1
|
Jacques C, Marchesi I, Fiorentino FP, Marchand F, Chatelais M, Floris I. Active Substances from the Micro-Immunotherapy Medicine 2LC1 ® Show In Vitro Anti-Cancer Properties in Colon, Prostate, and Breast Cancer Models and Immune-Enhancing Capabilities in Human Macrophages. Int J Mol Sci 2025; 26:4300. [PMID: 40362536 PMCID: PMC12072473 DOI: 10.3390/ijms26094300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in cancer regulation by influencing tumor growth, metastasis, and the immune microenvironment. By providing low doses and ultra-low doses (ULD) of immune regulators to the organism, micro-immunotherapy (MI) medicines (MIM) could be seen as valuable adjuvant drugs in the context of a wide range of pathological conditions, including cancers. Thus, these MIM could target TAMs, affecting their phenotype and activities. In this study, the anti-tumor and the immune-stimulatory effects of four capsules out of the ten composing the Labo'life's MIM 2LC1® (2LC1-1, 2LC1-6, 2LC1-7, and 2LC1-8), as well as the specific nucleic acid (SNA®) sequence SNA-MYC present at ULD in this medicine have been evaluated in vitro, in several cancer models, and in human monocyte-derived macrophages. Our results showed that the tested MI formulations increased the tumor cell death of spheroids from HCT-116 colon cancer cells, while reducing the spheroid volume. Moreover, the treatments impaired the clonogenic capabilities of two cancer cell lines from epithelial origin, the LNCaP prostate cancer and the MCF-7 breast cancer cells. Interestingly, ULD of the SNA-MYC shared similar anti-cancer capabilities in those models, and it led to a significant reduction in the expression of C-MYC when evaluated in a model of human M2 macrophages. In the same model, the MI formulations also increased the expression of CD86 and HLA-DR, two markers of M1 anti-tumor macrophages. In addition, the tested items modulated the secretion of a panel of chemokines related to macrophage activity and immune cell recruitment. Finally, our results showed that 2LC1-8 increased the phagocytosis capabilities of human monocyte-derived macrophages, thus possibly contributing to sustaining the immune functions of M1, which are crucial in the context of cancer. Even if more research is needed to uncover their exact mechanism of action, these results suggest that the tested capsules of 2LC1 as well as ULD of SNA-MYC display both anti-tumor and immune-enhancing effects.
Collapse
Affiliation(s)
- Camille Jacques
- Preclinical Research Department, Labo’life France, Pescalis-Les Magnys, 79320 Moncoutant-sur-Sevre, France;
| | - Irene Marchesi
- Kitos Biotech s.r.l.s., Porto Conte Ricerche, S.P. 55 Porto Conte-Capo Caccia, Km 8,400 Loc. Tramariglio, 07041 Alghero, Italy; (I.M.); (F.P.F.)
| | - Francesco Paolo Fiorentino
- Kitos Biotech s.r.l.s., Porto Conte Ricerche, S.P. 55 Porto Conte-Capo Caccia, Km 8,400 Loc. Tramariglio, 07041 Alghero, Italy; (I.M.); (F.P.F.)
| | - Flora Marchand
- ProfileHIT, 7 Rue du Buisson, 44680 Sainte-Pazanne, France; (F.M.); (M.C.)
| | - Mathias Chatelais
- ProfileHIT, 7 Rue du Buisson, 44680 Sainte-Pazanne, France; (F.M.); (M.C.)
| | - Ilaria Floris
- Preclinical Research Department, Labo’life France, Pescalis-Les Magnys, 79320 Moncoutant-sur-Sevre, France;
| |
Collapse
|
2
|
Rausch L, Kallies A. Molecular Mechanisms Governing CD8 T Cell Differentiation and Checkpoint Inhibitor Response in Cancer. Annu Rev Immunol 2025; 43:515-543. [PMID: 40279308 DOI: 10.1146/annurev-immunol-082223-044122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
CD8 T cells play a critical role in antitumor immunity. However, over time, they often become dysfunctional or exhausted and ultimately fail to control tumor growth. To effectively harness CD8 T cells for cancer immunotherapy, a detailed understanding of the mechanisms that govern their differentiation and function is crucial. This review summarizes our current knowledge of the molecular pathways that regulate CD8 T cell heterogeneity and function in chronic infection and cancer and outlines how T cells respond to therapeutic checkpoint blockade. We explore how T cell-intrinsic and -extrinsic factors influence CD8 T cell differentiation, fate choices, and functional states and ultimately dictate their response to therapy. Identifying cells that orchestrate long-term antitumor immunity and understanding the mechanisms that govern their development and persistence are critical steps toward improving cancer immunotherapy.
Collapse
Affiliation(s)
- Lisa Rausch
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| | - Axel Kallies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| |
Collapse
|
3
|
Zhang X, Lou Y, Zheng S, Chang X. HCC-derived CX3CL1 affects hepatocellular carcinoma prognosis and CX3CR1 + MDSC infiltration. Eur J Med Res 2025; 30:153. [PMID: 40051011 PMCID: PMC11884201 DOI: 10.1186/s40001-025-02410-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/26/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality worldwide, largely because of its ability to reshape the tumor microenvironment and evade immune surveillance. METHODS CX3CL1 expression in HCC tumor tissues was assessed via immunohistochemistry, while plasma levels were quantified using enzyme-linked immunosorbent assays (ELISAs). CX3CR1-positive immune cell infiltration was analyzed by immunofluorescence. The associations among CX3CL1 expression, CX3CR1-positive cell infiltration, and patient prognosis were examined. Additionally, cell-based assays were conducted to evaluate the impact of CX3CL1 amplification on the expression of myeloid-derived suppressor cell (MDSC)-recruiting factors. RESULTS Elevated CX3CL1 levels were significantly correlated with increased MDSC infiltration in the tumor microenvironment and poorer patient prognosis. CX3CL1 amplification led to the upregulation of MDSC-recruiting factors, suggesting a potential mechanism for immune evasion. CONCLUSIONS These findings highlight the possible involvement of CX3CL1 in HCC progression via MDSC recruitment, suggesting that it is a promising therapeutic target for promoting antitumor immunity. Further studies are needed to confirm these findings and explore potential therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Medical Oncology, Hangzhou First People's Hospital, Hangzhou, 310006, China
| | - Yidan Lou
- Department of Medical Oncology, Hangzhou First People's Hospital, Hangzhou, 310006, China
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Song Zheng
- Department of Medical Oncology, Hangzhou First People's Hospital, Hangzhou, 310006, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China.
| | - Xu Chang
- Department of Interventional Therapy II, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
4
|
Wu X, Han X, Zhu H, Li M, Gong L, Jing S, Xie W, Liu Z, Li C, Zhang Y. Single-cell transcriptomics identify a novel macrophage population associated with bone invasion in pituitary neuroendocrine tumors. J Exp Clin Cancer Res 2025; 44:27. [PMID: 39865310 PMCID: PMC11770939 DOI: 10.1186/s13046-025-03296-9] [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: 11/13/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Bone-invasive Pituitary Neuroendocrine Tumors (BI PitNETs) epitomize an aggressive subtype of pituitary tumors characterized by bone invasion, culminating in extensive skull base bone destruction and fragmentation. This infiltration poses a significant surgical risk due to potential damage to vital nerves and arteries. However, the mechanisms underlying bone invasion caused by PitNETs remain elusive, and effective interventions for PitNET-induced bone invasion are lacking in clinical practice. METHODS In this study, we performed single-cell (n = 87,287) RNA sequencing on 10 cases of bone-invasive PitNETs and 5 cases of non-bone-invasion PitNETs (Non-BI PitNETs). We identified various cell types and determined their interactions through cell-cell communication analysis, which was further validated experimentally. RESULTS We identified a novel TNF-α+ TAM macrophage subset. BI PitNETs showed increased IL-34 secretion, impacting TNF-α+ TAMs via the IL34/CSF1R axis, leading to TNF-α production. TNF-α+ TAMs, in turn, communicate with CD14+ monocytes to promote their differentiation into osteoclasts and leading to bone invasion. In addition, we defined a gene signature for TNF-α+ TAM to guide the clinical prognosis prediction of BI PitNETs. CONCLUSIONS Our study elucidates the tumor microenvironment changes in bone invasion and identifies the critical role of TNF-α+ TAMs in promoting bone invasion of PitNETs, laying a foundation for developing new molecular markers or therapeutic agents targeting BI PitNETs.
Collapse
Affiliation(s)
- Xinzhi Wu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China
| | - Xueshuai Han
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, 100101, China
| | - Haibo Zhu
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China
| | - Mingxuan Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China
| | - Lei Gong
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China
| | - Sicheng Jing
- Department of Biology, University of California San Diego, San Diego, CA, 92122, USA
| | - Weiyan Xie
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
| | - Zhaoqi Liu
- Beijing Institute of Genomics, China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100070, China.
- Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
5
|
Kajihara R, Long MD, Hoki T, Chen H, Yamauchi T, Kanemaru H, Segal BH, Dy GK, Ito F. Divergent transcriptional states and kinetics of circulating tumor-infiltrating lymphocyte repertoires with highly homologous T-cell receptor sequences in a patient during immunotherapy. J Immunother Cancer 2025; 13:e010092. [PMID: 39863301 PMCID: PMC11784231 DOI: 10.1136/jitc-2024-010092] [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: 07/17/2024] [Accepted: 12/12/2024] [Indexed: 01/27/2025] Open
Abstract
Evidence has shown that T-cell receptors (TCRs) that recognize the same epitopes may not be the exact TCR clonotypes but have slightly different TCR sequences. However, the changes in the genomic and transcriptomic signatures of these highly homologous T cells during immunotherapy remain unknown. Here, we examined the evolutionary features in circulating TCR clonotypes observed in tumors (tumor-infiltrating lymphocyte (TIL)-TCRs) by combining single-cell RNA/TCR sequencing of longitudinal blood samples and TCR sequencing of tumor tissue from a patient treated with anti-cytotoxic T-lymphocyte-associated protein 4/programmed cell death protein-1 therapy. We found frequent circulating CD8+ TIL-TCRs with identical complementarity determining region 3 (CDR3)α amino acid sequences but quasi-identical CDR3β and TCR α/β (TRA/TRB) sequences. Despite their sequence similarities, these highly homologous TIL-TCRs responded differently to immunotherapy, and exhibited distinct transcriptional signatures that were uniquely distinguished by the expression of GZMK Overall, the expression of IFNG in CD8+ T-cell subsets including highly homologous TIL-TCRs increased when the patient achieved a response, but gradually decreased as the patient developed acquired resistance. Our findings provide insight into the cross-talk between T cells in the tumor microenvironment and those in the blood, and highlight that CD8+ T cells with highly homologous TCR sequences might display divergent transcriptional states and kinetics in response to immunotherapy.
Collapse
Affiliation(s)
- Ryutaro Kajihara
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Hematology and Immunology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - Toshifumi Hoki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Merck Sharp and Dohme Corp, Tokyo, Japan
| | - Hongbin Chen
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Takayoshi Yamauchi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Surgery, University of Southern California, Los Angeles, California, USA
| | - Hisashi Kanemaru
- Department of Surgery, University of Southern California, Los Angeles, California, USA
| | - Brahm H Segal
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Grace K Dy
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Fumito Ito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Surgery, University of Southern California, Los Angeles, California, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Hamouda AEI, Filtjens J, Brabants E, Kancheva D, Debraekeleer A, Brughmans J, Jacobs L, Bardet PMR, Knetemann E, Lefesvre P, Allonsius L, Gontsarik M, Varela I, Crabbé M, Clappaert EJ, Cappellesso F, Caro AA, Gordún Peiró A, Fredericq L, Hadadi E, Estapé Senti M, Schiffelers R, van Grunsven LA, Aboubakar Nana F, De Geest BG, Deschoemaeker S, De Koker S, Lambolez F, Laoui D. Intratumoral delivery of lipid nanoparticle-formulated mRNA encoding IL-21, IL-7, and 4-1BBL induces systemic anti-tumor immunity. Nat Commun 2024; 15:10635. [PMID: 39639025 PMCID: PMC11621563 DOI: 10.1038/s41467-024-54877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Local delivery of mRNA-based immunotherapy offers a promising avenue as it enables the production of specific immunomodulatory proteins that can stimulate the immune system to recognize and eliminate cancer cells while limiting systemic exposure and toxicities. Here, we develop and employ lipid-based nanoparticles (LNPs) to intratumorally deliver an mRNA mixture encoding the cytokines interleukin (IL)-21 and IL-7 and the immunostimulatory molecule 4-1BB ligand (Triplet LNP). IL-21 synergy with IL-7 and 4-1BBL leads to a profound increase in the frequency of tumor-infiltrating CD8+ T cells and their capacity to produce granzyme B and IFN-γ, leading to tumor eradication and the development of long-term immunological memory. Mechanistically, the efficacy of the Triplet LNP depends on tumor-draining lymph nodes to tumor CD8+ T-cell trafficking. Moreover, we highlight the therapeutic potential of the Triplet LNP in multiple tumor models in female mice and its superior therapeutic efficacy to immune checkpoint blockade. Ultimately, the expression of these immunomodulators is associated with better overall survival in patients with cancer.
Collapse
Affiliation(s)
- Ahmed E I Hamouda
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | | | - Daliya Kancheva
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Ayla Debraekeleer
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jan Brughmans
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Pauline M R Bardet
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Elisabeth Knetemann
- Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Pierre Lefesvre
- Department of Anatomo-Pathology, Universitair Ziekenhuis Brussel (UZB), Brussels, Belgium
| | - Lize Allonsius
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Mark Gontsarik
- Department of Pharmaceutics, University of Ghent, Ghent, Belgium
| | | | | | - Emile J Clappaert
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Federica Cappellesso
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Aarushi A Caro
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Alícia Gordún Peiró
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Tumor Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Luna Fredericq
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Eva Hadadi
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | | | | | - Leo A van Grunsven
- Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Frank Aboubakar Nana
- Institut de Duve, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, University of Ghent, Ghent, Belgium
| | - Sofie Deschoemaeker
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | | | - Damya Laoui
- Lab of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium.
