1
|
Dong D, Yu X, Liu H, Xu J, Guo J, Guo W, Li X, Wang F, Zhang D, Liu K, Sun Y. Study of immunosenescence in the occurrence and immunotherapy of gastrointestinal malignancies. Semin Cancer Biol 2025; 111:16-35. [PMID: 39929408 DOI: 10.1016/j.semcancer.2025.01.007] [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: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 02/25/2025]
Abstract
In human beings heterogenous, pervasive and lethal malignancies of different parts of the gastrointestinal (GI) tract viz., tumours of the oesophagus, stomach, small intestine, colon, and rectum, represent gastrointestinal malignancies. Primary treatment modality for gastric cancer includes chemotherapy, surgical interventions, radiotherapy, monoclonal antibodies and inhibitors of angiogenesis. However, there is a need to improve upon the existing treatment modality due to associated adverse events and the development of resistance towards treatment. Additionally, age has been found to contribute to increasing the incidence of tumours due to immunosenescence-associated immunosuppression. Immunosenescence is the natural process of ageing, wherein immune cells as well as organs begin to deteriorate resulting in a dysfunctional or malfunctioning immune system. Accretion of senescent cells in immunosenescence results in the creation of a persistent inflammatory environment or inflammaging, marked with elevated expression of pro-inflammatory and immunosuppressive cytokines and chemokines. Perturbation in the T-cell pools and persistent stimulation by the antigens facilitate premature senility of the immune cells, and senile immune cells exacerbate inflammaging conditions and the inefficiency of the immune system to identify the tumour antigen. Collectively, these conditions contribute positively towards tumour generation, growth and eventually proliferation. Thus, activating the immune cells to distinguish the tumour cells from normal cells and invade them seems to be a logical strategy for the treatment of cancer. Consequently, various approaches to immunotherapy, viz., programmed death ligand-1 (PD-1) inhibitors, Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors etc are being extensively evaluated for their efficiency in gastric cancer. In fact, PD-1 inhibitors have been sanctioned as late late-line therapy modality for gastric cancer. The present review will focus on deciphering the link between the immune system and gastric cancer, and the alterations in the immune system that incur during the development of gastrointestinal malignancies. Also, the mechanism of evasion by tumour cells and immune checkpoints involved along with different approaches of immunotherapy being evaluated in different clinical trials will be discussed.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Haoran Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Jiayan Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Fei Wang
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Dongyong Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Kaiwei Liu
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
2
|
Lu Z, Lyu Z, Dong P, Liu Y, Huang L. N6-methyladenosine RNA modification in stomach carcinoma: Novel insights into mechanisms and implications for diagnosis and treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167793. [PMID: 40088577 DOI: 10.1016/j.bbadis.2025.167793] [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: 11/21/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
N6-methyladenosine (m6A) RNA methylation is crucially involved in the genesis and advancement of gastric cancer (GC) by controlling various pathobiological aspects including gene expression, signal transduction, metabolism, cell death, epithelial-mesenchymal transition, angiogenesis, and exosome function. Despite its importance, the exact mechanisms by which m6A modification influences GC biology remain inadequately explored. This review consolidates the latest advances in uncovering the mechanisms and diverse roles of m6A in GC and proposes new research and translational directions. Key regulators (writers, readers, and erasers) of m6A, such as METTL3/14/16 and WTAP, significantly affect cancer progression, anticancer immune response, and treatment outcomes. m6A modification also impacts immune cell infiltration and the tumor microenvironment, highlighting its potential as a diagnostic and prognostic marker. Interactions between m6A methylation and non-coding RNAs offer further novel insights into GC development and therapeutic targets. Targeting m6A regulators could enhance immunotherapy response, overcome treatment resistance, and improve oncological and clinical outcomes. Models based on m6A can precisely predict treatment response and prognosis in GC. Additional investigation is needed to fully understand the mechanisms of m6A methylation and its potential clinical applications and relevance (e.g., as precise markers for early detection, prediction of outcome, and response to therapy and as therapeutic targets) in GC. Future research should focus on in vivo studies, potential clinical trials, and the examination of m6A modification in other types of cancers.
Collapse
Affiliation(s)
- Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Zhaojie Lyu
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, Shanghai Institute of Pancreatic Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China; National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
3
|
Lara B, Snyder M, Fimbres J, Yang E, Song G, Duggineni VK, Wang Z, Sherr DH. The AhR regulates IFN-induced immune checkpoints in lung cancer cells through HNRNPH1, an RNA-binding protein, and INCR1, a novel long non-coding RNA. J Biol Chem 2025:110316. [PMID: 40449595 DOI: 10.1016/j.jbc.2025.110316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/28/2025] [Accepted: 05/17/2025] [Indexed: 06/03/2025] Open
Abstract
Although immune checkpoint inhibitors show great promise, not all patients respond and many do not achieve durable responses. Consequently, further investigations into potentially targetable molecules that regulate immune checkpoints are warranted. Previous studies in several cancers demonstrated that interferons produced by tumor-infiltrating leukocytes regulate immunosuppressive PD-L1, PD-L2 and IDO1 through JAK/STAT signaling. Here, we investigated a novel role for an immunosuppressive environmental chemical receptor, previously implicated in smoking-related cancers, in IFN signaling in human lung adenocarcinoma (LUAD) cells. Deletion of the aryl hydrocarbon receptor (AhR) from A549 LUAD cells significantly decreased baseline JAK2, STAT1, STAT3, IRF1 (a JAK/STAT target), PD-L1, PD-L2, and IDO1 expression. IFNγ and IFNα increased expression of JAK/STAT and immune checkpoint genes and proteins, but these increases were significantly diminished or absent in AhR-knockout cells. The AhR similarly controls IFN-induced, JAK/STAT-driven increases in multiple MHC class I- and class II-related genes. AhR control of type I and type II interferon signaling is mediated through up-regulation of a lncRNA, the IFN-stimulated non-coding RNA 1 (INCR1), and through repression of an RNA-binding protein, heterogeneous nuclear ribonucleoprotein H1 (HNRNPH1), which sequesters JAK/STAT-related and immune checkpoint gene transcripts. The data suggest that the AhR is a key mediator of tumor immunosuppression through regulation of IFN-induced INCR1 and JAK/STAT signaling and, thereby, expression of immune checkpoints. However, that immunosuppression may be tempered by AhR control of MHC expression. Given the multiple roles of JAK/STAT signaling in the immune system, the results also suggest multiple levels on which the AhR may affect tumor immunity.
Collapse
Affiliation(s)
- Brian Lara
- Department of Environmental Health, Boston University School of Public Health
| | | | - Jocelyn Fimbres
- Department of Environmental Health, Boston University School of Public Health
| | - Eric Yang
- Department of Environmental Health, Boston University School of Public Health
| | - Gang Song
- Department of Environmental Health, Boston University School of Public Health
| | | | - Zhongyan Wang
- Department of Environmental Health, Boston University School of Public Health
| | - David H Sherr
- Department of Environmental Health, Boston University School of Public Health.
| |
Collapse
|
4
|
Kim YS. Gastric Carcinoma. Curr Top Microbiol Immunol 2025. [PMID: 40423781 DOI: 10.1007/82_2025_303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Epstein-Barr virus (EBV)-associated gastric cancers (EBVaGCs) account for about 10% of gastric cancers globally, with higher prevalence in East Asia and Latin America. These cancers develop through a "gastritis-infection-cancer sequence" and are characterized by unique molecular signatures, including CpG island methylator phenotype and mutations in ARID1A and PIK3CA genes. EBVaGCs typically present in the proximal stomach with diffuse-type histology and dense lymphocytic infiltration. Key viral proteins EBNA1 and LMP2A drive oncogenesis by altering cellular processes and immune responses. The IFN-γ signature and extensive epigenetic modifications contribute to their distinct profile. Despite often presenting at advanced stages, EBVaGCs generally have a more favorable prognosis. EBV employs sophisticated strategies to evade immune detection, utilizing latent proteins and noncoding RNAs. Paradoxically, despite an immune-hot environment, EBVaGCs demonstrate effective immune evasion, partly due to the expression of immune checkpoint molecules like PD-L1 and LAG3. Treatment approaches vary based on disease stage, from endoscopic resection for early-stage cancers to systemic therapies for advanced cases. Immunotherapy, particularly PD-1/PD-L1 inhibitors, shows promising results. Emerging research suggests combining these with LAG3 inhibitors may enhance efficacy. Ongoing research and advanced genomic techniques continue to reveal new insights, paving the way for personalized therapies and novel diagnostic approaches.
Collapse
Affiliation(s)
- Young-Sik Kim
- Department of Pathology, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Sun X, Tong J, Fang X, Lu M, Rao C, Li Y. Comprehensive Multi-Omics Analysis of Copper Metabolism Related Molecular Subtypes and Prognostic Risk Stratification in Colon Adenocarcinoma. J Cell Mol Med 2025; 29:e70591. [PMID: 40391581 PMCID: PMC12089994 DOI: 10.1111/jcmm.70591] [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: 02/06/2025] [Revised: 04/21/2025] [Accepted: 04/26/2025] [Indexed: 05/22/2025] Open
Abstract
Colon adenocarcinoma (COAD) is the most common subtype of colorectal cancer, originating from glandular cells in the colon. Despite diagnostic and therapeutic advances, its prognosis remains poor. Copper, an essential micronutrient, is involved in tumorigenesis and other biological processes. In this study, we identified copper metabolism-related genes (CMRG) associated with COAD prognosis from TCGA and GEO databases and constructed a CMRG-based risk model. We assessed its clinical relevance through analyses of immune infiltration, immunotherapy response, and drug sensitivity. Single-cell sequencing revealed the spatial and cellular distribution of CMRG in COAD tissues, providing insight into their roles in the tumour microenvironment. COX19 was selected for further validation, and in vitro experiments (western blot, PCR, siRNA, colony formation, and Transwell assays) confirmed its role in promoting COAD cell invasion and proliferation. These findings highlight the involvement of copper metabolism in COAD progression and suggest potential targets for therapy.
Collapse
Affiliation(s)
- Xi Sun
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Jingfei Tong
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Xiaojie Fang
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Miaojiong Lu
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Chunhui Rao
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| | - Yanyan Li
- Department of Anorectal SurgeryHangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
6
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
7
|
Nihira NT, Kudo R, Ohta T. Inflammation and tumor immune escape in response to DNA damage. Semin Cancer Biol 2025; 110:36-45. [PMID: 39938581 DOI: 10.1016/j.semcancer.2025.02.005] [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: 07/30/2024] [Revised: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Senescent and cancer cells share common inflammatory characteristics, including factors of the senescence-associated secretory phenotype (SASP) and the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Inflammation in the tumor microenvironment not only provides an opportunity for immune cells to attack cancer cells, but also promotes cancer invasion and metastasis. Immune checkpoint molecule PD-L1 is transcriptionally induced by inflammation, and the immunological state of PD-L1-positive tumors influences the efficacy of Immune checkpoint inhibitors (ICIs). ICIs are effective against the PD-L1-positive "hot" tumors; however, the non-immunoactive "cold" tumors that express PD-L1 rarely respond to ICIs, suggesting that converting PD-L1-positive "cold" tumors into "hot" tumors would improve the efficacy of ICIs. To eliminate cancer via the innate immune system, a therapeutic strategy for manipulating inflammatory responses must be established. To date, the molecular mechanisms of inflammation-induced tumorigenesis are not yet fully understood. However, it is becoming clear that the regulatory mechanisms of inflammation in cancer via the cGAS-STING pathway play an important role in both cancer and sensescent cells. In this review, we focus on inflammation and immune escape triggered by DNA damage in cancer and senescent cells.
Collapse
Affiliation(s)
- Naoe Taira Nihira
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Rei Kudo
- Division of Cancer RNA Research, National Cancer Center Research Institute, Tokyo, Japan
| | - Tomohiko Ohta
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, Japan.
| |
Collapse
|
8
|
Lee SW, Yun JS, Kim YJ, Jeong S, Noh JE, Kim HO, Cho HJ, Park CK, Oh IJ, Cho JH. Progressive accumulation of circulating CD27 -CD28 - effector/memory CD8 + T cells in patients with lung cancer blunts responses to immune checkpoint inhibitor therapy. Exp Mol Med 2025:10.1038/s12276-025-01448-7. [PMID: 40307573 DOI: 10.1038/s12276-025-01448-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 05/02/2025] Open
Abstract
Suppression of tumor-reactive CD8+ T cells is common within the tumor microenvironment. However, little is known about how tumors systemically affect the overall CD8+ T cell compartment. Here we demonstrate that peripheral blood CD8+ T cells from patients with lung cancer showed altered compositions particularly within CD45RA-CCR7- effector memory subpopulation. Specifically, patients with lung cancer exhibited increased frequency of more differentiated effector memory cells, which are less susceptible to T cell-receptor-induced proliferation. Further analysis using single-cell RNA sequencing revealed that these alterations were correlated with reduced quiescence and increased spontaneous activation at a systemic level, indicative of homeostatic dysregulation of the entire CD8+ T cell population. This phenomenon was found to be correlated with a poor clinical response to immune checkpoint inhibitor therapy across four independent cohorts, consisting of a total of 224 patients with lung cancer. These findings suggest that lung cancers continue to counteract potentially tumor-reactive CD8+ T cells by inducing homeostatic dysregulation of the entire CD8+ T cell compartment systematically.
