1
|
Li J, Zhang G, Sun Z, Jiang M, Jia G, Liu H, Liu N, Shi L, Zhang L, Nie L, Zhang Y, Fu Y. Immunogenic cuproptosis in cancer immunotherapy via an in situ cuproptosis-inducing system. Biomaterials 2025; 319:123201. [PMID: 40020502 DOI: 10.1016/j.biomaterials.2025.123201] [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: 12/14/2024] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
Cell death-based therapies combined with immunotherapy have great potential in cancer therapy. To further explore and apply the combined therapies, the immunogenicity of different cell death modes in colorectal cancer (CRC) was evaluated by a cause-and-effect framework encompassing 12 cell death modes. Results show robust correlations among cuproptosis, immunogenic cell death (ICD) and immunity in CRC, as observed in our in-house and other independent cohorts, which are substantiated by in vitro and in vivo experiments. Subsequent investigations demonstrate that cuproptosis induces endoplasmic reticulum stress, leading to the release of damage-associated molecular patterns from CRC cells and triggering the maturation of antigen-presenting cells. Moreover, for in vivo therapeutic approaches, an in situ cuproptosis-inducing system was devised, which can further strengthen the effects of immune cells. Through the combined analysis including single-cell RNA sequencing, cuproptosis is shown to mobilize cytotoxic T lymphocytes and M1 macrophages within the tumor microenvironment (TME). Additionally, co-treatment with Imiquimod, the TLR7 agonist, augments the anti-tumor immune responses induced by cuproptosis. Overall, we provide compelling evidence that cuproptosis induces ICD thus fostering an inflammatory TME, and the cuproptosis-based delivery system further promotes this inflammatory environment, demonstrating considerable potential for enhancing tumor therapy efficacy.
Collapse
Affiliation(s)
- Jiehan Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Ge Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Zhao Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Meimei Jiang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Guiyun Jia
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Hao Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Nannan Liu
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Liyang Shi
- College of Biology, Hunan University, Changsha, 410082, China
| | - Lingling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Liming Nie
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Yingjie Zhang
- School of Biomedical Sciences, Hunan University, Changsha, 410082, China.
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, 450052, China.
| |
Collapse
|
2
|
Fu G, Liu Y, Qian C, Yang Z, Luo M, Cai W. SMARCD1 is a dual regulator of PD-L1 expression and cell proliferation facilitating tumor evasion. Pathol Res Pract 2025; 270:155975. [PMID: 40228401 DOI: 10.1016/j.prp.2025.155975] [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: 08/26/2024] [Revised: 03/04/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
PURPOSE Cancer cells often evade immune responses by overexpressing immune checkpoint regulators, such as programmed cell death ligand 1 (PD-L1). Identifying targets that regulate PD-L1 is a promising approach for anti-tumor therapy. METHODS Based on our previous CRISPR-Cas9 screening, we identified SMARCD1, a subunit of the mating-type switching/sucrose fermentation (SWI/SNF) complex, as a factor that promotes tumor evasion by inducing PD-L1-mediated immune checkpoint responses. Immunohistochemical staining (IHC) was used to assess SMARCD1 expression levels in colorectal cancer (CRC) and normal tissues. CRISPR-Cas9 technology was employed to generate SMARCD1 knockout (KO) cell lines. Western blotting and flow cytometry were used to evaluate PD-L1 expression. Cell proliferation, invasion, migration, and apoptosis were also assessed. A tumor model was established to examine the in vivo effects of SMARCD1. RNA-seq and ChIP-seq analyses were conducted to investigate the potential mechanisms. RESULTS SMARCD1 was significantly upregulated in CRC tissues. In vitro, SMARCD1 regulated PD-L1 expression and significantly promoted tumor growth. The SWI/SNF inhibitor FHT-1015 reversed the effects of SMARCD1 knockout. Mechanistically, SMARCD1 may maintain chromatin accessibility at the PD-L1 transcriptional regulatory element and promote cancer cell proliferation via the PI3K-Akt signaling pathway. CONCLUSION SMARCD1 regulates PD-L1 transcription and facilitates tumor cell proliferation, making it a promising target for CRC treatment.
Collapse
Affiliation(s)
- Gang Fu
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine affiliated Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, China
| | - Yutong Liu
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
| | - Chen Qian
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine affiliated Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, China
| | - Zheyu Yang
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine affiliated Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, China.
| | - Maocai Luo
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine affiliated Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, China.
| | - Wei Cai
- Department of General Surgery, Shanghai Jiao Tong University School of Medicine affiliated Ruijin Hospital, 197 Ruijin 2nd Road, Shanghai, China.
| |
Collapse
|
3
|
Liu WM, Li XB. East meets West: The winning combination against BRAF V600E metastatic colorectal cancer. World J Clin Oncol 2025; 16:102223. [DOI: 10.5306/wjco.v16.i5.102223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 05/19/2025] Open
Abstract
Metastatic colorectal cancer (mCRC) patients with BRAF V600E mutation have a poor prognosis despite the implementation of multiple treatment strategies. The integration of traditional Chinese medicine with Western medicine in treating BRAF mutant mCRC has garnered increasing attention. Recent studies indicate that combining traditional Chinese and modern Western medical approaches not only extend survival but also reduces the risk of mortality in patients with BRAF V600E mutant mCRC. This approach is particularly effective for colorectal cancer patients who have right-sided colon involvement, liver metastasis, or a history of radiotherapy or chemotherapy. In this treatment combination, traditional Chinese medicine may offer symptomatic relief and improve quality of life, while Western medicine targets the disease more aggressively with advanced pharmacological agents. Ongoing research is crucial to further elucidate the mechanisms underlying these benefits and to optimize treatment protocols.
Collapse
Affiliation(s)
- Wen-Ming Liu
- Department of Gastrointestinal Surgery, The First People’s Hospital of Tianmen, Tianmen 431700, Hubei Province, China
| | - Xiao-Bing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, Hubei Province, China
| |
Collapse
|
4
|
Huang X, Yang F, Gao B, Ge W, Gao L, Wu J, Tu S, Wang F. Free carrier-enhanced Bi/Bi 2S 3 nanoparticles enable precise OCT catheter-guided interventional photothermal therapy for colorectal cancer. Acta Biomater 2025; 198:401-412. [PMID: 40228616 DOI: 10.1016/j.actbio.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Current clinical colorectal cancer treatments usually possess unsatisfactory effects, mainly because of unavoidable surgical trauma and multidrug resistance. Precise and minimally invasive theragnostic technology has advanced through miniaturized catheter intervention with imaging-guided treatment methods; however, previously reported approaches cannot simultaneously perform in situ real-time imaging and therapy. We proposed a strategy of 0.9 mm catheter-based optical coherence tomography imaging-guided interventional photothermal therapy at 1310 nm for orthotopic colorectal cancer. Specifically, folate-modified Bi/Bi2S3 nanoparticles showed intense scattering signals and local hyperpyrexia under 1310 nm laser irradiation in vitro and in vivo due to the localized surface plasmon resonance effect, enabling imaging-guided precise tumor treatment. Histopathological and blood biochemistry analyses confirmed the high biosafety and negligible long-term toxicity of Bi/Bi2S3 nanoparticles. This new method offers a feasible methodology for catheter-based precise interventional photon theragnostics. STATEMENT OF SIGNIFICANCE: Emerging minimally invasive techniques have been explored for the treatment of colorectal cancer (CRC); however, these reported approaches cannot reach the requirement of precise orthotopic cancer treatment due to the lack of in situ real-time imaging guidance. This study proposes a 0.9 mm catheter-based OCT imaging-guided interventional photothermal therapy (IPTT) strategy at 1310 nm for treating orthotopic CRC. Folate-modified plasmonic Bi/Bi2S3 nanoparticles enable real-time imaging-guided IPTT by providing strong scattering signals and local hyperthermia. This approach allows simultaneous transmission of imaging and therapy light in the same optical fiber, offering a promising method for precise CRC theragnostics and addressing the gap of in situ real-time imaging during IPTT.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Yang
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Beibei Gao
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wei Ge
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lu Gao
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jigang Wu
- Biophotonics Laboratory, University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shengxian Tu
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Fu Wang
- Biomedical Instrument Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China; Shenzhen Research Institute, Shanghai Jiao Tong University, Shenzhen 518057, China.
| |
Collapse
|
5
|
Liu Z, Jiang X, Ke Z, Wang W, Tang J, Dai Y. PAR2 deficiency impairs antitumor immunity and attenuates anti-PD1 efficacy in colorectal cancer. Pharmacol Res 2025; 215:107721. [PMID: 40174816 DOI: 10.1016/j.phrs.2025.107721] [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: 11/05/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
A T cell-inflamed tumor microenvironment is predictive of better prognosis and clinical response to immunotherapy. Proteinase-activated receptor 2 (PAR2), a member of G-protein coupled receptors is involved in inflammatory process and the progression of various cancers. However, the role of PAR2 in modulating the tumor microenvironment remains unclear. Here, we found that PAR2 high-expression was associated with a favorable prognosis in patients with colorectal cancer. Intriguingly, PAR2 expression in human colorectal cancer was mainly confined to tumor cells and was significantly associated with CD8+ T cell infiltration. Tumor-intrinsic PAR2 deficiency blunted antitumor immune responses to promote tumor growth and attenuated the therapeutic efficacy of anti-PD1 in a mouse model of colon cancer. Tumors with downregulated PAR2 showed decreased CD8+ T cell infiltration and impaired effector function. Mechanistically, PAR2 activation in tumor cells induced CXCL9 and CXCL10 production via PI3K/AKT/mTOR signaling, thereby enhancing CD8+ T cell recruitment in the tumor microenvironment. In addition, PAR2 was essential for dendritic cell activation and differentiation towards conventional type 1 subset. PAR2 deficiency in dendritic cells markedly impaired their ability to prime CD8+ T cells and control tumor growth in vivo. Thus, our findings identify new roles for PAR2 in promoting antitumor immunity and provide a promising target to improve immunotherapy efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Zilin Liu
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Xuehui Jiang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Weihong Wang
- Department of Gastroenterology, Peking University First Hospital, Beijing, China
| | - Jianqiang Tang
- Department of General Surgery, Peking University First Hospital, Beijing, China.
| | - Yun Dai
- Department of Gastroenterology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
6
|
Teixeira N, Baião A, Dias S, Sarmento B. The progress and challenges in modeling colorectal cancer and the impact on novel drug discovery. Expert Opin Drug Discov 2025; 20:565-574. [PMID: 39282980 DOI: 10.1080/17460441.2024.2404238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/10/2024] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Colorectal cancer (CRC) remains one of the leading causes of cancer-related morbidity and mortality worldwide. This disease is complex and heterogeneous, influenced by a variety of genetic, epigenetic, and environmental factors that drive CRC initiation and progression. Despite advances in therapeutic strategies, the five-year survival rate for metastatic CRC is alarmingly low. Traditional two-dimensional (2D) cell culture systems have been the foundation of cancer research, but their inability to replicate the complex tumor microenvironment (TME) limits their effectiveness. AREAS COVERED This paper explores the evolution of CRC models, starting with the limitations of traditional 2D cell culture systems and the significant advancements offered by 3D models. Additionally, it highlights 3D bioprinting and on-chip CRC models, which have enhanced the ability to mimic in vivo conditions. EXPERT OPINION The transition to advanced 3D models represents a pivotal shift in CRC research, offering considerable improvements over the established 2D models. These models hold promise for the development of patient-specific models that better mimic in vivo conditions. However, the inherent complexity of CRC continues to pose challenges in developing models that can fully capture the disease's multifaceted nature. This complexity and high costs associated with these technologies, along with the need for standardized protocols, pose significant challenges to their widespread adoption.
Collapse
Affiliation(s)
- Natália Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Baião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sofia Dias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- CESPU - IUCS - Instituto Universitário de Ciências da Saúde, Gandra, Portugal
| |
Collapse
|
7
|
Xu X, Zhang S, Luo Z, Zheng Y, Kong T, Huang C, Qiu Z. Frontiers and Controversies in De Novo Gastrointestinal Tumors After Organ Transplantation: Current Progress and Future Directions. Ann Surg Oncol 2025; 32:3392-3405. [PMID: 40035907 DOI: 10.1245/s10434-025-16975-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: 11/06/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025]
Abstract
The increasing success of organ transplantation has significantly improved survival for patients with end-stage diseases, yet it introduces a complex dilemma: the elevated risk for the development of de novo gastrointestinal (GI) tumors. The sustained immunosuppression required to maintain graft function paradoxically undermines the body's natural defenses against cancer, leading to a higher incidence, aggressive progression, and atypical presentations of GI tumors among transplant recipients compared with the general population. This presents a pressing challenge: balancing the dual imperatives of preventing graft rejection and effectively managing malignancies. Current treatment paradigms, including surgical approaches, chemotherapy, radiation therapy, and the emerging role of immunotherapy, are fraught with complexities due to the altered immune landscape in these patients. This review underscores the critical need to understand the multifaceted relationship between post-transplantation immunosuppression and tumorigenesis, providing a comprehensive exploration of epidemiologic shifts, pathophysiologic insights, and the intricacies of the tumor microenvironment in this unique patient population. Understanding and managing GI tumors in transplant recipients is not only a clinical challenge, but also a necessary frontier in transplant oncology, promising to refine therapeutic strategies and improve the longevity and quality of life for this growing patient cohort.