- Lab of Cellular and Molecular Immunology, Brussels Center for Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.
| |
Collapse
|
7
|
Abdalla AM, Miao Y, Ming N, Ouyang C. ADAM10 modulates the efficacy of T-cell-mediated therapy in solid tumors. Immunol Cell Biol 2024; 102:907-923. [PMID: 39417304 DOI: 10.1111/imcb.12826] [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: 04/23/2024] [Revised: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
T-cell-mediated therapeutic strategies are the most potent effectors of cancer immunotherapy. However, an essential barrier to this therapy in solid tumors is disrupting the anti-cancer immune response, cancer-immunity cycle, T-cell priming, trafficking and T-cell cytotoxic capacity. Thus, reinforcing the anti-cancer immune response is needed to improve the effectiveness of T-cell-mediated therapy. Tumor-associated protease ADAM10, endothelial cells (ECs) and cytotoxic CD8+ T cells engage in complex communication via adhesion, transmigration and chemotactic mechanisms to facilitate an anti-cancer immune response. The precise impact of ADAM10 on the intricate mechanisms underlying these interactions remains unclear. This paper broadly explores how ADAM10, through different routes, influences the efficacy of T-cell-mediated therapy. ADAM10 cleaves CD8+ T-cell-targeting genes and impacts their expression and specificity. In addition, ADAM10 mediates the interactions of adhesion molecules with T cells and influences CD8+ T-cell activity and trafficking. Thus, understanding the role of ADAM10 in these events may lead to innovative strategies for advancing T-cell-mediated therapies.
Collapse
Affiliation(s)
- Ahmed Me Abdalla
- School of Biological Sciences and Technology, University of Jinan, Jinan, China
- Department of Biochemistry, College of Applied Science, University of Bahri, Khartoum, Sudan
| | - Yu Miao
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, Gansu, China
- Department of Phase 1 Clinical and Research Ward, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Ning Ming
- School of Biological Sciences and Technology, University of Jinan, Jinan, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Lv P, Wang Z, Si X, Su J, Yu Z, Yu H, Ji G, Song W. Biopolymer immune implants co-loaded with TMZ, R848 and IOX1 for perioperative therapy of glioblastoma. Acta Biomater 2024; 189:143-154. [PMID: 39368721 DOI: 10.1016/j.actbio.2024.09.053] [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: 05/13/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma (GBM), a prevalent and aggressive brain tumor, poses significant treatment challenges due to its rapid progression and the difficulty in achieving complete surgical resection. The current treatment regime, primarily surgery followed by radiotherapy and chemotherapy, offers limited success, with a five-year survival rate of less than 10 %. For addressing the challenges faced in the treatment of GBM, an approach using a biopolymer implant constructed with dynamic reversible covalent bonds, was designed to achieve controlled and constant-rate release of chemotherapy drug (Temozolomide, TMZ), immune adjuvant (Resiquimod, R848) and checkpoint inhibitor (5-carboxy-8-hydroxyquinoline, IOX1). The safety evaluation demonstrated the biocompatibility of the implants, with no significant inflammatory response or adverse effects on various systemic organs. In vivo antitumor study showed that the local delivery of drug combination via this implant significantly inhibited tumor recurrence of orthotopic GBM. Immune analysis revealed that the combination of the three drugs effectively activated systemic antitumor immune responses and induced memory effects. The synergistic mechanism of the drug combination was further validated by RNA whole sequencing. The innovative approach of combining chemotherapy and immunotherapy in biopolymer immune implants for GBM treatment showed promising and opens new avenues for treating GBM, particularly in addressing postoperative recurrence. STATEMENT OF SIGNIFICANCE: Our research introduces a pioneering approach in treating orthotopic brain glioblastoma (GBM), characterized by inevitable tumor recurrence, poor immune infiltration and the restrictive nature of the blood-brain barrier. To break the impasse of ineffective treatment for GBM, the innovative use of dynamically reversible covalent bonds in polymer matrix ensures the controlled, stable and sustained release of drug combinations of the chemotherapeutic agent temozolomide, immune adjuvants and checkpoint inhibitors, which maintains the optimal concentration in the tumor, overcoming problems associated with conventional chemotherapy such as systemic toxicity and low tumor targeting. Empirical evidence from in vivo experiments on the rat GBM model demonstrates significant outcomes: 90 % tumor size reduction and prolonged survival with over 70 % tumor cure rate.
Collapse
Affiliation(s)
- Pinxin Lv
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Zhanfeng Wang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| | - Jing Su
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China
| | - Zhifei Yu
- The Second Norman Bethune College of Clinical Medicine, Jilin University, Changchun 130021, China
| | - Hongquan Yu
- Department of Oncological Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Guofeng Ji
- Xuanwu Hospital, Capital Medical University, Beijing 100010, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China.
| |
Collapse
|
9
|
Morgan DM, Horton BL, Bhandarkar V, Van R, Dinter T, Zagorulya M, Love JC, Spranger S. Expansion of tumor-reactive CD8 + T cell clonotypes occurs in the spleen in response to immune checkpoint blockade. Sci Immunol 2024; 9:eadi3487. [PMID: 39270006 PMCID: PMC11580689 DOI: 10.1126/sciimmunol.adi3487] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/11/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024]
Abstract
Immune checkpoint blockade (ICB) enhances T cell responses against cancer, leading to long-term survival in a fraction of patients. CD8+ T cell differentiation in response to chronic antigen stimulation is highly complex, and it remains unclear precisely which T cell differentiation states at which anatomic sites are critical for the response to ICB. We identified an intermediate-exhausted population in the white pulp of the spleen that underwent substantial expansion in response to ICB and gave rise to tumor-infiltrating clonotypes. Increased systemic antigen redirected differentiation of this population toward a more circulatory exhausted KLR state, whereas a lack of cross-presented tumor antigen reduced its differentiation in the spleen. An analogous population of exhausted KLR CD8+ T cells in human blood samples exhibited diminished tumor-trafficking ability. Collectively, our data demonstrate the critical role of antigen density within the spleen for the differentiation and expansion of T cell clonotypes in response to ICB.
Collapse
Affiliation(s)
| | | | | | - Richard Van
- Koch Institute at MIT
- Department of Biology, MIT
| | | | | | | | | |
Collapse
|
10
|
Rodriguez C, Chocarro L, Echaide M, Ausin K, Escors D, Kochan G. Fractalkine in Health and Disease. Int J Mol Sci 2024; 25:8007. [PMID: 39125578 PMCID: PMC11311528 DOI: 10.3390/ijms25158007] [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: 06/18/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
CX3CL1 is one of the 50 up-to-date identified and characterized chemokines. While other chemokines are produced as small, secreted proteins, CX3CL1 (fractalkine) is synthetized as a transmembrane protein which also leads to a soluble form produced as a result of proteolytic cleavage. The membrane-bound protein and the soluble forms exhibit different biological functions. While the role of the fractalkine/CX3CR1 signaling axis was described in the nervous system and was also related to the migration of leukocytes to sites of inflammation, its actions are controversial in cancer progression and anti-tumor immunity. In the present review, we first describe the known biology of fractalkine concerning its action through its cognate receptor, but also its role in the activation of different integrins. The second part of this review is dedicated to its role in cancer where we discuss its role in anti-cancer or procarcinogenic activities.
Collapse
Grants
- FIS PI23/00196 Instituto de Salud Carlos III-FEDER
- FIS PI20/00010 Instituto de Salud Carlos III-FEDER
- BMED 036-2023 Departamento de Salud del Gobierno de Navarra-FEDER, Spain
- LINTERNA, Ref. 0011-1411-2020-000033 Departamento de Industria, Gobierno de Navarra, Spain
- ARNMUNE, 0011-1411-2023-000111 Departamento de Industria, Gobierno de Navarra, Spain
- ISOLDA project, under grant agreement ID: 848166. Horizon 2020, European Union
- PFIS, FI21/00080 Instituto de Salud Carlos III-FEDER
Collapse
Affiliation(s)
| | | | | | | | - David Escors
- Oncoimmunology Unit, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), 31008 Pamplona, Spain; (C.R.); (L.C.); (M.E.); (K.A.)
| | - Grazyna Kochan
- Oncoimmunology Unit, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), 31008 Pamplona, Spain; (C.R.); (L.C.); (M.E.); (K.A.)
| |
Collapse
|
11
|
Ishigaki H, Yamauchi T, Long MD, Hoki T, Yamamoto Y, Oba T, Ito F. Generation, Transcriptomic States, and Clinical Relevance of CX3CR1+ CD8 T Cells in Melanoma. CANCER RESEARCH COMMUNICATIONS 2024; 4:1802-1814. [PMID: 38881188 PMCID: PMC11267618 DOI: 10.1158/2767-9764.crc-24-0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
Recent progress in single-cell profiling technologies has revealed significant phenotypic and transcriptional heterogeneity in tumor-infiltrating CD8+ T cells. However, the transition between the different states of intratumoral antigen-specific CD8+ T cells remains elusive. Here, we sought to examine the generation, transcriptomic states, and the clinical relevance of melanoma-infiltrating CD8+ T cells expressing a chemokine receptor and T-cell differentiation marker, CX3C chemokine receptor 1 (CX3CR1). Analysis of single-cell datasets revealed distinct human melanoma-infiltrating CD8+ T-cell clusters expressing genes associated with effector T-cell function but with distinguishing expression of CX3CR1 or PDCD1. No obvious impact of CX3CR1 expression in melanoma on the response to immune checkpoint inhibitor therapy was observed while increased pretreatment and on-treatment frequency of a CD8+ T-cell cluster expressing high levels of exhaustion markers was associated with poor response to the treatment. Adoptively transferred antigen-specific CX3CR1- CD8+ T cells differentiated into the CX3CR1+ subset in mice treated with FTY720, which inhibits lymphocyte egress from secondary lymphoid tissues, suggesting the intratumoral generation of CX3CR1+ CD8+ T cells rather than their trafficking from secondary lymphoid organs. Furthermore, analysis of adoptively transferred antigen-specific CD8+ T cells, in which the Cx3cr1 gene was replaced with a marker gene confirmed that CX3CR1+ CD8+ T cells could directly differentiate from the intratumoral CX3CR1- subset. These findings highlight that tumor antigen-specific CX3CR1- CD8+ T cells can fully differentiate outside the secondary lymphoid organs and generate CX3CR1+ CD8+ T cells in the tumor microenvironment, which are distinct from CD8+ T cells that express markers of exhaustion. SIGNIFICANCE Intratumoral T cells are composed of heterogeneous subpopulations with various phenotypic and transcriptional states. This study illustrates the intratumoral generation of antigen-specific CX3CR1+ CD8+ T cells that exhibit distinct transcriptomic signatures and clinical relevance from CD8+ T cells expressing markers of exhaustion.
Collapse
Affiliation(s)
- Hirohito Ishigaki
- Department of Surgery, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, California.
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan.
| | - Takayoshi Yamauchi
- Department of Surgery, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, California.
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| | - Toshifumi Hoki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
- Oncology Science Unit, MSD Japan, Tokyo, Japan.
| | - Yuta Yamamoto
- Department of Surgery, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, California.
- Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
- Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Fumito Ito
- Department of Surgery, University of Southern California, Norris Comprehensive Cancer Center, Los Angeles, California.
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| |
Collapse
|
12
|
Benmelech S, Le T, McKay M, Nam J, Subramaniam K, Tellez D, Vlasak G, Mak M. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions. APL Bioeng 2024; 8:021502. [PMID: 38572312 PMCID: PMC10990568 DOI: 10.1063/5.0195244] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The tumor microenvironment (TME), composed of and influenced by a heterogeneous set of cancer cells and an extracellular matrix, plays a crucial role in cancer progression. The biophysical aspects of the TME (namely, its architecture and mechanics) regulate interactions and spatial distributions of cancer cells and immune cells. In this review, we discuss the factors of the TME-notably, the extracellular matrix, as well as tumor and stromal cells-that contribute to a pro-tumor, immunosuppressive response. We then discuss the ways in which cells of the innate and adaptive immune systems respond to tumors from both biochemical and biophysical perspectives, with increased focus on CD8+ and CD4+ T cells. Building upon this information, we turn to immune-based antitumor interventions-specifically, recent biophysical breakthroughs aimed at improving CAR-T cell therapy.
Collapse
Affiliation(s)
- Shoham Benmelech
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Thien Le
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Maggie McKay
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Jungmin Nam
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Krupakar Subramaniam
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA
| | - Daniela Tellez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Grace Vlasak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
13
|
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells 2024; 13:844. [PMID: 38786066 PMCID: PMC11120175 DOI: 10.3390/cells13100844] [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: 03/23/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell's ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
Collapse
Affiliation(s)
- Allison T. Ryan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
14
|
Gargett T, Truong NTH, Gardam B, Yu W, Ebert LM, Johnson A, Yeo ECF, Wittwer NL, Tapia Rico G, Logan J, Sivaloganathan P, Collis M, Ruszkiewicz A, Brown MP. Safety and biological outcomes following a phase 1 trial of GD2-specific CAR-T cells in patients with GD2-positive metastatic melanoma and other solid cancers. J Immunother Cancer 2024; 12:e008659. [PMID: 38754916 PMCID: PMC11097842 DOI: 10.1136/jitc-2023-008659] [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: 04/28/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapies specific for the CD19 and B-cell maturation antigen have become an approved standard of care worldwide for relapsed and refractory B-cell malignancies. If CAR-T cell therapy for non-hematological malignancies is to achieve the same stage of clinical development, then iterative early-phase clinical testing can add value to the clinical development process for evaluating CAR-T cell products containing different CAR designs and manufactured under differing conditions. METHODS We conducted a phase 1 trial of third-generation GD2-specific CAR-T cell therapy, which has previously been tested in neuroblastoma patients. In this study, the GD2-CAR-T therapy was evaluated for the first time in metastatic melanoma patients in combination with BRAF/MEK inhibitor therapy, and as a monotherapy in patients with colorectal cancer and a patient with fibromyxoid sarcoma. Feasibility and safety were determined and persistence studies, multiplex cytokine arrays on sera and detailed immune phenotyping of the original CAR-T products, the circulating CAR-T cells, and, in select patients, the tumor-infiltrating CAR-T cells were performed. RESULTS We demonstrate the feasibility of manufacturing CAR-T products at point of care for patients with solid cancer and show that a single intravenous infusion was well tolerated with no dose-limiting toxicities or severe adverse events. In addition, we note significant improvements in CAR-T cell immune phenotype, and expansion when a modified manufacturing procedure was adopted for the latter 6 patients recruited to this 12-patient trial. We also show evidence of CAR-T cell-mediated immune activity and in some patients expanded subsets of circulating myeloid cells after CAR-T cell therapy. CONCLUSIONS This is the first report of third-generation GD2-targeting CAR-T cells in patients with metastatic melanoma and other solid cancers such as colorectal cancer, showing feasibility, safety and immune activity, but limited clinical effect. TRIAL REGISTRATION NUMBER ACTRN12613000198729.