Collapse
Affiliation(s)
- Sung-Woo Lee
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ju Sik Yun
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - Young Ju Kim
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Saei Jeong
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jeong Eun Noh
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee-Ok Kim
- Selecxine Inc., Seoul, Republic of Korea
| | - Hyun-Ju Cho
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - Cheol-Kyu Park
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea.
| | - Jae-Ho Cho
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea.
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea.
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea.
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
9
|
Zeng F, Zhang Q, Tsui YM, Ma H, Tian L, Husain A, Lu J, Lee JMF, Zhang VX, Li PM, Cheung GCH, Cheung TT, Ho DWH, Ng IOL. Multimodal sequencing of neoadjuvant nivolumab treatment in hepatocellular carcinoma reveals cellular and molecular immune landscape for drug response. Mol Cancer 2025; 24:110. [PMID: 40205519 PMCID: PMC11980310 DOI: 10.1186/s12943-025-02314-w] [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: 10/15/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
A striking characteristic of liver cancer is its extensive heterogeneity, particularly with regard to its varied response to immunotherapy. In this study, we employed multimodal sequencing approaches to explore the various aspects of neoadjuvant nivolumab treatment in liver cancer patients. We used spatially-resolved transcriptomics, single- and bulk-cell transcriptomics, and TCR clonotype analyses to examine the spatiotemporal dynamics of the effects of nivolumab. We observed a significantly higher clonal expansion of T cells in the tumors of patients who responded to the treatment, while lipid accumulation was detected in those of non-responders, likely due to inherent differences in lipid metabolic processes. Furthermore, we found a preferential enrichment of T cells, which was associated with a better drug response. Our results also indicate a functional antagonism between tumor-associated macrophages (TAMs) and CD8 cells and their spatial separation. Notably, we identified a UBASH3B/NR1I2/CEACAM1/HAVCR2 signaling axis, highlighting the intense communication among TAMs, tumor cells, and T-cells that leads to pro-tumorigenic outcomes resulting in poorer nivolumab response. In summary, using integrative multimodal sequencing investigations, combined with the multi-faceted exploration of pre- and post-treatment samples of neoadjuvant nivolumab-treated HCC patients, we identified useful mechanistic determinants of therapeutic response. We also reconstructed the spatiotemporal model that recapitulates the physiological restoration of T cell cytotoxicity by anti-PD1 blockade. Our findings could provide important biomarkers and explain the mechanistic basis differentiating the responders and non-responders.
Collapse
Grants
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- T12-704/16-R and T12-716/22-R the Hong Kong Research Grants Council Theme-based Research Scheme
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- ITC PD/17-9 Innovation and Technology Commission grant to State Key Laboratory of Liver Research
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- 17100021 & 17117019 RGC General Research Fund
- Health and Medical Research Fund (10212956 & 07182546), RGC General Research Fund
- University Development Fund of The University of Hong Kong
Collapse
Affiliation(s)
- Fanhong Zeng
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Qingyang Zhang
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yu-Man Tsui
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Huanhuan Ma
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Lu Tian
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Abdullah Husain
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Jingyi Lu
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Joyce Man-Fong Lee
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Vanilla Xin Zhang
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Po-Man Li
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Gary Cheuk-Hang Cheung
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tan-To Cheung
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong
| | - Daniel Wai-Hung Ho
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong.
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| | - Irene Oi-Lin Ng
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong.
- State Key Laboratory of Liver Research, The University of Hong Kong, Pokfulam, Hong Kong.
| |
Collapse
|
10
|
Hagbi-Levi S, Abraham M, Gamaev L, Mishaelian I, Hay O, Zorde-Khevalevsky E, Wald O, Wald H, Olam D, Weiss L, Peled A. Identification of Dinaciclib and Ganetespib as anti-inflammatory drugs using a novel HTP screening assay that targets IFNγ-dependent PD-L1. Front Immunol 2025; 16:1502094. [PMID: 40264756 PMCID: PMC12011776 DOI: 10.3389/fimmu.2025.1502094] [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: 09/26/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction IFNγ plays both positive and negative roles in the regulation of innate and adaptive immune responses against tumors and virally infected tissues by upregulating CXCL10 and PD-L1 expression. Methods To identify novel pathways and drugs that regulate the IFNγ-dependent PD-L1, we expressed GFP under the control of mouse PD-L1 promoter in mouse cancer cells that up regulate PD-L1 and CXCL10 in response to IFNγ stimulation. Using these cells, we screened an FDA approved library of 1496 small molecules known for their ability to inhibit IFNγ-dependent increase in PD-L1. Results We identified 46 drugs that up regulated and 4 that down regulated IFNγ-dependent PD-L1 expression. We discovered that in addition to the known JAK inhibitors Ruxolitinib and Baricitinib, Dinaciclib, a CDK1/2/5/9 inhibitor, and Ganetespib, a Hsp90 inhibitor, significantly inhibit both PD-L1 and CXCL10 expression in the model cells. Furthermore, both drugs suppressed IFNγ-dependent CXCL10 and PD-L1 expression in-vitro in primary human lung cells and human cancer cells. These drugs also significantly inhibited delayed-type hypersensitivity (DTH) in-vivo in an inflammation mouse model. Discussion Our novel screening platform can therefore be used in the future to identify novel immunomodulators and pathways in cancer and inflammation, expanding therapeutic horizons.
Collapse
Affiliation(s)
- Shira Hagbi-Levi
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Lika Gamaev
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Inbal Mishaelian
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ophir Hay
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Elina Zorde-Khevalevsky
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ori Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hanna Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Olam
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
11
|
Song J, Zhu J, Jiang Y, Guo Y, Liu S, Qiao Y, Du Y, Li J. Advancements in immunotherapy for gastric cancer: Unveiling the potential of immune checkpoint inhibitors and emerging strategies. Biochim Biophys Acta Rev Cancer 2025; 1880:189277. [PMID: 39938663 DOI: 10.1016/j.bbcan.2025.189277] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 01/08/2025] [Accepted: 02/04/2025] [Indexed: 02/14/2025]
Abstract
Gastric cancer (GC) is linked to high morbidity and mortality rates. Approximately two-thirds of GC patients are diagnosed at an advanced or metastatic stage. Conventional treatments for GC, including surgery, radiotherapy, and chemotherapy, offer limited prognostic improvement. Recently, immunotherapy has gained attention for its promising therapeutic effects in various tumors. Immunotherapy functions by activating and regulating the patient's immune cells to target and eliminate tumor cells, thereby reducing the tumor burden in the body. Among immunotherapies, immune checkpoint inhibitors (ICIs) are the most advanced. ICIs disrupt the inhibitory protein-small molecule (PD-L1, CTLA4, VISTA, TIM-3 and LAG3) interactions produced by immune cells, reactivating these cells to recognize and attack tumor cells. However, adverse reactions and resistance to ICIs hinder their further clinical and experimental development. Therefore, a comprehensive understanding of the advancements in ICIs for GC is crucial. This article discusses the latest developments in clinical trials of ICIs for GC and examines combination therapies involving ICIs (targeted therapy, chemotherapy, radiotherapy), alongside ongoing clinical trials. Additionally, the review investigates the tumor immune microenvironment and its role in non-responsiveness to ICIs, highlighting the function of tumor immune cells in ICI efficacy. Finally, the article explores the prospects and limitations of new immunotherapy-related technologies, such as tumor vaccines, nanotechnologies, and emerging therapeutic strategies, aiming to advance research into personalized and optimized immunotherapy for patients with locally advanced gastric cancer.
Collapse
Affiliation(s)
- Jiawei Song
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China
| | - Jun Zhu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yu Jiang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yajie Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Shuai Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yihuan Qiao
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Yongtao Du
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China
| | - Jipeng Li
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi'an 710038, China; Department of Experimental Surgery, Xijing Hospital, Xi'an 710038, China.
| |
Collapse
|
12
|
Subbarayan K, Bieber H, Massa C, Rodríguez FAE, Hossain SMAA, Neuder L, Wahbi W, Salo T, Tretbar S, Al-Samadi A, Seliger B. Link of TMPRSS2 expression with tumor immunogenicity and response to immune checkpoint inhibitors in cancers. J Transl Med 2025; 23:294. [PMID: 40055791 PMCID: PMC11887338 DOI: 10.1186/s12967-025-06177-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: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND SARS-CoV-2 and other viruses rely on the protease function of the TMPRSS2 protein to invade host cells. Despite cancer patients often experience poorer outcomes following SARS-CoV-2 infection, the role of TMPRSS2 in different cancer types has not yet been analyzed in detail. Therefore, the aim of the study was to determine the expression, function and clinical relevance of TMPRSS2 in tumors. METHODS Publicly accessible RNA sequencing data from tumors, adjacent tissues and whole blood samples of COVID-19 patients as well as data from human tumor epithelial and endothelial cells infected with SARS-CoV-2 were analyzed for TMPRSS2 expression and correlated to the expression of immune-relevant genes and clinical parameters. In vitro models of cells transfected with TMPRSS2 (TMPRSS2high), siTMPRSS2 or mock controls (TMPRSS2low cells) were analyzed by qPCR, flow cytometry, ELISA and Western blot for the expression of immune response-relevant molecules. Co-cultures of TMPRSS2 model systems with blood peripheral mononuclear cells were employed to evaluate immune cell migration, cytotoxicity and cytokine release. RESULTS Higher expression levels of TMPRSS2 were found in blood from patients infected with SARS-CoV-2, while TMPRSS2 expression levels significantly varied between the tumor types analyzed. TMPRSS2high tumor cells exhibit increased activity of the interferon (IFN) signal pathway accompanied by an increased expression of class I human leukocyte antigens (HLA-I) and programmed cell death ligand 1 (PD-L1) elevated interleukin 6 (IL-6) secretion and reduced NK cell-mediated cytotoxicity compared to TMPRSS2low mock controls. Treatment with a Janus kinase (JAK) 2 inhibitor or TMPRSS2-specific siRNA decreased TMPRSS2 expression. Co-cultures of the in vitro TMPRSS2 models with peripheral blood mononuclear cells in the presence of the immune checkpoint inhibitor nivolumab resulted in a significantly increased migration and infiltration of immune cells towards TMPRSS2high cells and a reduced release of the innate immunity-related cytokines CCL2 and CCL3. CONCLUSIONS This study provides novel insights into the role of TMPRSS2 in various tumor systems and the impact of SARS-CoV-2 infection on the host immunogenicity via the activation of immune-relevant pathways. These findings were linked to the efficacy of immune checkpoint inhibitor therapy, offering a potential alternative strategy to mitigate the severity of COVID-19.
Collapse
Affiliation(s)
| | - Helena Bieber
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chiara Massa
- Institute of Translational Immunology, Faculty of Health Sciences, Brandenburg Medical School "Theodor Fontane", Brandenburg an der Havel, Germany
| | - Felipe Adonis Escalona Rodríguez
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Center for Protein Studies, Faculty of Biology, University of Havana (UH), Havana, Cuba
- NanoCancer, Molecular Immunology Center (CIM), Havana, Cuba
| | - S M Al Amin Hossain
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Lisa Neuder
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, 90014, Finland
| | - Sandy Tretbar
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ahmed Al-Samadi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Institute of Translational Immunology, Faculty of Health Sciences, Brandenburg Medical School "Theodor Fontane", Brandenburg an der Havel, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
| |
Collapse
|
13
|
Majocchi S, Lloveras P, Nouveau L, Legrand M, Viandier A, Malinge P, Charreton M, Raymond C, Pace EA, Millard BL, Svensson LA, Kelpšas V, Anceriz N, Salgado-Pires S, Daubeuf B, Magistrelli G, Gueneau F, Moine V, Masternak K, Shang L, Fischer N, Ferlin WG. NI-3201 Is a Bispecific Antibody Mediating PD-L1-Dependent CD28 Co-stimulation on T Cells for Enhanced Tumor Control. Cancer Immunol Res 2025; 13:365-383. [PMID: 39760515 PMCID: PMC11876958 DOI: 10.1158/2326-6066.cir-24-0298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025]
Abstract
Despite advances in cancer immunotherapy, such as targeting the PD-1/PD-L1 axis, a substantial number of patients harbor tumors that are resistant or relapse. Selective engagement of T-cell co-stimulatory molecules with bispecific antibodies may offer novel therapeutic options by enhancing signal 1-driven activation occurring via T-cell receptor engagement. In this study, we report the development and preclinical characterization of NI-3201, a PD-L1×CD28 bispecific antibody generated on the κλ-body platform that was designed to promote T-cell activity and antitumor function through a dual mechanism of action. We confirmed that NI-3201 blocks the PD-L1/PD-1 immune checkpoint pathway and conditionally provides T-cell co-stimulation via CD28 (signal 2) when engaging PD-L1+ tumors or immune cells. In systems with signal 1-primed T cells, NI-3201 enhanced potent effector functionality: in vitro through antigen-specific recall assays with cytomegalovirus-specific T cells and in vivo by inducing tumor regression and immunologic memory in tumor-associated antigen-expressing MC38 syngeneic mouse models. When T-cell engagers were used to provide synthetic signal 1, the combination with NI-3201 resulted in synergistic T cell-dependent cytotoxicity and potent antitumor activity in two humanized mouse tumor models. Nonhuman primate safety assessments showed favorable tolerability and pharmacokinetics at pharmacologically active doses. Quantitative systems pharmacology modeling predicted that NI-3201 exposure results in antitumor activity in patients, but this remains to be investigated. Overall, this study suggests that by combining PD-L1 blockade with safe and effective CD28 co-stimulation, NI-3201 has the potential to improve cancer immunotherapy outcomes, and the clinical development of NI-3201 for PD-L1+ solid tumors is planned.