Collapse
Affiliation(s)
- Ximo Xu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaopeng Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zai Luo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zheng
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Kong
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
8
|
Chai X, Zhang Y, Shi Z, Yang R, Liu X, Zhou Y, Li C, Li Z. An Overview of Predictive Biomarkers and Detection Approaches for Immunotherapy Response in GI Malignancies. J Gastroenterol Hepatol 2025; 40:1059-1069. [PMID: 40074558 DOI: 10.1111/jgh.16930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
This review provides an in-depth exploration of the evolving role of immunotherapy in gastrointestinal (GI) cancers, with a particular focus on immune checkpoint inhibitors (ICIs) and their associated predictive biomarkers. We present a detailed analysis of established biomarkers, such as PD-L1, microsatellite instability (MSI), tumor mutational burden (TMB), and the tumor microenvironment (TME), as well as emerging biomarkers, including gut microbiota and Epstein-Barr virus (EBV). The predictive value of these biomarkers in guiding clinical decision-making and optimizing immunotherapy outcomes is thoroughly discussed. Additionally, we highlight recent advancements in biomarker evaluation technologies, including next-generation sequencing (NGS), multiplex immunohistochemistry, and artificial intelligence (AI)-driven models. These technologies are instrumental in advancing precision medicine by enhancing the accuracy and efficiency of biomarker detection and facilitating personalized treatment approaches. The integration of these predictive biomarkers with advanced detection technologies has significantly improved the clinical efficacy of immunotherapy in GI cancers by addressing challenges such as tumor heterogeneity, immune evasion, and variable patient responses. By providing a deeper understanding of tumor biology and patient-specific factors, these tools offer the potential to optimize patient selection, treatment regimens, and, ultimately, clinical outcomes. This review underscores the transformative impact of combining predictive biomarkers with cutting-edge technologies, marking a significant step forward in the field of precision oncology for GI cancer treatment.
Collapse
Affiliation(s)
- Xinyu Chai
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiwen Zhang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhihui Shi
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruiling Yang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xumin Liu
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yueting Zhou
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Caiyang Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhenhui Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Wang YY, Lin JF, Wu WW, Fu Z, Cao F, Chen YX, Mo HY, Sheng H, Liu ZX, Zeng ZL, Guan XY, Ju HQ, Liao K, Xu RH. Inhibition of MBTPS1 enhances antitumor immunity and potentiates anti-PD-1 immunotherapy. Nat Commun 2025; 16:4047. [PMID: 40307212 PMCID: PMC12043911 DOI: 10.1038/s41467-025-59193-4] [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: 06/29/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Despite advances in cancer immunotherapy, colorectal cancer patients exhibit limited therapeutic responses. Therefore, the exploration of strategies combining immunotherapy with adjuvant approaches to enhance adaptive immune responses is in demand. Here, we perform a customized in vivo CRISPR-Cas9 screen to target genes encoding membrane and secreted proteins in CRC mouse models with different immune characteristics. We observe that loss of membrane-bound transcription factor site-1 protease (MBTPS1) in tumor cells enhances antitumor immunity and potentiates anti-PD-1 therapy. Mechanistic studies reveal that tumor cell-intrinsic MBTPS1 competes with USP13 for binding to STAT1, thereby disrupting the USP13-dependent deubiquitination-mediated STAT1 stabilization. The upregulated STAT1-transcribed chemokines including CXCL9, CXCL10, and CXCL11, promote CXCR3+CD8+ T cell infiltration. Notably, the regulatory role of MBTPS1 in antitumor immunity operates independently of its classic function in cleaving membrane-bound transcription factors. Collectively, our results provide a theoretical basis for MBTPS1 as a potential immunotherapy target.
Collapse
Affiliation(s)
- Yi-Yu Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Jin-Fei Lin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Wen-Wei Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Zhe Fu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Fen Cao
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Yan-Xing Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Hai-Yu Mo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Hui Sheng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Ze-Xian Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Zhao-Lei Zeng
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, P. R. China
| | - Huai-Qiang Ju
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China.
| | - Kun Liao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China.
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University, Guangzhou, 510060, P. R. China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, P. R. China.
| |
Collapse
|
10
|
Gong W, Wen S, Chen Y, Wu F, Yang M, Sun P, Guo X, Li M, Chen D, Zhao H, Wang L. Deciphering ERR family genes as prognostic and immunological biomarkers through pan-cancer analysis with validation in gallbladder cancer. Front Oncol 2025; 15:1525635. [PMID: 40356747 PMCID: PMC12066295 DOI: 10.3389/fonc.2025.1525635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
Background The estrogen-related receptor family genes (ERRs), including ESRRA, ESRRB, and ESRRG, have been implicated in a few tumors, exhibiting distinct roles through diverse mechanisms. The purpose of our research is to explore the commonalities and underlying mechanism of ERRs in malignancies from a pan-cancer perspective and to validate the role and mechanisms of ESRRG in gallbladder cancer (GBC). Methods We leveraged public databases such as TCGA and GTEx to systematically investigate the potential functions of ERRs in malignancies. ESRRG expression was analyzed through immunohistochemical staining in gallbladder cancer and cholecystitis tissues. For functional validation, ESRRG was knocked down in GBC cell lines, followed by CCK-8, colony formation, scratch wound healing, Transwell migration, and invasion assays. Western blot, qPCR, and immunofluorescence were performed to evaluate the relationship between ESRRG, PD-L1, and CD8+ T cells. Results Compared to adjacent normal tissues, ESRRA is overexpressed in most tumors, ESRRB is generally underexpressed, and ESRRG exhibits significant expression alterations across various tumors. All three ERRs demonstrate significant prognostic value across different cancers. Notably, the strong associations of ERRs with key immunological features-stromal scores, immune cell infiltration, microsatellite instability (MSI), and tumor mutational burden (TMB)-suggest their involvement in immune evasion and their potential utility in guiding immunotherapy strategies. All three ERRs display a positive correlation with advanced tumor stages in cholangiocarcinoma (CHOL). Specifically, in CHOL, ESRRG expression is closely associated with lymphatic metastasis, poorer overall survival, reduced immune infiltration, elevated PD-L1 expression, epithelial-mesenchymal transition (EMT), and DNA damage response. In GBC tissues, we subsequently confirmed that ESRRG expression positively correlates with pathological staging and PD-L1 expression, while negatively correlating with prognosis and CD8+ T cell infiltration. Knockdown of ESRRG in gallbladder cancer cells results in decreased proliferation, migration, and invasion. Moreover, the expression of PD-L1, MSH2, BRCA1, MMP2, and VIMENTIN decreased with ESRRG knockdown. Conclusion Our pan-cancer analysis reveals ERRs as critical regulators of tumor immunity and progression, with ESRRG emerging as a key oncogenic driver in GBC. The mechanistic link between ESRRG and PD-L1/EMT suggests its potential as a therapeutic target to enhance immunotherapy efficacy. These findings underscore the need for tissue-specific targeting strategies for ERR family members in precision oncology.
Collapse
Affiliation(s)
- Wanwan Gong
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Sijia Wen
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yu Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Fan Wu
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Mengmeng Yang
- Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, China
| | - Ping Sun
- Department of Pathology, Jiangnan University Medical Center, Wuxi, China
| | - Xingmei Guo
- Department of Pathology, Jiangnan University Medical Center, Wuxi, China
| | - Meiqin Li
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Daozhen Chen
- Research Institute for Reproductive Health and Genetic Diseases, Wuxi Maternal and Child Health Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Hui Zhao
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Lei Wang
- Department of Hepatopancreatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
- Department of Hepatopancreatobiliary Surgery, The Affiliated Wuxi No.2 People’s Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
11
|
Li L, Chen M, Reis RL, Kundu SC, Xiao B, Shi X. Advancements of nanoscale drug formulations for combination treatment of colorectal cancer. Int J Pharm 2025; 674:125508. [PMID: 40132771 DOI: 10.1016/j.ijpharm.2025.125508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Combination chemotherapy is widely utilized in treating colorectal cancer (CRC), particularly for patients who are ineligible for surgery or those with metastatic CRC (mCRC). While this therapeutic method has demonstrated efficacy in managing CRC and mCRC, its broader clinical application is limited due to the unique physical properties, mechanisms of action, and pharmacokinetics of different chemotherapeutic drugs. Consequently, achieving satisfactory treatment outcomes proves to be challenging. Nanotechnology has given rise to innovative drug systems that are precise, controllable, and highly efficient in drug delivery. These nanoscale drug delivery systems can integrate the advantageous aspects of various therapeutic modalities, including chemotherapy, gene therapy, and immunotherapy. This review aims to explain the application of nano-drug delivery system in the treatment of colorectal cancer. Through its unique physical/chemical properties and biological functions, it can solve the limitations of traditional therapy and achieve more accurate, efficient and safe treatment. The advantages/disadvantages, physical and chemical characteristics of various drug delivery systems are described in detail, and suggestions on selecting reasonable NDDSs according to different drug combination methods are given to achieve the best therapeutic effect. This review paper presents an exhaustive summary of the diverse range of drugs utilized in chemotherapy, in addition to outlining strategies for effectively integrating chemotherapy with other treatment modalities. Furthermore, it delves into the principle of selecting carriers for various drug combinations.
Collapse
Affiliation(s)
- Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Maohua Chen
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Bo Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
12
|
Shen H, Jin J, Yu N, Liu T, Nie Y, Wan Z, Chen Y, Cao K, Xu Y, Huang Y, Feng C, Huang R, Yang Y, Gao F. PRKCSH enhances colorectal cancer radioresistance via IRE1α/XBP1s-mediated DNA repair. Cell Death Dis 2025; 16:258. [PMID: 40189587 PMCID: PMC11973196 DOI: 10.1038/s41419-025-07582-4] [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: 09/10/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
Neoadjuvant radiotherapy is the standard treatment for locally advanced rectal cancer, but resistance to this therapy remains a significant clinical challenge. Understanding the molecular mechanisms of radioresistance and developing strategies to enhance radiosensitivity are crucial for improving treatment outcomes. This study investigated the role of PRKCSH in colorectal cancer radioresistance and its underlying mechanisms. Our results demonstrate that PRKCSH is upregulated in colorectal cancer cells following ionizing radiation. Inhibiting PRKCSH sensitized these cells to radiation by reducing clonogenic survival, promoting apoptosis, and impairing DNA damage repair. Mechanistically, PRKCSH inhibition reduced p53 ubiquitination and degradation by activating the ER stress IRE1α/XBP1s pathway after radiation exposure, which enhanced DNA repair and contributed to radioresistance. In preclinical CRC models, PRKCSH depletion suppressed tumor growth and increased radiosensitivity. Similarly, in patient-derived organoid models, PRKCSH knockdown reduced organoid growth post-radiotherapy. In rectal cancer patients receiving neoadjuvant radiotherapy, higher PRKCSH expression in post-treatment samples correlated with reduced tumor regression. These findings suggest that targeting PRKCSH diminishes radioresistance by impairing DNA repair through the modulation of ER stress. Furthermore, PRKCSH expression may serve as a biomarker for evaluating radiotherapy efficacy and clinical outcomes in rectal cancer patients undergoing neoadjuvant therapy.
Collapse
Affiliation(s)
- Hui Shen
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
- Department of Central Laboratory, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jing Jin
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi' an, China
| | - Nanxi Yu
- Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Tingting Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi' an, China
| | - Zhijie Wan
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Yuanyuan Chen
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Kun Cao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Ying Xu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Yijuan Huang
- Department of Central Laboratory, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chao Feng
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Hunan, Changsha, China.
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China.
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China.
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China.
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China.
| |
Collapse
|
13
|
Shi F, Li GJ, Liu Y, Zhou HM, Zhang Y, Wei SY, Zan BJ, Gao M, Chen FS, Li BX, Wang BQ, Dong MY, Du RL, Zhang XD. USP19 deficiency enhances T-cell-mediated antitumor immunity by promoting PD-L1 degradation in colorectal cancer. Pharmacol Res 2025; 214:107668. [PMID: 40020887 DOI: 10.1016/j.phrs.2025.107668] [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: 12/02/2024] [Revised: 01/26/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Colorectal cancer (CRC) is characterized by a highly immunosuppressive tumor microenvironment, which limits the effectiveness of current immunotherapies. Identifying strategies to overcome this resistance is critical for improving treatment outcomes. In this study, we discovered that USP19 plays a pivotal role in regulating T-cell-mediated antitumor immunity through a CRISPR/Cas9 sgRNA library screen and co-culture assays with activated T cells. We demonstrated that USP19 deficiency significantly enhances the susceptibility to T cell-mediated cytotoxicity in CRC cells, organoids, and mouse models. Transcriptomic sequencing (RNA-seq) revealed activation of the PD-1 pathway in tumor with USP19-deficiency cells. Mechanistic investigations revealed that USP19 directly stabilizes PD-L1 by binding to its intracellular domain and preventing its degradation via K48-linked ubiquitination and proteasomal pathways. Clinically, USP19 expression was found to be significantly elevated in CRC tissues and was positively associated with PD-L1 levels, advanced tumor grade, poor differentiation, and TP53 mutations, highlighting its potential as a biomarker for aggressive CRC. Importantly, in vivo experiments demonstrated that targeting USP19, in combination with αPD-L1 therapy, synergistically suppressed CRC progression. This combination not only reduced PD-L1 levels but also enhanced CD8+ T-cell activation and GzmB infiltration, resulting in robust antitumor effects. These findings establish USP19 as a key driver of immune evasion in CRC and suggest that targeting USP19 could enhance the efficacy of immunotherapy, providing a promising new avenue for CRC treatment.