Collapse
Affiliation(s)
- Tessa Gargett
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Nga T H Truong
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Bryan Gardam
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Wenbo Yu
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Lisa M Ebert
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Amy Johnson
- Flinders University, Adelaide, South Australia, Australia
| | - Erica C F Yeo
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
| | - Nicole L Wittwer
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Gonzalo Tapia Rico
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Jesikah Logan
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Purany Sivaloganathan
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Maria Collis
- Surgical Pathology, SA Pathology, Adelaide, South Australia, Australia
| | - Andrew Ruszkiewicz
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Surgical Pathology, SA Pathology, Adelaide, South Australia, Australia
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, South Australia, Australia
| | - Michael P Brown
- University of South Australia, Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology, Rundle Mall, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
15
|
Natsuki S, Yoshii M, Tanaka H, Mori T, Deguchi S, Miki Y, Tamura T, Toyokawa T, Lee S, Maeda K. Involvement of CX3CR1 + cells appearing in the abdominal cavity in the immunosuppressive environment immediately after gastric cancer surgery. World J Surg Oncol 2024; 22:74. [PMID: 38433196 PMCID: PMC10910822 DOI: 10.1186/s12957-024-03353-1] [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: 11/29/2023] [Accepted: 02/24/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Gastric cancer is primarily treated by surgery; however, little is known about the changes in the intraperitoneal immune environment and the prognostic impact of surgery. Surgical stress and cancer-associated inflammation cause immune cells to mobilize into the abdominal cavity via numerous cytokines. One such cytokine, CX3CR1, has various immune-related functions that remain to be fully explained. We characterized the intraperitoneal immune environment by investigating CX3CR1+ cells in intraperitoneal lavage fluid during gastric cancer surgery. METHODS Lavage fluid samples were obtained from a total of 41 patients who underwent gastrectomy. The relative expression of various genes was analyzed using quantitative real-time PCR. The association of each gene expression with clinicopathological features and surgical outcomes was examined. The fraction of CX3CR1+ cells was analyzed by flow cytometry. Cytokine profiles in lavage fluid samples were investigated using a cytometric beads array. RESULTS CX3CR1high patients exhibited higher levels of perioperative inflammation in blood tests and more recurrences than CX3CR1low patients. CX3CR1high patients tended to exhibit higher pathological T and N stage than CX3CR1low patients. CX3CR1 was primarily expressed on myeloid-derived suppressor cells and tumor-associated macrophages. In particular, polymorphonuclear myeloid-derived suppressor cells were associated with perioperative inflammation, pathological N, and recurrences. These immunosuppressive cells were associated with a trend toward unfavorable prognosis. Moreover, CX3CR1 expression was correlated with programmed death-1 expression. CONCLUSIONS Our results suggest that CX3CR1+ cells are associated with an acute inflammatory response, tumor-promotion, and recurrence. CX3CR1 expression could be taken advantage of as a beneficial therapeutic target for improving immunosuppressive state in the future. In addition, analysis of intra-abdominal CX3CR1+ cells could be useful for characterizing the immune environment after gastric cancer surgery.
Collapse
Affiliation(s)
- Seiji Natsuki
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Mami Yoshii
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan.
| | | | - Takuya Mori
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Sota Deguchi
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Yuichiro Miki
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Tatsuro Tamura
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Takahiro Toyokawa
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Shigeru Lee
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| | - Kiyoshi Maeda
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Asahi-machi 1-4-3, Abeno-ku, Osaka, Japan
| |
Collapse
|
16
|
Yi H, Qin L, Ye X, Song J, Ji J, Ye T, Li J, Li L. Progression of radio-labeled molecular imaging probes targeting chemokine receptors. Crit Rev Oncol Hematol 2024; 195:104266. [PMID: 38232861 DOI: 10.1016/j.critrevonc.2024.104266] [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: 03/13/2023] [Revised: 12/31/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
Chemokine receptors are significantly expressed in the surface of most inflammatory cells and tumor cells. Guided by chemokines, inflammatory cells which express the relevant chemokine receptors migrate to inflammatory lesions and participate in the evolution of inflammation diseases. Similarly, driven by chemokines, immune cells infiltrate into tumor lesions not only induces alterations in the tumor microenvironment, disrupting the efficacy of tumor therapies, but also has the potential to selectively target tumoral cells and diminish tumor progression. Chemokine receptors, which are significantly expressed on the surface of tumor cell membranes, are regulated by chemokines and initiate tumor-associated signaling pathways within tumor cells, playing a complex role in tumor progression. Based on the antagonists targeting chemokine receptors, radionuclide-labeled molecular imaging probes have been developed for the emerging application of molecular imaging in diseases such as tumors and inflammation. The value and limitations of molecular probes in disease imaging are worth reviewing.
Collapse
Affiliation(s)
- Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
| | - Lilin Qin
- Second Clinical Medical College of Zhejiang Chinese Medical University, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Xuemei Ye
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Jinling Song
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Jianfeng Ji
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Ting Ye
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Juan Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Dongfang Street 150, Hangzhou, Zhejiang 310022, China.
| | - Linfa Li
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
17
|
Loredan DG, Devlin JC, Khanna KM, Loke P. Recruitment and Maintenance of CX3CR1+CD4+ T Cells during Helminth Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:632-644. [PMID: 38180236 PMCID: PMC10954162 DOI: 10.4049/jimmunol.2300451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024]
Abstract
Distinct subsets of T lymphocytes express CX3CR1 under inflammatory conditions, but little is known about CX3CR1+CD4+ T cells during type 2 inflammation in helminth infections. In this study, we used a fate-mapping mouse model to characterize CX3CR1+CD4+ T cells during both acute Nippostrongylus brasiliensis and chronic Schistosoma mansoni murine models of helminth infections, revealing CX3CR1+CD4+ T cells to be an activated tissue-homing subset with varying capacity for cytokine production. Tracking these cells over time revealed that maintenance of CX3CR1 itself along with a TH2 phenotype conferred a survival advantage in the inflamed tissue. Single-cell RNA sequencing analysis of fate-mapped CX3CR1+CD4+ T cells from both the peripheral tissue and the spleen revealed a considerable level of diversity and identified a distinct population of BCL6+TCF-1+PD1+CD4+ T cells in the spleen during helminth infections. Conditional deletion of BCL6 in CX3CR1+ cells resulted in fewer CX3CR1+CD4+ T cells during infection, indicating a role in sustaining CD4+ T cell responses to helminth infections. Overall, our studies revealed the behavior and heterogeneity of CX3CR1+CD4+ T cells during type 2 inflammation in helminth infections and identified BCL6 to be important in their maintenance.
Collapse
Affiliation(s)
- Denis G. Loredan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Vilcek Institute of Graduate Biomedical Sciences, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Joseph C. Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Kamal M. Khanna
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Perlmutter Cancer Center, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - P’ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
18
|
Abstract
For our immune system to contain or eliminate malignant solid tumours, both myeloid and lymphoid haematopoietic cells must not only extravasate from the bloodstream into the tumour tissue but also further migrate to various specialized niches of the tumour microenvironment to functionally interact with each other, with non-haematopoietic stromal cells and, ultimately, with cancer cells. These interactions regulate local immune cell survival, proliferative expansion, differentiation and their execution of pro-tumour or antitumour effector functions, which collectively determine the outcome of spontaneous or therapeutically induced antitumour immune responses. None of these interactions occur randomly but are orchestrated and critically depend on migratory guidance cues provided by chemokines, a large family of chemotactic cytokines, and their receptors. Understanding the functional organization of the tumour immune microenvironment inevitably requires knowledge of the multifaceted roles of chemokines in the recruitment and positioning of its cellular constituents. Gaining such knowledge will not only generate new insights into the mechanisms underlying antitumour immunity or immune tolerance but also inform the development of biomarkers (or 'biopatterns') based on spatial tumour tissue analyses, as well as novel strategies to therapeutically engineer immune responses in patients with cancer. Here we will discuss recent observations on the role of chemokines in the tumour microenvironment in the context of our knowledge of their physiological functions in development, homeostasis and antimicrobial responses.
Collapse
Affiliation(s)
- Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Julia K Lill
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lukas M Altenburger
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Xu H, Jia Z, Liu F, Li J, Huang Y, Jiang Y, Pu P, Shang T, Tang P, Zhou Y, Yang Y, Su J, Liu J. Biomarkers and experimental models for cancer immunology investigation. MedComm (Beijing) 2023; 4:e437. [PMID: 38045830 PMCID: PMC10693314 DOI: 10.1002/mco2.437] [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: 05/30/2023] [Revised: 11/01/2023] [Accepted: 11/10/2023] [Indexed: 12/05/2023] Open
Abstract
The rapid advancement of tumor immunotherapies poses challenges for the tools used in cancer immunology research, highlighting the need for highly effective biomarkers and reproducible experimental models. Current immunotherapy biomarkers encompass surface protein markers such as PD-L1, genetic features such as microsatellite instability, tumor-infiltrating lymphocytes, and biomarkers in liquid biopsy such as circulating tumor DNAs. Experimental models, ranging from 3D in vitro cultures (spheroids, submerged models, air-liquid interface models, organ-on-a-chips) to advanced 3D bioprinting techniques, have emerged as valuable platforms for cancer immunology investigations and immunotherapy biomarker research. By preserving native immune components or coculturing with exogenous immune cells, these models replicate the tumor microenvironment in vitro. Animal models like syngeneic models, genetically engineered models, and patient-derived xenografts provide opportunities to study in vivo tumor-immune interactions. Humanized animal models further enable the simulation of the human-specific tumor microenvironment. Here, we provide a comprehensive overview of the advantages, limitations, and prospects of different biomarkers and experimental models, specifically focusing on the role of biomarkers in predicting immunotherapy outcomes and the ability of experimental models to replicate the tumor microenvironment. By integrating cutting-edge biomarkers and experimental models, this review serves as a valuable resource for accessing the forefront of cancer immunology investigation.
Collapse
Affiliation(s)
- Hengyi Xu
- State Key Laboratory of Molecular OncologyNational Cancer Center /National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Ziqi Jia
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Fengshuo Liu
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiayi Li
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yansong Huang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yiwen Jiang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pengming Pu
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tongxuan Shang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pengrui Tang
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yongxin Zhou
- Eight‐year MD ProgramSchool of Clinical Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yufan Yang
- School of MedicineTsinghua UniversityBeijingChina
| | - Jianzhong Su
- Oujiang LaboratoryZhejiang Lab for Regenerative Medicine, Vision, and Brain HealthWenzhouZhejiangChina
| | - Jiaqi Liu
- State Key Laboratory of Molecular OncologyNational Cancer Center /National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Breast Surgical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
20
|
Lee H, Joo J, Song J, Kim H, Kim YH, Park HR. Immunological link between periodontitis and type 2 diabetes deciphered by single-cell RNA analysis. Clin Transl Med 2023; 13:e1503. [PMID: 38082425 PMCID: PMC10713875 DOI: 10.1002/ctm2.1503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/19/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (DM) is a complex metabolic disorder that causes various complications, including periodontitis (PD). Although a bidirectional relationship has been reported between DM and PD, their immunological relationship remains poorly understood. Therefore, this study aimed to compare the immune response in patients with PD alone and in those with both PD and DM (PDDM) to expand our knowledge of the complicated connection between PD and DM. METHODS Peripheral blood mononuclear cells were collected from 11 healthy controls, 10 patients with PD without DM, and six patients with PDDM, followed by analysis using single-cell RNA sequencing. The differences among groups were then compared based on intracellular and intercellular perspectives. RESULTS Compared to the healthy state, classical monocytes exhibited the highest degree of transcriptional change, with elevated levels of pro-inflammatory cytokines in both PD and PDDM. DM diminished the effector function of CD8+ T and natural killer (NK) cells as well as completely modified the differentiation direction of these cells. Interestingly, a prominent pathway, RESISTIN, which is known to increase insulin resistance and susceptibility to diabetes, was found to be activated under both PD and PDDM conditions. In particular, CAP1+ classical monocytes from patients with PD and PDDM showed elevated nuclear factor kappa B-inducing kinase activity. CONCLUSIONS Overall, this study elucidates how the presence of DM contributes to the deterioration of T/NK cell immunity and the immunological basis connecting PD to DM.