Collapse
Affiliation(s)
- Sara Majocchi
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Lise Nouveau
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | | | | | - Maud Charreton
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | - Cecile Raymond
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | | | | | | | - Nadia Anceriz
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Bruno Daubeuf
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Franck Gueneau
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | - Valéry Moine
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | - Limin Shang
- Light Chain Bioscience – Novimmune SA, Geneva, Switzerland
| | | | | |
Collapse
|
14
|
Snyder M, Wang Z, Lara B, Fimbres J, Pichardo T, Mazzilli S, Khan MM, Duggineni VK, Monti S, Sherr DH. The aryl hydrocarbon receptor controls IFN-γ-induced immune checkpoints PD-L1 and IDO via the JAK/STAT pathway in lung adenocarcinoma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae023. [PMID: 40073102 DOI: 10.1093/jimmun/vkae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/12/2024] [Indexed: 03/14/2025]
Abstract
While immunotherapy has shown some efficacy in lung adenocarcinoma (LUAD) patients, many respond only partially or not at all. One limitation in improving outcomes is the lack of a complete understanding of immune checkpoint regulation. Here, we investigated a possible link between an environmental chemical receptor implicated in lung cancer and immune regulation, the AhR, a known but counterintuitive mediator of immunosuppression (interferon (IFN)-γ), and regulation of two immune checkpoints (PD-L1 and IDO). AhR gene-edited LUAD cell lines, a syngeneic LUAD mouse model, bulk and scRNA sequencing of LUADs and tumor-infiltrating T cells were used to map out a signaling pathway leading from IFN-γ through the AhR to JAK/STAT, PD-L1, IDO, and tumor-mediated immunosuppression. The data demonstrate that: (1) IFN-γ activation of the JAK/STAT pathway leading to PD-L1 and IDO1 up-regulation is mediated by the AhR in murine and human LUAD cells, (2) AhR-driven IDO1 induction results in the production of Kynurenine (Kyn), an AhR ligand, which likely mediates an AhR→IDO1→Kyn→AhR amplification loop, (3) transplantation of AhR-knockout LUAD cells results in long-term tumor immunity in most recipients. (4) The 23% of AhR-knockout tumors that do grow do so at a much slower pace than controls and exhibit higher densities of CD8+ T cells expressing markers of immunocompetence, increased activity, and increased cell-cell communication. The data definitively link the AhR to IFN-γ-induced JAK/STAT pathway and immune checkpoint-mediated immunosuppression and support the targeting of the AhR in the context of LUAD.
Collapse
Affiliation(s)
- Megan Snyder
- Graduate Program in Genetics and Genomics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Zhongyan Wang
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, United States
| | - Brian Lara
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, United States
| | - Jocelyn Fimbres
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, United States
| | - Táchira Pichardo
- Department of Medicine, Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Sarah Mazzilli
- Department of Medicine, Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Mohammed Muzamil Khan
- Department of Medicine, Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - Vinay K Duggineni
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, United States
| | - Stefano Monti
- Department of Medicine, Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | - David H Sherr
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, United States
| |
Collapse
|
15
|
Mastrolonardo EV, Nunes KL, Llerena P, Nikitina A, Sobol A, Scott ER, Tuluc M, Davitt CJH, Scher J, Tekumalla S, Mann D, Henao C, Jegede V, Gargano S, Harshyne LA, Alnemri A, Tyshevich A, Kushnarev V, Chasse M, Sookiasian D, Axelrod R, Zhan T, Leiby BE, Old M, Seim N, Mahoney MG, Martinez-Outschoorn U, Cognetti DM, Curry JM, Prendergast G, Argiris A, South AP, Linnenbach AJ, Johnson JM, Luginbuhl AJ. Response-Adaptive Surgical Timing in Neoadjuvant Immunotherapy Demonstrates Enhanced Pathologic Treatment Response in Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 2025; 31:515-528. [PMID: 39585339 PMCID: PMC11973698 DOI: 10.1158/1078-0432.ccr-24-0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/28/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE We evaluated whether indoleamine 2,3-dioxygenase (IDO1) inhibitor (IDOi) BMS986205 + PD-1 inhibitor nivolumab enhanced T-cell activity and augmented immune-mediated antitumor responses in untreated, resectable head and neck squamous cell carcinoma (HNSCC). We employed response-adaptive surgical timing to identify responders to immunotherapy and enhance their response. PATIENTS AND METHODS Patients with HNSCC were 3:1 randomized to receive nivolumab with or without BMS986205 orally daily (NCT03854032). In the combination arm, BMS986205 was initiated 7 days prior to nivolumab. Patients were stratified by human papillomavirus (HPV) status. Response-adaptive surgical timing involved response assessment by radiographic criteria 4 weeks after treatment with nivolumab in both arms. Nonresponders underwent surgical resection, whereas responders received 4 more weeks of randomized therapy before surgery. Biomarker analysis utilized pathologic treatment response (pTR) and RNA sequencing. RESULTS Forty-two patients were enrolled, and the addition of IDOi to nivolumab did not result in greater rate of radiographic response (P = 0.909). Treatment was well tolerated, with only 2 (5%) patients experiencing grade 3 immune-related adverse events. The addition of IDOi augmented rates of pTR in patients with high baseline IDO1 RNA expression (P < 0.05). Response-adaptive surgical timing demonstrated reliability in differentiating pathologic responders versus nonresponders (P = 0.009). A pretreatment NK cell signature, PD-L1 status, and IFN-γ expression in the HPV- cohort correlated with response. The HPV+ cohort found B-cell and cancer-associated fibroblast signatures predictive of response/nonresponse. CONCLUSIONS Response-adaptive surgical timing enhanced treatment response. IDOi BMS986205 augmented pTR in patients with high IDO1 expression in baseline samples, indicating a need for identifying and targeting resistant nodes to immunotherapy. HPV status-dependent signatures predicting response to immunotherapy in HNSCC warrant further study.
Collapse
Affiliation(s)
- Eric V Mastrolonardo
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Kathryn L Nunes
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Pablo Llerena
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | - E Reilly Scott
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Madalina Tuluc
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | - Sruti Tekumalla
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Derek Mann
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Camilo Henao
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Victor Jegede
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Stacey Gargano
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Larry A Harshyne
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Angela Alnemri
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | | | | | - Rita Axelrod
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Tingting Zhan
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Benjamin E Leiby
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Matthew Old
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio
| | - Nolan Seim
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University, Columbus, Ohio
| | - My G Mahoney
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ubaldo Martinez-Outschoorn
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - David M Cognetti
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Joseph M Curry
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | - Athanassios Argiris
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew P South
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Alban J Linnenbach
- Department of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jennifer M Johnson
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Adam J Luginbuhl
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Comprehensive Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
16
|
Tanaka Y, Hu Q, Kawazoe T, Tajiri H, Nakanishi R, Zaitsu Y, Nakashima Y, Ota M, Oki E, Oda Y, Yoshizumi T. The clinical significance of signal regulatory protein alpha expression in the immune environment of gastric cancer. Int J Clin Oncol 2025; 30:330-339. [PMID: 39589589 DOI: 10.1007/s10147-024-02666-1] [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: 06/25/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Signal regulatory protein alpha (SIRPα) inhibits phagocytosis by macrophages by interacting with CD47. Despite its known role in various cancers, the clinical significance of SIRPα in gastric cancer (GC) remains unclear. This study aimed to elucidate the clinical implications of SIRPα in GC, exploring its relevance to immunotherapy efficacy and the tumor microenvironment. METHODS Two cohorts were studied: a gastrectomy cohort (137 patients) and an immune checkpoint inhibitor (ICI)-treated cohort (19 patients with unresectable advanced GC who received nivolumab). Immunohistochemistry was used to assess SIRPα, CD80, CD163, CD8, and PD-L1 expressions. Kaplan-Meier curves and Cox models were used to analyze the clinical outcomes. In vitro experiments used peripheral blood mononuclear cells and THP-1 macrophage cell lines to examine SIRPα responses to interferon-γ (IFN-γ). RESULTS In the gastrectomy cohort, high SIRPα expression correlated with advanced tumor invasion, distant metastasis, and poor recurrence-free and overall survival. SIRPα expression was also significantly associated with macrophage and CD8 + T cells infiltration and PD-L1 expression. In the ICI-treated cohort, high SIRPα expression was associated with better overall survival after nivolumab induced. Moreover, in vitro IFN-γ stimulation upregulated SIRPα expression on monocytes in peripheral blood mononuclear cells and THP-1 cells, suggesting high SIRPα expression may reflect an active immune microenvironment. CONCLUSION SIRPα expression is not only a poor prognostic factor for GC, possibly through inhibition of the CD47-SIRP⍺ pathway, but may also be involved in the efficacy of ICI therapy in GC.
Collapse
Affiliation(s)
- Yasushi Tanaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Qingjiang Hu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Tetsuro Kawazoe
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Hirotada Tajiri
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Ryota Nakanishi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yoko Zaitsu
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yuichiro Nakashima
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Mitsuhiko Ota
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
17
|
Wei LJ, Wu ZY, Wu LY, Wu YW, Liang HY, Luo RZ, Liu LL. Prognostic implications of immune classification based on PD-L1 expression and tumor-infiltrating lymphocytes in endocervical adenocarcinoma. Transl Oncol 2025; 52:102265. [PMID: 39736213 PMCID: PMC11750284 DOI: 10.1016/j.tranon.2024.102265] [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: 08/27/2024] [Revised: 11/24/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND Endocervical adenocarcinoma (ECA) comprises a heterogeneous group of diseases whose incidence has increased significantly in recent decades. ECA can be histologically classified into human papillomavirus-associated (HPVA) and non-HPVA (NHPVA) types. Given the variability in pathological features and clinical behavior between the subtypes, evaluating their respective immune microenvironments is essential. They can be categorized into distinct tumor microenvironment immune types (TMIT). METHODS A total of 540 surgically resected ECA samples were classified into HPVA and NHPVA subgroups. Tumor-infiltrating immune markers were assessed using immunohistochemistry. We categorized ECA into four TMIT based on PD-L1 and CD8+ tumor-infiltrating lymphocytes (TILs) expression, and analyzed their prognostic significance. RESULTS PD-L1 positivity was observed in 319 out of 464 (68.8%) HPVA and 55 out of 76 (72.4%) NHPVA. Across the entire cohort, high CD8+ TILs expression was significantly associated with improved disease-free survival (DFS, p=0.018) and overall survival (OS, p=0.031). A total of 177 samples (32.8%) were classified as TMIT I (high PD-L1 and high CD8+ TILs), exhibiting markedly denser immune cell infiltration compared to the other TMIT groups. In NHPVA subgroup, TMIT was significantly associated with both DFS (p=0.005) and OS (p=0.003). Multivariate analysis identified TMIT as an independent prognostic factor for DFS in the NHPVA group, with TMIT I indicating a more favorable prognosis (p=0.042). CONCLUSIONS TMIT I group within the NHPVA population is most likely to benefit from PD-L1/PD-1 blockade immunotherapies. The immune classification of ECA demonstrates significant prognostic value, suggesting its potential utility in guiding clinical stratification and therapeutic decision-making.
Collapse
Affiliation(s)
- Li-Jun Wei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zi-Yun Wu
- Department of Urology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - Li-Yan Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying-Wen Wu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hao-Yu Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Rong-Zhen Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Li-Li Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
18
|
Cozac-Szőke AR, Cozac DA, Negovan A, Tinca AC, Vilaia A, Cocuz IG, Sabău AH, Niculescu R, Chiorean DM, Tomuț AN, Cotoi OS. Immune Cell Interactions and Immune Checkpoints in the Tumor Microenvironment of Gastric Cancer. Int J Mol Sci 2025; 26:1156. [PMID: 39940924 PMCID: PMC11818890 DOI: 10.3390/ijms26031156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Gastric cancer (GC) ranks as the fifth most prevalent malignant neoplasm globally, with an increased death rate despite recent advancements in research and therapeutic options. Different molecular subtypes of GC have distinct interactions with the immune system, impacting the tumor microenvironment (TME), prognosis, and reaction to immunotherapy. Tumor-infiltrating lymphocytes (TILs) in the TME are crucial for preventing tumor growth and metastasis, as evidenced by research showing that patients with GC who have a significant density of TILs have better survival rates. But cancer cells have evolved a variety of mechanisms to evade immune surveillance, both sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) and Programmed Death-Ligand 1 (PD-L1) playing a pivotal role in the development of an immunosuppressive TME. They prevent T cell activation and proliferation resulting in a decrease in the immune system's capacity to recognize and eliminate malignant cells. These immune checkpoint molecules function via different but complementary mechanisms, the expression of Siglec-15 being mutually exclusive with PD-L1 and, therefore, providing a different therapeutic approach. The review explores how TILs affect tumor growth and patient outcomes in GC, with particular emphasis on their interactions within the TME and potential targeting of the PD-L1 and Siglec-15 pathways for immunotherapy.
