1
|
Fu Y, Wen Z, Fan J. Interaction of low-density neutrophils with other immune cells in the mechanism of inflammation. Mol Med 2025; 31:133. [PMID: 40205584 PMCID: PMC11983930 DOI: 10.1186/s10020-025-01187-5] [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: 07/27/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Low-density neutrophils (LDNs) are a unique subpopulation of neutrophils, play a significant role in regulating innate and adaptive immunity in various inflammation-related diseases. Emerging evidence suggests that LDNs play a significant role in the pathogenesis and progression of various diseases, including infections, autoimmune disorders, and cancer. In this review, we address the origin, development, and heterogeneity of LDNs, and the roles of LDNs in system homeostasis and diseases. We will focus on the findings of the interaction between LDNs and other immune cells. We will then discuss potential novel therapeutic strategies of intervention in diseases by targeting LDNs.
Collapse
Affiliation(s)
- Yu Fu
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, 15213, USA.
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Jie Fan
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, 15213, USA.
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, 15213, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
2
|
Gomatou G, Charpidou A, Li P, Syrigos N, Gkiozos I. Mechanisms of primary resistance to immune checkpoint inhibitors in NSCLC. Clin Transl Oncol 2025; 27:1426-1437. [PMID: 39307892 DOI: 10.1007/s12094-024-03731-x] [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: 05/24/2024] [Accepted: 09/10/2024] [Indexed: 04/16/2025]
Abstract
Immune checkpoint inhibitors (ICIs) redefined the therapeutics of non-small cell lung cancer (NSCLC), leading to significant survival benefits and unprecedented durable responses. However, the majority of the patients develop resistance to ICIs, either primary or acquired. Establishing a definition of primary resistance to ICIs in different clinical scenarios is challenging and remains a work in progress due to the changing landscape of ICI-based regimens, mainly in the setting of early-stage NSCLC. The mechanisms of primary resistance to ICIs in patients with NSCLC include a plethora of pathways involving a cross-talk of the tumor cells, the tumor microenvironment and the host, leading to the development of an immunosuppressive phenotype. The optimal management of patients with NSCLC following primary resistance to ICIs represents a significant challenge in current thoracic oncology. Research in this field includes exploring other immunotherapeutic approaches, such as cancer vaccines, and investigating novel antibody-drug conjugates in patients with NSCLC.
Collapse
Affiliation(s)
- Georgia Gomatou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andriani Charpidou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Nikolaos Syrigos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Gkiozos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
3
|
Yao J, Ji L, Wang G, Ding J. Effect of neutrophils on tumor immunity and immunotherapy resistance with underlying mechanisms. Cancer Commun (Lond) 2025; 45:15-42. [PMID: 39485719 PMCID: PMC11758154 DOI: 10.1002/cac2.12613] [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: 05/05/2024] [Revised: 09/08/2024] [Accepted: 09/17/2024] [Indexed: 11/03/2024] Open
Abstract
Neutrophils are key mediators of the immune response and play essential roles in the development of tumors and immune evasion. Emerging studies indicate that neutrophils also play a critical role in the immunotherapy resistance in cancer. In this review, firstly, we summarize the novel classification and phenotypes of neutrophils and describe the regulatory relationships between neutrophils and tumor metabolism, flora microecology, neuroendocrine and tumor therapy from a new perspective. Secondly, we review the mechanisms by which neutrophils affect drug resistance in tumor immunotherapy from the aspects of the immune microenvironment, tumor antigens, and epigenetics. Finally, we propose several promising strategies for overcoming tumor immunotherapy resistance by targeting neutrophils and provide new research ideas in this area.