Collapse
Affiliation(s)
- Hansong Lee
- Medical Research InstitutePusan National UniversityYangsanRepublic of Korea
| | - Ji‐Young Joo
- Department of PeriodontologySchool of Dentistry, Pusan National UniversityYangsanRepublic of Korea
| | - Jae‐Min Song
- Department of Oral and Maxillofacial SurgerySchool of Dentistry, Pusan National UniversityYangsanRepublic of Korea
| | - Hyun‐Joo Kim
- Department of PeriodontologyDental and Life Science Institute, School of Dentistry, Pusan National UniversityYangsanRepublic of Korea
- Department of Periodontology and Dental Research InstitutePusan National University Dental HospitalYangsanRepublic of Korea
- Periodontal Disease Signaling Network Research CenterSchool of Dentistry, Pusan National UniversityYangsanRepublic of Korea
| | - Yun Hak Kim
- Periodontal Disease Signaling Network Research CenterSchool of Dentistry, Pusan National UniversityYangsanRepublic of Korea
- Department of Biomedical Informatics, School of MedicinePusan National UniversityYangsanRepublic of Korea
- Department of AnatomySchool of Medicine, Pusan National UniversityYangsanRepublic of Korea
| | - Hae Ryoun Park
- Department of Periodontology and Dental Research InstitutePusan National University Dental HospitalYangsanRepublic of Korea
- Periodontal Disease Signaling Network Research CenterSchool of Dentistry, Pusan National UniversityYangsanRepublic of Korea
- Department of Oral PathologyDental and Life Science Institute, Pusan National UniversityYangsanRepublic of Korea
| |
Collapse
|
21
|
Ben-Mordechai T, Lawrence YR, Symon Z, Shimoni-Sebag A, Amit U. CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer. Cancers (Basel) 2023; 15:5472. [PMID: 38001732 PMCID: PMC10669975 DOI: 10.3390/cancers15225472] [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: 09/22/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Chemokine (C-X3-C Motif) Receptor 1 (CX3CR1) is present in a subset of the immune cells in the tumor microenvironment (TME) and plays an essential and diverse role in cancer progression. However, its potential function in the irradiated TME remains unknown. MATERIALS AND METHODS A mouse lung cancer model was performed by subcutaneously inoculating Lewis Lung Carcinoma (LLC) cells expressing luciferase (Luc-2) and mCherry cells in CX3CR1GFP/GFP, CX3CR1DTR/+, and wild-type (WT) mice. Bioluminescence imaging, clonogenic assay, and flow cytometry were used to assess tumor progression, proliferation, and cell composition after radiation. RESULTS Radiation provoked a significant influx of CX3CR1-expressing immune cells, notably monocytes and macrophages, into the TME. Co-culturing irradiated LLC cells with CX3CR1-deficient monocytes, and macrophages resulted in reduced clonogenic survival and increased apoptosis of the cancer cells. Interestingly, deficiency of CX3CR1 in macrophages led to a redistribution of the irradiated LLC cells in the S-phase, parallel to increased expression of cyclin E1, required for cell cycle G1/S transition. In addition, the deficiency of CX3CR1 expression in macrophages altered the cytokine secretion with a decrease in interleukin 6, a crucial mediator of cancer cell survival and proliferation. Next, LLC cells were injected subcutaneously into CX3CR1DTR/+ mice, sensitive to diphtheria toxin (DT), and WT mice. After injection, tumors were irradiated with 8 Gy, and mice were treated with DT, leading to conditional ablation of CX3CR1-expressing cells. After three weeks, CX3CR1-depleted mice displayed reduced tumor progression. Furthermore, combining the S-phase-specific chemotherapeutic gemcitabine with CX3CR1 cell ablation resulted in additional attenuation of tumor progression. CONCLUSIONS CX3CR1-expressing mononuclear cells invade the TME after radiation therapy in a mouse lung cancer model. CX3CR1 cell depletion attenuates tumor progression following radiation and sensitizes the tumor to S-phase-specific chemotherapy. Thus, we propose a novel strategy to improve radiation sensitivity by targeting the CX3CR1-expressing immune cells.
Collapse
Affiliation(s)
- Tamar Ben-Mordechai
- Radiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, Israel; (T.B.-M.); (Y.R.L.); (Z.S.); (A.S.-S.)
| | - Yaacov R. Lawrence
- Radiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, Israel; (T.B.-M.); (Y.R.L.); (Z.S.); (A.S.-S.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Zvi Symon
- Radiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, Israel; (T.B.-M.); (Y.R.L.); (Z.S.); (A.S.-S.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ariel Shimoni-Sebag
- Radiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, Israel; (T.B.-M.); (Y.R.L.); (Z.S.); (A.S.-S.)
| | - Uri Amit
- Radiation Oncology Department, Tel Aviv Medical Center, Tel Aviv 64239, Israel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, TRC 2 West Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
Li X, He L, Ou Y, Wang S, Hu Y, Niu H. Oxymatrine inhibits melanoma development by modulating the immune microenvironment and targeting the MYC/PD-L1 pathway. Int Immunopharmacol 2023; 124:111000. [PMID: 37788594 DOI: 10.1016/j.intimp.2023.111000] [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: 06/22/2023] [Revised: 09/13/2023] [Accepted: 09/24/2023] [Indexed: 10/05/2023]
Abstract
Oxymatrine, also known as ammothamnine or oxysophoridine, is a natural compound isolated from Sophora flavescens (in Chinese, Kushen), and many previous researchers have characterized its anti-inflammatory, anti-fibrotic and anti-tumor properties. However, the underlying anti-tumor immunological mechanism of oxymatrine remains elusive. In this study, we carried out experiments both in vitro and in vivo and investigated the anti-tumor effect of oxymatrine to inhibit the proliferation and migration of melanoma B16 cells, while promoting apoptosis. Oxymatrine upregulated CD4+ T, CD8+ T and NKT cells, downregulated Treg cells, promoted TNF-α secretion, and successfully modulated the immune microenvironment and ultimately suppressed melanoma development in subcutaneous tumor models established in mice. Evidence from network pharmacology and RNAseq suggested that possible targets of oxymatrine for melanoma treatment included PD-L1 and MYC. We observed oxymatrine inhibited PD-L1 and MYC expression in melanoma cells via qRT-PCR and western blotting analysis, and found MYC potentially regulated PD-L1 to mediate anti-tumor effects. These findings provide insight into the mechanism by which oxymatrine inhibits melanoma and enhances the anti-tumor immune effect. In summary, our study proposes a novel approach to suppress melanoma by targeting the MYC/PD-L1 pathway using oxymatrine, which may develop into a less toxic and more efficient anti-tumor agent for melanoma treatment.
Collapse
Affiliation(s)
- Xin Li
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China
| | - Lun He
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China
| | - Yanhua Ou
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China
| | - Shanshan Wang
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China; School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Yaqian Hu
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China; School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Haitao Niu
- School of Medicine, Jinan University, Guangzhou, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Ministry of Education, Guangzhou, China; Guangzhou Key Laboratory for Germ-Free Animals and Microbiome Application, Guangzhou, China; School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, China.
| |
Collapse
|
23
|
Li Q, Lin L, Shou P, Liu K, Xue Y, Hu M, Ling W, Huang Y, Du L, Zheng C, Wang X, Zheng F, Zhang T, Wang Y, Shao C, Melino G, Shi Y, Wang Y. MHC class Ib-restricted CD8 + T cells possess strong tumoricidal activities. Proc Natl Acad Sci U S A 2023; 120:e2304689120. [PMID: 37856544 PMCID: PMC10614629 DOI: 10.1073/pnas.2304689120] [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: 03/22/2023] [Accepted: 08/28/2023] [Indexed: 10/21/2023] Open
Abstract
The importance of classical CD8+ T cells in tumor eradication is well acknowledged. However, the anti-tumor activity of MHC (major histocompatibility complex) Ib-restricted CD8+ T (Ib-CD8+ T) cells remains obscure. Here, we show that CX3CR1-expressing Ib-CD8+ T cells (Ib-restricted CD8+ T cells) highly express cytotoxic factors, austerely resist exhaustion, and effectively eliminate various tumors. These Ib-CD8+ T cells can be primed by MHC Ia (MHC class Ia molecules) expressed on various cell types for optimal activation in a Tbet-dependent manner. Importantly, MHC Ia does not allogeneically activate Ib-CD8+ T cells, rather, sensitizes these cells for T cell receptor activation. Such effects were observed when MHC Ia+ cells were administered to tumor-bearing Kb-/-Db-/-mice. A similar population of tumoricidal CX3CR1+CD8+ T cells was identified in wild-type mice and melanoma patients. Adoptive transfer of Ib-CD8+ T cells to wild-type mice inhibited tumor progression without damaging normal tissues. Taken together, we demonstrate that MHC class Ia can prime Ib-CD8+ T cells for robust tumoricidal activities.
Collapse
Affiliation(s)
- Qing Li
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Liangyu Lin
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Peishun Shou
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Keli Liu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Yueqing Xue
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Mingyuan Hu
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Weifang Ling
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Yin Huang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Liming Du
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Chunxing Zheng
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Xuefeng Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Fanjun Zheng
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Tao Zhang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Yu Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| | - Changshun Shao
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu215123, China
| | - Gerry Melino
- Department of Experimental Medicine, Tor Vergata Oncoscience Research, University of Rome Tor Vergata, Rome00133, Italy
| | - Yufang Shi
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
- The Third Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, Jiangsu215123, China
| | - Ying Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
| |
Collapse
|
24
|
Lee EJ, Choi JG, Han JH, Kim YW, Lim J, Chung HS. Single-Cell RNA Sequencing Reveals Immuno-Oncology Characteristics of Tumor-Infiltrating T Lymphocytes in Photodynamic Therapy-Treated Colorectal Cancer Mouse Model. Int J Mol Sci 2023; 24:13913. [PMID: 37762216 PMCID: PMC10531263 DOI: 10.3390/ijms241813913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Photodynamic therapy (PDT) has shown promise in reducing metastatic colorectal cancer (CRC); however, the underlying mechanisms remain unclear. Modulating tumor-infiltrating immune cells by PDT may be achieved, which requires the characterization of immune cell populations in the tumor microenvironment by single-cell RNA sequencing (scRNA-seq). Here, we determined the effect of Chlorin e6 (Ce6)-mediated PDT on tumor-infiltrating T cells using scRNA-seq analysis. We used a humanized programmed death-1/programmed death ligand 1 (PD-1/PD-L1) MC38 cell allograft mouse model, considering its potential as an immunogenic cancer model and in combination with PD-1/PD-L1 immune checkpoint blockade. PDT treatment significantly reduced tumor growth in mice containing hPD-1/PD-L1 MC38 tumors. scRNA-seq analysis revealed that the PDT group had increased levels of CD8+ activated T cells and CD8+ cytotoxic T cells, but decreased levels of exhausted CD8+ T cells. PDT treatment also enhanced the infiltration of CD8+ T cells into tumors and increased the production of key effector molecules, including granzyme B and perforin 1. These findings provide insight into immune-therapeutic modulation for CRC patients and highlight the potential of PDT in overcoming immune evasion and enhancing antitumor immunity.
Collapse
Affiliation(s)
- Eun-Ji Lee
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
| | - Jang-Gi Choi
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
| | - Jung Ho Han
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
| | - Yong-Wan Kim
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu 41061, Republic of Korea; (Y.-W.K.); (J.L.)
| | - Junmo Lim
- Dongsung Cancer Center, Dongsung Biopharmaceutical, Daegu 41061, Republic of Korea; (Y.-W.K.); (J.L.)
| | - Hwan-Suck Chung
- Korean Medicine Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea; (E.-J.L.); (J.-G.C.); (J.H.H.)
- Korean Convergence Medical Science Major, University of Science and Technology (UST), KIOM Campus, Daegu 41062, Republic of Korea
| |
Collapse
|
25
|
Chen Y, Xu Z, Sun H, Ouyang X, Han Y, Yu H, Wu N, Xie Y, Su B. Regulation of CD8 + T memory and exhaustion by the mTOR signals. Cell Mol Immunol 2023; 20:1023-1039. [PMID: 37582972 PMCID: PMC10468538 DOI: 10.1038/s41423-023-01064-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/02/2023] [Indexed: 08/17/2023] Open
Abstract
CD8+ T cells are the key executioners of the adaptive immune arm, which mediates antitumor and antiviral immunity. Naïve CD8+ T cells develop in the thymus and are quickly activated in the periphery after encountering a cognate antigen, which induces these cells to proliferate and differentiate into effector cells that fight the initial infection. Simultaneously, a fraction of these cells become long-lived memory CD8+ T cells that combat future infections. Notably, the generation and maintenance of memory cells is profoundly affected by various in vivo conditions, such as the mode of primary activation (e.g., acute vs. chronic immunization) or fluctuations in host metabolic, inflammatory, or aging factors. Therefore, many T cells may be lost or become exhausted and no longer functional. Complicated intracellular signaling pathways, transcription factors, epigenetic modifications, and metabolic processes are involved in this process. Therefore, understanding the cellular and molecular basis for the generation and fate of memory and exhausted CD8+ cells is central for harnessing cellular immunity. In this review, we focus on mammalian target of rapamycin (mTOR), particularly signaling mediated by mTOR complex (mTORC) 2 in memory and exhausted CD8+ T cells at the molecular level.
Collapse
Affiliation(s)
- Yao Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ziyang Xu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinxing Ouyang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Tumor Biology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuheng Han
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haihui Yu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiting Xie
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and The Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Department of Tumor Biology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Key Laboratory of Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
26
|
Strati P, Li X, Deng Q, Marques-Piubelli ML, Henderson J, Watson G, Deaton L, Cain T, Yang H, Ravanmehr V, Fayad LE, Iyer SP, Nastoupil LJ, Hagemeister FB, Parra ER, Saini N, Takahashi K, Fowler NH, Westin JR, Steiner RE, Nair R, Flowers CR, Wang L, Ahmed S, Al-Atrash G, Vega F, Neelapu SS, Green MR. Prolonged cytopenia following CD19 CAR T cell therapy is linked with bone marrow infiltration of clonally expanded IFNγ-expressing CD8 T cells. Cell Rep Med 2023; 4:101158. [PMID: 37586321 PMCID: PMC10439270 DOI: 10.1016/j.xcrm.2023.101158] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 06/13/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Autologous anti-CD19 chimeric antigen receptor T cell (CAR T) therapy is highly effective in relapsed/refractory large B cell lymphoma (rrLBCL) but is associated with toxicities that delay recovery. While the biological mechanisms of cytokine release syndrome and neurotoxicity have been investigated, the pathophysiology is poorly understood for prolonged cytopenia, defined as grade ≥3 cytopenia lasting beyond 30 days after CAR T infusion. We performed single-cell RNA sequencing of bone marrow samples from healthy donors and rrLBCL patients with or without prolonged cytopenia and identified significantly increased frequencies of clonally expanded CX3CR1hi cytotoxic T cells, expressing high interferon (IFN)-γ and cytokine signaling gene sets, associated with prolonged cytopenia. In line with this, we found that hematopoietic stem cells from these patients expressed IFN-γ response signatures. IFN-γ deregulates hematopoietic stem cell self-renewal and differentiation and can be targeted with thrombopoietin agonists or IFN-γ-neutralizing antibodies, highlighting a potential mechanism-based approach for the treatment of CAR T-associated prolonged cytopenia.
