1
|
Yu Y, Zhu N, Zhang Y, Zeng Y, An D, Zhang J, Yi Q, Wu Y. Monitoring melanoma immunotherapeutic efficacy through successive metabolic labeling and PD-L1-confined signal amplification based on membrane characteristics of the newly generated circulating tumor cells. Biosens Bioelectron 2025; 280:117403. [PMID: 40179698 DOI: 10.1016/j.bios.2025.117403] [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: 12/31/2024] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025]
Abstract
Unlike conventional detection, monitoring membrane characteristics of circulating tumor cells (CTCs) within a specific time frame can effectively indicate tumor progression, yet the challenge lies in selectively isolating CTCs generated during this period and precisely identifying subtle progression-related changes. This study focuses on CTCs newly generated during melanoma immunotherapy, utilizing a strategic combination of tumor-specific glycometabolic engineering, phenotypic protein-confined biotinylation and lanthanide luminescence to enrich and detect these spatiotemporally specific CTCs. First, Ac4ManNAz-associated cellular glycometabolic engineering selectively developes azide groups on the membranes of these CTCs, providing clickable artificial tags to distinguish them from pre-existing CTCs and blood cells. Subsequently, Fe3O4/lip-DBCO nanoparticles enrichs this CTC population through efficient click chemistry, achieving a capture efficiency exceeding 90 % for various types of CTCs, regardless of their phenotype, tumor type or species. Following this, a PD-L1-confined biotinylation is adopted to generate a significant number of biotin moieties in close proximity to the membrane PD-L1 of the captured CTCs, resulting in a 9-fold increase in the number of active sites available for introducing detection signal sources, compared to traditional immunofluorescent labeling methods. Further introduction of streptavidin-functionalized europium (Eu)-lanthanide nanoparticles (EuNPs-SA) quantifies the captured CTCs in a time-resolved manner and assesses CTC-related therapeutic response, while eliminating interferences from background biological substances. Specifically, significant advancements in detection performance have enabled the assessment of immunotherapeutic efficacy within melanoma model. Notably, substantial differences in lanthanide luminescent signals between the treated and untreated groups p = 0.000149), with the observed trends closely correlating with treatment outcomes.
Collapse
Affiliation(s)
- Yue Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Nanhang Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Yujia Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China; HC Scientific (Chengdu) L.L.C., Chengdu, 610000, PR China
| | - Yating Zeng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Di An
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Jinyu Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China
| | - Qiangying Yi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China.
| | - Yao Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, PR China.
| |
Collapse
|
2
|
Gao H, Sun L, Wang H, Ji X, Shen Q, Chen D, Jiao Y, Ni D, Zheng X, Bao Z. In situ non-canonical activation and sensitization of cGAS-STING pathway with manganese telluride nanosheets. Biomaterials 2025; 318:123170. [PMID: 39933314 DOI: 10.1016/j.biomaterials.2025.123170] [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: 01/02/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
Immune checkpoint blockade (ICB) has achieved encouraging outcome in various malignant tumors. However, the low immunogenicity and insufficient infiltration of T cells within tumors severely limit the curative effects. Herein, we reported synthesis and experimental evaluation of H2O2-responsive MnTe2 nanosheets (NSs) for improving anti-tumor immune responses. Within the tumor microenvironment characterized by high level of H2O2, the MnTe2 NSs were degraded to release Mn2+ and TeO42- which subsequently induced cellular endoplasmic reticulum (ER) stress and non-canonically activated the cGAS-STING pathway. Moreover, the cellular Mn2+ ions enhanced the sensitivity of cGAS-STING pathway and the maturation of dendritic cells (DCs) concurrently. Ultimately, the MnTe2 NSs exhibited favorable in vivo anti-tumor immune effects, especially in combination with PD-L1 checkpoint inhibitors. These findings provided compelling evidence for exploration and utilization of nanomedicine to leverage innate immune system for better tumor immunotherapy.
Collapse
Affiliation(s)
- Hongbo Gao
- Department of Radiation Oncology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Li Sun
- Department of Radiation Oncology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Xiuru Ji
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Qianwen Shen
- Department of Radiation Oncology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Di Chen
- Department of Radiation Oncology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Yuxin Jiao
- Department of Radiation Oncology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China.
| | - Xiangpeng Zheng
- Department of Radiation Oncology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China.
| | - Zhijun Bao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, PR China; Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, 200040, PR China.