Collapse
Affiliation(s)
- Jiali Yao
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| | - Linlin Ji
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| | - Guang Wang
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| | - Jin Ding
- Clinical Cancer InstituteCenter for Translational MedicineNaval Medical UniversityShanghaiChina
| |
Collapse
|
4
|
Huang S, Chung JYF, Li C, Wu Y, Qiao G, To KF, Tang PMK. Cellular dynamics of tumor microenvironment driving immunotherapy resistance in non-small-cell lung carcinoma. Cancer Lett 2024; 604:217272. [PMID: 39326553 DOI: 10.1016/j.canlet.2024.217272] [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: 07/06/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have profoundly reshaped the treatment paradigm for non-small cell lung cancer (NSCLC). Despite these advancements, primary and secondary resistance to ICIs remain prevalent challenges in managing advanced NSCLC. Recent studies have highlighted the significant role of the tumor microenvironment (TME) in modulating treatment responses. This review aims to comprehensively examine the interactive roles of immune/stromal cells-such as T cells, B cells, neutrophils, macrophages, and CAFs within the TME, elucidating how these diverse cellular interactions contribute to immunotherapy resistance. It focuses on the dynamic interactions among diverse cell types such as the varying states of T cells under the influence of TME constituents like immune cells and cancer-associated fibroblasts (CAFs). By exploring the mechanisms involved in the complex cellular interactions, we highlight novel therapeutic targets and strategies aimed at overcoming resistance, thereby enhancing the efficacy of ICIs in NSCLC. Our synthesis of recent research provides critical insights into the multifaceted mechanisms of resistance and paves the way for the development of more effective, personalized treatment approaches.
Collapse
Affiliation(s)
- Shujie Huang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jeff Yat-Fai Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chunjie Li
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Wu
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong; Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
5
|
Marcos Rubio Á, Oh S, Roelandt S, Stevens D, Van Damme E, Vermaelen K, De Preter K, Everaert C. Defining the optimal setting for transcriptomic analyses on blood samples for response prediction in immunotherapy-treated NSCLC patients. Sci Rep 2024; 14:26026. [PMID: 39472635 PMCID: PMC11522423 DOI: 10.1038/s41598-024-76982-x] [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: 05/21/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Transcriptomic profiling of blood immune cells offers a promising alternative to invasive, sampling bias-prone tissue-based biomarker assays for predicting immune checkpoint inhibitor (ICI) therapy response in non-small cell lung cancer (NSCLC) patients. However, the optimal analytical approach to identify systemic correlates of response still needs to be explored. We collected peripheral blood mononuclear cells and whole blood (WB) samples from 33 ICI-treated NSCLC patients before ICI treatment and at the first response evaluation. After bulk polyadenylated RNA-sequencing, we assessed differences in gene expression profiles between non-responders and responders using differential expression analysis, single sample gene set enrichment analysis (ssGSEA), and cell type deconvolution. We evaluated gene expression values, ssGSEA scores, and deconvolved cell type proportions to distinguish non-responders from responders via ROC curve (AUC) analysis, training a logistic regression classification model. Gene expression values and deconvolved proportions yielded the best results with WB samples after treatment (AUC = 0.87 and 0.85, respectively). Overall, ssGSEA scores showed superior classification performance across all sample types and timepoints (AUC > 0.7). In conclusion, transcriptomic analysis through ssGSEA demonstrated the best performance as a non-invasive biomarker for predicting clinical benefit in ICI-treated NSCLC patients, with gene expression and deconvolution on post-treatment WB samples also showing promising results.
Collapse
Affiliation(s)
- Álvaro Marcos Rubio
- Department of Biomolecular Medicine, VIB-UGent Center for Medical Biotechnology, Ghent University, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Seoyeon Oh
- Department of Biomolecular Medicine, VIB-UGent Center for Medical Biotechnology, Ghent University, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Sofie Roelandt
- Department of Biomolecular Medicine, VIB-UGent Center for Medical Biotechnology, Ghent University, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Dieter Stevens
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Pulmonary Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Ghent, Belgium
| | - Eufra Van Damme
- Department of Biomolecular Medicine, VIB-UGent Center for Medical Biotechnology, Ghent University, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Karim Vermaelen
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
- Department of Pulmonary Medicine and Immuno-Oncology Network Ghent, Ghent University Hospital, Ghent, Belgium
| | - Katleen De Preter
- Department of Biomolecular Medicine, VIB-UGent Center for Medical Biotechnology, Ghent University, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Celine Everaert
