
- Richard A. O’Connor
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Begoña Roman Martinez
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Lilian Koppensteiner
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Layla Mathieson
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Ahsan R. Akram
- Centre for Inflammation Research, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Das S, Valton J, Duchateau P, Poirot L. Stromal depletion by TALEN-edited universal hypoimmunogenic FAP-CAR T cells enables infiltration and anti-tumor cytotoxicity of tumor antigen-targeted CAR-T immunotherapy. Front Immunol 2023; 14:1172681. [PMID: 37251405 PMCID: PMC10213512 DOI: 10.3389/fimmu.2023.1172681] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
Adoptive cell therapy based on chimeric antigen receptor (CAR)-engineered T-cells has proven to be lifesaving for many cancer patients. However, its therapeutic efficacy has so far been restricted to only a few malignancies, with solid tumors proving to be especially recalcitrant to efficient therapy. Poor intra-tumor infiltration by T cells and T cell dysfunction due to a desmoplastic, immunosuppressive microenvironment are key barriers for CAR T-cell success against solid tumors. Cancer-associated fibroblasts (CAFs) are critical components of the tumor stroma, evolving specifically within the tumor microenvironment (TME) in response to tumor cell cues. The CAF secretome is a significant contributor to the extracellular matrix and a plethora of cytokines and growth factors that induce immune suppression. Together they form a physical and chemical barrier which induces a T cell-excluding 'cold' TME. CAF depletion in stroma rich solid tumors can thus provide an opportunity to convert immune evasive tumors susceptible to tumor-antigen CAR T-cell cytotoxicity. Using our TALEN-based gene editing platform we engineered non-alloreactive, immune evasive CAR T-cells (termed UCAR T-cells) targeting the unique CAF marker Fibroblast Activation Protein, alpha (FAP). In an orthotopic mouse model of triple-negative breast cancer (TNBC) composed of patient derived-CAFs and tumor cells, we demonstrate the efficacy of our engineered FAP UCAR T-cells in CAF depletion, reduction of desmoplasia and successful tumor infiltration. Furthermore, while previously resistant, pre-treatment with FAP UCAR T-cells now sensitized these tumors to Mesothelin (Meso) UCAR T-cell infiltration and anti-tumor cytotoxicity. Combination therapy of FAP UCAR, Meso UCAR T cells and the checkpoint inhibitor anti-PD-1 significantly reduced tumor burden and prolonged mice survival. Our study thus proposes a novel treatment paradigm for successful CAR T-cell immunotherapy against stroma-rich solid tumors.
Collapse
Affiliation(s)
- Shipra Das
- Cellectis Inc, New York, NY, United States
| | | | | | | |
Collapse
|
21
|
Moeckel C, Bakhl K, Georgakopoulos-Soares I, Zaravinos A. The Efficacy of Tumor Mutation Burden as a Biomarker of Response to Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24076710. [PMID: 37047684 PMCID: PMC10095310 DOI: 10.3390/ijms24076710] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/21/2023] [Accepted: 03/31/2023] [Indexed: 04/14/2023] Open
Abstract
Cancer is one of the leading causes of death in the world; therefore, extensive research has been dedicated to exploring potential therapeutics, including immune checkpoint inhibitors (ICIs). Initially, programmed-death ligand-1 was the biomarker utilized to predict the efficacy of ICIs. However, its heterogeneous expression in the tumor microenvironment, which is critical to cancer progression, promoted the exploration of the tumor mutation burden (TMB). Research in various cancers, such as melanoma and lung cancer, has shown an association between high TMB and response to ICIs, increasing its predictive value. However, the TMB has failed to predict ICI response in numerous other cancers. Therefore, future research is needed to analyze the variations between cancer types and establish TMB cutoffs in order to create a more standardized methodology for using the TMB clinically. In this review, we aim to explore current research on the efficacy of the TMB as a biomarker, discuss current approaches to overcoming immunoresistance to ICIs, and highlight new trends in the field such as liquid biopsies, next generation sequencing, chimeric antigen receptor T-cell therapy, and personalized tumor vaccines.