Collapse
Affiliation(s)
- Feng Shi
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Guang-Jing Li
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, China
| | - Yi Liu
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Hai-Meng Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Yue Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Si-Yi Wei
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Bo-Jun Zan
- Medical Laboratory College, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Meng Gao
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Fei-Shan Chen
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Bo-Xin Li
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Bai-Qi Wang
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Ming-You Dong
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Run-Lei Du
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China.
| | - Xiao-Dong Zhang
- National Health Commission Key Laboratory of Birth Defect Research and Prevention & MOE Key Lab of Rare Pediatric Diseases, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
14
|
Dai F, Cao Y, Zhu C, Li Y, Ma X, Wang S, Liu H, Xie X, Gao L, Wang Y, Wang C. Design, Synthesis, and Biological Evaluation of Naphthalimide-Polyamine Conjugate as a Potential Anti-Colorectal Cancer Agent. Chem Biodivers 2025; 22:e202401873. [PMID: 39632400 DOI: 10.1002/cbdv.202401873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
Colorectal cancer is the second most common cause of cancer-related death worldwide, with a rising incidence, highlighting an urgent need for novel therapeutics. In this study, we developed several polyamine conjugates. Compound 6 (C6) was selected as the lead compound and was evaluated for anticancer activity in vitro and in vivo. In vitro experiments showed that C6 inhibited cell proliferation, colony formation, migration, and invasion of colorectal cancer cells while inducing apoptosis, pyroptosis, and autophagosome formation. Genetic or pharmacological inhibition of autophagy weakened C6-induced apoptosis and gasdermin E (GSDME)-dependent pyroptosis. Inactivation of caspase 3 activity by AC-DEVD-CHO decreased the levels of N-terminal GSDME induced by C6. Furthermore, animal models exhibited suppressed tumor growth and dissemination after treatment with C6. Taken together, our findings highlight C6 as a potential drug against colorectal cancer.
Collapse
Affiliation(s)
- Fujun Dai
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Yue Cao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Chenguang Zhu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Yibing Li
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Xiaoxuan Ma
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Senzhen Wang
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Haizhen Liu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Xiaoya Xie
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Lei Gao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| | - Yanming Wang
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, Henan, China
| | - Chaojie Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan, China
| |
Collapse
|
15
|
Chang Y, Long M, Shan H, Liu L, Zhong S, Luo JL. Combining gut microbiota modulation and immunotherapy: A promising approach for treating microsatellite stable colorectal cancer. Crit Rev Oncol Hematol 2025; 208:104629. [PMID: 39864533 DOI: 10.1016/j.critrevonc.2025.104629] [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: 12/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent and lethal cancers worldwide, ranking third in incidence and second in mortality. While immunotherapy has shown promise in patients with deficient mismatch repair (dMMR) or high microsatellite instability (MSI-H), its effectiveness in proficient mismatch repair (pMMR) or microsatellite stable (MSS) CRC remains limited. Recent advances highlight the gut microbiota as a potential modulator of anti-tumor immunity. The gut microbiome can significantly influence the efficacy of immune checkpoint inhibitors (ICIs), especially in pMMR/MSS CRC, by modulating immune responses and systemic inflammation. This review explores the role of the gut microbiota in pMMR/MSS CRC, the mechanisms by which it may enhance immunotherapy, and current strategies for microbiota modulation. We discuss the potential benefits of combining microbiota-targeting interventions with immunotherapy to improve treatment outcomes for pMMR/MSS CRC patients.
Collapse
Affiliation(s)
- Yujie Chang
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Min Long
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Hanguo Shan
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Logen Liu
- Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China
| | - Shangwei Zhong
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China
| | - Jun-Li Luo
- The Cancer Research Institute and the Second Affiliated Hospital, Hengyang Medical School, University of South China (USC), Hunan 421001, China; Hunan Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, USC, Hunan 421001, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, USC, Hunan 421001, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, USC, Hunan 410008, China.
| |
Collapse
|
16
|
Naveed M, Mughal MS, Aziz T, Makhdoom SI, Jamil H, Ali Khan A, Al-Hoshani N, Al-Joufi FA, Tahir Kassim RM, Alwethaynani MS. Exploration of mRNA-modifying METTL3 oncogene as momentous prognostic biomarker responsible for colorectal cancer development. Open Med (Wars) 2025; 20:20251167. [PMID: 40177651 PMCID: PMC11964186 DOI: 10.1515/med-2025-1167] [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: 04/19/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025] Open
Abstract
Background Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide, emphasizing the need for improved prognostic biomarkers. Recent studies have identified the mRNA-modifying METTL3 oncogene as a potential biomarker in CRC progression. Objective This study aimed to investigate the expression patterns of METTL3 in CRC, assess its association with clinical outcomes, identify interacting proteins and biological pathways, and explore its correlation with immune cell infiltration. Methods Comprehensive analyses were conducted using public datasets, including transcriptome profiles from The Cancer Genome Atlas and the GSE103512 dataset. Protein-protein interaction (PPI) networks, pathway enrichment, and immune infiltration analyses were performed to elucidate METTL3's role in CRC progression. Results METTL3 expression was significantly higher in CRC tissues compared to normal tissues (p < 0.001). Mutations in METTL3 were detected in approximately 6% of CRC cases, with fusion events involving the SRPK2 gene. PPI analysis identified ten interacting proteins, including METTL4, EIF3H, RBM15B1, CBLL1, WTAP, NCBP1, RBM15, ZC3H13, METTL14, and KIAA1429. METTL3 expression showed a positive correlation with METTL4, METTL14, NCBP1, and WTAP expression (R > 0.5, p < 0.001). Higher METTL3 expression was associated with immunosuppressive phenotypes, such as increased infiltration of tumor-associated macrophages, regulatory T cells, and cancer-associated fibroblasts (p < 0.001). Pathway enrichment analysis revealed METTL3's involvement in crucial pathways, including the cell cycle and renal cell carcinoma (p < 0.01). Gene ontology analysis highlighted its role in mRNA and RNA-related processes. Conclusion The study supports the potential of METTL3 as a prognostic biomarker in CRC and highlights its involvement in immune modulation and cancer progression. These findings lay the groundwork for future studies aimed at developing targeted therapies and improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Muhammad Saad Mughal
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health Food Hygiene and Quality, University of Ioannina, Arta, 47132, Greece
| | - Syeda Izma Makhdoom
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Hamza Jamil
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Ayaz Ali Khan
- Department of Biotechnology, University of Malakand, Chakdara, 18800, Pakistan
| | - Nawal Al-Hoshani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia
| | - Fakhria A. Al-Joufi
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Aljouf, Saudi Arabia
| | | | - Maher S. Alwethaynani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Alquwayiyah, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Liu C, Huang Z, Li Z, Dai X, Chen Q, Chen M, Xu Y, Chen X, Wang F, Chen Q, Wei J. A self-sustainable DNA amplification circuit for sensitive microRNA imaging. Anal Chim Acta 2025; 1343:343689. [PMID: 39947797 DOI: 10.1016/j.aca.2025.343689] [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: 11/12/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Early detection of cancer biomarkers such as microRNA-21 (miR-21), a small RNA molecule, can facilitate earlier diagnosis and potentially lead to earlier treatment. The detection of trace miRNA in complicated cellular environments necessitates the construction of self-sustainable DNA circuitry boasting high signal gain and robust anti-interference capabilities. However, current self-sustainable DNA circuits suffer from complex designs and severe signal leakage. Therefore, it is essential to develop a highly efficient and reliable strategy to improve sensing performance and accurately detect trace biomolecules in intricate biological matrices. RESULTS We engineered a general and high-performance self-sustainable DNA amplification (SDA) circuit for reliable bioimaging inside cells. The autocatalytic SDA system was composed of the catalytic DNA assembly (CDA) and the rolling circle amplification (RCA) module. Upon input of the initiator, it stimulated the self-driven cross-invasion of the CDA and RCA amplicon, facilitating the successive replication of initiator sequences and resulting in synergistically accelerated and exponential signal amplification, as systematically investigated by various experimental studies. Due to its highly efficient amplification capability and universal applicability, the self-driven SDA system enabled reliable determination of miR-21 in buffer and serum and achieved a low detection limit of 8.9 pM. As a powerful imaging strategy, the SDA circuit realized accurate miR-21 imaging within cells, highlighting its potential for clinical diagnosis. SIGNIFICANCE Reciprocal reinforcement of the CDA and RCA amplifiers accelerates the entire reaction process, facilitating the generation of an exponentially amplified FRET signal for reliable detection of analytes. The proposed SDA strategy achieves the one-step determination of DNA or miRNA with simple design, high signal gain, low signal leakage, and single-base specificity, and furthermore enables reliable miRNA localization inside cells, highlighting its potential to monitor significant biomolecules and substantially expanding the toolkits for clinical diagnosis.
Collapse
Affiliation(s)
- Chuanyi Liu
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Ziling Huang
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Zhigang Li
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Xiaojiao Dai
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Qingmin Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Min Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Yi Xu
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Xiaomei Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China
| | - Fuan Wang
- College of Chemistry and Molecular Sciences, Wuhan University, 430072, Wuhan, PR China
| | - Quansheng Chen
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China.
| | - Jie Wei
- College of Ocean Food and Biological Engineering, Jimei University, Fujian Provincial Key Laboratory of Food Microbiology and Enzyme Engineering, 361021, Xiamen, PR China; College of Chemistry and Molecular Sciences, Wuhan University, 430072, Wuhan, PR China.
| |
Collapse
|
18
|
Kang HS, Lim HK, Jang WY, Cho JY. Anti-Colorectal Cancer Activity of Panax and Its Active Components, Ginsenosides: A Review. Int J Mol Sci 2025; 26:2593. [PMID: 40141242 PMCID: PMC11941759 DOI: 10.3390/ijms26062593] [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: 02/16/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Colorectal cancer (CRC) poses a significant health burden worldwide and necessitates novel treatment approaches with fewer side effects than conventional chemotherapy. Many natural compounds have been tested as possible cancer treatments. Plants in the genus Panax have been widely studied due to their therapeutic potential for various diseases such as inflammatory disorders and cancers. Extracts from plants of genus Panax activate upstream signals, including those related to autophagy and the generation of reactive oxygen species, to induce intrinsic apoptosis in CRC cells. The root extract of Panax notoginseng (P. notoginseng) regulated the gut microbiota to enhance the T-cell-induced immune response against CRC. Protopanaxadiol (PPD)-type ginsenosides, especially Rh2, Rg3, Rb1, and Rb2, significantly reduced proliferation of CRC cells and tumor size in a xenograft mouse model, as well as targeting programmed death (PD)-1 to block the immune checkpoint of CRC cells. Moreover, modified nanocarriers with ginsenosides upregulated drug efficacy, showing that ginsenosides can also be utilized as drug carriers. An increasing body of studies has demonstrated the potential of the genus Panax in curing CRC. Ginsenosides are promising active compounds in the genus Panax, which can also support the activity of conventional cancer therapies.
Collapse
Affiliation(s)
| | | | | | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; (H.S.K.); (H.K.L.); (W.Y.J.)
| |
Collapse
|
19
|
Wang T, Chen Z, Wang W, Wang H, Li S. Single-cell and spatial transcriptomic analysis reveals tumor cell heterogeneity and underlying molecular program in colorectal cancer. Front Immunol 2025; 16:1556386. [PMID: 40145096 PMCID: PMC11936967 DOI: 10.3389/fimmu.2025.1556386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background Colorectal cancer (CRC) is a highly heterogeneous tumor, with significant variation in malignant cells, posing challenges for treatment and prognosis. However, this heterogeneity offers opportunities for personalized therapy. Methods The consensus non-negative matrix factorization algorithm was employed to analyze single-cell transcriptomic data from CRC, which helped identify malignant cell expression programs (MCEPs). Subsequently, a crosstalk network linking MCEPs with immune/stromal cell trajectory development was constructed using Monocle3 and NicheNet. Additionally, bulk RNA-seq data were utilized to systematically explore the relationships between MCEPs, clinical features, and genetic mutations. A prognostic model was then established through Lasso and Cox regression analyses, integrating clinical data into a nomogram for personalized risk prediction. Furthermore, key genes associated with MCEPs and their potential therapeutic targets were identified using protein-protein interaction networks, followed by molecular docking to predict drug-binding affinity. Results We classified CRC malignant cell transcriptional states into eight distinct MCEPs and successfully constructed crosstalk networks between these MCEPs and immune or stromal cells. A prognostic model containing 15 genes was developed, demonstrating an AUC greater than 0.8 for prognostic evaluation over 1 to 10 years when combined with clinical features. A key drug target gene TIMP1 was identified, and several potential targeted drugs were discovered. Conclusion This study demonstrated that characterization of the malignant cell transcriptional programs could effectively reveal the biological features of highly heterogeneous tumors like CRC and exhibit significant potential in tumor prognosis assessment. Our research provides new theoretical and practical directions for CRC prognosis and targeted therapy.