Collapse
Affiliation(s)
- Paolo Strati
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xubin Li
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Deng
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mario L Marques-Piubelli
- Department Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jared Henderson
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grace Watson
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurel Deaton
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Taylor Cain
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haopeng Yang
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vida Ravanmehr
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luis E Fayad
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Swaminathan P Iyer
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Loretta J Nastoupil
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederick B Hagemeister
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neeraj Saini
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koichi Takahashi
- Department Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nathan H Fowler
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason R Westin
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Raphael E Steiner
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ranjit Nair
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher R Flowers
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sairah Ahmed
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michael R Green
- Department of Lymphoma & Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
27
|
Marcos Rubio A, Everaert C, Van Damme E, De Preter K, Vermaelen K. Circulating immune cell dynamics as outcome predictors for immunotherapy in non-small cell lung cancer. J Immunother Cancer 2023; 11:e007023. [PMID: 37536935 PMCID: PMC10401220 DOI: 10.1136/jitc-2023-007023] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2023] [Indexed: 08/05/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) continues to transform the therapeutic landscape of non-small cell lung cancer (NSCLC), with these drugs now being evaluated at every stage of the disease. In contrast to these advances, little progress has been made with respect to reliable predictive biomarkers that can inform clinicians on therapeutic efficacy. All current biomarkers for outcome prediction, including PD-L1, tumor mutational burden or complex immune gene expression signatures, require access to tumor tissue. Besides the invasive nature of the sampling procedure, other disadvantages of tumor tissue biopsies are the inability to capture the complete spatial heterogeneity of the tumor and the difficulty to perform longitudinal follow-up on treatment. A concept emerges in which systemic immune events developing at a distance from the tumor reflect local response or resistance to immunotherapy. The importance of this cancer 'macroenvironment', which can be deciphered by comprehensive analysis of peripheral blood immune cell subsets, has been demonstrated in several cutting-edge preclinical reports, and is corroborated by intriguing data emerging from ICI-treated patients. In this review, we will provide the biological rationale underlying the potential of blood immune cell-based biomarkers in guiding treatment decision in immunotherapy-eligible NSCLC patients. Finally, we will describe new techniques that will facilitate the discovery of more immune cell subpopulations with potential to become predictive biomarkers, and reflect on ways and the remaining challenges to bring this type of analysis to the routine clinical care in the near future.
Collapse
Affiliation(s)
- Alvaro Marcos Rubio
- VIB UGent Center for Medical Biotechnology, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
| | - Celine Everaert
- VIB UGent Center for Medical Biotechnology, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
| | - Eufra Van Damme
- VIB UGent Center for Medical Biotechnology, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
| | - Katleen De Preter
- VIB UGent Center for Medical Biotechnology, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
| | - Karim Vermaelen
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
- Tumor Immunology Laboratory, Department of Pulmonary Medicine, University Hospital Ghent, Ghent, Belgium
| |
Collapse
|
28
|
Bocanegra A, Fernández-Hinojal G, Ajona D, Blanco E, Zuazo M, Garnica M, Chocarro L, Alfaro-Arnedo E, Piñeiro-Hermida S, Morente P, Fernández L, Remirez A, Echaide M, Martinez-Aguillo M, Morilla I, Tavira B, Roncero A, Gotera C, Ventura A, Recalde N, Pichel JG, Lasarte JJ, Montuenga L, Vera R, Pio R, Escors D, Kochan G. Plasma fractalkine contributes to systemic myeloid diversity and PD-L1/PD-1 blockade in lung cancer. EMBO Rep 2023:e55884. [PMID: 37366231 PMCID: PMC10398648 DOI: 10.15252/embr.202255884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 05/17/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Recent studies highlight the importance of baseline functional immunity for immune checkpoint blockade therapies. High-dimensional systemic immune profiling is performed in a cohort of non-small-cell lung cancer patients undergoing PD-L1/PD-1 blockade immunotherapy. Responders show high baseline myeloid phenotypic diversity in peripheral blood. To quantify it, we define a diversity index as a potential biomarker of response. This parameter correlates with elevated activated monocytic cells and decreased granulocytic phenotypes. High-throughput profiling of soluble factors in plasma identifies fractalkine (FKN), a chemokine involved in immune chemotaxis and adhesion, as a biomarker of response to immunotherapy that also correlates with myeloid cell diversity in human patients and murine models. Secreted FKN inhibits lung adenocarcinoma growth in vivo through a prominent contribution of systemic effector NK cells and increased tumor immune infiltration. FKN sensitizes murine lung cancer models refractory to anti-PD-1 treatment to immune checkpoint blockade immunotherapy. Importantly, recombinant FKN and tumor-expressed FKN are efficacious in delaying tumor growth in vivo locally and systemically, indicating a potential therapeutic use of FKN in combination with immunotherapy.
Collapse
Affiliation(s)
- Ana Bocanegra
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | | | - Daniel Ajona
- Program in Solid Tumors, CIMA-University of Navarre-IdISNA, Pamplona, Spain
- CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra-IdISNA, Pamplona, Spain
| | - Ester Blanco
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
- Program in Gene Therapy and Regulation of Gene Expression, CIMA-University of Navarra-IdISNA, Pamplona, Spain
| | - Miren Zuazo
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Maider Garnica
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Luisa Chocarro
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Elvira Alfaro-Arnedo
- Lung Cancer and Respiratory Diseases Unit, Center for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
| | - Sergio Piñeiro-Hermida
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Pilar Morente
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Leticia Fernández
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Ana Remirez
- Program in Solid Tumors, CIMA-University of Navarre-IdISNA, Pamplona, Spain
| | - Miriam Echaide
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | | | - Idoia Morilla
- Department of Oncology, Hospital Universitario de Navarra-IdISNA, Pamplona, Spain
| | - Beatriz Tavira
- Program in Solid Tumors, CIMA-University of Navarre-IdISNA, Pamplona, Spain
- Cancer Center University of Navarra (CCUN), Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra-IdISNA, Pamplona, Spain
| | - Alejandra Roncero
- Pathological Anatomy Service, Hospital Universitario San Pedro, Rioja Salud, Logroño, Spain
- Pneumology Service, Rioja Salud, Logroño, Spain
| | | | | | | | - José G Pichel
- Lung Cancer and Respiratory Diseases Unit, Center for Biomedical Research of La Rioja (CIBIR), Fundación Rioja Salud, Logroño, Spain
- Spanish Biomedical Research Networking Centre, CIBERES, Madrid, Spain
| | - Juan José Lasarte
- Cancer Center University of Navarra (CCUN), Pamplona, Spain
- Program in Immunology and Immunotherapy, CIMA-University of Navarra-IdISNA, Pamplona, Spain
| | - Luis Montuenga
- Program in Solid Tumors, CIMA-University of Navarre-IdISNA, Pamplona, Spain
- CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra-IdISNA, Pamplona, Spain
| | - Ruth Vera
- Department of Oncology, Hospital Universitario de Navarra-IdISNA, Pamplona, Spain
| | - Ruben Pio
- Program in Solid Tumors, CIMA-University of Navarre-IdISNA, Pamplona, Spain
- CIBERONC, Centro de Investigación Biomédica en Red de Cáncer, Madrid, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra-IdISNA, Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Navarrabiomed, Hospital Universitario de Navarra, Universidad Publica de Navarra (UPNA), IdISNA, Pamplona, Spain
| |
Collapse
|
29
|
Sarkar J, Cortes Gomez E, Oba T, Chen H, Dy GK, Segal BH, Ernstoff MS, Ito F. Fluctuations in Gut Microbiome Composition During Immune Checkpoint Inhibitor Therapy. World J Oncol 2023; 14:178-187. [PMID: 37350807 PMCID: PMC10284636 DOI: 10.14740/wjon1587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/01/2023] [Indexed: 06/24/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) such as programmed cell death protein-1 (PD-1) inhibitors or PD-1 ligand-1 (PD-L1) inhibitors have led to remarkable improvement in outcomes of non-small cell lung cancer (NSCLC). Unfortunately, the significant benefits of ICI therapy are frequently limited by resistance to treatment and adverse effects, and the predictive value of pre-treatment tumor tissue PD-L1 expression is limited. Development of less invasive biomarkers that could identify responders and non-responders in early on-treatment could markedly improve the treatment regimen. Accumulating evidence suggests that baseline gut microbiota profile is associated with response to PD-1/PD-L1 blockade therapy. However, change in the gut microbiome composition during PD-1/PD-L1 blockade therapy and its relation to response remain unclear. Methods Here, we analyzed pre- and on-treatment fecal samples from five NSCLC patients receiving anti-PD-1 immunotherapy, alone or in tandem with chemotherapy, and performed 16S rRNA sequencing. Results The overall alpha diversity of the baseline gut microbiome was similar between three responders and two non-responders. While the gut microbiome composition remained stable overall during treatment (R2 = 0.145), responders showed significant changes in microbiome diversity between pre- and on-treatment samples during anti-PD-1 therapy compared to non-responders (P = 0.0274). Within the diverse microbiota, responders showed decreases in the abundance of genera Odoribacter, Gordonibacter, Candidatus Stoquefichus, Escherichia-Shigella, and Collinsella, and increase in abundance of Clostridium sensu stricto 1. In contrast, non-responders demonstrated on-treatment increases in genera Prevotella, Porphyromonas, Streptococcus, and Escherichia-Shigella, and decrease in abundance of Akkermansia. Conclusions This pilot study identified a substantial change in gut microbiome diversity between pre- and on-treatment samples in NSCLC patients responding to anti-PD-1 therapy compared to non-responders. Our findings highlight the potential utility of gut microbiota dynamics as a noninvasive biomarker to predict response to PD-1/PD-L1 blockade therapy for a wide variety of malignancies, which sets a path for future investigation in larger prospective studies.
Collapse
Affiliation(s)
- Joy Sarkar
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- These authors contributed equally to the study
| | - Eduardo Cortes Gomez
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Biostatistics, School of Public Health and Health Professions, SUNY at Buffalo, NY, USA
- These authors contributed equally to the study
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hongbin Chen
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA
| | - Grace K. Dy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA
| | - Brahm H. Segal
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marc S. Ernstoff
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, NY, USA
- National Cancer Institute, Division of Cancer Treatment and Diagnosis, Bethesda, Maryland, USA
| | - Fumito Ito
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Surgery, Norris Comprehensive Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
30
|
Trinh T, Adams WA, Calescibetta A, Tu N, Dalton R, So T, Wei M, Ward G, Kostenko E, Christiansen S, Cen L, McLemore A, Reed K, Whitting J, Gilvary D, Blanco NL, Segura CM, Nguyen J, Kandell W, Chen X, Cheng P, Wright GM, Cress WD, Liu J, Wright KL, Wei S, Eksioglu EA. CX3CR1 deficiency-induced TIL tumor restriction as a novel addition for CAR-T design in solid malignancies. iScience 2023; 26:106443. [PMID: 37070068 PMCID: PMC10105289 DOI: 10.1016/j.isci.2023.106443] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 11/15/2022] [Accepted: 03/14/2023] [Indexed: 04/19/2023] Open
Abstract
Advances in the understanding of the tumor microenvironment have led to development of immunotherapeutic strategies, such as chimeric antigen receptor T cells (CAR-Ts). However, despite success in blood malignancies, CAR-T therapies in solid tumors have been hampered by their restricted infiltration. Here, we used our understanding of early cytotoxic lymphocyte infiltration of human lymphocytes in solid tumors in vivo to investigate the receptors in normal, adjacent, and tumor tissues of primary non-small-cell lung cancer specimens. We found that CX3CL1-CX3CR1 reduction restricts cytotoxic cells from the solid-tumor bed, contributing to tumor escape. Based on this, we designed a CAR-T construct using the well-established natural killer group 2, member D (NKG2D) CAR-T expression together with overexpression of CX3CR1 to promote their infiltration. These CAR-Ts infiltrate tumors at higher rates than control-activated T cells or IL-15-overexpressing NKG2D CAR-Ts. This construct also had similar functionality in a liver-cancer model, demonstrating potential efficacy in other solid malignancies.
Collapse
Affiliation(s)
- ThuLe Trinh
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - William A. Adams
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alexandra Calescibetta
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nhan Tu
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert Dalton
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Tina So
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Max Wei
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Grace Ward
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology PhD Program, University of South Florida and H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Elena Kostenko
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sean Christiansen
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ling Cen
- Bioinformatics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Amy McLemore
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kayla Reed
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Junmin Whitting
- Cancer Biology PhD Program, University of South Florida and H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Danielle Gilvary
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Neale Lopez Blanco
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Carlos Moran Segura
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jonathan Nguyen
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Wendy Kandell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology PhD Program, University of South Florida and H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Xianghong Chen
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Pingyan Cheng
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Gabriela M. Wright
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - W. Douglas Cress
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jinghong Liu
- Department of Anesthesiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Kenneth L. Wright
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sheng Wei
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Erika A. Eksioglu
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
31
|
Zhou J, Chen G, Wang J, Zhou B, Sun X, Wang J, Tang S, Xing X, Hu X, Zhao Y, Peng Y, Shi W, Zhao T, Wu Y, Zhong H, Hong N, Ruan Z, Zhang Y, Jin W. Anti-PD-1 therapy achieves favorable outcomes in HBV-positive non-liver cancer. Oncogenesis 2023; 12:22. [PMID: 37080999 PMCID: PMC10119302 DOI: 10.1038/s41389-023-00468-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Anti-PD-1 therapy has shown promising outcomes in the treatment of different types of cancer. It is of fundamental interest to analyze the efficacy of anti-PD-1 therapy in cancer patients infected with hepatitis B virus (HBV) since the comorbidity of HBV and cancer is widely documented. We designed a multicenter retrospective study to evaluate the efficacy of anti-PD-1 therapy on non-liver cancer patients infected with HBV. We found anti-PD-1 therapy achieved much better outcomes in HBV+ non-liver cancer patients than their HBV- counterparts. We performed single-cell RNA sequencing (scRNA-seq) on peripheral blood mononuclear cells (PBMCs) from esophageal squamous cell carcinoma (ESCC) patients. We found both cytotoxicity score of T cells and MHC score of B cells significantly increased after anti-PD-1 therapy in HBV+ ESCC patients. We also identified CX3CR1high TEFF, a subset of CD8+ TEFF, associated with better clinical outcome in HBV+ ESCC patients. Lastly, we found CD8+ TEFF from HBV+ ESCC patients showing higher fraction of Exhaustionhi T than their HBV- counterpart. In summary, anti-PD-1 therapy on HBV+ non-liver cancer patients is safe and achieves better outcomes than that on HBV- non-liver cancer patients, potentially because HBV+ patients had higher fraction of Exhaustionhi T, which made them more efficiently respond to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Jie Zhou
- Chongqing International Institute for Immunology, 400030, Chongqing, China
- Department of Oncology, Southwest Hospital, Army Medical University, 400038, Chongqing, China
| | - Guanming Chen
- School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Jiuling Wang
- Chongqing International Institute for Immunology, 400030, Chongqing, China
- Institute of Immunology, PLA, Army Medical University, 400038, Chongqing, China
| | - Bo Zhou
- School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Xuemin Sun
- Chongqing International Institute for Immunology, 400030, Chongqing, China
- Institute of Immunology, PLA, Army Medical University, 400038, Chongqing, China
| | - Jinsong Wang
- Chongqing International Institute for Immunology, 400030, Chongqing, China
- Institute of Immunology, PLA, Army Medical University, 400038, Chongqing, China
| | - Shu Tang
- Institute of Cancer, Xinqiao Hospital, Army Medical University, 400038, Chongqing, China
| | - Xiangju Xing
- Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Chongqing Medical University, 400038, Chongqing, China
| | - Xiaofei Hu
- Department of Radiology, Southwest Hospital, Army Medical University, 400038, Chongqing, China
| | - Yang Zhao
- Department of Oncology, Southwest Hospital, Army Medical University, 400038, Chongqing, China
| | - Yu Peng
- Department of Oncology, Southwest Hospital, Army Medical University, 400038, Chongqing, China
| | - Wenjiong Shi
- Chongqing International Institute for Immunology, 400030, Chongqing, China
| | - Tingting Zhao
- Chongqing International Institute for Immunology, 400030, Chongqing, China
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 400054, Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, PLA, Army Medical University, 400038, Chongqing, China
| | - Hanbing Zhong
- School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Ni Hong
- School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Zhihua Ruan
- Department of Oncology, Southwest Hospital, Army Medical University, 400038, Chongqing, China.
| | - Yi Zhang
- Chongqing International Institute for Immunology, 400030, Chongqing, China.