Collapse
Affiliation(s)
- Andreea-Raluca Cozac-Szőke
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Dan Alexandru Cozac
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Emergency Institute for Cardiovascular Diseases and Transplantation Targu Mures, 540142 Targu Mures, Romania
| | - Anca Negovan
- Department of Clinical Science-Internal Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Andreea Cătălina Tinca
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Alexandra Vilaia
- Department of Infectious Diseases I, Doctoral School of Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Iuliu-Gabriel Cocuz
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Adrian Horațiu Sabău
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Raluca Niculescu
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Diana Maria Chiorean
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Alexandru Nicușor Tomuț
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Ovidiu Simion Cotoi
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| |
Collapse
|
19
|
Xia Y, Huang C, Zhong M, Zhong H, Ruan R, Xiong J, Yao Y, Zhou J, Deng J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun Signal 2025; 23:46. [PMID: 39856684 PMCID: PMC11762533 DOI: 10.1186/s12964-025-02033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor (HGF) along with its receptor (c-MET) are crucial in preserving standard cellular physiological activities, and imbalances in the c-MET signaling pathway can lead to the development and advancement of tumors. It has been extensively demonstrated that immune checkpoint inhibitors (ICIs) can result in prolonged remission in certain patients. Nevertheless, numerous preclinical studies have shown that MET imbalance hinders the effectiveness of anti-PD-1/PD-L1 treatments through various mechanisms. Consequently, clarifying the link between the c-MET signaling pathway and the tumor microenvironment (TME), as well as uncovering the effects of anti-MET treatment on ICI therapy, is crucial for enhancing the outlook for tumor patients. In this review, we examine the impact of abnormal activation of the HGF/c-MET signaling pathway on the control of the TME and the processes governing PD-L1 expression in cancer cells. The review thoroughly examines both clinical and practical evidence regarding the use of c-MET inhibitors alongside PD-1/PD-L1 inhibitors, emphasizing that focusing on c-MET with immunotherapy enhances the effectiveness of treating MET tumors exhibiting elevated PD-L1 expression.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Min Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jing Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
20
|
Xu SX, Wang L, Ip P, Randhawa RR, Benatar T, Prosser SL, Lal P, Khan AN, Nitya-Nootan T, Thakor G, MacGregor H, Hayes DL, Vucicevic A, Mathew P, Sengupta S, Helsen CW, Bader AG. Preclinical Development of T Cells Engineered to Express a T-Cell Antigen Coupler Targeting Claudin 18.2-Positive Solid Tumors. Cancer Immunol Res 2025; 13:35-46. [PMID: 39404622 PMCID: PMC11712040 DOI: 10.1158/2326-6066.cir-24-0138] [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: 02/09/2024] [Revised: 05/29/2024] [Accepted: 10/01/2024] [Indexed: 01/11/2025]
Abstract
The T-cell antigen coupler (TAC) is a chimeric receptor that facilitates tumor antigen-specific activation of T cells by co-opting the endogenous T-cell receptor complex in the absence of tonic signaling. Previous data demonstrate that the TAC affords T cells with the ability to induce durable and safe antitumor responses in preclinical models of hematologic and solid tumors. In this study, we describe the preclinical pharmacology and safety of an autologous Claudin 18.2 (CLDN18.2)-directed TAC T-cell therapy, TAC01-CLDN18.2, in preparation for a phase I/II clinical study in subjects with CLDN18.2-positive solid tumors. Following a screen of putative TAC constructs, the specificity, activity, and cytotoxicity of TAC T cells expressing the final CLDN18.2-TAC receptor were evaluated in vitro and in vivo using gastric, gastroesophageal, and pancreatic tumor models as well as human cells derived from normal tissues. CLDN18.2-specific activity and cytotoxicity of CLDN18.2-TAC T cells were observed in coculture with various 2D tumor cultures naturally expressing CLDN18.2 as well as tumor spheroids. These effects occurred in models with low antigen levels and were positively associated with increasing CLDN18.2 expression. CLDN18.2-TAC T cells effectively eradicated established tumor xenografts in mice in the absence of observed off-target or on-target/off-tumor effects, elicited durable efficacy in recursive killing and tumor rechallenge experiments, and remained unreactive in coculture with human cells representing vital organs. Thus, the data demonstrate that CLDN18.2-TAC T cells can induce a specific and long-lasting antitumor response in various CLDN18.2-positive solid tumor models without notable TAC-dependent toxicities, supporting the clinical development of TAC01-CLDN18.2.
Collapse
MESH Headings
- Humans
- Animals
- Claudins/metabolism
- Claudins/genetics
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Neoplasms/therapy
- Neoplasms/immunology
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Immunotherapy, Adoptive/methods
- Female
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
Collapse
Affiliation(s)
- Stacey X. Xu
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Ling Wang
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Philbert Ip
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Ritu R. Randhawa
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Tania Benatar
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Suzanna L. Prosser
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Prabha Lal
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Alima Naim Khan
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Thanyashanthi Nitya-Nootan
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Gargi Thakor
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Heather MacGregor
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Danielle L Hayes
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Andrea Vucicevic
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Princy Mathew
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Sadhak Sengupta
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Christopher W. Helsen
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| | - Andreas G. Bader
- Triumvira Immunologics, Inc., headquartered in Austin, Texas, with research facilities in Hamilton, Canada
| |
Collapse
|
21
|
Angelicola S, Giunchi F, Ruzzi F, Frascino M, Pitzalis M, Scalambra L, Semprini MS, Pittino OM, Cappello C, Siracusa I, Chillico IC, Di Noia M, Turato C, De Siervi S, Lescai F, Ciavattini T, Lopatriello G, Bertoli L, De Jonge H, Iamele L, Altimari A, Gruppioni E, Ardizzoni A, Rossato M, Gelsomino F, Lollini PL, Palladini A. PD-L1 and IFN-γ modulate Non-Small Cell Lung Cancer (NSCLC) cell plasticity associated to immune checkpoint inhibitor (ICI)-mediated hyperprogressive disease (HPD). J Transl Med 2025; 23:2. [PMID: 39748404 PMCID: PMC11697469 DOI: 10.1186/s12967-024-06023-8] [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: 08/03/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Non-Small Cell Lung Cancer (NSCLC) is the leading cause of cancer death worldwide. Although immune checkpoint inhibitors (ICIs) have shown remarkable clinical efficacy, they can also induce a paradoxical cancer acceleration, known as hyperprogressive disease (HPD), whose causative mechanisms are still unclear. METHODS This study investigated the mechanisms of ICI resistance in an HPD-NSCLC model. Two primary cell cultures were established from samples of a NSCLC patient, before ICI initiation ("baseline", NSCLC-B) and during HPD ("hyperprogression", NSCLC-H). The cell lines were phenotypically and molecularly characterized through immunofluorescence, Western Blotting and RNA-Seq analysis. To assess cell plasticity and aggressiveness, cellular growth patterns were evaluated both in vitro and in vivo through 2D and 3D cell growth assays and patient-derived xenografts establishment. In vitro investigations, including the evaluation of cell sensitivity to interferon-gamma (IFN-γ) and cell response to PD-L1 modulation, were conducted to explore the influence of these factors on cell plasticity regulation. RESULTS NSCLC-H exhibited increased expression of specific CD44 isoforms and a more aggressive phenotype, including organoid formation ability, compared to NSCLC-B. Plastic changes in NSCLC-H were well described by a deep transcriptome shift, that also affected IFN-γ-related genes, including PD-L1. IFN-γ-mediated cell growth inhibition was compromised in both 2D-cultured NSCLC-B and NSCLC-H cells. Further, the cytokine induced a partial activation of both type I and type II IFN-pathway mediators, together with a striking increase in NSCLC-B growth in 3D cell culture systems. Finally, low IFN-γ doses and PD-L1 modulation both promoted plastic changes in NSCLC-B, increasing CD44 expression and its ability to produce spheres. CONCLUSIONS Our findings identified plasticity as a relevant hallmark of ICI-mediated HPD by demonstrating that ICIs can modulate the IFN-γ and PD-L1 pathways, driving tumor cell plasticity and fueling HPD development.
Collapse
Affiliation(s)
- Stefania Angelicola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | | - Mary Pitzalis
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Laura Scalambra
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Olga Maria Pittino
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Chiara Cappello
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Irene Siracusa
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Martina Di Noia
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Silvia De Siervi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Francesco Lescai
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | | | - Luca Bertoli
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Hugo De Jonge
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Luisa Iamele
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Ardizzoni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
- Genartis S.R.L., Verona, Italy
| | - Francesco Gelsomino
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Arianna Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Unità Operativa di Oncologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
22
|
Kallenberger EM, Khandelwal A, Nath P, Nguyen SA, DiGiovanni J, Nathan CA. FGFR2 in the Development and Progression of Cutaneous Squamous Cell Cancer. Mol Carcinog 2025; 64:5-13. [PMID: 39466044 DOI: 10.1002/mc.23835] [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: 10/04/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is an increasingly common malignancy of the skin and the leading cause of death from skin cancer in adults over the age of 85. Fibroblast growth factor receptor 2 (FGFR2) has been identified as an important effector of signaling pathways that lead to the growth and development of cSCC. In recent years, there have been numerous studies evaluating the role FGFR2 plays in multiple cancers, its contribution to resistance to anticancer therapy, and new drugs that may be used to inhibit FGFR2. This review will provide an overview of our current understanding of FGFR2 and potential mechanisms in which we can target FGFR2 in cSCC. The goals of this review are the following: (1) to highlight our current knowledge of the role of FGFR2 in healthy skin and contrast this with its role in the development of cancer; (2) to further explain the specific molecular mechanisms that FGFR2 uses to promote tumorigenesis; (3) to describe how FGFR2 contributes to more invasive disease; (4) to describe its immunosuppressive effects in skin; and (5) to evaluate its effect on current anticancer therapy and discuss therapies on the horizon to target FGFR2 related malignancy.
Collapse
Affiliation(s)
- Ethan M Kallenberger
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alok Khandelwal
- Department of Oto/HNS, Health Sciences Center, Louisiana State University, Shreveport, Louisiana, USA
| | - Priyatosh Nath
- Department of Oto/HNS, Health Sciences Center, Louisiana State University, Shreveport, Louisiana, USA
| | - Shaun A Nguyen
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - John DiGiovanni
- Department of Pharmacology, University of Texas, Austin, Texas, USA
| | - Cherie-Ann Nathan
- Department of Oto/HNS, Health Sciences Center, Louisiana State University, Shreveport, Louisiana, USA
| |
Collapse
|
23
|
Kim A, Lee EJ, Han JH, Chung HS. Caryophylli Cortex Suppress PD-L1 Expression in Cancer Cells and Potentiates Anti-Tumor Immunity in a Humanized PD-1/PD-L1 Knock-In MC-38 Colon Cancer Mouse Model. Nutrients 2024; 16:4415. [PMID: 39771037 PMCID: PMC11679492 DOI: 10.3390/nu16244415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Immune checkpoints are essential for regulating excessive autoimmune responses and maintaining immune homeostasis. However, in the tumor microenvironment, these checkpoints can lead to cytotoxic T cell exhaustion, allowing cancer cells to evade immune surveillance and promote tumor progression. The expression of programmed death-ligand 1 (PD-L1) in cancer cells is associated with poor prognoses, reduced survival rates, and lower responses to therapies. Consequently, downregulating PD-L1 expression has become a key strategy in developing immune checkpoint inhibitors (ICIs). Caryophylli cortex (CC), derived from the bark of the clove tree Syzygium aromaticum, possesses antioxidant and cytotoxic properties against cancer cells, yet its potential as an ICI remains unclear. METHODS In this study, we aimed to investigate whether CC extract modulates PD-L1 expression in cancer cells and activates T cell immunity through a co-culture system of cancer cells and T cells, as well as in hPD-L1/MC-38 tumor-bearing animal models. RESULTS Our findings indicate that CC extract significantly downregulated both constitutive and inducible PD-L1 expression at non-toxic concentrations for cancer cells while simultaneously enhancing cancer cell mortality and T cell activity in the co-culture system. Furthermore, the administration of CC extract to hPD-L1/MC-38 tumor-bearing mice resulted in a greater than 70% reduction in tumor growth and increased infiltration of CD8+ T cells within the tumor microenvironment. Principal component analysis identified bergenin, chlorogenic acid, and ellagic acid as active ICIs. CONCLUSIONS These findings suggest that CC extract exerts a potent antitumor effect as an immune checkpoint blocker by inhibiting PD-L1 expression in cancer cells and disrupting the PD-1/PD-L1 interaction.
Collapse
Affiliation(s)
- Aeyung Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-J.L.); (J.H.H.)
| | | | | | - Hwan-Suck Chung
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea; (E.-J.L.); (J.H.H.)
| |
Collapse
|
24
|
Godiyal Y, Maheshwari D, Taniguchi H, Zinzuwadia SS, Morera-Díaz Y, Tewari D, Bishayee A. Role of PD-1/PD-L1 signaling axis in oncogenesis and its targeting by bioactive natural compounds for cancer immunotherapy. Mil Med Res 2024; 11:82. [PMID: 39690423 DOI: 10.1186/s40779-024-00586-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024] Open
Abstract
Cancer is a global health problem and one of the leading causes of mortality. Immune checkpoint inhibitors have revolutionized the field of oncology, emerging as a powerful treatment strategy. A key pathway that has garnered considerable attention is programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1). The interaction between PD-L1 expressed on tumor cells and PD-1 reduces the innate immune response and thus compromises the capability of the body's immune system. Furthermore, it controls the phenotype and functionality of innate and adaptive immune components. A range of monoclonal antibodies, including avelumab, atezolizumab, camrelizumab, dostarlimab, durvalumab, sinitilimab, toripalimab, and zimberelimab, have been developed for targeting the interaction between PD-1 and PD-L1. These agents can induce a broad spectrum of autoimmune-like complications that may affect any organ system. Recent studies have focused on the effect of various natural compounds that inhibit immune checkpoints. This could contribute to the existing arsenal of anticancer drugs. Several bioactive natural agents have been shown to affect the PD-1/PD-L1 signaling axis, promoting tumor cell apoptosis, influencing cell proliferation, and eventually leading to tumor cell death and inhibiting cancer progression. However, there is a substantial knowledge gap regarding the role of different natural compounds targeting PD-1 in the context of cancer. Hence, this review aims to provide a common connection between PD-1/PD-L1 blockade and the anticancer effects of distinct natural molecules. Moreover, the primary focus will be on the underlying mechanism of action as well as the clinical efficacy of bioactive molecules. Current challenges along with the scope of future research directions targeting PD-1/PD-L1 interactions through natural substances are also discussed.