Collapse
Affiliation(s)
- Teng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zhaoming Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Wang Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, China
| | - Heng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Shenglong Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
20
|
Sculthorpe DJ, Denton A, Fadhil W, Rusnita D, Ilyas M, Mukherjee A. High α-SMA expression in the tumor stroma is associated with adverse clinical parameters in mismatch repair-proficient colorectal cancers only. Am J Clin Pathol 2025; 163:464-472. [PMID: 39495028 PMCID: PMC11890275 DOI: 10.1093/ajcp/aqae145] [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: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
OBJECTIVES As mismatch repair status confers differential prognosis in colorectal cancers, this study aimed to determine associations of α-smooth muscle actin (α-SMA) protein expression in mismatch repair-proficient (pMMR) and mismatch repair-deficient (dMMR) colorectal tumors with clinicopathologic and prognostic features. METHODS Tissue microarrays from patients with colorectal cancer, immunostained with α-SMA, were assessed through digital image analysis. Total (n = 962), pMMR (n = 782), and dMMR (n = 156) stromal H-scores were assessed for associations with clinicopathologic and survival data. RESULTS Higher α-SMA expression was correlated with pMMR status (P = 5.2223 × 10-8). In the pMMR subgroup, higher α-SMA stromal expression at the tumor periphery was correlated with higher T stage (P = .002), perineural invasion (P = .038), infiltrative tumor edge (P = .01), involved nodal status (P = .036), metastases (P = .013), synchronous metastases (P = .007), recurrence (P = .004), and both 3-year and 5-year survival (P = .018). dMMR tumors showed no significant correlations with α-SMA staining. CONCLUSIONS The findings highlight that immunostaining with α-SMA in pMMR colorectal tumors, especially at the tumor periphery, has the potential to identify patients with adverse prognostic features. Digital assessment of α-SMA may offer improved objectivity, accuracy, economy of time, and risk stratification for management.
Collapse
Affiliation(s)
- Declan J Sculthorpe
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Amy Denton
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Wakkas Fadhil
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Dewi Rusnita
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Mohammad Ilyas
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Queen’s Medical Centre, Nottingham, UK
| | - Abhik Mukherjee
- Molecular Pathology Research, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Queen’s Medical Centre, Nottingham, UK
| |
Collapse
|
21
|
Zhang C, Wang Y, Cheng L, Cao X, Liu C. Gut microbiota in colorectal cancer: a review of its influence on tumor immune surveillance and therapeutic response. Front Oncol 2025; 15:1557959. [PMID: 40110192 PMCID: PMC11919680 DOI: 10.3389/fonc.2025.1557959] [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: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Colorectal cancer (CRC) poses a significant global health burden, with gut microbiota emerging as a crucial modulator of CRC pathogenesis and therapeutic outcomes. This review synthesizes current evidence on the influence of gut microbiota on tumor immune surveillance and responses to immunotherapies and chemotherapy in CRC. We highlight the role of specific microbial taxa in promoting or inhibiting tumor growth and the potential of microbiota-based biomarkers for predicting treatment efficacy. The review also discusses the implications of microbiota modulation strategies, including diet, probiotics, and fecal microbiota transplantation, for personalized CRC management. By critically evaluating the literature, we aim to provide a comprehensive understanding of the gut microbiota's dual role in CRC and to inform future research directions in this field.
Collapse
Affiliation(s)
- Chunlei Zhang
- Department of Colorectal and Anus Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yong Wang
- Department of Hepatobiliary Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Lei Cheng
- Department of Colorectal and Anus Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xiansheng Cao
- Department of Gastrointestinal Surgery, Hernia and Abdominal Wall Surgery I, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Chunyuan Liu
- Department of Colorectal and Anus Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| |
Collapse
|
22
|
Li Z, Liu S, Liu D, Yang K, Xiong J, Fang Z. Multiple mechanisms and applications of tertiary lymphoid structures and immune checkpoint blockade. J Exp Clin Cancer Res 2025; 44:84. [PMID: 40038799 PMCID: PMC11881293 DOI: 10.1186/s13046-025-03318-6] [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: 10/22/2024] [Accepted: 02/05/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) inhibits tumor immune escape and has significantly advanced tumor therapy. However, ICB benefits only a minority of patients treated and may lead to many immune-related adverse events. Therefore, identifying factors that can predict treatment outcomes, enhance synergy with ICB, and mitigate immune-related adverse events is urgently needed. MAIN TEXT Tertiary lymphoid structures (TLS) are ectopic lymphoid tissues that arise from the tumor periphery. They have been found to be associated with better prognosis and improved clinical outcomes after ICB therapy. TLS may help address the problems associated with ICB. The multiple mechanisms of action between TLS and ICB remain unknown. This paper described potential mechanisms of interaction between the two and explored their potential applications.
Collapse
Affiliation(s)
- Zelin Li
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shuhan Liu
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Deyu Liu
- Department of Clinical Medicine, Queen Mary School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kangping Yang
- The 2st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jing Xiong
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Department of General Practice, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Ziling Fang
- The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Department of Oncology, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
23
|
Yang Y, Xiao Z, Ouyang J, Guo Y. Synergistic efficacy of orthokeratology and 0.01% atropine in controlling pediatric myopia progression: A retrospective analysis. J Investig Med 2025; 73:290-299. [PMID: 39679531 DOI: 10.1177/10815589241308819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
This study investigates the combined efficacy of orthokeratology lenses and 0.01% atropine in controlling the progression of pediatric progressive myopia. The study, conducted retrospectively on 33 children aged 8-14, measured key parameters, including axial length growth, uncorrected visual acuity (UCVA), intraocular pressure (IOP), tear film breakup time (TBUT), and pupil diameter. The results revealed a significant reduction in axial length growth with the combined treatment compared to orthokeratology alone, while UCVA and IOP remained stable. TBUT decreased, and pupil diameter increased post-treatment. The findings suggest that combining orthokeratology with low-dose atropine offers a safe and effective strategy for managing pediatric myopia, particularly in younger patients.
Collapse
|
24
|
Qiao W, Li S, Luo L, Chen M, Zheng X, Ye J, Liang Z, Wang Q, Hu T, Zhou L, Wang J, Ge X, Feng G, Hu F, Liu R, Li J, Yang J. Ce6-GFFY is a novel photosensitizer for colorectal cancer therapy. Genes Dis 2025; 12:101441. [PMID: 39759121 PMCID: PMC11697048 DOI: 10.1016/j.gendis.2024.101441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 07/10/2024] [Accepted: 10/21/2024] [Indexed: 01/07/2025] Open
Abstract
Photodynamic therapy is an "old" strategy for cancer therapy featuring clinical safety and rapid working, but suitable photosensitizers for colorectal cancer therapy remain lacking. This study synthesized a novel photosensitizer termed Ce6-GFFY based on a self-assembling peptide GFFY and a photo-responsive molecule chlorin e6 (Ce6). Ce6-GFFY forms macroparticles with a diameter of ∼160 nm and possesses a half-life of 10 h, as well as an ideal tumor-targeting ability in mouse models. Ce6-GFFY effectively penetrates cells and generates numerous reactive oxygen species upon 660 nm laser irradiation. The reactive oxygen species promotes the accumulation of cytotoxic T cells and decrease of myeloid-derived suppressor cells in the tumor microenvironment through immunogenic cell death, thus prohibiting the growth of both primary and metastatic tumors after once treatment. This study not only provides a strategy for photosensitizer development but also confirms a promising application of Ce6-GFFY for colorectal cancer therapy.
Collapse
Affiliation(s)
- Wei Qiao
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Shuxin Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Linna Luo
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Meiling Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaobin Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
- Department of Nuclear Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jiacong Ye
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Zhaohui Liang
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Qiaoli Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Ting Hu
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Ling Zhou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jing Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaosong Ge
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Guokai Feng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Fang Hu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Rongbin Liu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Jianjun Li
- Department of Endoscopy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Jie Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| |
Collapse
|
25
|
Xie Z, Yang T, Zhou C, Xue Z, Wang J, Lu F. Integrative Bioinformatics Analysis and Experimental Study of NLRP12 Reveal Its Prognostic Value and Potential Functions in Ovarian Cancer. Mol Carcinog 2025; 64:383-398. [PMID: 39601513 DOI: 10.1002/mc.23854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024]
Abstract
NLRP12 plays a significant role in cellular functional behavior and immune homeostasis, influencing inflammation, tumorigenesis, and prognosis. This study aimed to explore its specific effects on the tumor microenvironment (TME) and its contribution to heterogeneity in ovarian cancer (OV) through bioinformatics analysis and experimental verification. Utilizing various bioinformatics databases and clinical specimens, we investigated NLRP12 expression and its relationship with OV prognosis and immune infiltration. In vitro assays were conducted to assess the impact of NLRP12 on the proliferation and invasion of OV cells. Our findings indicate that NLRP12 is upregulated in OV, with high expression correlating with a negative prognosis. Furthermore, NLRP12 expression demonstrated a positive correlation with the infiltration of various immune cells and the expression of immune checkpoint molecules in OV. Analysis of The Cancer Immunome Atlas (TCIA) database revealed that OV patients with lower NLRP12 expression may exhibit an enhanced response to immunotherapy, particularly CTLA4 blockers, a finding validated in animal experiments. Additionally, the study emphasized the role of NLRP12 in influencing the prognosis of OV patients by promoting epithelial-mesenchymal transition (EMT) in ovarian cancer cells. Finally, we identified a potential therapeutic compound, Schisandrin B (Schi B), which decreases NLRP12 expression in ovarian cancer cells by binding to the transcription factor SPI1 associated with NLRP12. Our findings suggest that NLRP12 serves as a crucial immune-related biomarker predicting poor outcomes in OV, and targeting NLRP12 may represent a promising therapeutic approach for OV patients in the future.
Collapse
Affiliation(s)
- Zhihui Xie
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Tiantian Yang
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Chuchu Zhou
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Zixin Xue
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| | - Jianjun Wang
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Feng Lu
- Department of Medical Oncology, Huaihe Hospital of Henan University, Kaifeng, China
- Department of Immunology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Joint National Laboratory for Antibody Drug Engineering, Medical School, Henan University, Kaifeng, China
| |
Collapse
|
26
|
Jiao J, Wu Y, Wu S, Jiang J. Enhancing Colorectal Cancer Treatment Through VEGF/VEGFR Inhibitors and Immunotherapy. Curr Treat Options Oncol 2025; 26:213-225. [PMID: 40045029 DOI: 10.1007/s11864-025-01306-8] [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] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
OPINION STATEMENT Colorectal cancer, ranking as the third most prevalent malignancy globally, substantially benefits from both immunotherapy and VEGF/VEGFR inhibitors. Nevertheless, the use of monotherapy proves inadequate in effectively tackling the heterogeneity of tumors and the intricacies of their microenvironment, frequently leading to drug resistance and immune evasion. This situation underscores the pressing need for innovative strategies aimed at augmenting the effectiveness and durability of treatments. Clinical research demonstrates that the combination of VEGF/VEGFR inhibitors (primarily including VEGF/VEGFR-targeted drugs and multi-kinase inhibitors) with immune checkpoint inhibitors creates a synergistic effect in the treatment of colorectal cancer. Our analysis explores how VEGF/VEGFR inhibitors recalibrate the tumor microenvironment, modulate immune cell functions, and influence the expression of immune checkpoints and cytokines. Furthermore, we critically evaluate the preclinical and clinical feasibility of these combined therapeutic approaches. Despite the potential for toxicity, the significant benefits and prospective applications of these strategies warrant thorough exploration. Exploring the synergistic mechanisms of these combined treatments has the potential to inaugurate a new paradigm in oncology, enabling more personalized and efficacious treatment modalities. Additionally, the synergy between VEGF/VEGFR inhibitors and nascent immunotherapies emerges as a promising field of inquiry.
Collapse
Affiliation(s)
- Jing Jiao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - You Wu
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Jingting Jiang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Department of Tumor Biological Treatment, Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Juqian Road №185, Changzhou, 213003, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
27
|
Muta Y, Nakanishi Y. Mouse colorectal cancer organoids: Lessons from syngeneic and orthotopic transplantation systems. Eur J Cell Biol 2025; 104:151478. [PMID: 39919450 DOI: 10.1016/j.ejcb.2025.151478] [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: 08/19/2024] [Revised: 01/01/2025] [Accepted: 02/04/2025] [Indexed: 02/09/2025] Open
Abstract
Colorectal cancer (CRC) organoids provide more accurate and tissue-relevant models compared to conventional two-dimensional cultured cell cultures. Mouse CRC organoids, in particular, offer unique advantages over their human counterparts, as they can be transplanted into immunocompetent mice. These syngeneic transplantation models create a robust system for studying cancer biology in the immunocompetent tumor microenvironment (TME). This article discusses the development and applications of these organoid systems, emphasizing their capacity to faithfully recapitulate in vivo tumor progression, metastasis, and the immune landscape.