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 400054, Chongqing, China.
| | - Wenfei Jin
- School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China.
| |
Collapse
|
32
|
Abdelfatah E, Long MD, Kajihara R, Oba T, Yamauchi T, Chen H, Sarkar J, Attwood K, Matsuzaki J, Segal BH, Dy GK, Ito F. Predictive and Prognostic Implications of Circulating CX3CR1 + CD8 + T Cells in Non-Small Cell Lung Cancer Patients Treated with Chemo-Immunotherapy. CANCER RESEARCH COMMUNICATIONS 2023; 3:510-520. [PMID: 37009132 PMCID: PMC10060186 DOI: 10.1158/2767-9764.crc-22-0383] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 02/20/2023] [Indexed: 02/25/2023]
Abstract
Lack of reliable predictive biomarkers is a major limitation of combination therapy with chemotherapy and anti-programmed cell death protein 1/programmed death-ligand 1 (anti-PD-1/PD-L1) therapy (chemo-immunotherapy). We previously observed that the increase of peripheral blood CD8+ T cells expressing CX3CR1, a marker of differentiation, correlates with response to anti-PD-1 therapy; however, the predictive and prognostic value of T-cell CX3CR1 expression during chemo-immunotherapy is unknown. Here, we evaluated the utility of circulating CX3CR1+CD8+ T cells as a predictive correlate of response to chemo-immunotherapy in patients with non-small cell lung cancer (NSCLC). At least 10% increase of the CX3CR1+ subset in circulating CD8+ T cells from baseline (CX3CR1 score) was associated with response to chemo-immunotherapy as early as 4 weeks with 85.7% overall accuracy of predicting response at 6 weeks. Furthermore, at least 10% increase of the CX3CR1 score correlated with substantially better progression-free (P = 0.0051) and overall survival (P = 0.0138) on Kaplan-Meier analysis. Combined single-cell RNA/T-cell receptor (TCR) sequencing of circulating T cells from longitudinally obtained blood samples and TCR sequencing of tumor tissue from the same patient who received a long-term benefit from the treatment demonstrated remarkable changes in genomic and transcriptomic signatures of T cells as well as evolution of TCR clonotypes in peripheral blood containing highly frequent tumor-infiltrating lymphocyte repertoires overexpressing CX3CR1 early after initiation of the treatment despite stable findings of the imaging study. Collectively, these findings highlight the potential utility of T-cell CX3CR1 expression as a dynamic blood-based biomarker during the early course of chemo-immunotherapy and a marker to identify frequent circulating tumor-infiltrating lymphocyte repertoires. Significance Current approaches to combined chemotherapy and anti-PD-1/PD-L1 therapy (chemo-immunotherapy) for patients with NSCLC are limited by the lack of reliable predictive biomarkers. This study shows the utility of T-cell differentiation marker, CX3CR1, as an early on-treatment predictor of response and changes in genomic/transcriptomic signatures of circulating tumor-infiltrating lymphocyte repertoires in patients with NSCLC undergoing chemo-immunotherapy.
Collapse
Affiliation(s)
- Eihab Abdelfatah
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Mark D. Long
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Ryutaro Kajihara
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Biomedical Laboratory Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takayoshi Yamauchi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Surgery, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Hongbin Chen
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, New York
| | - Joy Sarkar
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Kristopher Attwood
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Junko Matsuzaki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Brahm H. Segal
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, New York
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Grace K. Dy
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Fumito Ito
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Surgery, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, the State University of New York, Buffalo, New York
| |
Collapse
|
33
|
Schaible P, Bethge W, Lengerke C, Haraszti RA. RNA Therapeutics for Improving CAR T-cell Safety and Efficacy. Cancer Res 2023; 83:354-362. [PMID: 36512627 PMCID: PMC7614194 DOI: 10.1158/0008-5472.can-22-2155] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/02/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Autologous chimeric antigen receptor (CAR) T cells have recently emerged as potent tools in the fight against cancer, with promising therapeutic efficacy against hematological malignancies. However, several limitations hamper their widespread clinical use, including availability of target antigen, severe toxic effects, primary and secondary resistance, heterogeneous quality of autologous T cells, variable persistence, and low activity against solid tumors. Development of allogeneic off-the-shelf CAR T cells could help address some of these limitations but is impeded by alloimmunity with either rejection and limited expansion of allo-CAR T cells or CAR T cells versus host reactions. RNA therapeutics, such as small interfering RNAs, microRNAs, and antisense oligonucleotides, are able to silence transcripts in a sequence-specific and proliferation-sensitive way, which may offer a way to overcome some of the challenges facing CAR T-cell development and clinical utility. Here, we review how different RNA therapeutics or a combination of RNA therapeutics and genetic engineering could be harnessed to improve the safety and efficacy of autologous and allogeneic CAR T-cell therapy.
Collapse
Affiliation(s)
- Philipp Schaible
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Wolfgang Bethge
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Claudia Lengerke
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Reka Agnes Haraszti
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
34
|
Atreya I, Neurath MF. How the Tumor Micromilieu Modulates the Recruitment and Activation of Colorectal Cancer-Infiltrating Lymphocytes. Biomedicines 2022; 10:biomedicines10112940. [PMID: 36428508 PMCID: PMC9687992 DOI: 10.3390/biomedicines10112940] [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: 10/23/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
The successful treatment of advanced colorectal cancer disease still represents an insufficiently solved clinical challenge, which is further complicated by the fact that the majority of malignant colon tumors show only relatively low immunogenicity and therefore have only limited responsiveness to immunotherapeutic approaches, such as, for instance, the use of checkpoint inhibitors. As it has been well established over the past two decades that the local tumor microenvironment and, in particular, the quantity, quality, and activation status of intratumoral immune cells critically influence the clinical prognosis of patients diagnosed with colorectal cancer and their individual benefits from immunotherapy, the enhancement of the intratumoral accumulation of cytolytic effector T lymphocytes and other cellular mediators of the antitumor immune response has emerged as a targeted objective. For the future identification and clinical validation of novel therapeutic target structures, it will thus be essential to further decipher the molecular mechanisms and cellular interactions in the intestinal tumor microenvironment, which are crucially involved in immune cell recruitment and activation. In this context, our review article aims at providing an overview of the key chemokines and cytokines whose presence in the tumor micromilieu relevantly modulates the numeric composition and antitumor capacity of tumor-infiltrating lymphocytes.
Collapse
Affiliation(s)
- Imke Atreya
- Department of Medicine 1, Erlangen University Hospital, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Erlangen University Hospital, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen University Hospital, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8535204; Fax: +49-9131-8535209
| |
Collapse
|
35
|
Systemic CD4 Immunity and PD-L1/PD-1 Blockade Immunotherapy. Int J Mol Sci 2022; 23:ijms232113241. [PMID: 36362027 PMCID: PMC9655397 DOI: 10.3390/ijms232113241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/25/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
PD-L1/PD-1 blockade immunotherapy has changed the therapeutic approaches for the treatment of many cancers. Nevertheless, the mechanisms underlying its efficacy or treatment failure are still unclear. Proficient systemic immunity seems to be a prerequisite for efficacy, as recently shown in patients and in mouse models. It is widely accepted that expansion of anti-tumor CD8 T cell populations is principally responsible for anti-tumor responses. In contrast, the role of CD4 T cells has been less studied. Here we review and discuss the evidence supporting the contribution of CD4 T cells to anti-tumor immunity, especially recent advances linking CD4 T cell subsets to efficacious PD-L1/PD-1 blockade immunotherapy. We also discuss the role of CD4 T cell memory subsets present in peripheral blood before the start of immunotherapies, and their utility as predictors of response.
Collapse
|
36
|
Chen Q, Ma J, Wang X, Zhu X. Identification of prognostic candidate signatures by systematically revealing transcriptome characteristics in lung adenocarcinoma with differing tumor microenvironment immune phenotypes. Aging (Albany NY) 2022; 14:4786-4818. [PMID: 35675043 PMCID: PMC9217709 DOI: 10.18632/aging.204112] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 05/24/2022] [Indexed: 12/09/2022]
Abstract
Accumulated evidence shows that tumor microenvironment plays crucial roles in predicting clinical outcomes of lung adenocarcinoma (LUAD). The current study aimed to identify some potentially prognostic signatures by systematically revealing the transcriptome characteristics in LUADs with differing immune phenotypes. LUAD gene expression data were retrieved from the public TCGA and GEO databases, and the transcriptome characteristics were systematically revealed using a comprehensive bioinformatics method including single-sample gene set enrichment analysis, differentially expressed gene (DEG) analysis, protein and protein interaction (PPI) network construction, competitive endogenous RNA (ceRNA) network construction, weighted gene coexpression network analysis and prognostic model establishment. Finally, 1169 key DEGs associated with LUAD immune phenotype, including 88 immune DEGs, were excavated. Five essential and eight immune essential DEGs were separately identified by constructing two PPI networks based on the above DEGs. Totals of 1085 key DElncRNAs and 45 key DEmiRNAs were excavated and one ceRNA network consisting of 26 DEmRNAs, 3 DEmiRNAs and 57 DElncRNAs were established. The most significant gene coexpression module (cor=0.63 and p=3e-55) associated with LUAD immune phenotypes and three genes (FGR, BTK, SPI1) related to the immune cell infiltration were identified. Three robust prognostic signatures including a 9-lncRNA, an 8-lncRNA and an 8-mRNA were established. The areas under the curves of 5-year correlated with overall survival rate were separately 0.7319, 0.7228 and 0.713 in the receiver operating characteristic curve. The findings provide novel insights into the immunological mechanism in LUAD biology and in predicting the prognosis of LUAD patients.
Collapse
Affiliation(s)
- Qiang Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jiakang Ma
- Department of Medical Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyi Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Xiangqing Zhu
- Basic Medical Laboratory, The 920th Hospital of Joint Logistics Support Force of PLA, Kunming, China
| |
Collapse
|
37
|
Wang J, Shen K, Mu W, Li W, Zhang M, Zhang W, Li Z, Ge T, Zhu Z, Zhang S, Chen C, Xing S, Zhu L, Chen L, Wang N, Huang L, Li D, Xiao M, Zhou J. T Cell Defects: New Insights Into the Primary Resistance Factor to CD19/CD22 Cocktail CAR T-Cell Immunotherapy in Diffuse Large B-Cell Lymphoma. Front Immunol 2022; 13:873789. [PMID: 35572515 PMCID: PMC9094425 DOI: 10.3389/fimmu.2022.873789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/21/2022] [Indexed: 12/05/2022] Open
Abstract
Despite impressive progress, a significant portion of patients still experience primary or secondary resistance to chimeric antigen receptor (CAR) T-cell immunotherapy for relapsed/refractory diffuse large B-cell lymphoma (r/r DLBCL). The mechanism of primary resistance involves T-cell extrinsic and intrinsic dysfunction. In the present study, a total of 135 patients of DLBCL treated with murine CD19/CD22 cocktail CAR T-therapy were assessed retrospectively. Based on four criteria (maximal expansion of the transgene/CAR-positive T-cell levels post-infusion [Cmax], initial persistence of the transgene by the CAR transgene level at +3 months [Tlast], CD19+ B-cell levels [B-cell recovery], and the initial response to CAR T-cell therapy), 48 patients were included in the research and divided into two groups (a T-normal group [n=22] and a T-defect [n=26] group). According to univariate and multivariate regression analyses, higher lactate dehydrogenase (LDH) levels before leukapheresis (hazard ratio (HR) = 1.922; p = 0.045) and lower cytokine release syndrome (CRS) grade after CAR T-cell infusion (HR = 0.150; p = 0.026) were independent risk factors of T-cell dysfunction. Moreover, using whole-exon sequencing, we found that germline variants in 47 genes were significantly enriched in the T-defect group compared to the T-normal group (96% vs. 41%; p<0.0001), these genes consisted of CAR structure genes (n=3), T-cell signal 1 to signal 3 genes (n=13), T cell immune regulation- and checkpoint-related genes (n=9), cytokine- and chemokine-related genes (n=13), and T-cell metabolism-related genes (n=9). Heterozygous germline UNC13D mutations had the highest intergroup differences (26.9% vs. 0%; p=0.008). Compound heterozygous CX3CR1I249/M280 variants, referred to as pathogenic and risk factors according to the ClinVar database, were enriched in the T-defect group (3 of 26). In summary, the clinical characteristics and T-cell immunodeficiency genetic features may help explain the underlying mechanism of treatment primary resistance and provide novel insights into CAR T-cell immunotherapy.