Collapse
Affiliation(s)
- Yogesh Godiyal
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Drishti Maheshwari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Magdalenka, Poland
- African Genome Center, Mohammed VI Polytechnic University, Hay Moulay Rachid, 43150, Ben Guerir, Morocco
| | - Shweta S Zinzuwadia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Yanelys Morera-Díaz
- Clinical Investigation and Biomedical Research Directions, Center for Genetic Engineering and Biotechnology, 11600, Havana, Cuba
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
25
|
Mendonca P, Kaur S, Kirpal B, Soliman KFA. Cardamonin anticancer effects through the modulation of the tumor immune microenvironment in triple-negative breast cancer cells. Am J Cancer Res 2024; 14:5644-5664. [PMID: 39803666 PMCID: PMC11711538 DOI: 10.62347/anxs3815] [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: 08/17/2024] [Accepted: 10/21/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor immune microenvironment (TIME) plays a critical role in cancer development and response to immunotherapy. Immune checkpoint inhibitors aim to reverse the immunosuppressive effects of the TIME, but their success has been limited. Immunotherapy directed at PD-1/PD-L1 has been widely employed, yielding positive results. Unfortunately, the gradual emergence of resistance to PD-1/PD-L1 inhibition has diminished the effectiveness of this immunotherapy in cancer patients, emphasizing the need for new compounds that will be more effective in managing immunotherapy. This study investigated the effect of the natural compound cardamonin on PD-L1 expression and its ability to modulate the TIME, which could overcome immunotherapy resistance in triple-negative breast cancer (TNBC). This investigation used two genetically distinct triple-negative breast cancer cell lines, MDA-MB-231 (MDA-231) and MDA-MB-468 (MDA-468). The results show that TNBC cell treatment with cardamonin inhibited PD-L1 expression and reduced JAK1 and STAT3 levels in MDA-231 cells, while it increased JAK1 expression in MDA-468 cells. Also, cardamonin increased the expression of Nrf2 in both cell lines. In addition, cardamonin decreased MUC1, NF-κB1, and NF-κB2 expression in MDA-MB-231 cells and selectively reduced NF-κB1 expression in MDA-468 cells. Furthermore, cardamonin very potently reduced the inflammatory cytokine CCL2 levels. The decrease in CCL2 release reduces the chemoattraction of macrophages in the tumor microenvironment, which may increase the effectiveness of PD-1/PD-L1 inhibition and allow T-cell infiltration. These findings suggest that the cardamonin modulation of TIME holds promise in reversing resistance of PD-1/PD-L1 inhibition when it is used along with immunotherapy in TNBC treatment.
Collapse
Affiliation(s)
- Patricia Mendonca
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M UniversityTallahassee, FL 32307, The United States
- Department of Biology, College of Science and Technology, Florida A&M UniversityTallahassee, FL 32307, The United States
| | - Sukhmandeep Kaur
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M UniversityTallahassee, FL 32307, The United States
| | - Bhonesa Kirpal
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M UniversityTallahassee, FL 32307, The United States
| | - Karam FA Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M UniversityTallahassee, FL 32307, The United States
| |
Collapse
|
26
|
Hu M, Zhang Y, Liu J, Chen Y, Kang J, Zhong J, Lin S, Liang Y, Cen R, Zhu X, Zhang C. B2M or CIITA knockdown decreased the alloimmune response of dental pulp stem cells: an in vitro study. Stem Cell Res Ther 2024; 15:425. [PMID: 39538338 PMCID: PMC11562604 DOI: 10.1186/s13287-024-04023-5] [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: 06/19/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Dental pulp stem cells (DPSCs) have acquired noteworthy attention for their application in treating ischemic diseases and facilitating tissue regeneration. However, the host's immune response following allogenic DPSC transplantation often handicaps the long-term survival of transplanted cells, thereby limiting the application of DPSCs in cell therapy. This study aims to investigate whether genetic modification can alleviate the immunogenicity of DPSCs. METHODS Beta 2-microglobulin (B2M) and the class II histocompatibility complex transactivator (CIITA) were individually knocked down in DPSCs by lentiviral particles encoding short hairpin (sh) RNAs. The self-renewal capacity and pluripotency of DPSCs-shB2M (B2M silenced DPSCs) and DPSCs-shCIITA (CIITA silenced DPSCs) were evaluated by CCK8 and differentiation assays including osteogenesis, adipogenesis, and neurogenesis. The expression of HLA-I and HLA-II in DPSCs-shB2M and DPSCs-shCIITA after IFN-γ treatment were analyzed by western blotting, immunofluorescence, and flow cytometry. The function of genetically modified cells was assessed by leukocyte-mediated cytotoxicity and T-cell proliferation assays. RESULTS Western blotting, immunofluorescence, and flow cytometry revealed that DPSCs-shB2M and DPSCs-shCIITA exhibited impaired IFN-γ inducible HLA-I and HLA-II expression. There were no significant differences in the self-renewal capacity and pluripotency among DPSCs-shB2M, DPSCs-shCIITA, and control groups (p > 0.05). Lower leukocyte-mediated cytotoxicity and higher cell survival rates were found in DPSCs-shB2M and DPSCs-shCIITA groups compared to the control (p < 0.05). T cell proliferation was significantly inhibited in both DPSCs-shB2M and DPSCs-shCIITA groups (p < 0.05). CONCLUSION Genetic knockdown of B2M or CIITA in DPSCs substantially reduced their immunogenicity without compromising their stemness, thereby broadening the clinical application of DPSCs in cell therapy and tissue regeneration.
Collapse
Affiliation(s)
- Mingxin Hu
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Yuchen Zhang
- Obstetrics Department, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junqing Liu
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Yihan Chen
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Jun Kang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Jialin Zhong
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Shulan Lin
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Ye Liang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Rong Cen
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China
| | - Xiaofei Zhu
- Department of Endodontics, Henry M. Goldman School of Dental Medicine, Boston University, Boston, USA
| | - Chengfei Zhang
- Restorative Dental Sciences, Endodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, SAR, China.
| |
Collapse
|
27
|
Huang T, Jiang K, Li L, Li G, Cao Y, Huang X. Hsa_circ_0000423 promotes colorectal cancer EMT and immune escape by competitive adsorption of miR-369-3p mediating CCND1 expression. Discov Oncol 2024; 15:634. [PMID: 39520607 PMCID: PMC11550305 DOI: 10.1007/s12672-024-01501-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND This investigation evaluated the mechanism of hsa_circ_0000423 in colorectal cancer (CRC). METHODS The hsa_circ_0000423 gene was identified by bioinformatics analyses of GEO circRNA microarrays, and its expression in CRC was investigated. Based on this, in vitro experiments were conducted. Assays with dual luciferase reporter and RIP were conducted to detect interactions between hsa_circ_0000423, miR-369-3p and CCND1. Cell proliferation was measured by MTT and colony formation assay assays, apoptosis was detected by flow cytometry, migration and invasion were detected by Transwell, and expression of EMT-related proteins was detected by Western Blot. SW480 cells and T cells were co-cultured to assess immune escape. RESULTS hsa_circ_0000423 and CCND1 were elevated in CRC while miR-369-3p was downregulated Silencing hsa_circ_0000423 resulted in reduced CCND1 expression by upregulating miR-369-3p. Overexpressing CCND1 or down-regulating miR-369-3p both interrupted the anti-tumor role of silencing hsa_circ_0000423 on CRC cells. CONCLUSION Hsa_circ_0000423 promotes CCND1 expression through competitive binding of miR-369-3p and promotes CRC cell development and immune escape.
Collapse
Affiliation(s)
- TianFu Huang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Youjiang Medical Universityfor Nationalities, Baise, 533000, Guangxi Zhuang, China
| | - KaiHai Jiang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2Nd Road, Baise, 533000, Guangxi Zhuang, China
| | - LinTao Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2Nd Road, Baise, 533000, Guangxi Zhuang, China
| | - GuangSheng Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2Nd Road, Baise, 533000, Guangxi Zhuang, China
| | - YuSheng Cao
- Department of General Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi Zhuang, China
| | - XuSen Huang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2Nd Road, Baise, 533000, Guangxi Zhuang, China.
| |
Collapse
|
28
|
Zhang G, Liu J, Li S, Wang T, Chen L, Li H, Ding Q, Li X, Zhu S, Tang X. Cytochalasin H enhances sensitivity to gefitinib in non-small-cell lung cancer cells through inhibiting EGFR activation and PD-L1 expression. Sci Rep 2024; 14:25276. [PMID: 39455693 PMCID: PMC11512071 DOI: 10.1038/s41598-024-76060-2] [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: 06/14/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
In our previous study, we have isolated cytochalasin H (CyH) from endophytic fungus derived from mangrove plant and found that CyH inhibited the proliferation of non-small cell lung cancer (NSCLC) cells. Recently, epidermal growth factor receptor (EGFR) activation and programmed cell death 1 ligand (PD-L1) expression have been demonstrated to mediate NSCLC resistance to gefitinib, first-generation EGFR tyrosine kinase inhibitor (EGFR-TKI). Here, we further investigated the effect of CyH on EGFR activation, PD-L1 expression, and gefitinib sensitivity in NSCLC cell lines, A549 (wild-type EGFR), HCC827 (EGFR mutation), and NCI-H1975 (dual EGFR mutations and acquired gefitinib resistance) and animal model. Our results showed that CyH significantly inhibited EGFR activation and PD-L1 expression in NSCLC cells. Additionally, CyH dramatically promoted the inhibitory effect of gefitinib on the proliferation of A549 and HCC827 cells, and enhanced the sensitivity to gefitinib in NCI-H1975 cells. Moreover, CyH increased the inhibitory effect of gefitinib on EGFR activation and PD-L1 expression in HCC827 and NCI-H1975 cells. Animal experiments further demonstrated that CyH significantly promoted the inhibitory effect of gefitinib on the growth of NSCLC and the expression of Ki-67, p-EGFR, and PD-L1 in NCI-H1975 NSCLC xenograft tumors of nude mice. Furthermore, CyH inhibited the activation of JAK3/STAT signaling pathway. Taken together, our findings suggest that CyH promotes the sensitivity to gefitinib in NSCLC cells through the inhibition of EGFR activation and PD-L1 expression.
Collapse
Affiliation(s)
- Guihong Zhang
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Jiao Liu
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Sanzhong Li
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Tianyu Wang
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Li Chen
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Huan Li
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Qingkai Ding
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Xiangyong Li
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan, 523808, People's Republic of China
| | - Shaoping Zhu
- Center for Animal, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China
| | - Xudong Tang
- Collaborative Innovation Center for Antitumor Active Substance Research and Development, Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, People's Republic of China.
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Medical University, Dongguan, 523808, People's Republic of China.
| |
Collapse
|
29
|
Mori S, Nakamura K, Shimamura M, Ohe K. Use of Upadacitinib to Treat a Severe Flare-Up of Rheumatoid Arthritis During Anti-PD-1 Immune Checkpoint Inhibitor Therapy for Stage IV Squamous Cell Carcinoma of the Lung. J Clin Med 2024; 13:6257. [PMID: 39458207 PMCID: PMC11508340 DOI: 10.3390/jcm13206257] [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: 09/23/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Immune checkpoint inhibitor (ICI) therapy is becoming the standard of care for the treatment of advanced non-small-cell lung cancer. However, T-cell activation by ICIs frequently induces a flare-up of preexisting autoimmune diseases such as rheumatoid arthritis (RA). Janus kinase (JAK) inhibitors are increasingly used in the treatment of RA, but they could interfere with the efficacy of ICIs by inhibiting interferon signaling. Case Report: Here, we describe a case in which upadacitinib, a JAK1-selective inhibitor, was used to manage a severe RA flare-up occurring during ICI therapy with pembrolizumab, an anti-programmed cell death protein-1 antibody. A 54-year-old man with RA was diagnosed with grade IV lung squamous cell carcinoma. The patient had maintained RA remission for 4 years at the time of lung cancer diagnosis. After seven cycles of pembrolizumab therapy, the size of the primary tumor was markedly reduced, but a severe RA flare-up and organizing pneumonia (OP)-like pulmonary lesions occurred. Considering the severity of the flare-up, pembrolizumab was discontinued. Upadacitinib induced swift recovery from the RA flare-up and OP. Eleven months after the last pembrolizumab use, almost all metastatic lesions in the body had disappeared. We did not observe recurrence of lung cancer for more than 1 year during upadacitinib therapy. Conclusions: Upadacitinib could be a safe and effective option to treat severe RA flare-ups occurring during anti-PD-1 ICI therapy.