Collapse
Affiliation(s)
- Yu Muta
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| |
Collapse
|
28
|
Gan L, Yang C, Zhao L, Wang S, Ye Y, Gao Z. The expression of ERAP1 is favorable for the prognosis and immunotherapy in colorectal cancer: a study based on the bioinformatic and immunohistochemical analysis. Clin Transl Oncol 2025; 27:549-566. [PMID: 39009862 DOI: 10.1007/s12094-024-03520-6] [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: 01/28/2024] [Accepted: 05/12/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Endoplasmic reticulum aminopeptidase 1 (ERAP1) is an emerging pharmacological target in cancer immunotherapy. This study was set out to examine the expression profiles and implications for prognosis and immunotherapy of ERAP1 in CRC. METHODS Based on bioinformatics and immunohistochemical analysis, we analyzed ERAP1 for potential diagnostic and prognostic significance in CRC. Functional enrichment analysis was conducted to detect the pathways associated with ERAP1, thus determining possible mechanisms. ESTIMATE, TIMER, and CIBESORT probed the links between ERAP1 and tumor-infiltrating immune cells. Lastly, we examined how ERAP1 expression correlated with the sensitivity to immunotherapy. RESULTS Tumor tissues had decreased levels of ERAP1 expression relative to normal tissues. Patients whose ERAP1 expression was low suffered a worse chance of survival. Besides, it was shown that ERAP1 expression was associated with the advanced M stage and pathologic stage. Survival analysis revealed that low ERAP1 expression, age, pathologic stage, T stage, and M stage were independent indicators for unfavorable CRC patients' prognoses. The 1-, 3-, and 5-year OS calibration curves all fit well with the ideal model, suggesting that the age-ERAP1-T-stage-M-stage nomogram is a reliable predictor of OS. Additionally, we discovered that ERAP1 expression was associated with immune response and infiltration of various immune cells, such as down-regulated inhibitory immune cells and up-regulated stimulating immune cells. Sensitivity to PD-1 and CTLA4 inhibitors was associated with high ERAP1 levels. CONCLUSIONS In summary, ERAP1 has potential as a diagnostic and prognostic biological marker, highlighting new insights into the study of CRC and the design of effective therapies.
Collapse
Affiliation(s)
- Lin Gan
- Department of Gastroenterological Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People's Republic of China
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Changjiang Yang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Long Zhao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China
| | - Zhidong Gao
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, 100044, People's Republic of China.
| |
Collapse
|
29
|
Liu H, Zhang C, Peng S, Yin Y, Xu Y, Wu S, Wang L, Fu Y. Prognostic models of immune-related cell death and stress unveil mechanisms driving macrophage phenotypic evolution in colorectal cancer. J Transl Med 2025; 23:127. [PMID: 39875913 PMCID: PMC11776142 DOI: 10.1186/s12967-025-06143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Tumor microenvironment (TME), particularly immune cell infiltration, programmed cell death (PCD) and stress, has increasingly become a focal point in colorectal cancer (CRC) treatment. Uncovering the intricate crosstalk between these factors can enhance our understanding of CRC, guide therapeutic strategies, and improve patient prognosis. METHODS We constructed an immune-related cell death and stress (ICDS) prognostic model utilizing machine learning methodologies. Furthermore, we performed enrichment analyses and deconvolution algorithms to elucidate the complex interactions between immune cell infiltration and the processes of PCD and stress within a substantial array of transcriptomic data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus data base (GEO) related to CRC. Single-cell sequencing and biochemical experiments were used to validate the interaction between the model genes and programmed cell death in tumor cells. RESULTS The ICDS prognostic model exhibited robust predictive performance in seven independent cohorts, revealing an inverse correlation between model scores and patient prognosis. Meanwhile, the ICDS index was positively correlated with clinical stage. Model analysis indicated that patient subgroups with low ICDS index exhibited heightened immune activation features and elevated activity in PCD and stress pathways. Single-cell analysis further revealed that macrophages were the central drivers of immune characteristics underlying prognostic differences within the ICDS prognostic model. Pseudotime analysis and cellular experiments indicated that the model gene GAL3ST4 promotes the transition of macrophages toward an M2 pro-tumor phenotype. Furthermore, cell communication analysis and experimental validation revealed that the cuproptosis in tumor cells suppress GAL3ST4 expression, thereby inhibiting M2-like macrophage polarization. CONCLUSION In summary, we constructed the ICDS prognostic model and uncovered the mechanism by which tumor cells downregulate GAL3ST4 expression via cuproptosis to inhibit M2-like macrophage polarization, providing new targets and biomarkers for CRC treatment and prognosis evaluation.
Collapse
Affiliation(s)
- Hao Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Chuhan Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sanfei Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhan Yin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yishi Xu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sihan Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Liping Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
30
|
Yan Y, Duan T, Xue X, Yang X, Liu M, Ma B, Duan X, Su C. LBP-CD155 Liposome Nanovaccine Efficiently Resist Colorectal Cancer and Enhance ICB Therapy. Int J Nanomedicine 2025; 20:1047-1063. [PMID: 39877587 PMCID: PMC11773180 DOI: 10.2147/ijn.s492734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/25/2024] [Indexed: 01/31/2025] Open
Abstract
Background Colorectal cancer (CRC) is a highly malignant and aggressive gastrointestinal tumor. Due to its weak immunogenicity and limited immune, cell infiltration lead to ineffective clinical outcomes. Therefore, to improve the current prophylaxis and treatment scheme, offering a favorable strategy efficient against CRC is urgently needed. Methods Here, we developed a nanovaccine (LBP-CD155L NVs) loaded with CD155 gene in liposome, which was modified by Lycium barbarum polysaccharides (LBP) through electrostatic interaction. The nanovaccine was characterized by transmission electron microscopy and Zetasizer. It was evaluated in vitro, where NVs facilitated the endocytosis and maturation of DCs, and in vivo, where NVs improved the efficacy of prophylaxis and therapy. In addition, further confirmed the mechanisms by how TLR4 and MGL synergistic pathway endow the nanovaccines towards dendritic cells (DCs). Finally, the safety and tumor immunosuppressive microenvironment were evaluated in the CRC tumor-bearing mouse model. Results We successfully developed a nanovaccine that facilitates the endocytosis and maturation of DCs via a synergistic pathway involving TLR4 and MGL, which endow the nanovaccines towards dendritic cells (DCs) and promote the differentiation, thereby enhancing the cytotoxicity of CD8+T cells. Consequently, LBP-CD155L NVs can potentiate the efficacy of prophylactic and therapeutic administration in a mouse CRC model, as evidenced by decreased infiltration of myeloid-derived suppressor cells (MDSCs) and Tregs, reprogrammed the macrophage phenotypes, which promoted polarization from M2-like macrophages to M1-like macrophages, increased infiltration of effector T cells. Prophylactic and therapeutic combination regimens with anti-PD-1 treatment demonstrate synergism that stimulates T-cell infiltration into tumors and counteracts immunosuppression, leading to remarkable tumor remission and enhancing the efficacy of immune checkpoint therapy in solid tumors. Conclusion Our work provided that LBP-CD155L NVs may serve as a promising tool for reversing tumor immunosuppressive microenvironment and enhancing ICB therapy in CRC.
Collapse
Affiliation(s)
- Yajuan Yan
- School of Basic Medicine, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Ting Duan
- School of Basic Medicine, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Xiaonan Xue
- School of Basic Medicine, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Xiaojuan Yang
- School of Inspection, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Miao Liu
- School of Inspection, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Bin Ma
- Department of Surgery, The First People’s Hospital of Yinchuan, Yinchuan, Ningxia, People’s Republic of China
- The Second School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, People’s Republic of China
| | - Xiangguo Duan
- School of Inspection, Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Chunxia Su
- School of Basic Medicine, Ningxia Medical University, Yinchuan, People’s Republic of China
| |
Collapse
|
31
|
Wang H, Zhai M, Li M, Han C, Liu L, Huang C, Zhao L, Yu D, Tao K, Ren J, Lin Z, Zhang T. Phenotypic plasticity and increased infiltration of peripheral blood-derived TREM1 + mono-macrophages following radiotherapy in rectal cancer. Cell Rep Med 2025; 6:101887. [PMID: 39793571 PMCID: PMC11866444 DOI: 10.1016/j.xcrm.2024.101887] [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: 10/29/2024] [Accepted: 12/05/2024] [Indexed: 01/13/2025]
Abstract
In our previously reported phase 2 and phase 3 studies, the combination of short-course radiotherapy and neoadjuvant immunochemotherapy (SIC) is established as effective cancer therapies for locally advanced rectal cancer (LARC). Here, we apply multi-omic analyses to paired pre- and post-treatment LARC specimens undergoing SIC. The peripheral blood-derived TREM1+ mono-macrophage subsets that display a pro-inflammatory phenotype are identified and correlate with complete response to SIC. Mechanically, ionizing radiation (IR) induces peripheral TREM1+ mono-macrophage expansion in tumors. Following IR, the loss of TREM1 in mono-macrophages undermines antitumor immunity by altering mono-macrophage differentiation and inhibiting CD8+ T cell infiltration and activation. The TREM1+ mono-macrophage response may rely on activation of key inflammatory pathways, including nuclear factor κB (NF-κB) signaling and Toll-like receptor pathway. Pharmacological inhibition of TREM1 signaling abolishes IR-induced immunoactivation and reduces combined IR and/or anti-PD-1 treatment. Thus, we establish a crucial role of a mono-macrophage state in mediating effective cancer therapy.
Collapse
Affiliation(s)
- Haihong Wang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Menglan Zhai
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Mingjie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Chaoqun Han
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lichao Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Chuying Huang
- Hubei Selenium and Human Health Institute, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China; Hubei Provincial Key Lab of Selenium Resources and Bioapplications, Enshi, China
| | - Lei Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, China.
| |
Collapse
|
32
|
Li W, Li K, Chen Y, Wang S, Xu K, Ye S, Zhao B, Yuan H, Li Z, Shen Y, Mou T, Wang Y, Zhou W, Ma W. IRF1 transcriptionally up-regulates CXCL10 which increases CD8 + T cells infiltration in colorectal cancer. Int Immunopharmacol 2025; 144:113678. [PMID: 39591825 DOI: 10.1016/j.intimp.2024.113678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
Tumor-infiltrating CD8+ T cell is a robust predictor of outcome and immunotherapy response in patients with CRC. However, limited introduction of intratumoral CD8+ T cells remains a barrier for treatment of CRC. One of the most effective but difficult therapy for CD8+ T cells entering the tumor is activating chemokine receptors. This study observed a decrease in the expression level of interferon regulator factor 1(IRF1) in CRC tumor tissues compared to matched non-tumor tissues. Furthermore, it found a positive correlation between low IRF1 expression and unfavorable prognosis in CRC patients. The present study also demonstrated that overexpression of IRF1 attenuated tumor growth by promoting the accumulation of facilitating CD8+T cells at the tumor site in mouse models. Additionally, this study identified IRF1 response elements in the promoter region of CXCL10 and show that the binding of IRF1 promoted the transcription of CXCL10. Of note, it was discovered that an increase in CXCL10 was positively associated with improved survival in CRC. These findings strongly suggest that IRF1 serves as a key transcription factor for CXCL10, highlighting its potential as a therapeutic target for CRC.
Collapse
Affiliation(s)
- Wenyi Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Kejun Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yuehong Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shunyi Wang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Ke Xu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Shengzhi Ye
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Bohou Zhao
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Haitao Yuan
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Zhenghao Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yunhao Shen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Tingyu Mou
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China
| | - Yanan Wang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China.
| | - Weijie Zhou
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China.
| | - Wenhui Ma
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Clinical Research Academy of Chinese Medicine, Jichang Road No. 16, Guangzhou 510405, China; State Key Laboratory of Organ Failure Research, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, No. 1838 Guangzhou Avenue North, Guangzhou 510515, China; Department of Gastrointestinal and Hernia Surgery, Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
33
|
Chen X, Zeng Q, Yin L, Yan B, Wu C, Feng J, Wu Y, He J, Ding W, Zhong J, Shen Y, Zu X. Enhancing immunotherapy efficacy in colorectal cancer: targeting the FGR-AKT-SP1-DKK1 axis with DCC-2036 (Rebastinib). Cell Death Dis 2025; 16:8. [PMID: 39788945 PMCID: PMC11718245 DOI: 10.1038/s41419-024-07263-8] [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: 07/09/2024] [Revised: 11/17/2024] [Accepted: 11/26/2024] [Indexed: 01/12/2025]
Abstract
This research demonstrates that DCC-2036 (Rebastinib), a potent third-generation tyrosine kinase inhibitor (TKI), effectively suppresses tumor growth in colorectal cancer (CRC) models with functional immune systems. The findings underscore the capacity of DCC-2036 to enhance both the activation and cytotoxic functionality of CD8+ T cells, which are crucial for facilitating anti-tumor immune responses. Through comprehensive multi-omics investigations, significant shifts in both gene and protein expression profiles were detected, notably a marked decrease in DKK1 levels. This reduction in DKK1 was linked to diminished CD8+ T cell effectiveness, correlating with decreased FGR expression. Moreover, our findings identify FGR as a pivotal modulator that influences DKK1 expression via the PI3K-AKT-SP1 signaling cascade. Correlative analysis of clinical specimens supports the experimental data, showing that increased levels of FGR and DKK1 in CRC tissues are associated with inferior clinical outcomes and reduced efficacy of immunotherapeutic interventions. Consequently, targeting the FGR-AKT-SP1-DKK1 pathway with DCC-2036 could potentiate immunotherapy by enhancing CD8+ T cell functionality and their tumor infiltration. This strategy may contribute significantly to the refinement of therapeutic approaches for CRC, potentially improving patient prognoses.