Collapse
Affiliation(s)
- Jiachen Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Kefeng Shen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Weigang Li
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meilan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Wei Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Zhe Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Tong Ge
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | | | | | - Caixia Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Shugang Xing
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Li Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Na Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Dengju Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, China
| |
Collapse
|
38
|
Dhanwani R, Lima-Junior JR, Sethi A, Pham J, Williams G, Frazier A, Xu Y, Amara AW, Standaert DG, Goldman JG, Litvan I, Alcalay RN, Peters B, Sulzer D, Arlehamn CSL, Sette A. Transcriptional analysis of peripheral memory T cells reveals Parkinson's disease-specific gene signatures. NPJ Parkinsons Dis 2022; 8:30. [PMID: 35314697 PMCID: PMC8938520 DOI: 10.1038/s41531-022-00282-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a multi-stage neurodegenerative disorder with largely unknown etiology. Recent findings have identified PD-associated autoimmune features including roles for T cells. To further characterize the role of T cells in PD, we performed RNA sequencing on PBMC and peripheral CD4 and CD8 memory T cell subsets derived from PD patients and age-matched healthy controls. When the groups were stratified by their T cell responsiveness to alpha-synuclein (α-syn) as a proxy for an ongoing inflammatory autoimmune response, the study revealed a broad differential gene expression profile in memory T cell subsets and a specific PD associated gene signature. We identified significant enrichment of transcriptomic signatures previously associated with PD, including for oxidative stress, phosphorylation, autophagy of mitochondria, cholesterol metabolism and inflammation, and the chemokine signaling proteins CX3CR1, CCR5, and CCR1. In addition, we identified genes in these peripheral cells that have previously been shown to be involved in PD pathogenesis and expressed in neurons, such as LRRK2, LAMP3, and aquaporin. Together, these findings suggest that features of circulating T cells with α-syn-specific responses in PD patients provide insights into the interactive processes that occur during PD pathogenesis and suggest potential intervention targets.
Collapse
Affiliation(s)
- Rekha Dhanwani
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - João Rodrigues Lima-Junior
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Ashu Sethi
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - John Pham
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Gregory Williams
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - April Frazier
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Yaqian Xu
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.,Department of Neurology, Columbia University, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Amy W Amara
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.,Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - David G Standaert
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.,Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Jennifer G Goldman
- Shirley Ryan AbilityLab, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Irene Litvan
- Department of Neuroscience, University of California San Diego, La Jolla, CA, 92093, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - David Sulzer
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.,Department of Neurology, Columbia University, Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA.,Departments of Psychiatry and Pharmacology, Columbia University, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Cecilia S Lindestam Arlehamn
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA. .,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA. .,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA. .,Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
39
|
Hezaveh K, Shinde RS, Klötgen A, Halaby MJ, Lamorte S, Ciudad MT, Quevedo R, Neufeld L, Liu ZQ, Jin R, Grünwald BT, Foerster EG, Chaharlangi D, Guo M, Makhijani P, Zhang X, Pugh TJ, Pinto DM, Co IL, McGuigan AP, Jang GH, Khokha R, Ohashi PS, O’Kane GM, Gallinger S, Navarre WW, Maughan H, Philpott DJ, Brooks DG, McGaha TL. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022; 55:324-340.e8. [PMID: 35139353 PMCID: PMC8888129 DOI: 10.1016/j.immuni.2022.01.006] [Citation(s) in RCA: 335] [Impact Index Per Article: 111.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/19/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a sensor of products of tryptophan metabolism and a potent modulator of immunity. Here, we examined the impact of AhR in tumor-associated macrophage (TAM) function in pancreatic ductal adenocarcinoma (PDAC). TAMs exhibited high AhR activity and Ahr-deficient macrophages developed an inflammatory phenotype. Deletion of Ahr in myeloid cells or pharmacologic inhibition of AhR reduced PDAC growth, improved efficacy of immune checkpoint blockade, and increased intra-tumoral frequencies of IFNγ+CD8+ T cells. Macrophage tryptophan metabolism was not required for this effect. Rather, macrophage AhR activity was dependent on Lactobacillus metabolization of dietary tryptophan to indoles. Removal of dietary tryptophan reduced TAM AhR activity and promoted intra-tumoral accumulation of TNFα+IFNγ+CD8+ T cells; provision of dietary indoles blocked this effect. In patients with PDAC, high AHR expression associated with rapid disease progression and mortality, as well as with an immune-suppressive TAM phenotype, suggesting conservation of this regulatory axis in human disease.
Collapse
Affiliation(s)
- Kebria Hezaveh
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,These authors contributed equally,Present address: Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceutical R&D, Astra Zeneca, Gothenburg, 431 50, Sweden
| | - Rahul S. Shinde
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,These authors contributed equally,Present address: Immunology, Microenvironment, and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andreas Klötgen
- Department of Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig 38124, Germany
| | - Marie Jo Halaby
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Sara Lamorte
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - M. Teresa Ciudad
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Rene Quevedo
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Luke Neufeld
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zhe Qi Liu
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Robbie Jin
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Barbara T. Grünwald
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | | | - Danica Chaharlangi
- Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mengdi Guo
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Priya Makhijani
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Xin Zhang
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Trevor J. Pugh
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada,The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Devanand M. Pinto
- National Research Council, Human Health Therapeutics, Halifax, NS B3H 3Z1, Canada
| | - Ileana L. Co
- Institute of Biomedical Engineering, The University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Alison P. McGuigan
- Institute of Biomedical Engineering, The University of Toronto, Toronto, ON M5S 3G9, Canada,Department of Chemical Engineering and Applied Chemistry, The University of Toronto, Toronto, ON M5S 3E5, Canada
| | - Gun Ho Jang
- The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Rama Khokha
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada,Department of Medical Biophysics, The University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Pamela S. Ohashi
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Grainne M. O’Kane
- The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada,Division of Medical Oncology, Department of Medicine, The University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Steven Gallinger
- The Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada,Department of Laboratory Medicine and Pathobiology, The University of Toronto, Toronto, ON M5S 1A8, Canada,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - William W. Navarre
- Department of Molecular Genetics, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Dana J. Philpott
- Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David G. Brooks
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tracy L. McGaha
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada,Department of Immunology, The University of Toronto, Toronto, ON M5S 1A8, Canada,Lead contact,Correspondence:
| |
Collapse
|
40
|
Wolf SP, Wen FT, Schreiber H. Criteria to make animal studies more relevant to treating human cancer. Curr Opin Immunol 2022; 74:25-31. [PMID: 34619458 PMCID: PMC8901458 DOI: 10.1016/j.coi.2021.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/28/2021] [Accepted: 09/11/2021] [Indexed: 02/03/2023]
Abstract
Certain aspects of experimental tumor models in mice most accurately reflect the biology and immunology of cancer in patients. A survey of experimental cancer immunotherapy papers published in 2020 shows most do not achieve cancer shrinkage although treatment is initiated at an early time point after cancer cell injection, which does not reflect cancer immunotherapy in patients. Even then, few current experimental approaches eradicate the injected malignant cells, most only delay outgrowth. The value of targeting mutation-encoded tumor-specific antigens becomes increasingly evident while problems of finding normal gene-encoded tumor-associated antigens as safe, effective targets persist. It might be time to refocus on realistic experimental settings and truly cancer-specific targets. These antigens are associated with the least risk of side effects.
Collapse
Affiliation(s)
- Steven P Wolf
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA; David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL, 60637, USA
| | - Frank T Wen
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | - Hans Schreiber
- Department of Pathology, The University of Chicago, Chicago, IL, 60637, USA; David and Etta Jonas Center for Cellular Therapy, The University of Chicago, Chicago, IL, 60637, USA; Committee on Cancer Biology and Committee on Immunology, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
41
|
Yue Y, Zhang Q, Sun Z. CX3CR1 Acts as a Protective Biomarker in the Tumor Microenvironment of Colorectal Cancer. Front Immunol 2022; 12:758040. [PMID: 35140706 PMCID: PMC8818863 DOI: 10.3389/fimmu.2021.758040] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) plays an important role in the pathogenesis of many cancers. We aimed to screen the TME-related hub genes of colorectal adenoma (CRAD) and identify possible prognostic biomarkers. The gene expression profiles and clinical data of 464 CRAD patients in The Cancer Genome Atlas (TCGA) database were downloaded. The Estimation of STromal and Immune cells in MAlignant Tumours using Expression data (ESTIMATE) algorithm was performed to calculate the ImmuneScore, StromalScore, and EstimateScore. Thereafter, differentially expressed genes (DEGs) were screened. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and protein–protein interaction (PPI) analysis were performed to explore the roles of DEGs. Furthermore, univariate and multivariate Cox analyses were accomplished to identify independent prognostic factors of CRAD. CX3CR1 was selected as a hub gene, and the expression was confirmed in colorectal cancer (CRC) patients and cell lines. The correlations between CX3CR1 and tumor-infiltrating immune cells were estimated by Tumor IMmune Estimation Resource database (TIMER) and CIBERSORT analysis. Besides, we investigated the effects of coculture with THP-1-derived macrophages with HCT8 cells with low CX3CR1 expression on immune marker expression, cell viability, and migration. There were significant differences in the ImmuneScore and EstimateScore among different stages. Patients with low scores presented significantly lower lifetimes than those in the high-score group. Moreover, we recognized 1,578 intersection genes in ImmuneScore and StromalScore, and these genes were mainly enriched in numerous immune-related biological processes. CX3CR1 was found to be associated with immune cell infiltration levels, immune marker expression, and macrophage polarization. Simultaneous silencing of CX3CR1 and coculture with THP-1 cells further regulated macrophage polarization and promoted the cell proliferation and migration of CRC cells. CX3CR1 was decreased in CRAD tissues and cell lines and was related to T and N stages, tumor differentiation, and prognosis. Our results suggest that CX3CR1 contributes to the recruitment and regulation of immune-infiltrating cells and macrophage polarization in CRC and TAM-induced CRC progression. CX3CR1 may act as a prognostic biomarker in CRC.
Collapse
Affiliation(s)
- Yuanyi Yue
- Department of Gastroenterology Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiang Zhang
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhengrong Sun
- BioBank, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Zhengrong Sun,
| |
Collapse
|
42
|
Yamauchi T, Hoki T, Oba T, Kajihara R, Attwood K, Cao X, Ito F. CD40 and CD80/86 signaling in cDC1s mediate effective neoantigen vaccination and generation of antigen-specific CX3CR1 + CD8 + T cells. Cancer Immunol Immunother 2022; 71:137-151. [PMID: 34037810 PMCID: PMC8715856 DOI: 10.1007/s00262-021-02969-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/12/2021] [Indexed: 01/03/2023]
Abstract
The use of tumor mutation-derived neoantigen represents a promising approach for cancer vaccines. Preclinical and early phase human clinical studies have shown the successful induction of tumor neoepitope-directed responses; however, overall clinical efficacy of neoantigen vaccines has been limited. One major obstacle of this strategy is the prevailing lack of sufficient understanding of the mechanism underlying the generation of neoantigen-specific CD8+ T cells. Here, we report a correlation between antitumor efficacy of neoantigen/toll-like receptor 3 (TLR3)/CD40 agonists vaccination and an increased frequency of circulating antigen-specific CD8+ T cells expressing CX3C chemokine receptor 1 (CX3CR1) in a preclinical model. Mechanistic studies using mixed bone marrow chimeras identified that CD40 and CD80/86, but not CD70 signaling in Batf3-dependent conventional type 1 dendritic cells (cDC1s) is required for the antitumor efficacy of neoantigen vaccine and generation of neoantigen-specific CX3CR1+ CD8+ T cells. Although CX3CR1+ CD8+ T cells exhibited robust in vitro effector function, in vivo depletion of this subset did not alter the antitumor efficacy of neoantigen/TLR3/CD40 agonists vaccination. These findings indicate that the vaccine-primed CX3CR1+ subset is dispensable for antitumor CD8+ T cell responses, but can be used as a blood-based T-cell biomarker for effective priming of CD8+ T cells as post-differentiated T cells. Taken together, our results reveal a critical role of CD40 and CD80/86 signaling in cDC1s in antitumor efficacy of neoantigen-based therapeutic vaccines, and implicate the potential utility of CX3CR1 as a circulating predictive T-cell biomarker in vaccine therapy.
Collapse
Affiliation(s)
- Takayoshi Yamauchi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Toshifumi Hoki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ryutaro Kajihara
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Xuefang Cao
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fumito Ito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, State University of New York At Buffalo, Buffalo, NY, 14263, USA.
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
43
|
Rational administration sequencing of immunochemotherapy elicits powerful anti-tumor effect. J Control Release 2021; 341:769-781. [PMID: 34952044 DOI: 10.1016/j.jconrel.2021.12.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/02/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022]
Abstract
As a research hotspot, immune checkpoint inhibitors (ICIs) is often combined with other therapeutics in order to exert better clinical efficacy. To date, extensive laboratory and clinical investigations into the combination of ICIs and chemotherapy have been carried out, demonstrating augmented effectiveness and broad application prospects in anti-tumor therapy. However, the administration of these two treatment modalities is usually randomized or fixed to a given chronological order. Nevertheless, the pharmacological effect of drug is closely related to its exposure behavior in vivo, which may consequently affect the synergistic outcomes of a combined therapy. In this study, we prepared a lipid nanoparticle encapsulating docetaxel (DTX-VNS), and associated it with the immune checkpoint inhibitor anti-PD-1 antibody (αPD-1) for the treatment of malignant tumors. To identify the optimum timing and sequencing for chemotherapy and immunotherapy, we designed three administration regimes, including the simultaneous delivery of DTX-VNS and αPD-1(DTX-VNS@αPD-1), DTX-VNS delivery before (DTX-VNS plus αPD-1) or post (αPD-1 plus DTX-VNS) PD-1 blockade with an interval of two days. Analysis from mass spectrometry, multi-factor detection and other techniques indicated that DTX-VNS plus αPD-1 initiated a powerful anti-tumor response in multiple tumor models, contributing to a remarkably reshaped tumor microenvironment landscape, which may attribute to the maximum therapeutic additive effects arise from a concomitant exposure of DTX-VNS and αPD-1 at the tumor site. By profiling the exposure kinetics of nanoparticles and αPD-1 in vivo, we defined the administration schedule with utmost therapeutic benefits, which may provide a valuable clinical reference for the rational administration of immunochemotherapy.