- Department of Biomolecular Medicine, VIB-UGent Center for Medical Biotechnology, Ghent University, Technologiepark-Zwijnaarde 75, 9052, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Medical Research Building 2 (MRB2) - UZ Gent - Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
6
|
Kim G, Kim EY, Lee H, Shin SH, Lee SH, Sohn KY, Kim JW, Lee JS. 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol treatment inhibits abnormal tumor growth by regulating neutrophil infiltration in a non-small cell lung carcinoma mouse model. Biomed Pharmacother 2024; 178:117269. [PMID: 39137654 DOI: 10.1016/j.biopha.2024.117269] [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: 05/07/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Excessive neutrophil infiltration into the tumor microenvironment (TME) is an important factor that contributes to tumor overgrowth and limited immunotherapy efficacy. Neutrophils activate various receptors involved in tumor progression, while suppressing the infiltration and activity of cytotoxic T cells and creating optimal conditions for tumor growth. Therefore, the appropriate control of neutrophil infiltration is an effective strategy for tumor treatment. In the present study, 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) inhibited tumor overgrowth by suppressing excessive neutrophil infiltration, resulting in >74.97 % reduction in tumor size in a Lewis lung carcinoma (LLC-1) mouse model. All subjects in the positive control group died during the 90-day survival period, whereas only four subjects in the PLAG treatment group survived. PLAG had a significantly higher tumor growth inhibitory effect and survival rate than other neutrophil infiltration-targeting inhibitors (e.g., Navarixin, lymphocyte antigen 6 complex locus G6D antibody [aLy6G]). The ability of PLAG to regulate neutrophil infiltration and inhibit tumor growth depends on thioredoxin-interacting protein (TXNIP). In tumors lacking TXNIP expression, PLAG failed to control neutrophil infiltration and infiltration-related factor release, and the inhibitory effect of PLAG on tumor growth was reduced. PLAG-mediated inhibition of neutrophil infiltration enhances the efficacy of immune checkpoint inhibitors (ICIs), increasing the antitumor efficacy and survival rate by 30 %. In conclusion, PLAG could be a novel alternative to anti-tumor drugs that effectively targets excessive neutrophil infiltration into cancer tissues.
Collapse
Affiliation(s)
- Guentae Kim
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea; Biotoxtech, 53 Yeongudanji-ro, Ochang-eup, Cheongju-si, South Korea
| | - Eun Young Kim
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Hyowon Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Su-Hyun Shin
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Se Hee Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Ki-Young Sohn
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea
| | - Jae Wha Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Kwahak-ro, Daejeon, South Korea
| | - Jae Sam Lee
- Enzychem Lifesciences, 14F aT Center 27 Gangnam-daero, Seoul, South Korea.
| |
Collapse
|
7
|
He C, Guo Y, Zhou N, Wang Z, Liu T, Xu X, Wang F, Zhu H, Yang Z, Yang X, Xia L. Construction and Application of a PD-L1-Targeted Multimodal Diagnostic and Dual-Functional Theranostics Nanoprobe. Int J Nanomedicine 2024; 19:5479-5492. [PMID: 38863646 PMCID: PMC11166151 DOI: 10.2147/ijn.s461701] [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: 01/28/2024] [Accepted: 05/29/2024] [Indexed: 06/13/2024] Open
Abstract
Background In recent years, PD-L1 has been primarily utilized as an immune checkpoint marker in cancer immunotherapy. However, due to tumor heterogeneity, the response rate to such therapies often falls short of expectations. In addition to its role in immunotherapy, PD-L1 serves as a specific target on the surface of tumor cells for targeted diagnostic and therapeutic interventions. There is an absence of a fully developed PD-L1-targeted diagnostic and therapeutic probe for clinical use, which constrains the exploration and clinical exploitation of this target. Methods and Results In this study, we engineered a PD-L1-targeted probe with multimodal imaging and dual therapeutic functionalities utilizing organic melanin nanoparticles. Functionalization with the WL12-SH peptide endowed the nanoprobe with specific targeting capabilities. Subsequent radiolabeling with 89Zr (half-life: 100.8 hours) and chelation of Mn2+ ions afforded the probe the capacity for simultaneous PET and MRI imaging modalities. Cellular uptake assays revealed pronounced specificity, with -positive cells exhibiting significantly higher uptake than -negative counterparts (p < 0.05). Dual-modal PET/MRI imaging delineated rapid and sustained accumulation at the neoplastic site, yielding tumor-to-non-tumor (T/NT) signal ratios at 24 hours post-injection of 16.67±3.45 for PET and 6.63±0.64 for MRI, respectively. We conjugated the therapeutic radionuclide 131I (half-life: 8.02 days) to the construct and combined low-dose radiotherapy and photothermal treatment (PTT), culminating in superior antitumor efficacy while preserving a high safety profile. The tumors in the cohort receiving the dual-modality therapy exhibited significantly reduced volume and weight compared to those in the control and monotherapy groups. Conclusion We developed and applied a novel -targeted multimodal theranostic nanoprobe, characterized by its high specificity and superior imaging capabilities as demonstrated in PET/MRI modalities. Furthermore, this nanoprobe facilitates potent therapeutic efficacy at lower radionuclide doses when used in conjunction with PTT.