Collapse
Affiliation(s)
- Camille Moeckel
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Katrina Bakhl
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ilias Georgakopoulos-Soares
- Department of Biochemistry and Molecular Biology, Institute for Personalized Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Apostolos Zaravinos
- Department of Life Sciences, European University Cyprus, Diogenis Str., 6, Nicosia 2404, Cyprus
- Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia 1516, Cyprus
| |
Collapse
|
22
|
Geng S, Xiang T, Zhang Y, Guo P, Zhang H, Zhang Z, Gu M, Zhang K, Song H, Shi J, Liu J. Safe engineering of cancer-associated fibroblasts enhances checkpoint blockade immunotherapy. J Control Release 2023; 356:272-287. [PMID: 36870541 DOI: 10.1016/j.jconrel.2023.02.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
Abundant cancer-associated fibroblasts (CAFs) in highly fibrotic breast cancer constitute an immunosuppressive barrier for T cell activity and are closely related to the failure of immune checkpoint blockade therapy (ICB). Inspired by the similar antigen-processing capacity of CAFs to professional antigen-presenting cells (APCs), a "turning foes to friends" strategy is proposed by in situ engineering immune-suppressed CAFs into immune-activated APCs for improving response rates of ICB. To achieve safe and specific CAFs engineering in vivo, a thermochromic spatiotemporal photo-controlled gene expression nanosystem was developed by self-assembly of molten eutectic mixture, chitosan andfusion plasmid. After photoactivatable gene expression, CAFs could be engineered as APCs via co-stimulatory molecule (CD86) expression, which effectively induced activation and proliferation of antigen-specific CD8 + T cells. Meanwhile, engineered CAFs could also secrete PD-L1 trap protein in situ for ICB, avoiding potential autoimmune-like disorders caused by "off-target" effects of clinically applied PD-L1 antibody. The study demonstrated that the designed nanosystem could efficiently engineer CAFs, significantly enhance the percentages of CD8+ T cells (4-folds), result in about 85% tumor inhibition rate and 83.3% survival rate at 60 days in highly fibrotic breast cancer, further inducing long-term immune memory effects and effectively inhibiting lung metastasis.
Collapse
Affiliation(s)
- Shizhen Geng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Tingting Xiang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yunya Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pengke Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Hongling Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Mengchao Gu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China
| | - Haiwei Song
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore 138673, Singapore.
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Henan Province, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou 450001, China.
| |
Collapse
|
23
|
Pakravan K, Mossahebi-Mohammadi M, Ghazimoradi MH, Cho WC, Sadeghizadeh M, Babashah S. Monocytes educated by cancer-associated fibroblasts secrete exosomal miR-181a to activate AKT signaling in breast cancer cells. J Transl Med 2022; 20:559. [PMID: 36463188 PMCID: PMC9719191 DOI: 10.1186/s12967-022-03780-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/17/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), one of the major components of the tumor stroma, contribute to an immunosuppressive tumor microenvironment (TME) through the induction and functional polarization of protumoral macrophages. We have herein investigated the contribution of CAFs to monocyte recruitment and macrophage polarization. We also sought to identify a possible paracrine mechanism by which CAF-educated monocytes affect breast cancer (BC) cell progression. METHODS Monocytes were educated by primary CAFs and normal fibroblast (NF); the phenotypic alterations of CAF- or NF-educated monocytes were measured by flow cytometry. Exosomes isolated from the cultured conditioned media of the educated monocytes were characterized. An in vivo experiment using a subcutaneous transplantation tumor model in athymic nude mice was conducted to uncover the effect of exosomes derived from CAF- or NF-educated monocytes on breast tumor growth. Gain- and loss-of-function experiments were performed to explore the role of miR-181a in BC progression with the involvement of the AKT signaling pathway. Western blotting, enzyme-linked immunosorbent assay, RT-qPCR, flow cytometry staining, migration assay, immunohistochemical staining, and bioinformatics analysis were performed to reveal the underlying mechanisms. RESULTS We illustrated that primary CAFs recruited monocytes and established pro-tumoral M2 macrophages. CAF may also differentiate human monocyte THP-1 cells into anti-inflammatory M2 macrophages. Besides, we revealed that CAFs increased reactive oxygen species (ROS) generation in THP-1 monocytes, as differentiating into M2 macrophages requires a level of ROS for proper polarization. Importantly, T-cell proliferation was suppressed by CAF-educated monocytes and their exosomes, resulting in an immunosuppressive TME. Interestingly, CAF-activated, polarized monocytes lost their tumoricidal abilities, and their derived exosomes promoted BC cell proliferation and migration. In turn, CAF-educated monocyte exosomes exhibited a significant promoting effect on BC tumorigenicity in vivo. Of clinical significance, we observed that up-regulation of circulating miR-181a in BC was positively correlated with tumor aggressiveness and found a high level of this miRNA in CAF-educated monocytes and their exosomes. We further clarified that the pro-oncogenic effect of CAF-educated monocytes may depend in part on the exosomal transfer of miR-181a through modulating the PTEN/Akt signaling axis in BC cells. CONCLUSIONS Our findings established a connection between tumor stromal communication and tumor progression and demonstrated an inductive function for CAF-educated monocytes in BC cell progression. We also proposed a supporting model in which exosomal transfer of miR-181a from CAF-educated monocytes activates AKT signaling by regulating PTEN in BC cells.