Collapse
Affiliation(s)
- Shunsuke Mori
- Department of Rheumatology, Clinical Research Center for Rheumatic Diseases, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| | - Kazuyoshi Nakamura
- Department of Respiratory Medicine, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| | - Minori Shimamura
- Department of Respiratory Medicine, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| | - Kouhei Ohe
- Department of Respiratory Medicine, National Hospital Organization (NHO) Kumamoto Saishun Medical Center, Kohshi 861-1196, Kumamoto, Japan
| |
Collapse
|
30
|
Li F, Yang Z, Savage TM, Vincent RL, de los Santos-Alexis K, Ahn A, Rouanne M, Mariuzza DL, Danino T, Arpaia N. Programmable bacteria synergize with PD-1 blockade to overcome cancer cell-intrinsic immune resistance mechanisms. Sci Immunol 2024; 9:eadn9879. [PMID: 39423284 PMCID: PMC11984541 DOI: 10.1126/sciimmunol.adn9879] [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: 01/10/2024] [Revised: 06/07/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
Interferon-γ (IFN-γ) is a potent cytokine critical for response to immunotherapy, yet conventional methods to systemically deliver this cytokine have been hindered by severe dose-limiting toxicities. Here, we engineered a strain of probiotic bacteria that home to tumors and locally release IFN-γ. A single intratumoral injection of these IFN-γ-producing bacteria was sufficient to drive systemic tumor antigen-specific antitumor immunity, without observable toxicity. Although cancer cells use various resistance mechanisms to evade immune responses, bacteria-derived IFN-γ overcame primary resistance to programmed cell death 1 (PD-1) blockade via activation of cytotoxic Foxp3-CD4+ and CD8+ T cells. Moreover, by activating natural killer (NK) cells, bacteria-derived IFN-γ also overcame acquired resistance mechanisms to PD-1 blockade, specifically loss-of-function mutations in IFN-γ signaling and antigen presentation pathways. Collectively, these results demonstrate the promise of combining IFN-γ-producing bacteria with PD-1 blockade as a therapeutic strategy for overcoming immunotherapy-resistant, locally advanced, and metastatic disease.
Collapse
Affiliation(s)
- Fangda Li
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Zaofeng Yang
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Thomas M. Savage
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Rosa L. Vincent
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | | - Alexander Ahn
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Mathieu Rouanne
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Dylan L. Mariuzza
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| | - Nicholas Arpaia
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
31
|
Tsai CL, Tang YH, Yang LY, Chao A, Wang CJ, Lin CY, Lai CH. Inhibition of nucleophosmin/B23 sensitizes ovarian cancer cells to immune check-point blockade via PD-L1 in ovarian cancer. J Formos Med Assoc 2024; 123:1045-1056. [PMID: 38821736 DOI: 10.1016/j.jfma.2024.05.020] [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: 05/15/2023] [Revised: 03/05/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) that against programmed cell death protein-1 (PD-1) and its ligand PD-L1 have been approved as a promising treatment of many human cancers. However, the responses to these ICIs were limited in patients with ovarian cancer. Studies have indicated that the response to PD-1/PD-L1 blockade might be correlated with the PD-L1 expression level in cancer cells. Nucleophosmin (NPM/B23) was found to be a potential target for immunotherapy. Whether NPM/B23 plays a role in cancer-associated immunity, such as PD-1/PD-L1 axis, and its underlying mechanisms remain largely unknown in ovarian cancer. METHODS We applied ovarian cancer cell lines as research models. The effect of modulating PD-L1 by NPM/B23 was subsequently confirmed via Western blot, flow cytometry, qRT-PCR, luciferase reporter assays, and immunoprecipitation. Protein stability and ubiquitin assay assays were used to analyze the interplay between NPM/B23 and NF-ĸB/p65 in PD-L1 regulation. The MOSEC/Luc xenograft mouse model was used to validate the role of NPM/B23-PD-L1 through tumor growth in vivo. RESULTS Our results revealed that NPM/B23 negatively regulates PD-L1 expression via a protein complex with NF-κB/p65 and through an IFN-γ pathway. Moreover, NPM/B23 inhibitor/modulator sensitized ovarian cancer cells to the anti-PD-1 antibody by regulating PD-L1 expression in the immunocompetent mouse model. Compared to anti-PD-1 antibody alone, a combination of anti-PD-1 antibody and NPM/B23 inhibitor/modulator showed reduced tumorigenesis and increased CD8+ T-cell expansion, thus contributing to prolonged survival on MOSEC/Luc-bearing mouse model. CONCLUSION Targeting NPM/B23 is a novel and potential therapeutic approach to sensitize ovarian cancer cells to immunotherapy.
Collapse
Affiliation(s)
- Chia-Lung Tsai
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Yun-Hsin Tang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taiwan
| | - Lan-Yan Yang
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan; Biostatics Unit, and Clinical Trial Center, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taoyuan, Taiwan; Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Angel Chao
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taiwan
| | - Chin-Jung Wang
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taiwan
| | - Chiao-Yun Lin
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taiwan.
| | - Chyong-Huey Lai
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital, Linkou Branch, and Chang Gung University, College of Medicine, Taoyuan, Taiwan; Gynecologic Cancer Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taiwan.
| |
Collapse
|
32
|
Snyder M, Wang Z, Lara B, Fimbres J, Pichardo T, Mazzilli S, Khan MM, Duggineni VK, Monti S, Sherr DH. The Aryl Hydrocarbon Receptor Controls IFNγ-Induced Immune Checkpoints PD-L1 and IDO via the JAK/STAT Pathway in Lung Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607602. [PMID: 39185148 PMCID: PMC11343147 DOI: 10.1101/2024.08.12.607602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
While immunotherapy has shown efficacy in lung adenocarcinoma (LUAD) patients, many respond only partially or not at all. One limitation in improving outcomes is the lack of a complete understanding of immune checkpoint regulation. Here, we investigated a possible link between an environmental chemical receptor implicated in lung cancer and immune regulation, (the aryl hydrocarbon receptor/AhR), a known but counterintuitive mediator of immunosuppression (IFNγ), and regulation of two immune checkpoints (PD-L1 and IDO). AhR gene-edited LUAD cell lines, a syngeneic LUAD mouse model, bulk- and scRNA sequencing of LUADs and tumor-infiltrating leukocytes were used to map out a signaling pathway leading from IFNγ through the AhR to JAK/STAT, PD-L1, IDO, and tumor-mediated immunosuppression. The data demonstrate that: 1) IFNγ activation of the JAK/STAT pathway leading to PD-L1 and IDO1 upregulation is mediated by the AhR in murine and human LUAD cells, 2) AhR-driven IDO1 induction results in the production of Kynurenine (Kyn), an AhR ligand, which likely mediates an AhR→IDO1→Kyn→AhR amplification loop, 3) transplantation of AhR-knockout LUAD cells results in long-term tumor immunity in most recipients. 4) The 23% of AhR-knockout tumors that do grow do so at a much slower pace than controls and exhibit higher densities of CD8+ T cells expressing markers of immunocompetence, increased activity, and increased cell-cell communication. The data definitively link the AhR to IFNγ-induced JAK/STAT pathway and immune checkpoint-mediated immunosuppression and support the targeting of the AhR in the context of LUAD.
Collapse
Affiliation(s)
- Megan Snyder
- Graduate Program in Genetics and Genomics, Boston University School of Medicine
| | - Zhongyan Wang
- Department of Environmental Health, Boston University School of Public Health
| | - Brian Lara
- Department of Environmental Health, Boston University School of Public Health
| | - Jocelyn Fimbres
- Department of Environmental Health, Boston University School of Public Health
| | | | | | - Mohammed Muzamil Khan
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine
| | - Vinay K. Duggineni
- Department of Environmental Health, Boston University School of Public Health
| | - Stefano Monti
- Section of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine
| | - David H. Sherr
- Department of Environmental Health, Boston University School of Public Health
| |
Collapse
|
33
|
Wei LJ, Fu J, Yang HX, Yang X, Liang HY, Luo RZ, Liu LL. Evaluation of pathological response to neoadjuvant chemotherapy in locally advanced cervical cancer. J Transl Med 2024; 22:655. [PMID: 39004706 PMCID: PMC11247755 DOI: 10.1186/s12967-024-05482-3] [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: 04/12/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Neoadjuvant chemotherapy (NACT) is a viable therapeutic option for women diagnosed locally advanced cervical cancer (LACC). However, the factors influencing pathological response are still controversial. We collected pair specimens of 185 LACC patients before and after receiving NACT and conducted histological evaluation. 8 fresh tissues pre-treatment were selected from the entire cohort to conducted immune gene expression profiling. A novel pathological grading system was established by comprehensively assessing the percentages of viable tumor, inflammatory stroma, fibrotic stroma, and necrosis in the tumor bed. Then, 185 patients were categorized into either the good pathological response (GPR) group or the poor pathological response (PPR) group post-NACT, with 134 patients (72.4%, 134/185) achieving GPR. Increasing tumor-infiltrating lymphocytes (TILs) and tumor-infiltrating lymphocytes volume (TILV) pre-treatment were correlated with GPR, with TILV emerging as an independent predictive factor for GPR. Additionally, CIBERSORT analysis revealed noteworthy differences in the expression of immune makers between cPR and non-cPR group. Furthermore, a significantly heightened density of CD8 + T cells and a reduced density of FOXP3 + T cells were observed in GPR than PPR. Importantly, patients exhibiting GPR or inflammatory type demonstrated improved overall survival and disease-free survival. Notably, stromal type was an independent prognostic factor in multivariate analysis. Our study indicates the elevated TILV in pre-treatment specimens may predict a favorable response to NACT, while identifying stromal type in post-treatment specimens as an independent prognostic factor. Moreover, we proposed this pathological grading system in NACT patients, which may offer a more comprehensive understanding of treatment response and prognosis.
Collapse
Affiliation(s)
- Li-Jun Wei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, 510060, Guangdong, China
| | - Jia Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, 510060, Guangdong, China
| | - Hai-Xia Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, The Second Affiliated Hospital of Shenzhen University, Shenzhen, 518101, China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, 510060, Guangdong, China
| | - Hao-Yu Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, 510060, Guangdong, China
| | - Rong-Zhen Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, 510060, Guangdong, China.
| | - Li-Li Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, 651# Dong Feng Road East, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
34
|
Kono K, Nakajima S, Mimura K. Biomarker-oriented chemo-immunotherapy for advanced gastric cancer. Int J Clin Oncol 2024; 29:865-872. [PMID: 38647874 DOI: 10.1007/s10147-024-02525-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: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
The biomarker-oriented chemo-immunotherapy is useful and promising in the development of new anticancer agents, since the responders can be enriched by selecting patients with biomarkers. Compared to colorectal and lung cancers, the development of biomarker-driven molecular-targeted therapeutics for gastric cancers has been straggled. However, several new biomarkers in gastric cancers have been discovered and clinical trials in enrichment design with certain biomarkers have been conducted. Therefore, there are currently several treatment options to treat gastric cancer patients based on individual biomarker-oriented strategies. In the present review, we describe the useful biomarkers in gastric cancer, with focusing on HER2, PD-L1, and Claudin18.2, in relation to their clinical significance and associated targeted agents.
Collapse
Affiliation(s)
- Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan.
| | - Shotaro Nakajima
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Multidisciplinary Treatment of Cancer and Regional Medical Support, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima City, Fukushima, 960-1295, Japan
| | - Kosaku Mimura
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
- Department of Blood Transfusion and Transplantation Immunology, Fukushima Medical University School of Medicine, Fukushima City, Fukushima, 960-1295, Japan
| |
Collapse
|
35
|
Salminen A. The role of the immunosuppressive PD-1/PD-L1 checkpoint pathway in the aging process and age-related diseases. J Mol Med (Berl) 2024; 102:733-750. [PMID: 38600305 PMCID: PMC11106179 DOI: 10.1007/s00109-024-02444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024]
Abstract
The accumulation of senescent cells within tissues is a hallmark of the aging process. Senescent cells are also commonly present in many age-related diseases and in the cancer microenvironment. The escape of abnormal cells from immune surveillance indicates that there is some defect in the function of cytotoxic immune cells, e.g., CD8+ T cells and natural killer (NK) cells. Recent studies have revealed that the expression of programmed death-ligand 1 (PD-L1) protein is abundantly increased in senescent cells. An increase in the amount of PD-L1 protein protects senescent cells from clearance by the PD-1 checkpoint receptor in cytotoxic immune cells. In fact, the activation of the PD-1 receptor suppresses the cytotoxic properties of CD8+ T and NK cells, promoting a state of immunosenescence. The inhibitory PD-1/PD-L1 checkpoint pathway acts in cooperation with immunosuppressive cells; for example, activation of PD-1 receptor can enhance the differentiation of regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC), and M2 macrophages, whereas the cytokines secreted by immunosuppressive cells stimulate the expression of the immunosuppressive PD-L1 protein. Interestingly, many signaling pathways known to promote cellular senescence and the aging process are crucial stimulators of the expression of PD-L1 protein, e.g., epigenetic regulation, inflammatory mediators, mTOR-related signaling, cGAS-STING pathway, and AhR signaling. It seems that the inhibitory PD-1/PD-L1 immune checkpoint axis has a crucial role in the accumulation of senescent cells and thus it promotes the aging process in tissues. Thus, the blockade of the PD-1/PD-L1 checkpoint signaling might be a potential anti-aging senolytic therapy. KEY MESSAGES: Senescent cells accumulate within tissues during aging and age-related diseases. Senescent cells are able to escape immune surveillance by cytotoxic immune cells. Expression of programmed death-ligand 1 (PD-L1) markedly increases in senescent cells. Age-related signaling stimulates the expression of PD-L1 protein in senescent cells. Inhibitory PD-1/PD-L1 checkpoint pathway suppresses clearance of senescent cells.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
36
|
Lin X, Kang K, Chen P, Zeng Z, Li G, Xiong W, Yi M, Xiang B. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer 2024; 23:108. [PMID: 38762484 PMCID: PMC11102195 DOI: 10.1186/s12943-024-02023-w] [Citation(s) in RCA: 130] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Immune evasion contributes to cancer growth and progression. Cancer cells have the ability to activate different immune checkpoint pathways that harbor immunosuppressive functions. The programmed death protein 1 (PD-1) and programmed cell death ligands (PD-Ls) are considered to be the major immune checkpoint molecules. The interaction of PD-1 and PD-L1 negatively regulates adaptive immune response mainly by inhibiting the activity of effector T cells while enhancing the function of immunosuppressive regulatory T cells (Tregs), largely contributing to the maintenance of immune homeostasis that prevents dysregulated immunity and harmful immune responses. However, cancer cells exploit the PD-1/PD-L1 axis to cause immune escape in cancer development and progression. Blockade of PD-1/PD-L1 by neutralizing antibodies restores T cells activity and enhances anti-tumor immunity, achieving remarkable success in cancer therapy. Therefore, the regulatory mechanisms of PD-1/PD-L1 in cancers have attracted an increasing attention. This article aims to provide a comprehensive review of the roles of the PD-1/PD-L1 signaling in human autoimmune diseases and cancers. We summarize all aspects of regulatory mechanisms underlying the expression and activity of PD-1 and PD-L1 in cancers, including genetic, epigenetic, post-transcriptional and post-translational regulatory mechanisms. In addition, we further summarize the progress in clinical research on the antitumor effects of targeting PD-1/PD-L1 antibodies alone and in combination with other therapeutic approaches, providing new strategies for finding new tumor markers and developing combined therapeutic approaches.