Collapse
Affiliation(s)
- Chengxue He
- Medical College, Guizhou University, Guiyang, GuiZhou Province, People’s Republic of China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - YanHui Guo
- Department of Radiology, Peking University Third Hospital, Beijing, People’s Republic of China
| | - Nina Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Zhen Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Department of Hepato-Pancreato-Biliary Surgery, Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Teli Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Xiaoxia Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Feng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Zhi Yang
- Medical College, Guizhou University, Guiyang, GuiZhou Province, People’s Republic of China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| | - Xianteng Yang
- Medical College, Guizhou University, Guiyang, GuiZhou Province, People’s Republic of China
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, GuiZhou Province, People’s Republic of China
| | - Lei Xia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Berland L, Gabr Z, Chang M, Ilié M, Hofman V, Rignol G, Ghiringhelli F, Mograbi B, Rashidian M, Hofman P. Further knowledge and developments in resistance mechanisms to immune checkpoint inhibitors. Front Immunol 2024; 15:1384121. [PMID: 38903504 PMCID: PMC11188684 DOI: 10.3389/fimmu.2024.1384121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
The past decade has witnessed a revolution in cancer treatment, shifting from conventional drugs (chemotherapies) towards targeted molecular therapies and immune-based therapies, in particular immune-checkpoint inhibitors (ICIs). These immunotherapies release the host's immune system against the tumor and have shown unprecedented durable remission for patients with cancers that were thought incurable, such as metastatic melanoma, metastatic renal cell carcinoma (RCC), microsatellite instability (MSI) high colorectal cancer and late stages of non-small cell lung cancer (NSCLC). However, about 80% of the patients fail to respond to these immunotherapies and are therefore left with other less effective and potentially toxic treatments. Identifying and understanding the mechanisms that enable cancerous cells to adapt to and eventually overcome therapy can help circumvent resistance and improve treatment. In this review, we describe the recent discoveries on the onco-immunological processes which govern the tumor microenvironment and their impact on the resistance to PD-1/PD-L1 checkpoint blockade.
Collapse
Affiliation(s)
- Léa Berland
- Inserm U1081 Institute for Research on Cancer and Aging, Nice (IRCAN) Team 4, Université Côte d’Azur, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Nice, France
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Zeina Gabr
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
- School of Life Science, Ecole Polytechnique Federale de Lausanne (EPFL), Lausanne, Switzerland
| | - Michelle Chang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Marius Ilié
- Inserm U1081 Institute for Research on Cancer and Aging, Nice (IRCAN) Team 4, Université Côte d’Azur, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Nice, France
- Laboratory of Clinical and Experimental Pathology, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Pasteur Hospital, Université Côte d’Azur, Nice, France
- Institut Hospitalo Universitaire (IHU) RespirERA, Nice, France
- Hospital-Integrated Biobank (BB-0033–00025), Pasteur Hospital, Nice, France
| | - Véronique Hofman
- Inserm U1081 Institute for Research on Cancer and Aging, Nice (IRCAN) Team 4, Université Côte d’Azur, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Nice, France
- Laboratory of Clinical and Experimental Pathology, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Pasteur Hospital, Université Côte d’Azur, Nice, France
- Institut Hospitalo Universitaire (IHU) RespirERA, Nice, France
- Hospital-Integrated Biobank (BB-0033–00025), Pasteur Hospital, Nice, France
| | - Guylène Rignol
- Inserm U1081 Institute for Research on Cancer and Aging, Nice (IRCAN) Team 4, Université Côte d’Azur, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Nice, France
- Laboratory of Clinical and Experimental Pathology, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Pasteur Hospital, Université Côte d’Azur, Nice, France
- Institut Hospitalo Universitaire (IHU) RespirERA, Nice, France
| | - François Ghiringhelli
- Institut Hospitalo Universitaire (IHU) RespirERA, Nice, France
- Department of Biology and Pathology of Tumors, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France
| | - Baharia Mograbi
- Inserm U1081 Institute for Research on Cancer and Aging, Nice (IRCAN) Team 4, Université Côte d’Azur, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Nice, France
- Institut Hospitalo Universitaire (IHU) RespirERA, Nice, France
| | - Mohamad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Paul Hofman