Collapse
Affiliation(s)
- Xiguang Chen
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- The First Affiliated Hospital, Gastrointestinal Surgery Department, Hengyang, Hunan, 421001, PR China
| | - Qiting Zeng
- The First Affiliated Hospital, Department of Clinical Laboratory Medicine, Hengyang, Hunan, 421001, PR China
| | - Liyang Yin
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Bingru Yan
- Central Hospital of Hengyang City, Oncology Department, Hengyang, Hunan, 421001, PR China
| | - Chen Wu
- The First Affiliated Hospital, Department of Ultrasound Imaging, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jianbo Feng
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Ying Wu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jun He
- The Nanhua Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Wenjun Ding
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
| | - Jing Zhong
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, University of South China, Hengyang, Hunan, 421001, China
| | - Yingying Shen
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, University of South China, Hengyang, Hunan, 421001, China.
| | - Xuyu Zu
- The First Affiliated Hospital, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, PR China.
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
34
|
Chen Z, Xu J, Fang K, Jiang H, Leng Z, Wu H, Zhang Z, Wang Z, Li Z, Sun M, Zhao Z, Feng A, Zhang S, Chu Y, Ye L, Xu M, He L, Chen T. FOXC1-mediated serine metabolism reprogramming enhances colorectal cancer growth and 5-FU resistance under serine restriction. Cell Commun Signal 2025; 23:13. [PMID: 39773485 PMCID: PMC11708197 DOI: 10.1186/s12964-024-02016-8] [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: 08/05/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
Colorectal cancer (CRC) is the most common gastrointestinal malignancy, and 5-Fluorouracil (5-FU) is the principal chemotherapeutic drug used for its treatment. However, 5-FU resistance remains a significant challenge. Under stress conditions, tumor metabolic reprogramming influences 5-FU resistance. Serine metabolism plasticity is one of the crucial metabolic pathways influencing 5-FU resistance in CRC. However, the mechanisms by which CRC modulates serine metabolic reprogramming under serine-deprived conditions remain unknown. We found that exogenous serine deprivation enhanced the expression of serine synthesis pathway (SSP) genes, which in turn supported CRC cell growth and 5-FU resistance. Serine deprivation activate the ERK1/2-p-ELK1 signaling axis, leading to upregulated FOXC1 expression in CRC cells. Elevated FOXC1 emerged as a critical element, promoting the transcription of serine metabolism enzymes PHGDH, PSAT1, and PSPH, which in turn facilitated serine production, supporting CRC growth. Furthermore, through serine metabolism, FOXC1 influenced purine metabolism and DNA damage repair, thereby increasing 5-FU resistance. Consequently, combining dietary serine restriction with targeted therapy against the ERK1/2-pELK1-FOXC1 axis could be a highly effective strategy for treating CRC, enhancing the efficacy of 5-FU.
Collapse
Affiliation(s)
- Zhukai Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiacheng Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kang Fang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hanyu Jiang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhuyun Leng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Wu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zehua Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zeyu Wang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhaoxing Li
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Mingchuang Sun
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziying Zhao
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anqi Feng
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shihan Zhang
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan Chu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lechi Ye
- Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Meidong Xu
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Lingnan He
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Tao Chen
- Endoscopy Center, Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
35
|
Jang KY, Kim KM, Ha GW, Lee MR, Park HS, Chung MJ, Moon WS, Ahn AR. PD-L1 and ALK expressions in stages III and IV colorectal cancer and their correlation with clinicopathological features. Medicine (Baltimore) 2025; 104:e41228. [PMID: 40184080 PMCID: PMC11709163 DOI: 10.1097/md.0000000000041228] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 04/05/2025] Open
Abstract
Colorectal cancer (CRC) is a type of cancer with a high recurrence rate. Studies are in the progress to identify effective treatments for CRC patients. We aimed to compare the expression of programmed death-ligand 1 (PD-L1) and anaplastic lymphoma kinase (ALK) in stages III and IV CRC patients and evaluate the clinicopathological significance associated with their expression. A total of 169 stages III and IV CRC specimens was tested for ALK (D5F3) and PD-L1 (SP142 and SP263) expression using immunohistochemistry on formalin-fixed paraffin-embedded specimens. Clinicopathological characteristics were obtained through a review of the medical records and hematoxylin and eosin slides. Expression of PD-L1 SP142 and PD-L1 SP263 was detected in 17.8% and 28.4% of CRC patients, respectively. ALK D5F3 expression was detected in 4 cases. PD-L1 SP142 expression was significantly correlated with tumor site and serum carcinoembryonic antigen (CEA) level. PD-L1 SP263 expression was associated with serum tumor marker level and tumor-infiltrating lymphocytes. In univariate analysis, PD-L1 expression was correlated with shorter survival in CRC patients. PD-L1 SP263 expression was an independent indicator of shorter survival in multivariate analysis. PD-L1 expression was associated with poor prognostic factors, including shorter survival. Further investigation is needed to understand the mechanisms of the association between PD-L1 expression and unfavorable CRC prognosis.
Collapse
Affiliation(s)
- Kyu Yun Jang
- Department of Pathology, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Kyoung Min Kim
- Department of Pathology, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Gi Won Ha
- Department of General Surgery, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Min Ro Lee
- Department of General Surgery, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Ho Sung Park
- Department of Pathology, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Myoung Ja Chung
- Department of Pathology, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Woo Sung Moon
- Department of Pathology, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| | - Ae Ri Ahn
- Department of Pathology, Jeonbuk National University Medical School, Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, and Research Institute for Endocrine Sciences, Jeonju, Jeonbuk, Republic of Korea
| |
Collapse
|
36
|
Hu X, Huang Z, Li L. LDHB Mediates Histone Lactylation to Activate PD-L1 and Promote Ovarian Cancer Immune Escape. Cancer Invest 2025; 43:70-79. [PMID: 39587817 DOI: 10.1080/07357907.2024.2430283] [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: 09/11/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND To investigate the effects of LDHB on lactylation of programmed cell death 1 ligand (PD-L1) and immune evasion of ovarian cancer. METHODS Ovarian cancer cells were transfected with LDHB siRNA and cultured with primed T cells. Cell proliferation and viability were measured by cell counting kit 8 (CCK-8) and colony formation assay. The production of immune factors was detected by enzyme-linked immunosorbent assay (ELISA). The histone lactylation and activity of PD-L1 promoter were measured by chromatin immunoprecipitation (ChIP)-qPCR assay and luciferase reporter gene assay, respectively. RESULTS Knockdown of LDHB notably inhibited the growth, glucose uptake, lactate production, and ATP production of ovarian cancer cells. Knockdown of LDHB enhanced the killing effects of T cells, led to increased production of immune activation factors IL-2, TNF-α, and IFN-γ, as well as elevated the levels of granzyme B and perforin. Mechanical study identified that LDHB regulated the H3K18 lactylation (H3K18la) modification on PD-L1 promoter region to promote its expression. Overexpression of PD-L1 abolished the immune activation effects that induced by siLDHB. CONCLUSION The LDHB modulated lactate production and the histone lactylation on PD-L1 promoter, which ultimately regulated its expression and participated in the immune evasion of ovarian cancer cells.
Collapse
Affiliation(s)
- Xuemei Hu
- Department of Obstetrics and Gynecology, People's Hospital of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Zhenqiang Huang
- Department of Clinical Laboratory, People's Hospital of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| | - Lingyun Li
- Department of Clinical Laboratory, People's Hospital of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, China
| |
Collapse
|
37
|
Wang J, Hua D, Li M, Liu N, Zhang Y, Zhao Y, Jiang S, Hu X, Wang Y, Zhu H. The Role of Zuo Jin Wan in Modulating the Tumor Microenvironment of Colorectal Cancer. Comb Chem High Throughput Screen 2025; 28:523-532. [PMID: 38284730 DOI: 10.2174/0113862073281374231228041841] [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: 09/11/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024]
Abstract
INTRODUCTION Traditional Chinese medicine (TCM) can modulate the immune function of tumor patients in various ways. Zuojin Wan (ZJW, a 6:1 ratio of Huang Lian and Wu Zhu Yu) can modulate the microenvironment of ulcerative colitis, but its role in regulating the colorectal cancer (CRC) microenvironment remains unclear. Exploring the role of ZJW in CRC immunomodulation may improve the antitumor effect of existing immunotherapeutic strategies. MATERIAL AND METHODS The active compounds of each herb in ZJW were obtained from the HIT2.0 database with literature evidence. Single-cell RNA sequencing data of CRC were obtained from published studies (PMID: 32451460, 32103181, and 32561858). Pathway enrichment was analyzed using the reactome database, and intergenic correlation analysis was performed using the corrplot R software package. ZJW-regulated gene expression was verified by RT-qPCR. RESULTS Huang Lian and Wu Zhu Yu contain 19 and 4 compounds, respectively. Huang Lian targets 146 proteins, and Wu Zhu Yu targets 28 proteins based on evidence from the literature. ZJW regulates a range of biological processes associated with immune function, including cytokine signaling and Toll-Like Receptor 4 (TLR4) cascade. ZJW regulates malignant CRC cells, immune cells (including T-cells, B-cells, mast cells, NK/NKT cells, and myeloid cells), and other nonimmune cells (including endothelial cells, enteric glial cells, and pericytes). We confirmed that ZJW significantly downregulated the expression of TIMP1 and MTDHin CRC cell lines. CONCLUSIONS ZJW regulates a range of cells in the CRC microenvironment, including malignant CRC, immune cells, and stromal cells. In CRC cell lines, downregulation of TIMP1 and MTDH by ZJW may play an important role in the immunomodulation in CRC.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dongming Hua
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mengyao Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ningning Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yiyang Zhao
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shasha Jiang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xueqing Hu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huirong Zhu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
38
|
Li X, Ye C, Wang M, Kwan P, Tian X, Zhang Y. Crosstalk Between the Nervous System and Colorectal Cancer. Neurosci Bull 2025; 41:93-106. [PMID: 38879846 PMCID: PMC11748644 DOI: 10.1007/s12264-024-01238-7] [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: 11/27/2023] [Accepted: 03/22/2024] [Indexed: 01/05/2025] Open
Abstract
The nervous system is the dominant regulatory system in the human body. The traditional theory is that tumors lack innervation. However, an increasing number of studies have shown complex bidirectional interactions between tumors and the nervous system. Globally, colorectal cancer (CRC) is the third most common cancer. With the rise of tumor neuroscience, the role of nervous system imbalances in the occurrence and development of CRC has attracted increasing amounts of attention. However, there are still many gaps in the research on the interactions and mechanisms involved in the nervous system in CRC. This article systematically reviews emerging research on the bidirectional relationships between the nervous system and CRC, focusing on the following areas: (1) Effects of the nervous system on colon cancer. (2) Effects of CRC on the nervous system. (3) Treatment of CRC associated with the nervous system.
Collapse
Affiliation(s)
- Xi Li
- Jining Medical University, Jining, 272000, China
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Chunshui Ye
- Department of Gastrointestinal Surgery, Jining No. 1 People's Hospital, Jining, 272000, China
| | - Min Wang
- Department of Neurology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Patrick Kwan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
- Department of Neurology, Alfred Health, Melbourne, VIC, 3004, Australia.
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, VIC, 3004, Australia.
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, 400016, China.
| | - Yanke Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, 272000, China.