Collapse
Affiliation(s)
- Xin Lin
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Kuan Kang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Pan Chen
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- FuRong Laboratory, Changsha, 410078, Hunan, China
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China
| | - Mei Yi
- Department of Dermotology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Bo Xiang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- FuRong Laboratory, Changsha, 410078, Hunan, China.
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410008, Hunan, China.
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Central South University, Changsha, 410078, Hunan, China.
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
37
|
Deng P, Dong X, Wu Z, Hou X, Mao L, Guo J, Zhao W, Peng C, Zhang Z, Peng L. Development of Glycosylation-Modified DPPA-1 Compounds as Innovative PD-1/PD-L1 Blockers: Design, Synthesis, and Biological Evaluation. Molecules 2024; 29:1898. [PMID: 38675717 PMCID: PMC11054459 DOI: 10.3390/molecules29081898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
In the context of peptide drug development, glycosylation plays a pivotal role. Accordingly, L-type peptides were synthesized predicated upon the PD-1/PD-L1 blocker DPPA-1. Subsequent glycosylation resulted in the production of two distinct glycopeptides, D-glu-LPPA-1 and D-gal-LPPA-1, by using D-glucose (D-glu) and D-galactose (D-gal), respectively, during glycosylation. Both glycopeptides significantly inhibited the interaction between PD-1 and PD-L1, and the measured half maximal inhibitory concentrations (IC50s) were 75.5 μM and 101.9 μM for D-glu-LPPA-1 and D-gal-LPPA-1, respectively. Furthermore, D-gal-LPPA-1 displayed a pronounced ability to restore T-cell functionality. In an MC38 tumor-bearing mouse model, D-gal-LPPA-1 demonstrated a significant inhibitory effect. Notably, D-gal-LPPA-1 substantially augmented the abundance and functionality of CD8+ T cells in the tumor microenvironment. Additionally, in the lymph nodes and spleens, D-gal-LPPA-1 significantly increased the proportion of CD8+ T cells secreting interferon-gamma (IFN-γ). These strong findings position D-gal-LPPA-1 as a potent enhancer of the antitumor immune response in MC38 tumor-bearing mice, underscoring its potential as a formidable PD-1/PD-L1 blocking agent.
Collapse
Affiliation(s)
- Peng Deng
- Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Xiaodan Dong
- Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Ziyuan Wu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Road, Luoyang 471003, China
| | - Xixi Hou
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Longfei Mao
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, 263 Kaiyuan Road, Luoyang 471003, China
| | - Jingjing Guo
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao 999078, China;
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China;
| | - Chune Peng
- Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Zhe Zhang
- School of Sciences, Henan University of Technology, Zhengzhou 450001, China
| | - Lizeng Peng
- Key Laboratory of Novel Food Resources Processing Ministry of Agriculture, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| |
Collapse
|
38
|
Wang R, Zhao Y. Effects of Metformin on JNK Signaling Pathway and PD-L1 Expression in Triple Negative Breast Cancer. Cancer Manag Res 2024; 16:259-268. [PMID: 38585433 PMCID: PMC10998504 DOI: 10.2147/cmar.s454960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/27/2024] [Indexed: 04/09/2024] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer. Metformin has been shown to have the potential to inhibit the proliferation of malignant cells. This study aimed to investigate the regulatory effect of metformin on the expression of programmed death protein ligand 1(PD-L1) and mechanisms in TNBC. Methods Mouse breast cancer cell line 4T1 was co-cultured with metformin, and the effect of metformin on cell proliferation was detected by MTT assay. The effect of metformin on the expression of JNK, RSK2 and CREB was detected by MAPK pathway protein chip. BALB/c mice were inoculated with 4T1 cells with knockdown/overexpression of C-Jun N-terminal kinase (JNK), and administered with metformin. The weight of tumor tissue was observed at the end of the experiment. The expression of PD-L1 in tumor cells was observed by immunofluorescence staining and the level of INF-γwas quantitatively determined by ELISA. Results Metformin inhibited the viability of 4T1 cells and increased the phosphorylation of JNK to reduce the phosphorylation of RSK2 and CREB. Metformin and JNK knockdown reduced the expression of PD-L1 in tumor cells, but there was no significant difference in the weight of tumor tissue. Metformin can reduce the level of INF-γ in tumor tissues, but JNK has no effect. Conclusion Metformin can inhibit the expression of PD-L1 in triple-negative breast cancer mice and improve the tumor microenvironment, but does not reduce the size of the tumor.
Collapse
Affiliation(s)
- Ruibin Wang
- Department of Emergency, Beijing Shijitan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yanjie Zhao
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
39
|
Chen M, Wang S. Preclinical development and clinical studies of targeted JAK/STAT combined Anti-PD-1/PD-L1 therapy. Int Immunopharmacol 2024; 130:111717. [PMID: 38387193 DOI: 10.1016/j.intimp.2024.111717] [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: 11/13/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Programmed cell death protein 1 (PD-1) binds to its ligand to help tumours evade the immune system and promote tumour progression. Although anti-PD-1/PD-L1 therapies show powerful effects in some patients, most patients are unable to benefit from this treatment due to treatment resistance. Therefore, it is important to overcome tumour resistance to PD-1/PD-L1 blockade. There is substantial evidence suggesting that the JAK/STAT signalling pathway plays a significant role in PD-1/PD-L1 expression and anti-PD-1/PD-L1 treatment. Herein, we describe the effects of the JAK/STAT signalling pathway on PD-1/PD-L1. Subsequently, the relationship between molecular mutations in the JAK/STAT signalling pathway and immune resistance was analysed. Finally, the latest advancements in drugs targeting the JAK/STAT pathway combined with PD1/PD-L1 inhibitors are summarised.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
40
|
Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM, Wang H. RNA modifications in cellular metabolism: implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther 2024; 9:70. [PMID: 38531882 DOI: 10.1038/s41392-024-01777-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Cellular metabolism is an intricate network satisfying bioenergetic and biosynthesis requirements of cells. Relevant studies have been constantly making inroads in our understanding of pathophysiology, and inspiring development of therapeutics. As a crucial component of epigenetics at post-transcription level, RNA modification significantly determines RNA fates, further affecting various biological processes and cellular phenotypes. To be noted, immunometabolism defines the metabolic alterations occur on immune cells in different stages and immunological contexts. In this review, we characterize the distribution features, modifying mechanisms and biological functions of 8 RNA modifications, including N6-methyladenosine (m6A), N6,2'-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N4-acetylcytosine (ac4C), N7-methylguanosine (m7G), Pseudouridine (Ψ), adenosine-to-inosine (A-to-I) editing, which are relatively the most studied types. Then regulatory roles of these RNA modification on metabolism in diverse health and disease contexts are comprehensively described, categorized as glucose, lipid, amino acid, and mitochondrial metabolism. And we highlight the regulation of RNA modifications on immunometabolism, further influencing immune responses. Above all, we provide a thorough discussion about clinical implications of RNA modification in metabolism-targeted therapy and immunotherapy, progression of RNA modification-targeted agents, and its potential in RNA-targeted therapeutics. Eventually, we give legitimate perspectives for future researches in this field from methodological requirements, mechanistic insights, to therapeutic applications.
Collapse
Affiliation(s)
- Wei-Wei Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Clinical Medicine, Shandong University, Jinan, China
| | - Si-Qing Zheng
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Tian Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Yun-Fei Fei
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Chen Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Shuang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China
| | - Fei Wang
- Neurosurgical Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Guan-Min Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| | - Hao Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Core Unit of National Clinical Research Center for Laboratory Medicine, Hefei, China.
| |
Collapse
|
41
|
Zhao B, Zheng X, Wang Y, Cheng N, Zhong Y, Zhou Y, Huang J, Wang F, Qi X, Zhuang Q, Wang Y, Liu X. Lnc-CCNH-8 promotes immune escape by up-regulating PD-L1 in hepatocellular carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102125. [PMID: 38356866 PMCID: PMC10865404 DOI: 10.1016/j.omtn.2024.102125] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy with poor prognosis. In recent years, immune checkpoint inhibitors (ICIs) have enabled breakthroughs in the clinical treatment of patients with HCC, but the overall response rate to ICIs in HCC patients is still low, and no validated biomarker is available to guide clinical decision making. Here, we demonstrated that the long non-coding RNA Lnc-CCNH-8 is highly expressed in HCC and correlates with poor prognosis. Functionally, elevated Lnc-CCNH-8 inactivated co-cultured T cells in vitro and compromised antitumor immunity in an immunocompetent mouse model. Mechanistically, up-regulated Lnc-CCNH-8 can sponge microRNA (miR)-217 to regulate the expression of PD-L1. In addition, Lnc-CCNH-8 can also stabilize PD-L1 through miR-3173/PKP3 axis. Furthermore, mice bearing tumors with high Lnc-CCNH-8 expression had significant therapeutic sensitivity to anti-PD-L1 monoclonal antibody treatment. More important, HCC patients with high levels of plasma exosomal Lnc-CCNH-8 had a better therapeutic response to ICIs. Taken together, our results reveal the function of Lnc-CCNH-8 in inducing immune escape from CD8+ T-cell-mediated killing by up-regulating PD-L1 in a miR-217/miR-3173-dependent manner, which also reveals a novel mechanism of PD-L1 regulation in HCC, and exosomal Lnc-CCNH-8 can serve as a predictive marker for immunotherapy response in HCC.
Collapse
Affiliation(s)
- Bixing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Xiaoyuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Yang Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
| | - Niangmei Cheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Yue Zhong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
| | - Yang Zhou
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Jingyun Huang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Xin Qi
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
| | - Qiuyu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| |
Collapse
|
42
|
Asadi M, Zarredar H, Zafari V, Soleimani Z, Saeedi H, Caner A, Shanehbandi D. Immune Features of Tumor Microenvironment: A Genetic Spotlight. Cell Biochem Biophys 2024; 82:107-118. [PMID: 37870699 DOI: 10.1007/s12013-023-01192-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
A tumor represents a highly intricate tissue entity, characterized by an exceptionally complex microenvironment that starkly contrasts with the typical physiological surroundings of healthy tissues. Within this tumor microenvironment (TME), every component and factor assume paramount importance in the progression of malignancy and exerts a pivotal influence on a patient's clinical outcome. One of the remarkable aspects of the TME is its remarkable heterogeneity, not only across different types of cancers but even within the same histological category of tumors. In-depth research has illuminated the intricate interplay between specific immune cells and molecules and the dynamic characteristics of the TME. Recent investigations have yielded compelling evidence that several mutations harbored by tumor cells possess the capacity to instigate substantial alterations in the TME. These mutations, often acting as drivers of tumorigenesis, can orchestrate a cascade of events that remodel the TME, thereby influencing crucial aspects of cancer behavior, including its invasiveness, immune evasion, and response to therapies. It is within this nuanced context that the present study endeavors to provide a concise yet comprehensive summary of how specific mutations, within the genetic landscape of cancer cells, can instigate profound changes in TME features. By elucidating the intricate relationship between genetic mutations and the TME, this research aims to contribute to a deeper understanding of cancer biology. Ultimately, the knowledge gained from this study holds the potential to inform the development of more targeted and effective treatments, thereby offering new hope to patients grappling with the complexities of cancer.
Collapse
Affiliation(s)
- Milad Asadi
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Venus Zafari
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey
| | - Zahra Soleimani
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Saeedi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayse Caner
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Turkey.
- The University of Texas, MD Anderson Cancer Center, Houston, USA.