- Inserm U1081 Institute for Research on Cancer and Aging, Nice (IRCAN) Team 4, Université Côte d’Azur, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Nice, France
- Laboratory of Clinical and Experimental Pathology, Institut Hospitalo Universitaire (IHU) RespirERA, Federation Hospitalo Universitaire (FHU) OncoAge, Pasteur Hospital, Université Côte d’Azur, Nice, France
- Institut Hospitalo Universitaire (IHU) RespirERA, Nice, France
- Hospital-Integrated Biobank (BB-0033–00025), Pasteur Hospital, Nice, France
| |
Collapse
|
9
|
Yang F, Hua Q, Zhu X, Xu P. Surgical stress induced tumor immune suppressive environment. Carcinogenesis 2024; 45:185-198. [PMID: 38366618 DOI: 10.1093/carcin/bgae012] [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: 08/16/2023] [Revised: 01/25/2024] [Accepted: 02/14/2024] [Indexed: 02/18/2024] Open
Abstract
Despite significant advances in cancer treatment over the decades, surgical resection remains a prominent management approach for solid neoplasms. Unfortunately, accumulating evidence suggests that surgical stress caused by tumor resection may potentially trigger postoperative metastatic niche formation. Surgical stress not only activates the sympathetic-adrenomedullary axis and hypothalamic-pituitary-adrenocortical axis but also induces hypoxia and hypercoagulable state. These adverse factors can negatively impact the immune system by downregulating immune effector cells and upregulating immune suppressor cells, which contribute to the colonization and progression of postoperative tumor metastatic niche. This review summarizes the effects of surgical stress on four types of immune effector cells (neutrophils, macrophages, natural killer cells and cytotoxic T lymphocytes) and two types of immunosuppressive cells (regulatory T cells and myeloid-derived suppressor cells), and discusses the immune mechanisms of postoperative tumor relapse and progression. Additionally, relevant therapeutic strategies to minimize the pro-tumorigenic effects of surgical stress are elucidated.
Collapse
Affiliation(s)
- Fan Yang
- Department of Anesthesiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Research Center for Neuro-Oncology Interaction, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qing Hua
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaoyan Zhu
- Department of Physiology, Navy Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Pingbo Xu
- Department of Anesthesiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Research Center for Neuro-Oncology Interaction, Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
10
|
Tsai YT, Schlom J, Donahue RN. Blood-based biomarkers in patients with non-small cell lung cancer treated with immune checkpoint blockade. J Exp Clin Cancer Res 2024; 43:82. [PMID: 38493133 PMCID: PMC10944611 DOI: 10.1186/s13046-024-02969-1] [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: 12/05/2023] [Accepted: 01/30/2024] [Indexed: 03/18/2024] Open
Abstract
The paradigm of non-small cell lung cancer (NSCLC) treatment has been profoundly influenced by the development of immune checkpoint inhibitors (ICI), but the range of clinical responses observed among patients poses significant challenges. To date, analyses of tumor biopsies are the only parameter used to guide prognosis to ICI therapy. Tumor biopsies, however, are often difficult to obtain and tissue-based biomarkers are limited by intratumoral heterogeneity and temporal variability. In response, there has been a growing emphasis on the development of "liquid biopsy"‒ derived biomarkers, which offer a minimally invasive means to dynamically monitor the immune status of NSCLC patients either before and/or during the course of treatment. Here we review studies in which multiple blood-based biomarkers encompassing circulating soluble analytes, immune cell subsets, circulating tumor DNA, blood-based tumor mutational burden, and circulating tumor cells have shown promising associations with the clinical response of NSCLC patients to ICI therapy. These investigations have unveiled compelling correlations between the peripheral immune status of patients both before and during ICI therapy and patient outcomes, which include response rates, progression-free survival, and overall survival. There is need for rigorous validation and standardization of these blood-based assays for broader clinical application. Integration of multiple blood-based biomarkers into comprehensive panels or algorithms also has the potential to enhance predictive accuracy. Further research aimed at longitudinal monitoring of circulating biomarkers is also crucial to comprehend immune dynamics and resistance mechanisms and should be used alongside tissue-based methods that interrogate the tumor microenvironment to guide treatment decisions and may inform on the development of novel therapeutic strategies. The data reviewed here reinforce the opportunity to refine patient stratification, optimize treatments, and improve outcomes not only in NSCLC but also in the wider spectrum of solid tumors undergoing immunotherapy.