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
39
|
Ou QL, Chang YL, Liu JH, Yan HX, Chen LZ, Guo DY, Zhang SF. Mapping the intellectual structure and landscape of colorectal cancer immunotherapy: A bibliometric analysis. Hum Vaccin Immunother 2024; 20:2323861. [PMID: 38497584 PMCID: PMC10950274 DOI: 10.1080/21645515.2024.2323861] [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: 12/01/2023] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
Immunotherapy, particularly immune checkpoint inhibitor (ICIs) therapy, stands as an innovative therapeutic approach currently garnering substantial attention in cancer treatment. It has become a focal point of numerous studies, showcasing significant potential in treating malignancies, including lung cancer and melanoma. The objective of this research is to analyze publications regarding immunotherapy for colorectal cancer (CRC), investigating their attributes and identifying the current areas of interest and cutting-edge advancements. We took into account the publications from 2002 to 2022 included in the Web of Science Core Collection. Bibliometric analysis and visualization were conducted using CiteSpace, VOSviewer, R-bibliometrix, and Microsoft Excel. The quantity of publications associated with this domain has been steadily rising over the years, encompassing 3753 articles and 1498 reviews originating from 573 countries and regions, involving 19,166 institutions, 1011 journals, and 32,301 authors. In this field, China, the United States, and Italy are the main countries that come forward for publishing. The journal with the greatest impact factor is CA-A Cancer Journal for Clinicians. Romain Cohen leads in the number of publications, while Le Dt stands out as the most influential author. The immune microenvironment and immune infiltration are emerging as key hotspots and future research directions in this domain. This research carries out an extensive bibliometric examination of immunotherapy for colorectal cancer, aiding researchers in understanding current focal points, investigating possible avenues for research, and recognizing forthcoming development trends.
Collapse
Affiliation(s)
- Qin Ling Ou
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Yong Long Chang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jin Hui Liu
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Hai Xia Yan
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Zi Chen
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duan Yang Guo
- College of Integrated Traditional Chinese & Western Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Si Fang Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR, Duttaroy AK, Pathak S. Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective. Int J Colorectal Dis 2024; 40:1. [PMID: 39731596 DOI: 10.1007/s00384-024-04790-w] [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] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Metastatic colorectal cancer (mCRC) continues to present significant challenges, particularly in patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors. This narrative review aims to provide recent developments in immunotherapy for CRC treatment, focusing on its efficacy and challenges. METHODS This review discussed the various immunotherapeutic strategies for CRC treatment, including immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, combination therapies involving ICIs with other modalities, chimeric antigen receptor T-cell (CAR-T) cell therapy, and cancer vaccines. The role of the tumor microenvironment and immune evasion mechanisms was also explored to understand their impact on the effectiveness of these therapies. RESULTS This review provides a comprehensive update of recent advancements in immunotherapy for CRC, highlighting the potential of various immunotherapeutic approaches, including immune checkpoint inhibitors, combination therapies, CAR-T therapy, and vaccination strategies. The results of checkpoint inhibitors, particularly in patients with MSI-H/dMMR tumors, which have significant improvements in survival rates have been observed. Furthermore, this review also addresses the challenges faced in treating pMMR/MSS CRC, which remains resistant to immunotherapy. CONCLUSION Immunotherapy plays a significant role in the treatment of CRC, particularly in patients with MSI-H/dMMR tumors. However, many challenges remain, especially in treating pMMR/MSS CRC. This review discussed the need for further research into combination therapies, biomarker development, CAR-T cell therapy, and a deeper understanding of immune evasion mechanisms for CRC treatment.
Collapse
Affiliation(s)
- Vaishak Kaviyarasan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Dikshita Deka
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Biki Saha
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| | - Neeta Raj Sharma
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| |
Collapse
|
41
|
Pi JK, Chen XT, Zhang YJ, Chen XM, Wang YC, Xu JY, Zhou JH, Yu SS, Wu SS. Insight of immune checkpoint inhibitor related myocarditis. Int Immunopharmacol 2024; 143:113559. [PMID: 39536487 DOI: 10.1016/j.intimp.2024.113559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
As the understanding of immune-related mechanisms in the development and progression of cancer advances, immunotherapies, notably Immune Checkpoint Inhibitors (ICIs), have become integral in comprehensive cancer treatment strategies. ICIs reactivate T-cell cytotoxicity against tumors by blocking immune suppressive signals on T cells, such as Programmed Death-1 (PD-1) and Cytotoxic T-lymphocyte Antigen-4 (CTLA-4). Despite their beneficial effects, ICIs are associated with immune-related adverse events (irAEs), manifesting as autoimmune side effects across various organ systems. A particularly alarming irAE is life-threatening myocarditis. This rare but severe side effect of ICIs leads to significant long-term cardiac complications, including arrhythmias and heart failure, and has been observed to have a mortality rate of up to 50% in affected patients. This greatly limits the clinical application of ICI-based immunotherapy. In this review, we provide a comprehensive summary of the current knowledge regarding the diagnosis and management of ICI-related myocarditis. We also discuss the utility of preclinical mouse models in understanding and addressing this critical challenge.
Collapse
Affiliation(s)
- Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao-Ting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xue-Mei Chen
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yin-Chan Wang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jia-Yi Xu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jin-Han Zhou
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shuai-Shuai Yu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Si-Si Wu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
42
|
Luo Y, Liang G, Zhang Q, Luo B. The role of cGAS-STING signaling pathway in colorectal cancer immunotherapy: Mechanism and progress. Int Immunopharmacol 2024; 143:113447. [PMID: 39515043 DOI: 10.1016/j.intimp.2024.113447] [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: 09/06/2024] [Revised: 10/09/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
Colorectal cancer (CRC) is a common malignant tumor in the gastrointestinal tract, it is known as the "silent killer", which poses a serious threat to the lives of patients. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) signaling pathway responds to DNA by sensing, which plays an important role in anti-infection, autoimmune diseases and anti-tumor immunity. Recent studies have found that the activation of cGAS-STING pathway in CRC can induce the expression and secretion of type I interferon (IFN-I) and a variety of inflammatory factors, further activate anti-tumor CD8+ T cells, exert anti-tumor immune response, and inhibit the progression of CRC. Therefore, targeting the cGAS-STING pathway and developing drugs that can regulate the cGAS-STING pathway are of great significance for improving the therapeutic effect and prognosis of CRC patients. In this review, we introduce the cGAS-STING signaling pathway and the regulatory role of this signaling pathway in CRC immune microenvironment. In addition, we discussed the research progress of cGAS-STING pathway in CRC immunotherapy and the clinical research status of STING agonists developed against this pathway, emphasizing the clinical potential of CRC immunotherapy based on the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Yan Luo
- Department of Abdominal Radiotherapy, Hubei Provincial Cancer Hospital, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Hubei Province, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Wuhan, China.
| | - Gai Liang
- Department of Abdominal Radiotherapy, Hubei Provincial Cancer Hospital, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Hubei Province, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Wuhan, China
| | - Qu Zhang
- Department of Abdominal Radiotherapy, Hubei Provincial Cancer Hospital, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Hubei Province, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Wuhan, China
| | - Bo Luo
- Department of Abdominal Radiotherapy, Hubei Provincial Cancer Hospital, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Hubei Province, Wuhan, China; Colorectal Cancer Clinical Medical Research Center of Wuhan, China.
| |
Collapse
|
43
|
Sharif E, Nezafat N, Mohit E. Recombinant ClearColi™-derived outer membrane vesicles as an effective carrier for development of neoepitope-based vaccine candidate against colon carcinoma. Int Immunopharmacol 2024; 143:113283. [PMID: 39418728 DOI: 10.1016/j.intimp.2024.113283] [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: 07/27/2024] [Revised: 09/07/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Colorectal carcinoma (CRC) is the third most common cancer worldwide, with high clonal heterogeneity due to somatic mutations. Poly neoepitope vaccines can inhibit the tumor's escape from the immune system. However, they have rapid clearance and low immunogenicity. Bacteria-derived recombinant outer membrane vesicles (OMVs) have gained increased attention as ideal cancer vaccine candidates due to their unique adjuvant properties and ability to carry antigens. Herein, the benefits of OMV-based and polyneoepitope-based vaccines were combined to obtain a functional individualized cancer vaccine. METHODS OMVs and rOMVs displaying CT26 polytopes were isolated from ClearColi™ and recombinant ClearColi™ containing pET-22b (ClyA-CT26 polytope) by the AS (70 %) + UDF method. BALB/c mice were immunized with OMVs (40 µg) and rOMVs (20 and 40 µg) and subcutaneously challenged with CT26 cells. Then, IgG1 and IgG2a antibodies specific for CT26 M90 and CT26 polytope, the stimulated IFN-γ, TNF-α, and IL-10 cytokines and the stimulated CTL responses by measuring granzyme B were evaluated. To investigate whether pooled sera and pooled splenocytes are indicators of individual responses, pooled and individual methods for determining the elicited immunity were compared. Additionally, the ability of OMVs and rOMVs (20 and 40 µg) to prevent tumor growth against the CT26 challenge was investigated. RESULTS Immunization with rOMVs displaying CT26 polytopes induced a higher titer of CT26 polytope- and CT26 M90 peptide-specific IgG2a than IgG1 antibodies in a dose-dependent manner, thus directing immunity to Th1. The antibody responses determined by pooled sera can be used as indicators of individual responses. In addition, both OMVs and rOMVs displaying CT26 polytopes could induce tumor-suppressing cytokines (IFN-γ and TNF-α). The ability of rOMVs displaying CT26 polytopes to induce these cytokines was higher than OMVs in a dose-dependent way. The results of the granzyme B assay were also in agreement with the cytokine assay. The survival of mice after the CT26 challenge was 100 % in the OMVs and rOMVs groups, and inhibition of tumor growth was significantly higher by rOMVs (40 µg) compared to OMVs. CONCLUSION The bioengineered OMVs displaying CT26 neoepitopes have the potential for the development of personalized tumor vaccines. Our results can provide new insights for developing rOMV-based vaccines displaying polytopes against diseases containing highly variable antigens.
Collapse
Affiliation(s)
- Elham Sharif
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Mohit
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Zhang X, Ma J, Chen Y, Deng X, Zhang Y, Han Y, Tan J, Deng G, Ouyang Y, Zhou Y, Cai C, Zeng S, Shen H. FOS + B cells: Key mediators of immunotherapy resistance in diverse cancer types. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200895. [PMID: 39583007 PMCID: PMC11584611 DOI: 10.1016/j.omton.2024.200895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/02/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024]
Abstract
While immunotherapy has marked significant advances in cancer treatment, resistance remains a challenge. The complexity of the tumor microenvironment, particularly the role of B cell subpopulations, is a critical factor affecting treatment efficacy. In this study, we conducted analyses of single-cell RNA sequencing data from immunotherapy patients (n = 25) to explore the biomarker of immunotherapy resistance. Spatial transcriptome analysis, immunofluorescence analysis, and multi-cancer immunotherapy transcriptome analysis (n = 1,253) were used to validate our finding, and the potential mechanisms were explored. FOS+ B cells, identified across multiple cancer types, were associated with poor response to immunotherapy. FOS may form AP-1 (activator protein 1) with JUNB, thereby promoting the expression of Blimp-1 and subsequently facilitating the differentiation of B cells into immunosuppressive plasma cells. Furthermore, FOS+ B cells were linked to altered tumor necrosis factor signaling pathways, suggesting a mechanism for their immunosuppressive effects. Our findings highlight FOS+ B cells as important players in immunotherapy resistance, providing a novel biomarker for predicting treatment response. This study not only deepens our understanding of the immunological landscape influencing immunotherapy efficacy but also opens avenues for targeted interventions to overcome resistance.
Collapse
Affiliation(s)
- Xiangyang Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong 518057, China
| | - Jiayao Ma
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiangying Deng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yan Zhang
- Department of Oncology, Yueyang People’s Hospital, Yueyang Hospital Affiliated to Hunan Normal University, Yueyang 414022, Hunan, China
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Gongping Deng
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311 China
| | - Yanhong Ouyang
- Department of Emergency, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311 China
| | - Yulai Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008 China
| |
Collapse
|
45
|
Liang R, Ding D, Li Y, Lan T, Ryabtseva S, Huang S, Ren J, Huang H, Wei B. HDACi combination therapy with IDO1i remodels the tumor microenvironment and boosts antitumor efficacy in colorectal cancer with microsatellite stability. J Nanobiotechnology 2024; 22:753. [PMID: 39676171 DOI: 10.1186/s12951-024-02936-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/16/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Immunotherapy for colorectal cancer (CRC) with microsatellite stability (MSS) and mismatch repair proficiency (pMMR) has shown limited success in clinical trials. The combination of immunomodulators and immune checkpoint inhibitors (ICIs) is a potential strategy for treating CRC. METHODS Histone deacetylase (HDAC) and indoleamine 2,3-dioxygenase 1 (IDO1) expression in CRC tissues and adjacent normal tissues was analyzed via database analysis, immunohistochemistry, and western blotting. A nanodrug designated as NP-I/P was subsequently formulated, encapsulating an IDO1 inhibitor (IDO1i; namely, epacadostat) and an immunomodulatory HDAC inhibitor (HDACi; namely, panobinostat). The antitumor efficacy of the nanoparticles and their effects on tumor microenvironment features were evaluated via in vitro and in vivo experiments. RESULTS In the present study, we found that HDAC overexpression and IDO1 expression were attenuated in MSS/pMMR CRC. Thus, a nanodrug designated as NP-I/P was formulated to encapsulate epacadostat and panobinostat. In vitro, NP-I/P treatment promoted the apoptosis of tumor cells and induced the release of damage-associated molecular patterns, thereby leading to cell death-associated immune activation. The in vivo results revealed that NP-I/P treatment reversed the immunosuppressive phenotype of the microenvironment by inducing tumor immunogenic cell death (ICD), promoting CD8+ T cell infiltration, and reducing the numbers of Tregs, tumor-associated macrophages, and myeloid-derived suppressor cells. Finally, the results of the patient-derived xenograft and patient-derived organoid models demonstrated that NP-I/P treatment triggered tumor cell death and modulated the immune microenvironment in human CRC. CONCLUSION The combination of IDO1 and HDAC inhibitors represents a promising strategy for CRC treatment, and NP-I/P is a candidate for clinical trials.