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
43
|
Chen L, Song HY, Tan CY, Jiang HY, Gan XF, Bu L, Wei W, Lin SN. Mechanism for Anwei decoction to inhibit immune escape induced by programmed cell death protein 1/programmed cell death ligand 1 axis in rats with chronic atrophic gastritis. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:148-157. [DOI: 10.11569/wcjd.v32.i2.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
|
44
|
Huynh JC, Cho M, Monjazeb A, Al-Obeidi E, Singh A, Tam K, Lara F, Martinez A, Garcia L, Kim EJ. Phase I/II trial of BMS-986,205 and nivolumab as first line therapy in hepatocellular carcinoma. Invest New Drugs 2024; 42:35-43. [PMID: 38038862 PMCID: PMC10891185 DOI: 10.1007/s10637-023-01416-w] [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: 11/02/2023] [Accepted: 11/26/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Indoleamine-2,3-dioxygenase (IDO) helps orchestrate immune suppression and checkpoint inhibitor resistance in hepatocellular carcinoma (HCC). BMS-986,205 is a novel oral drug that potently and selectively inhibits IDO. This Phase I/II study evaluated the safety and tolerability of BMS-986,205 in combination with nivolumab as first-line therapy in advanced HCC. METHODS Adults with untreated, unresectable/metastatic HCC received BMS-986,205 at two dose levels (50-100 mg orally daily) in combination with fixed dose nivolumab (240mg/m2 IV on Day 1 of each 14-day cycle). The primary objective was to determine the safety and tolerability of this combination; secondary objectives were to obtain preliminary efficacy. RESULTS Eight patients received a total of 91 treatment cycles in the dose escalation phase. All patients were Child Pugh A and 6 patients had underlying viral hepatitis. In the 6 evaluable patients, no dose-limiting toxicities (DLTs) were observed. The most common treatment-related adverse events (TRAEs) were aspartate transaminase (AST) and alanine transaminase (ALT) elevation (3 patients) and diarrhea, maculopapular rash and increased alkaline phosphatase (2 patients each). Grade 3 events were diarrhea and AST elevation (1 patient), and hyperglycemia and pancreatitis requiring treatment discontinuation (1 patient). No grade 4-5 events occurred. Partial response was observed in 1 patient (12.5%) and stable disease in 3 patients (37.5%), yielding a disease control rate of 50%. Median PFS was 8.5 weeks; median OS was not reached. CONCLUSION Combination BMS-986,205 and nivolumab showed a manageable safety profile with durable benefit as first-line therapy in a meaningful subset of advanced HCC patients.
Collapse
Affiliation(s)
- Jasmine C Huynh
- Division of Hematology and Oncology, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - May Cho
- Division of Hematology and Oncology, Irvine Comprehensive Cancer Center, University of California, Orange, CA, 92868, USA
| | - Arta Monjazeb
- Department of Radiation Oncology, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - Ebaa Al-Obeidi
- Division of Hematology and Oncology, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - Amisha Singh
- Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA, 95817, USA
| | - Kit Tam
- Division of Hematology and Oncology, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - Frances Lara
- Office of Clinical Research, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - Anthony Martinez
- Office of Clinical Research, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - Leslie Garcia
- Office of Clinical Research, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA
| | - Edward J Kim
- Division of Hematology and Oncology, Davis Comprehensive Cancer Center, University of California, Sacramento, CA, 95817, USA.
| |
Collapse
|
45
|
Logghe T, van Zwol E, Immordino B, Van den Cruys K, Peeters M, Giovannetti E, Bogers J. Hyperthermia in Combination with Emerging Targeted and Immunotherapies as a New Approach in Cancer Treatment. Cancers (Basel) 2024; 16:505. [PMID: 38339258 PMCID: PMC10854776 DOI: 10.3390/cancers16030505] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in the development of novel therapies, cancer continues to stand as a prominent global cause of death. In many cases, the cornerstone of standard-of-care therapy consists of chemotherapy (CT), radiotherapy (RT), or a combination of both. Notably, hyperthermia (HT), which has been in clinical use in the last four decades, has proven to enhance the effectiveness of CT and RT, owing to its recognized potency as a sensitizer. Furthermore, HT exerts effects on all steps of the cancer-immunity cycle and exerts a significant impact on key oncogenic pathways. Most recently, there has been a noticeable expansion of cancer research related to treatment options involving immunotherapy (IT) and targeted therapy (TT), a trend also visible in the research and development pipelines of pharmaceutical companies. However, the potential results arising from the combination of these innovative therapeutic approaches with HT remain largely unexplored. Therefore, this review aims to explore the oncology pipelines of major pharmaceutical companies, with the primary objective of identifying the principal targets of forthcoming therapies that have the potential to be advantageous for patients by specifically targeting molecular pathways involved in HT. The ultimate goal of this review is to pave the way for future research initiatives and clinical trials that harness the synergy between emerging IT and TT medications when used in conjunction with HT.
Collapse
Affiliation(s)
- Tine Logghe
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Eke van Zwol
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Benoît Immordino
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy
| | | | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Johannes Bogers
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
46
|
Cho S, Kim W, Yoo D, Han Y, Hwang H, Kim S, Kim J, Park S, Park Y, Jo H, Pyun JC, Lee M. Impact of glucose metabolism on PD-L1 expression in sorafenib-resistant hepatocellular carcinoma cells. Sci Rep 2024; 14:1751. [PMID: 38243049 PMCID: PMC10798953 DOI: 10.1038/s41598-024-52160-x] [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: 08/10/2023] [Accepted: 01/15/2024] [Indexed: 01/21/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth leading cause of cancer-related mortality worldwide. Programmed cell death ligand-1 (PD-L1) is an immune checkpoint protein that binds to programmed cell death-1 (PD-1), which is expressed in activated T cells and other immune cells and has been employed in cancer therapy, including HCC. Recently, PD-L1 overexpression has been documented in treatment-resistant cancer cells. Sorafenib is a multikinase inhibitor and the only FDA-approved treatment for advanced HCC. However, several patients exhibit resistance to sorafenib during treatment. This study aimed to assess the effect of glucose deprivation on PD-L1 expression in HCC cells. We used PD-L1-overexpressing HepG2 cells and IFN-γ-treated SK-Hep1 cells to explore the impact of glycolysis on PD-L1 expression. To validate the correlation between PD-L1 expression and glycolysis, we analyzed data from The Cancer Genome Atlas (TCGA) and used immunostaining for HCC tissue analysis. Furthermore, to modulate PD-L1 expression, we treated HepG2, SK-Hep1, and sorafenib-resistant SK-Hep1R cells with rapamycin. Here, we found that glucose deprivation reduced PD-L1 expression in HCC cells. Additionally, TCGA data and immunostaining analyses confirmed a positive correlation between the expression of hexokinase II (HK2), which plays a key role in glucose metabolism, and PD-L1. Notably, rapamycin treatment decreased the expression of PD-L1 and HK2 in both high PD-L1-expressing HCC cells and sorafenib-resistant cells. Our results suggest that the modulation of PD-L1 expression by glucose deprivation may represent a strategy to overcome PD-L1 upregulation in patients with sorafenib-resistant HCC.
Collapse
Affiliation(s)
- Sua Cho
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Wonjin Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Dayoung Yoo
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Yeonju Han
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Hyemin Hwang
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Seunghwan Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Jimin Kim
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Sanghee Park
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Yusun Park
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - HanHee Jo
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Jae-Chul Pyun
- Department of Materials Science and Engineering, Yonsei University, 50 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Misu Lee
- Division of Life Sciences, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea.
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea.
| |
Collapse
|
47
|
Chen Z, Yao MW, Ao X, Gong QJ, Yang Y, Liu JX, Lian QZ, Xu X, Zuo LJ. The expression mechanism of programmed cell death 1 ligand 1 and its role in immunomodulatory ability of mesenchymal stem cells. Chin J Traumatol 2024; 27:1-10. [PMID: 38065706 PMCID: PMC10859298 DOI: 10.1016/j.cjtee.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 02/05/2024] Open
Abstract
Programmed cell death 1 ligand 1 (PD-L1) is an important immunosuppressive molecule, which inhibits the function of T cells and other immune cells by binding to the receptor programmed cell death-1. The PD-L1 expression disorder plays an important role in the occurrence, development, and treatment of sepsis or other inflammatory diseases, and has become an important target for the treatment of these diseases. Mesenchymal stem cells (MSCs) are a kind of pluripotent stem cells with multiple differentiation potential. In recent years, MSCs have been found to have a strong immunosuppressive ability and are used to treat various inflammatory insults caused by hyperimmune diseases. Moreover, PD-L1 is deeply involved in the immunosuppressive events of MSCs and plays an important role in the treatment of various diseases. In this review, we will summarize the main regulatory mechanism of PD-L1 expression, and discuss various biological functions of PD-L1 in the immune regulation of MSCs.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China; College of Basic Medical Sciences, Army Medical University, Chongqing, 400038, China
| | - Meng-Wei Yao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiang Ao
- Department of Orthopedics, 953 Hospital of PLA, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, Tibet Autonomous Region, China
| | - Qing-Jia Gong
- College of Basic Medical Sciences, Army Medical University, Chongqing, 400038, China
| | - Yi Yang
- Department of Rheumatology and Immunology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jin-Xia Liu
- Department of Obstetrics and Gynecology, Chongqing People's Hospital, Chongqing, 401121, China
| | - Qi-Zhou Lian
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Ling-Jing Zuo
- Department of Nuclear Medicine, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650034, China.
| |
Collapse
|
48
|
Minz AP, Mohapatra D, Dutta M, Sethi M, Parida D, Mohapatra AP, Mishra S, Kar S, Sasmal PK, Senapati S. Statins abrogate gemcitabine-induced PD-L1 expression in pancreatic cancer-associated fibroblasts and cancer cells with improved therapeutic outcome. Cancer Immunol Immunother 2023; 72:4261-4278. [PMID: 37926727 PMCID: PMC10992415 DOI: 10.1007/s00262-023-03562-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 10/17/2023] [Indexed: 11/07/2023]
Abstract
A combination of chemotherapy with immunotherapy has been proposed to have better clinical outcomes in Pancreatic Ductal Adenocarcinoma (PDAC). On the other hand, chemotherapeutics is known to have certain unwanted effects on the tumor microenvironment that may mask the expected beneficial effects of immunotherapy. Here, we have investigated the effect of gemcitabine (GEM), on two immune checkpoint proteins (PD-L1 and PD-L2) expression in cancer associated fibroblasts (CAFs) and pancreatic cancer cells (PCCs). Findings of in vitro studies conducted by using in-culture activated mouse pancreatic stellate cells (mPSCs) and human PDAC patients derived CAFs demonstrated that GEM significantly induces PD-L1 and PD-L2 expression in these cells. Moreover, GEM induced phosphorylation of STAT1 and production of multiple known PD-L1-inducing secretory proteins including IFN-γ in CAFs. Upregulation of PD-L1 in PSCs/CAFs upon GEM treatment caused T cell inactivation and apoptosis in vitro. Importantly, Statins suppressed GEM-induced PD-L1 expression both in CAFs and PCCs while abrogating the inactivation of T-cells caused by GEM-treated PSCs/CAFs. Finally, in an immunocompetent syngeneic orthotopic mouse pancreatic tumor model, simvastatin and GEM combination therapy significantly reduced intra-tumor PD-L1 expression and noticeably reduced the overall tumor burden and metastasis incidence. Together, the findings of this study have provided experimental evidence that illustrates potential unwanted side effects of GEM that could hamper the effectiveness of this drug as mono and/or combination therapy. At the same time the findings also suggest use of statins along with GEM will help in overcoming these shortcomings and warrant further clinical investigation.
Collapse
Affiliation(s)
- Aliva Prity Minz
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Debasish Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- CV Raman Global University, Bhubaneswar, Odisha, India
| | - Madhuri Dutta
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Manisha Sethi
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Deepti Parida
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Amlan Priyadarshee Mohapatra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Swayambara Mishra
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Salona Kar
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Prakash K Sasmal
- Department of General Surgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | | |
Collapse
|
49
|
Skacel J, Melichar B, Mohelnikova-Duchonova B, Lemstrova R. Hyperprogression on anti-PD-1 treatment. Is subsequent therapy feasible? A case report and review of the literature. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2023; 167:376-384. [PMID: 35703362 DOI: 10.5507/bp.2022.025] [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: 03/11/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Hyperprogressive disease (HPD) is a new phenomenon that has emerged in the immunotherapy era. HPD is defined as a rapid tumour growth with detrimental effect on the patient condition and disease course. The management and treatment following HPD is not defined. We present here the case report of patient with HPD and review of the literature on putative mechanisms of HPD and following disease management. METHODS AND RESULTS A 60-year old male patient with metastatic melanoma was indicated for systemic treatment with anti-programmed cell death (PD)-1 antibody. Rapid tumour growth and detrimental effect on the patient general condition after administration of a single dose of anti-PD-1 antibody met the criteria of HPD. The patient underwent the second line taxane-based chemotherapy with good tolerance and disease stabilization. The third line treatment with anti- cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibody ipilimumab was well tolerated and resulted in partial response. Re-challenge with anti-CTLA-4 antibody was feasible, but only with a modest clinical effect. CONCLUSION Prompt recognition of HPD and administration of salvage chemotherapy with taxane-based regimens may be crucial. HPD is rarely observed with ipilimumab treatment. Administration of ipilimumab as well as an ipilimumab re-challenge are feasible after HPD on anti-PD-1 antibodies. Investigation of new predictive biomarkers of HPD is warranted as well as new agents that potentiate the immune response in patients affected with this insidious complication.
Collapse
Affiliation(s)
- Jan Skacel
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| | - Beatrice Mohelnikova-Duchonova
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| | - Radmila Lemstrova
- Department of Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Zdravotniku 7, 779 00 Olomouc, Czech Republic
| |
Collapse
|
50
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [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: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|