Collapse
Affiliation(s)
- Yo-Ting Tsai
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Arrazubi V, Goñi S, González-Borja I, Hernandez-Garcia I, Arasanz H, Pérez-Sanz J, Bocanegra AI, Kochan G, Escors D, Ruiz de Azúa Y, Elizalde JM, Viúdez A, Vera R. Circulating low density neutrophils are associated with resistance to anti-PD1 immunotherapy in squamous head and neck cancer. Head Neck 2023; 45:3075-3085. [PMID: 37792371 DOI: 10.1002/hed.27536] [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: 04/11/2023] [Revised: 07/21/2023] [Accepted: 09/22/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND Identification of predictive biomarkers to Immune checkpoint inhibitors (ICIs) in head and neck cancer (HNSCC) is an unmet need. METHODS This was a prospective observational study including 25 patients with HNSCC treated with immunotherapy or chemotherapy after a prior platinum-based regimen. Low density neutrophils (LDNs) and serum markers were analyzed. RESULTS In the immunotherapy cohort, patients with high LDN levels had a shorter progression free survival (PFS) (1.8 months vs. 10.9 months; *p = 0.034). Also, progressors showed higher percentage of LDNs compared to non-progressors although significance was not reached (mean 20.68% vs. 4.095%, p = 0.0875). These findings were not replicated in patients treated with chemotherapy. High levels of interleukin-7 (IL7) were correlated with a significantly longer overall survival (OS) (13.47 months 3.51 vs. months, *p = 0.013). CONCLUSIONS High baseline circulating LDNs and low IL7 could identify a subset of patients intrinsically refractory to ICIs as monotherapy in HNSCC.
Collapse
Affiliation(s)
- Virginia Arrazubi
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Saioa Goñi
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Iranzu González-Borja
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Irene Hernandez-Garcia
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Hugo Arasanz
- Medical Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Jairo Pérez-Sanz
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Ana Isabel Bocanegra
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Grazyna Kochan
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | | | - Antonio Viúdez
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Ruth Vera
- Oncobiona Group, Navarrabiomed, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
| |
Collapse
|
12
|
Moreira RS, da Silva MM, de Melo Vasconcelos CF, da Silva TD, Cordeiro GG, Mattos-Jr LAR, da Rocha Pitta MG, de Melo Rêgo MJB, Pereira MC. Siglec 15 as a biomarker or a druggable molecule for non-small cell lung cancer. J Cancer Res Clin Oncol 2023; 149:17651-17661. [PMID: 37843557 DOI: 10.1007/s00432-023-05437-z] [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: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
Lung cancer has been the main cause of cancer mortality worldwide. Furthermore, lung cancer rates of new cases per year evidenced a large incidence of this neoplasm in both men and women. Because there is no biomarker for early detection, it is frequently detected late, at an advanced state. The introduction of multiple lines of tyrosine kinase inhibitors in patients with EGFR, ALK, ROS1, and NTRK mutations has modified the therapy of lung cancer. Immunotherapy advances have resulted in substantial improvements in overall survival and disease-free survival, making immune checkpoint inhibitors (ICIs) a potential option for lung cancer treatment. Current PD-1/PD-L1/CTLA-4 immunotherapies have resulted in important response and survival rates. However, existing medicines only function in around 20% of unselected, advanced NSCLC patients, and primary and acquired resistance remain unsolved obstacles. Therefore, precise predictive indicators must be identified to choose the best patients for ICI treatment. Thus, Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) stands out as a potential tumor biomarker, with distinctive expression in normal tissues, in tumor immune involvement, and a high structural similarity to PD-L1. Understanding the tumor immune response and the search for new therapeutic targets leads to the improvement of therapeutic pathways directed at the tumor microenvironment. The present review aims to analyze Siglec-15 potential as a diagnostic, prognostic, and response biomarker in lung cancer, considering its results evidenced in the current literature.