Collapse
Affiliation(s)
- Rongpu Liang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Dongbing Ding
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Yiquan Li
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China
| | - Tianyun Lan
- Central Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Svetlana Ryabtseva
- Center of Electronic and Light Microscopy, Institute of Physiology of the National Academy of Sciences of Belarus, Minsk, Belarus
| | - Shengxin Huang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China
| | - Jiannan Ren
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China
| | - He Huang
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China.
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China.
| | - Bo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, P.R. China.
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China.
| |
Collapse
|
46
|
Shan H, Wang M, Huang S, Liu H, Liu J, Du Q. Efficacy and Safety of Bevacizumab Biosimilar (Encoda) Compared With Reference Bevacizumab (Avastin) in Patients With Metastatic Colorectal Cancer: A Multicenter, Real-World Study. Clin Med Insights Oncol 2024; 18:11795549241303726. [PMID: 39669102 PMCID: PMC11635854 DOI: 10.1177/11795549241303726] [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: 06/14/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024] Open
Abstract
Background The bevacizumab biosimilar (Encoda), which was approved by the National Medical Products Administration (NMPA) in China in 2019, is a biosimilar of bevacizumab. Approval of bevacizumab biosimilar (Encoda) for metastatic colorectal cancer (mCRC) was based on the extrapolation principle of biosimilar. However, there is currently no available data regarding the efficacy and safety of both bevacizumab biosimilar (Encoda) and bevacizumab in patients with mCRC. Methods The present real-world study included patients with mCRC who received first-line therapy with either bevacizumab biosimilar (Encoda) or bevacizumab combined with backbone chemotherapy between April 2021 and December 2022. The overall response rate (ORR) was the primary endpoint of the study. Bevacizumab biosimilar (Encoda) would be considered equivalent to bevacizumab if it met any of the following criteria: the 90% CI for ORR risk ratio of bevacizumab biosimilar (Encoda): bevacizumab was included within the predefined equivalence range (0.75 to 1.33) as specified by the NMPA or within the predefined equivalence range (0.73 to 1.36) as specified by US Food and Drug Administration (FDA); the 95% CI for the ORR risk difference of 2 groups was included within the predefined equivalence range (-0.12 to 0.15) as specified by the European Medicines Agency (EMA). Results The study included a total of 436 patients, with 234 receiving bevacizumab biosimilar (Encoda) and 202 receiving bevacizumab. The ORR was 42.3% (95% CI: 35.9% to 48.9%) in the bevacizumab biosimilar (Encoda) group and 42.1% (95% CI: 35.2% to 49.2%) in the bevacizumab group. The ORR risk ratio and risk difference were 1.005 (90% CI: 0.836 to 1.210) and 0.002 (95% CI: -0.091 to 0.095), respectively, both included within the prespecified equivalence range. The overall safety profile was comparable between the 2 groups. Conclusions This real-world study is the first to demonstrate the efficacy equivalence of bevacizumab biosimilar (Encoda) and bevacizumab in first-line treatment of mCRC.
Collapse
Affiliation(s)
- Han Shan
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengmeng Wang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuohan Huang
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pharmacy, Zhongshan Hospital Fudan University, Shanghai, China
| | - Hongyue Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiyong Liu
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiong Du
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
47
|
Stukalin I, Gupta M, Buhler K, Forbes N, Heitman SJ, Ma C. Brief Report: Trends in Incidence, Mortality, and Disability-Adjusted Life Years for Early-Onset Colorectal Cancer in Canada Between 1990 and 2019. Curr Oncol 2024; 31:7765-7769. [PMID: 39727694 DOI: 10.3390/curroncol31120571] [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: 09/29/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Colorectal cancer is the third most common malignancy globally. Early-onset colorectal cancer (EOCRC) is becoming a growing healthcare focus globally, particularly in North America. We estimated trends in incidence, mortality, and disability-adjusted life years (DALYs) for EOCRC in Canada between 1990 and 2019. METHODS We used the Global Burden of Diseases Study to evaluate trends in incidence, mortality, and DALYs for EOCRC in Canada between 1990 and 2019. Rates were estimated per 100,000 persons at risk with associated uncertainty intervals (UIs). Annual percentage changes (APC) were estimated using joinpoint regression with 95% confidence intervals (CIs). RESULTS In 2019, the incidence, mortality, and DALYs rates for EOCRC were 10.89 (95% UI 8.09, 14.34), 2.24 (95% UI 2.00, 2.51), and 111.37 (95% UI 99.34, 124.78) per 100,000 individuals, respectively. Incidence increased during the study period by 1.12%/year (95% CI 1.03%, 1.22%; p < 0.001). The largest increase in incidence in EOCRC occurred between 1990 and 2007, with an APC of 2.23% (95% CI 2.09%, 2.37%; p < 0.001). Mortality (APC 2.95%, 95% CI 1.89%, 4.02%; p < 0.001) and DALY (APC 2.96%, 95% CI 1.84%, 4.09%; p < 0.001) rates increased for males between 2001 and 2006. CONCLUSIONS Our study reveals a substantial burden in EOCRC in Canada, with a significant increase in incidence.
Collapse
Affiliation(s)
- Igor Stukalin
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Mehul Gupta
- Division of Internal Medicine, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Katherine Buhler
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Nauzer Forbes
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Steven J Heitman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Christopher Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, AB T2N 4Z6, Canada
| |
Collapse
|
48
|
Patel H, Patel A, Vats M, Patel K. Albumin and Polysorbate-80 Coated Sterile Nanosuspensions of Mebendazole for Glioblastoma Therapy. AAPS PharmSciTech 2024; 25:271. [PMID: 39586850 DOI: 10.1208/s12249-024-02978-5] [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] [Accepted: 10/17/2024] [Indexed: 11/27/2024] Open
Abstract
The scarcity of existing and novel therapies for brain cancer has significantly affected the survival rate of glioblastoma patients. Mebendazole (MBZ), an antiparasitic agent demonstrated promising activity against brain cancer. However, poor solubility, multiple polymorphs, and insufficient permeability through blood-brain barrier (BBB) restricts its therapeutic efficacy through parenteral administration. The current study aimed to develop, optimize, and characterize sterile, injectable nanosuspension of mebendazole using parenterally acceptable stabilizers. Albumin and polysorbate 80 (PS-80) coated MBZ Nanosuspension (NS) was prepared using wet media milling technique. Design of experiment (DoE) approach was used to understand effect of drug loading versus stabilizer concentration. The optimized MBZ NS showed hydrodynamic diameter of 208.36 ± 0.24 nm with a poly dispersibility index (PDI) of 0.210 ± 0.03 and zeta potential of -20.41 ± 0.36 mV. The IC50 value of MBZ NS in U-87 MG and LN-229 cell lines were found to be 0.49 ± 0.02 μM and 0.48 ± 0.05 μM, respectively. Additionally, MBZ NS demonstrated a 2.65-fold decrease in colony-forming efficiency and a 1.16-fold reduction in migration of the bridging area compared to MBZ. In 3D spheroids of the U-87 MG glioma cell line, MBZ NS exhibited a 50% reduction in tumor growth and increased cell apoptosis compared to the control. MBZ NS formulations were sterilized by gamma irradiation and tested as per the USP sterility test. Albumin-PS 80 coated NS is rendered to be useful parenteral delivery of mebendazole for the treatment of brain cancer.
Collapse
Affiliation(s)
- Himaxi Patel
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA
| | - Akanksha Patel
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA
| | - Mukti Vats
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, St. Albert Hall, B-49, 8000 Utopia Parkway, Queens, New York, 11439, USA.
| |
Collapse
|
49
|
Yin X, Song Y, Deng W, Blake N, Luo X, Meng J. Potential predictive biomarkers in antitumor immunotherapy: navigating the future of antitumor treatment and immune checkpoint inhibitor efficacy. Front Oncol 2024; 14:1483454. [PMID: 39655071 PMCID: PMC11625675 DOI: 10.3389/fonc.2024.1483454] [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: 08/20/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment modality, offering promising outcomes for various malignancies. However, the efficacy of ICIs varies among patients, highlighting the essential need of accurate predictive biomarkers. This review synthesizes the current understanding of biomarkers for ICI therapy, and discusses the clinical utility and limitations of these biomarkers in predicting treatment outcomes. It discusses three US Food and Drug Administration (FDA)-approved biomarkers, programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), and microsatellite instability (MSI), and explores other potential biomarkers, including tumor immune microenvironment (TIME)-related signatures, human leukocyte antigen (HLA) diversity, non-invasive biomarkers such as circulating tumor DNA (ctDNA), and combination biomarker strategies. The review also addresses multivariable predictive models integrating multiple features of patients, tumors, and TIME, which could be a promising approach to enhance predictive accuracy. The existing challenges are also pointed out, such as the tumor heterogeneity, the inconstant nature of TIME, nonuniformed thresholds and standardization approaches. The review concludes by emphasizing the importance of biomarker research in realizing the potential of personalized immunotherapy, with the goal of improving patient selection, treatment strategies, and overall outcomes in cancer treatment.
Collapse
Affiliation(s)
- Xiangyu Yin
- Department of Biological Sciences, School of Science, AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Yunjie Song
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Wanglong Deng
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Neil Blake
- Institute of Infection, Veterinary & Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Xinghong Luo
- Jiangsu Simcere Diagnostics Co., Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Jia Meng
- Department of Biological Sciences, School of Science, AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Institute of Biomedical Research, Regulatory Mechanism and Targeted Therapy for Liver Cancer Shiyan Key Laboratory, Hubei Provincial Clinical Research Center for Precise Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
50
|
Gu Y, Li C, Yan Y, Ming J, Li Y, Chao X, Wang T. Comprehensive Analysis and Verification of the Prognostic Significance of Cuproptosis-Related Genes in Colon Adenocarcinoma. Int J Mol Sci 2024; 25:11830. [PMID: 39519383 PMCID: PMC11546850 DOI: 10.3390/ijms252111830] [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: 09/21/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Colon adenocarcinoma (COAD) is a frequently occurring and lethal cancer. Cuproptosis is an emerging type of cell death, and the underlying pathways involved in this process in COAD remain poorly understood. Transcriptomic and clinical data for COAD patients were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We investigated alterations in DNA and chromatin of cuproptosis-related genes (CRGs) in COAD. In order to identify predictive differentially expressed genes (DEGs) and various molecular subtypes, we used consensus cluster analysis. Through univariate, multivariate, and Lasso Cox regression analyses, four CRGs were identified. A risk prognostic model for cuproptosis characteristics was constructed based on four CRGs. This study also examined the association between the risk score and the tumor microenvironment (TME), the immune landscape, and drug sensitivity. We distinguished two unique molecular subtypes using consensus clustering analysis. We discovered that the clinical characteristics, prognosis, and TME cell infiltration characteristics of patients with multilayer CRG subtypes were all connected. The internal and external evaluations of the predicted accuracy of the prognostic model built using data derived from a cuproptosis risk score were completed at the same time. A nomogram and a clinical pathological analysis make it more useful in the field of medicine. A significant rise in immunosuppressive cells was observed in the high cuproptosis risk score group, with a correlation identified between the cuproptosis risk score and immune cell infiltration. Despite generally poor prognoses, the patients with a high cuproptosis risk but low tumor mutation burden (TMB), cancer stem cell (CSC) index, or microsatellite instability (MSI) may still benefit from immunotherapy. Furthermore, the cuproptosis risk score positively correlated with immune checkpoint gene expression. Analyzing the potential sensitivity to medications could aid in the development of clinical chemotherapy regimens and decision-making. CRGs are the subject of our in-depth study, which exposed an array of regulatory mechanisms impacting TME. In addition, we performed additional data mining into clinical features, prognosis effectiveness, and possible treatment medications. COAD's molecular pathways will be better understood, leading to more precise treatment options.
Collapse
Affiliation(s)
- Yixiao Gu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.G.); (C.L.); (Y.Y.)
| | - Chengze Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.G.); (C.L.); (Y.Y.)
| | - Yinan Yan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.G.); (C.L.); (Y.Y.)
| | - Jingmei Ming
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 100029, China; (J.M.); (Y.L.)
| | - Yuanhua Li
- School of Chinese Material Medical, Beijing University of Chinese Medicine, Beijing 100029, China; (J.M.); (Y.L.)
| | - Xiang Chao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (Y.G.); (C.L.); (Y.Y.)
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| |
Collapse
|