Collapse
|
44
|
Yokoi T, Oba T, Kajihara R, Abrams SI, Ito F. Local, multimodal intralesional therapy renders distant brain metastases susceptible to PD-L1 blockade in a preclinical model of triple-negative breast cancer. Sci Rep 2021; 11:21992. [PMID: 34754037 PMCID: PMC8578367 DOI: 10.1038/s41598-021-01455-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/25/2021] [Indexed: 12/31/2022] Open
Abstract
Despite recent progress in therapeutic strategies, prognosis of metastatic triple-negative breast cancer (TNBC) remains dismal. Evidence suggests that the induction and activation of tumor-residing conventional type-1 dendritic cells (cDC1s) is critical for the generation of CD8+ T cells that mediate the regression of mammary tumors and potentiate anti-PD-1/PD-L1 therapeutic efficacy. However, it remains unknown whether this strategy is effective against metastatic TNBC, which is poorly responsive to immunotherapy. Here, using a mouse model of TNBC, we established orthotopic mammary tumors and brain metastases, and treated mammary tumors with in situ immunomodulation (ISIM) consisting of intratumoral injections of Flt3L to mobilize cDC1s, local irradiation to induce immunogenic tumor cell death, and TLR3/CD40 stimulation to activate cDC1s. ISIM treatment of the mammary tumor increased circulating T cells with effector phenotypes, and infiltration of CD8+ T cells into the metastatic brain lesions, resulting in delayed progression of brain metastases and improved survival. Furthermore, although anti-PD-L1 therapy alone was ineffective against brain metastases, ISIM overcame resistance to anti-PD-L1 therapy, which rendered these tumor-bearing mice responsive to anti-PD-L1 therapy and further improved survival. Collectively, these results illustrate the therapeutic potential of multimodal intralesional therapy for patients with unresectable and metastatic TNBC.
Collapse
Affiliation(s)
- Toshihiro Yokoi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.,Department of Neurosurgery, Shiga University of Medical Science, Otsu, Japan
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.,Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Ryutaro Kajihara
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Fumito Ito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA. .,Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA. .,Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
45
|
Patel A, Oba T, Kajihara R, Yokoi T, Abrams SI, Ito F. Multimodal Intralesional Therapy for Reshaping the Myeloid Compartment of Tumors Resistant to Anti-PD-L1 Therapy via IRF8 Expression. THE JOURNAL OF IMMUNOLOGY 2021; 207:1298-1309. [PMID: 34362833 DOI: 10.4049/jimmunol.2100281] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 06/23/2021] [Indexed: 12/29/2022]
Abstract
Intralesional therapy is a promising approach for remodeling the immunosuppressive tumor microenvironment while minimizing systemic toxicities. A combinatorial in situ immunomodulation (ISIM) regimen with intratumoral administration of Fms-like tyrosine kinase 3 ligand (Flt3L), local irradiation, and TLR3/CD40 stimulation induces and activates conventional type 1 dendritic cells in the tumor microenvironment and elicits de novo adaptive T cell immunity in poorly T cell-inflamed tumors. However, the impact of ISIM on myeloid-derived suppressor cells (MDSCs), which may promote treatment resistance, remains unknown. In this study, we examined changes in the frequencies and heterogeneity of CD11b+Ly-6CloLy-6G+ polymorphonuclear (PMN)-MDSCs and CD11b+Ly-6ChiLy-6G- monocytic (M)-MDSCs in ISIM-treated tumors using mouse models of triple-negative breast cancer. We found that ISIM treatment decreased intratumoral PMN-MDSCs, but not M-MDSCs. Although the frequency of M-MDSCs remained unchanged, ISIM caused a substantial reduction of CX3CR1+ M-MDSCs that express F4/80. Importantly, these ISIM-induced changes in tumor-residing MDSCs were not observed in Batf3-/- mice. ISIM upregulated PD-L1 expression in both M-MDSCs and PMN-MDSCs and synergized with anti-PD-L1 therapy. Furthermore, ISIM increased the expression of IFN regulatory factor 8 (IRF8) in myeloid cells, a known negative regulator of MDSCs, indicating a potential mechanism by which ISIM decreases PMN-MDSC levels. Accordingly, ISIM-mediated reduction of PMN-MDSCs was not observed in mice with conditional deletion of IRF8 in myeloid cells. Altogether, these findings suggest that ISIM holds promise as a multimodal intralesional therapy to alter both lymphoid and myeloid compartments of highly aggressive poorly T cell-inflamed, myeloid-enriched tumors resistant to anti-PD-L1 therapy.
Collapse
Affiliation(s)
- Ankit Patel
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Ryutaro Kajihara
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Toshihiro Yokoi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY; and
| | - Fumito Ito
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY; .,Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY.,Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY; and.,Department of Surgery, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY
| |
Collapse
|
46
|
Collier JL, Weiss SA, Pauken KE, Sen DR, Sharpe AH. Not-so-opposite ends of the spectrum: CD8 + T cell dysfunction across chronic infection, cancer and autoimmunity. Nat Immunol 2021; 22:809-819. [PMID: 34140679 PMCID: PMC9197228 DOI: 10.1038/s41590-021-00949-7] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/29/2021] [Indexed: 02/05/2023]
Abstract
CD8+ T cells are critical mediators of cytotoxic effector function in infection, cancer and autoimmunity. In cancer and chronic viral infection, CD8+ T cells undergo a progressive loss of cytokine production and cytotoxicity, a state termed T cell exhaustion. In autoimmunity, autoreactive CD8+ T cells retain the capacity to effectively mediate the destruction of host tissues. Although the clinical outcome differs in each context, CD8+ T cells are chronically exposed to antigen in all three. These chronically stimulated CD8+ T cells share some common phenotypic features, as well as transcriptional and epigenetic programming, across disease contexts. A better understanding of these CD8+ T cell states may reveal novel strategies to augment clearance of chronic viral infection and cancer and to mitigate self-reactivity leading to tissue damage in autoimmunity.
Collapse
Affiliation(s)
- Jenna L Collier
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital
| | - Sarah A Weiss
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston MA.,Broad Institute of MIT and Harvard, Cambridge MA
| | - Kristen E Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital
| | - Debattama R Sen
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital.,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women’s Hospital.,Broad Institute of MIT and Harvard, Cambridge MA
| |
Collapse
|
47
|
Fan T, Liu Y, Liu H, Wang L, Tian H, Zheng Y, Zheng B, Xue L, Tan F, Xue Q, Gao S, Li C, He J. Comprehensive analysis of a chemokine- and chemokine receptor family-based signature for patients with lung adenocarcinoma. Cancer Immunol Immunother 2021; 70:3651-3667. [PMID: 33977344 DOI: 10.1007/s00262-021-02944-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/10/2021] [Indexed: 01/12/2023]
Abstract
The clinical significance and comprehensive features of chemokines and their receptors in lung adenocarcinoma (LUAD) have not been clarified. We aimed to characterize the expression profiles of chemokine and chemokine receptor family members and construct a chemokine- and chemokine receptor-based prognosis signature. A total of 1511 patients with LUAD from seven independent cohorts were included in the study. The training set collected from The Cancer Genome Atlas (TCGA) database containing 468 cases. The validation was performed on the basis of six different cohorts downloaded from Gene Expression Omnibus (GEO) database. A five-chemokine- and chemokine receptor-(CXCL2, CXCL13, CCL26, CCL20, CX3CR1) based prognosis signature was constructed with TCGA dataset using LASSO Cox regression and Cox proportional hazards regression analysis. A multivariate analysis verified that this signature was an independent prognostic factor. The predictive value of this signature was further verified by other six independent cohorts and multiple clinical subtypes. We performed immune cell infiltration analysis and biological pathway analysis which provided more insight into this signature-related immune and inflammatory landscape and clarified the intrinsic molecular mechanism by which this signature could be used to predict clinical prognosis. Furthermore, we explored the close relationship between this signature and tumor mutation burden (TMB), neoantigen burden, PD-1, PD-L1, CTLA4, TIDE score, T cell-inflamed score. This signature provided a robust prognostic biomarker for LUAD and could serve as a predictor for immunotherapy response, which may be used as an important supplement to immunotherapy to achieve individualized tumor treatment by optimizing the prognostic management and immunotherapy for patients with LUAD.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.,Department of Oncology, Renmin Hospital of Wuhan University, 238th Jiefang Road, Wuhan, 430060, China
| | - Yu Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengchang Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyu Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shungeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Department of Oncology, Renmin Hospital of Wuhan University, 238th Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
48
|
Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol 2021; 18:842-859. [PMID: 33139907 PMCID: PMC8115290 DOI: 10.1038/s41423-020-00565-9] [Citation(s) in RCA: 544] [Impact Index Per Article: 136.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
The clinical success of cancer immune checkpoint blockade (ICB) has refocused attention on tumor-infiltrating lymphocytes (TILs) across cancer types. The outcome of immune checkpoint inhibitor therapy in cancer patients has been linked to the quality and magnitude of T cell, NK cell, and more recently, B cell responses within the tumor microenvironment. State-of-the-art single-cell analysis of TIL gene expression profiles and clonality has revealed a remarkable degree of cellular heterogeneity and distinct patterns of immune activation and exhaustion. Many of these states are conserved across tumor types, in line with the broad responses observed clinically. Despite this homology, not all cancer types with similar TIL landscapes respond similarly to immunotherapy, highlighting the complexity of the underlying tumor-immune interactions. This observation is further confounded by the strong prognostic benefit of TILs observed for tumor types that have so far respond poorly to immunotherapy. Thus, while a holistic view of lymphocyte infiltration and dysfunction on a single-cell level is emerging, the search for response and prognostic biomarkers is just beginning. Within this review, we discuss recent advances in the understanding of TIL biology, their prognostic benefit, and their predictive value for therapy.
Collapse
Affiliation(s)
- Sterre T Paijens
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annegé Vledder
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
49
|
Yamauchi T, Hoki T, Oba T, Jain V, Chen H, Attwood K, Battaglia S, George S, Chatta G, Puzanov I, Morrison C, Odunsi K, Segal BH, Dy GK, Ernstoff MS, Ito F. T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors. Nat Commun 2021; 12:1402. [PMID: 33658501 PMCID: PMC7930182 DOI: 10.1038/s41467-021-21619-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized treatment for various cancers; however, durable response is limited to only a subset of patients. Discovery of blood-based biomarkers that reflect dynamic change of the tumor microenvironment, and predict response to ICI, will markedly improve current treatment regimens. Here, we investigate CX3C chemokine receptor 1 (CX3CR1), a marker of T-cell differentiation, as a predictive correlate of response to ICI therapy. Successful treatment of tumor-bearing mice with ICI increases the frequency and T-cell receptor clonality of the peripheral CX3CR1+CD8+ T-cell subset that includes an enriched repertoire of tumor-specific and tumor-infiltrating CD8+ T cells. Furthermore, an increase in the frequency of the CX3CR1+ subset in circulating CD8+ T cells early after initiation of anti-PD-1 therapy correlates with response and survival in patients with non-small cell lung cancer. Collectively, these data support T-cell CX3CR1 expression as a blood-based dynamic early on-treatment predictor of response to ICI therapy.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Animals
- Antibodies, Monoclonal, Humanized/pharmacology
- Biomarkers, Pharmacological/blood
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/physiology
- CX3C Chemokine Receptor 1/blood
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/mortality
- Cell Line, Tumor
- Female
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Ki-67 Antigen/blood
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/mortality
- Lymphocytes, Tumor-Infiltrating/drug effects
- Male
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Middle Aged
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Nivolumab/pharmacology
- Receptors, Antigen, T-Cell/metabolism
- Survival Rate
- Treatment Outcome
- Mice
Collapse
Affiliation(s)
- Takayoshi Yamauchi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Toshifumi Hoki
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Merck Sharp & Dohme, Tokyo, Japan
| | - Takaaki Oba
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Vaibhav Jain
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Hongbin Chen
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
| | - Kristopher Attwood
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Saby George
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
| | - Gurkamal Chatta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Carl Morrison
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Brahm H Segal
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Grace K Dy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marc S Ernstoff
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Medicine, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
- Division of Cancer Treatment and Diagnosis, Developmental Therapeutics Program, National Cancer Institute, Bethesda, MD, USA
| | - Fumito Ito
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA.
| |
Collapse
|
50
|
Nakajima H, Maeno K, Ito T, Kanai T, Oba T, Ono M, Takayama F, Uehara T, Ito KI. Concomitant use of 18F-FDG PET-CT SUVmax is useful in the assessment of Ki67 labeling index in core-needle biopsy specimens of breast cancer. Gland Surg 2021; 10:1-9. [PMID: 33633957 DOI: 10.21037/gs-20-485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Ki67 is a recognized proliferative and predictive marker in invasive breast cancer. However, results of Ki67 evaluation are affected by the method employed for sample fixation or biopsy, as well as by intratumor heterogeneity. Here, we aimed to compare the Ki67 labeling index (Ki67LI) between core-needle biopsy specimens (CNBSs) and surgically resected specimens (SRSs) of invasive breast cancer, and verify whether the discordance in Ki67LI can be reduced by analyzing the maximum standardized uptake value (SUVmax) obtained from pretreatment whole-body positron emission tomography/computed tomography (PET/CT) in combination with Ki67LI. Methods Tumor tissues were obtained from 118 patients with invasive breast cancer. Ki67LI was evaluated in CNBSs and SRSs by immunohistochemistry. First, we directly compared Ki67LI between CNBS and SRS, "allowing a tolerance margin of 5%." We divided the Ki67LI values into three groups (Low: 0≤ Ki67LI ≤10, Intermediate: 10< Ki67LI <30, and High: 30≤ Ki67LI) and the SUVmax into three groups (SUVmax ≤4, 4< SUVmax <8, and 8≤ SUVmax). We then verified the concordance rate between CNBS and SRS in each group in combination with the SUVmax obtained by PET/CT. Results The median Ki67LI was 17.8% (0.5-75.9%) and 17.0% (1.0-75.7%) in CNBS and SRS, respectively. The overall Ki67LI concordance rate between CNBS and SRS was 37.3% (44/118). The concordance was improved in the Low and High Ki67LI groups by applying SUVmax thresholds of 4 [82.6% (19/23), P=0.033 and 8 (92.3% (12/13), P=0.009], respectively. Conclusions Our results indicated that CNBS Ki67LI alone was not able to reflect SRS Ki67LI with sufficient accuracy. By dividing CNBS Ki67LI into three classes in combination with SUVmax, tumor proliferation could be predicted with higher accuracy in patients with invasive breast carcinoma.
Collapse
Affiliation(s)
- Hiroki Nakajima
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Kazuma Maeno
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Tokiko Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Toshiharu Kanai
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Takaaki Oba
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | - Mayu Ono
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| | | | - Tsuyoshi Uehara
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Ken-Ichi Ito
- Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Japan
| |
Collapse
|