Collapse
Affiliation(s)
- Rodrigo Santiago Moreira
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Marillya Morais da Silva
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Thiago Douberin da Silva
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Luiz Alberto Reis Mattos-Jr
- Department of Clinic Medicine, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Recife, PE, Brazil
| | - Maira Galdino da Rocha Pitta
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Michelly Cristiny Pereira
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
13
|
Liu L, Chen G, Gong S, Huang R, Fan C. Targeting tumor-associated macrophage: an adjuvant strategy for lung cancer therapy. Front Immunol 2023; 14:1274547. [PMID: 38022518 PMCID: PMC10679371 DOI: 10.3389/fimmu.2023.1274547] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The emergence of immunotherapy has revolutionized the treatment landscape for various types of cancer. Nevertheless, lung cancer remains one of the leading causes of cancer-related mortality worldwide due to the development of resistance in most patients. As one of the most abundant groups of immune cells in the tumor microenvironment (TME), tumor-associated macrophages (TAMs) play crucial and complex roles in the development of lung cancer, including the regulation of immunosuppressive TME remodeling, metabolic reprogramming, neoangiogenesis, metastasis, and promotion of tumoral neurogenesis. Hence, relevant strategies for lung cancer therapy, such as inhibition of macrophage recruitment, TAM reprograming, depletion of TAMs, and engineering of TAMs for drug delivery, have been developed. Based on the satisfactory treatment effect of TAM-targeted therapy, recent studies also investigated its synergistic effect with current therapies for lung cancer, including immunotherapy, radiotherapy, chemotherapy, anti-epidermal growth factor receptor (anti-EGFR) treatment, or photodynamic therapy. Thus, in this article, we summarized the key mechanisms of TAMs contributing to lung cancer progression and elaborated on the novel therapeutic strategies against TAMs. We also discussed the therapeutic potential of TAM targeting as adjuvant therapy in the current treatment of lung cancer, particularly highlighting the TAM-centered strategies for improving the efficacy of anti-programmed cell death-1/programmed cell death-ligand 1 (anti-PD-1/PD-L1) treatment.
Collapse
Affiliation(s)
| | | | | | | | - Chunmei Fan
- *Correspondence: Chunmei Fan, ; Rongfu Huang,
| |
Collapse
|
14
|
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
|
15
|
Jain S, Ma K, Morris LGT. CD66b as a prognostic and predictive biomarker in patients with non-small cell lung cancer treated with checkpoint blockade immunotherapy. Transl Cancer Res 2023; 12:447-451. [PMID: 36915573 PMCID: PMC10007870 DOI: 10.21037/tcr-22-2880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 02/24/2023]
Affiliation(s)
- Swati Jain
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin Ma
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc G T Morris
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
16
|
Machiraju D, Hassel JC. Targeting the cMET pathway to enhance immunotherapeutic approaches for mUM patients. Front Oncol 2023; 12:1068029. [PMID: 36761417 PMCID: PMC9902905 DOI: 10.3389/fonc.2022.1068029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
The liver is the most preferential initial site of metastasis for uveal melanoma (mUM), and this preference is associated with rapid mortality in mUM patients. Despite the significant clinical benefits of Immune checkpoint inhibitors (ICIs) in metastatic cutaneous melanoma patients, ICIs have shown little to no benefit in mUM patients. A potential reason for this inefficiency of ICI could be partly devoted to the involvement of the liver itself, thanks to its rich source of growth factors and immunosuppressive microenvironment. Uveal melanoma cells show increased expression of a transmembrane protein called cMET, which is known as the sole receptor for the Hepatocyte growth factor (HGF). Hyperactivation of cMET by HGF contributes to mUM development, and the liver, being the major source of HGF, may partially explain the metastasis of uveal melanoma cells to the liver. In addition, cMET/HGF signaling has also been shown to mediate resistance to ICI treatment, directly and indirectly, involving tumor and immune cell populations. Therefore, targeting the cMET/HGF interaction may enhance the efficacy of immunotherapeutic regimes for mUM patients. Hence in this minireview, we will discuss the rationale for combining cMET inhibitors/antibodies with leading immune checkpoint inhibitors for treating mUM. We will also briefly highlight the challenges and opportunities in targeting cMET in mUM.