Collapse
Affiliation(s)
- Katayoon Pakravan
- grid.412266.50000 0001 1781 3962Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Majid Mossahebi-Mohammadi
- grid.412266.50000 0001 1781 3962Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad H. Ghazimoradi
- grid.412266.50000 0001 1781 3962Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - William C. Cho
- grid.415499.40000 0004 1771 451XDepartment of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong China
| | - Majid Sadeghizadeh
- grid.412266.50000 0001 1781 3962Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Sadegh Babashah
- grid.412266.50000 0001 1781 3962Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| |
Collapse
|
24
|
Eskandari-Malayeri F, Rezaei M. Immune checkpoint inhibitors as mediators for immunosuppression by cancer-associated fibroblasts: A comprehensive review. Front Immunol 2022; 13:996145. [PMID: 36275750 PMCID: PMC9581325 DOI: 10.3389/fimmu.2022.996145] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/01/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor microenvironment (TME) is a significant contributor to cancer progression containing complex connections between cellular and chemical components and provides a suitable substrate for tumor growth and development. Growing evidence shows targeting tumor cells while ignoring the surrounding TME is not effective enough to overcome the cancer disease. Fibroblasts are essential sentinels of the stroma that due to certain conditions in TME, such as oxidative stress and local hypoxia, become activated, and play the prominent role in the physical support of tumor cells and the enhancement of tumorigenesis. Activated fibroblasts in TME, defined as cancer-associated fibroblasts (CAFs), play a crucial role in regulating the biological behavior of tumors, such as tumor metastasis and drug resistance. CAFs are highly heterogeneous populations that have different origins and, in addition to their role in supporting stromal cells, have multiple immunosuppressive functions via a membrane and secretory patterns. The secretion of different cytokines/chemokines, interactions that mediate the recruitment of regulatory immune cells and the reprogramming of an immunosuppressive function in immature myeloid cells are just a few examples of how CAFs contribute to the immune escape of tumors through various direct and indirect mechanisms on specific immune cell populations. Moreover, CAFs directly abolish the role of cytotoxic lymphocytes. The activation and overexpression of inhibitory immune checkpoints (iICPs) or their ligands in TME compartments are one of the main regulatory mechanisms that inactivate tumor-infiltrating lymphocytes in cancer lesions. CAFs are also essential players in the induction or expression of iICPs and the suppression of immune response in TME. Based on available studies, CAF subsets could modulate immune cell function in TME through iICPs in two ways; direct expression of iICPs by activated CAFs and indirect induction by production soluble and then upregulation of iICPs in TME. With a focus on CAFs’ direct and indirect roles in the induction of iICPs in TME as well as their use in immunotherapy and diagnostics, we present the evolving understanding of the immunosuppressive mechanism of CAFs in TME in this review. Understanding the complete picture of CAFs will help develop new strategies to improve precision cancer therapy.
Collapse
|
25
|
Wang Z, Zhang H, Huang C, Li K, Luo W, Zhang G, Li X. Predictive value of modified systemic inflammation score for postoperative unplanned ICU admission in patients with NSCLC. Front Surg 2022; 9:893555. [PMID: 35990092 PMCID: PMC9381959 DOI: 10.3389/fsurg.2022.893555] [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: 03/10/2022] [Accepted: 07/18/2022] [Indexed: 01/17/2023] Open
Abstract
BackgroundThe purpose of this study was to investigate the predictive value of the modified systemic inflammation score (mSIS) in postoperative unplanned admission to the intensive care unit (ICU) in patients with non-small-cell lung cancer (NSCLC).MethodsThe clinical data of 1,321 patients with NSCLC treated with thoracic surgery in our hospital from August 2019 to June 2021 were analyzed retrospectively. The preoperative mSIS, which takes into account the serum albumin (ALB) level and lymphocyte-to-monocyte ratio (LMR), was recorded as 0, 1 or 2 and then was used to identify high-risk patients with unplanned admission to the ICU. The independent risk factors for unplanned admission to the ICU in patients with NSCLC after surgery were identified by multivariate logistic regression analysis.ResultsA total of 1,321 patients, including 549 (41.6%) males and 772 (58.4%) females, were included. The median age was 57 years (range 16–95 years). The incidence of unplanned admission to the ICU in patients with mSIS = 2 was significantly higher than that in those with mSIS = 0 and mSIS = 1. The multivariate analysis showed that an mSIS of 2 (OR = 3.728; P = 0.004; 95% CI, 1.520–9.143), an alcohol consumption history (OR = 2.791, P = 0.011; 95% CI, 1.262–6.171), intraoperative infusion volume (OR = 1.001, P = 0.021; 95% CI, 1.000–1.001) and preoperative underlying diseases (OR = 3. 57, P = 0.004; 95% CI, 1.497–8.552) were independent risk factors for unplanned admission to the ICU after lung cancer surgery. In addition, the multivariate logistic regression model showed that the C-statistic value was 0.799 (95% CI: 0.726∼0.872, P < 0.001).ConclusionsThe mSIS scoring system can be used as a simplified and effective predictive tool for unplanned ICU admission in patients with NSCLC.
Collapse
Affiliation(s)
- Zhulin Wang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, ZhengzhouChina
| | - Hua Zhang
- Department of Cardiovascular surgery, Henan Provincial Chest Hospital, ZhengzhouChina
| | - Chunyao Huang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, ZhengzhouChina
| | - Kaiyuan Li
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, ZhengzhouChina
| | - Wenqing Luo
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, ZhengzhouChina
| | - Guoqing Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, ZhengzhouChina
- Correspondence: Xiangnan Li Guoqing Zhang
| | - Xiangnan Li
- Department of Thoracic Surgery, First Affiliated Hospital of Zhengzhou University, ZhengzhouChina
- Correspondence: Xiangnan Li Guoqing Zhang
| |
Collapse
|