Collapse
|
17
|
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
|
18
|
Echaide M, Labiano I, Delgado M, Fernández de Lascoiti A, Ochoa P, Garnica M, Ramos P, Chocarro L, Fernández L, Arasanz H, Bocanegra A, Blanco E, Piñeiro-Hermida S, Morente P, Vera R, Alsina M, Escors D, Kochan G. Immune Profiling Uncovers Memory T-Cell Responses with a Th17 Signature in Cancer Patients with Previous SARS CoV-2 Infection Followed by mRNA Vaccination. Cancers (Basel) 2022; 14:cancers14184464. [PMID: 36139625 PMCID: PMC9496802 DOI: 10.3390/cancers14184464] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/28/2022] Open
Abstract
Simple Summary Cancer patients are considered a high-risk group for infectious diseases including COVID-19. The protective effects of vaccination are unclear in oncologic patients, as well as their duration. In this study antibody, T-cell and myeloid cell immunity were evaluated in three cohorts of healthy donors and oncologic patients, including those infected with SARS-CoV-2, BNT162b2-vaccinated (mRNA vaccine), and with previous COVID-19 and subsequently vaccinated. We concluded that vaccination was a poor inductor of cellular immunity towards the S protein. Memory T-cells were only detected in patients and healthy donors with COVID-19 previous to vaccination but with an accentuated Th17 inflammatory profile, together with elevated numbers of circulating neutrophils. Abstract It is unclear whether patients with cancer present inherently impaired responses to COVID-19 and vaccination due to their treatments, neoplastic diseases or both. To address this question, immune profiling was performed in three cohorts of healthy donors and oncologic patients: infected with SARS-CoV-2, BNT162b2-vaccinated, and with previous COVID-19 disease and subsequently vaccinated. Cancer patients showed good antibody responses to vaccination, but poor induction of T-cell responses towards the S protein when compared to infection. Following natural infection, the major targets for T-cells were the SARS-CoV-2 structural proteins M and S, but not the N protein. Similar to antibody titers, the T-cell responses quickly decayed after six months post-vaccination. Significant memory T-cell expansion was observed in vaccinated donors only if previously diagnosed with COVID-19 before undergoing vaccination. Oncologic patients with previous COVID-19 followed by vaccination exhibited potent IL-17+ CD4 and CD8 T-cell responses and elevated numbers of circulating neutrophils in peripheral blood.
Collapse
Affiliation(s)
- Miriam Echaide
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Ibone Labiano
- Oncobiona Group-Navarrabiomed-UPNA-IdiSNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Marina Delgado
- Oncobiona Group-Navarrabiomed-UPNA-IdiSNA, Irunlarrea 3, 31008 Pamplona, Spain
- Department of Medical Oncology, Hospital Universitario de Navarra-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Angela Fernández de Lascoiti
- Oncobiona Group-Navarrabiomed-UPNA-IdiSNA, Irunlarrea 3, 31008 Pamplona, Spain
- Department of Medical Oncology, Hospital Universitario de Navarra-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Patricia Ochoa
- Department of Medical Oncology, Hospital Universitario de Navarra-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Maider Garnica
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Pablo Ramos
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Luisa Chocarro
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Leticia Fernández
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Hugo Arasanz
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
- Department of Medical Oncology, Hospital Universitario de Navarra-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Ana Bocanegra
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Ester Blanco
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Sergio Piñeiro-Hermida
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Pilar Morente
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Ruth Vera
- Oncobiona Group-Navarrabiomed-UPNA-IdiSNA, Irunlarrea 3, 31008 Pamplona, Spain
- Department of Medical Oncology, Hospital Universitario de Navarra-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - Maria Alsina
- Oncobiona Group-Navarrabiomed-UPNA-IdiSNA, Irunlarrea 3, 31008 Pamplona, Spain
- Department of Medical Oncology, Hospital Universitario de Navarra-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
| | - David Escors
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
- Correspondence: (D.E.); (G.K.)
| | - Grazyna Kochan
- Oncoimmunology Group, Navarrabiomed, Fundación Miguel Servet-Hospital Universitario de Navarra-UPNA-IdISNA, Irunlarrea 3, 31008 Pamplona, Spain
- Correspondence: (D.E.); (G.K.)
| |
Collapse
|