| |
Collapse
|
3
|
Sun J, Wu X, Zhang X, Huang W, Zhong X, Li X, Xue K, Liu S, Chen X, Li W, Liu X, Shen H, You J, He W, Jin Z, Yu L, Li Y, Zhang S, Zhang B. Radiomic Model Associated with Tumor Microenvironment Predicts Immunotherapy Response and Prognosis in Patients with Locoregionally Advanced Nasopharyngeal Carcinoma. RESEARCH (WASHINGTON, D.C.) 2025; 8:0749. [PMID: 40556946 PMCID: PMC12187091 DOI: 10.34133/research.0749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 05/31/2025] [Accepted: 06/01/2025] [Indexed: 06/28/2025]
Abstract
Background: No robust biomarkers have been identified to predict the efficacy of programmed cell death protein 1 (PD-1) inhibitors in patients with locoregionally advanced nasopharyngeal carcinoma (LANPC). We aimed to develop radiomic models using pre-immunotherapy MRI to predict the response to PD-1 inhibitors and the patient prognosis. Methods: This study included 246 LANPC patients (training cohort, n = 117; external test cohort, n = 129) from 10 centers. The best-performing machine learning classifier was employed to create the radiomic models. A combined model was constructed by integrating clinical and radiomic data. A radiomic interpretability study was performed with whole slide images (WSIs) stained with hematoxylin and eosin (H&E) and immunohistochemistry (IHC). A total of 150 patient-level nuclear morphological features (NMFs) and 12 cell spatial distribution features (CSDFs) were extracted from WSIs. The correlation between the radiomic and pathological features was assessed using Spearman correlation analysis. Results: The radiomic model outperformed the clinical and combined models in predicting treatment response (area under the curve: 0.760 vs. 0.559 vs. 0.652). For overall survival estimation, the combined model performed comparably to the radiomic model but outperformed the clinical model (concordance index: 0.858 vs. 0.812 vs. 0.664). Six treatment response-related radiomic features correlated with 50 H&E-derived (146 pairs, |r|= 0.31 to 0.46) and 2 to 26 IHC-derived NMF, particularly for CD45RO (69 pairs, |r|= 0.31 to 0.48), CD8 (84, |r|= 0.30 to 0.59), PD-L1 (73, |r|= 0.32 to 0.48), and CD163 (53, |r| = 0.32 to 0.59). Eight prognostic radiomic features correlated with 11 H&E-derived (16 pairs, |r|= 0.48 to 0.61) and 2 to 31 IHC-derived NMF, particularly for PD-L1 (80 pairs, |r|= 0.44 to 0.64), CD45RO (65, |r|= 0.42 to 0.67), CD19 (35, |r|= 0.44 to 0.58), CD66b (61, |r| = 0.42 to 0.67), and FOXP3 (21, |r| = 0.41 to 0.71). In contrast, fewer CSDFs exhibited correlations with specific radiomic features. Conclusion: The radiomic model and combined model are feasible in predicting immunotherapy response and outcomes in LANPC patients. The radiology-pathology correlation suggests a potential biological basis for the predictive models.
Collapse
Affiliation(s)
- Jie Sun
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xuewei Wu
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiao Zhang
- Medical AI Lab, The First Hospital of Hebei Medical University,
Hebei Medical University, Shijiazhuang, Hebei, China
- Hebei Provincial Engineering Research Center for AI-Based Cancer Treatment Decision-Making, The First Hospital of Hebei Medical University,
Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Oncology, The First Hospital of Hebei Medical University,
Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weiyuan Huang
- Department of Radiology,
Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, Hainan, China
| | - Xi Zhong
- Department of Medical Imaging, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xueyan Li
- Department of Radiology, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Kaiming Xue
- Department of Radiology,
The Third Bethune Hospital of Jilin University, Changchun, Jilin, China
| | - Shuyi Liu
- Department of Radiology, Guangzhou Women and Children’s Medical Center,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xianjie Chen
- Department of Radiology, The Affiliated Panyu Central Hospital,
Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenzhu Li
- Department of Radiology,
Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, Hainan, China
| | - Xin Liu
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Hui Shen
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Wenle He
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhe Jin
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lijuan Yu
- Department of Radiology,
Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), Haikou, Hainan, China
| | - Yuange Li
- Department of Radiology,
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shuixing Zhang
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Bin Zhang
- Department of Radiology,
The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Kolawole OP, Pramanik A, Kasani-Akula P, Rai S, Janorkar S, Edorodion Z, Gates K, Kundu S, Ray PC. Multifunctional Nanoplatform for Highly Accurate Profiling of Triple-Negative Breast Cancer-Derived Chemo- and Immunotherapy Resistance Exosomes. ACS APPLIED BIO MATERIALS 2025. [PMID: 40526975 DOI: 10.1021/acsabm.5c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most malignant breast cancer with a higher mortality rate, which is due to the lack of targeted therapies and the development of resistance to chemotherapy and immunotherapy. Since the ubiquitin carboxyl-terminal hydrolase L1 (UCH-L1) (+) exosome promotes chemoresistance and the programmed cell death ligand-1 (PD-L1) (+) exosome plays a crucial role in immunotherapy resistance, there is an urgent need in clinics to explore strategies for the rapid profiling of UCH-L1(+) and PD-L1(+) exosomes from blood to predict chemo- and immunotherapy treatment outcomes. Herein, we develop a multifunctional nanoplatform using antibody-conjugated green- and red-emissive carbon quantum dots attached to cobalt ferrite (CoFe2O4) magnetic nanoparticles for the highly accurate profiling of TNBC-derived UCH-L1(+) and PD-L1(+) exosomes. Specifically, by employing multiple microscopic, spectroscopic, and SQUID magnetometer techniques, we show that the multifunctional nanoplatform exhibits good chemical stability, high photostability, strong photoluminescence quantum yield, excellent superparamagnetic behavior, and biocompatibility. Moreover, by leveraging strategically designed anti-UCH-L1 antibody-attached red luminescence (660 nm) magnetic nanoplatform and anti-PD-L1 antibody-attached green luminescence (530 nm) magnetic nanoplatform, the reported data demonstrate that the multifunctional nanoplatform has the capability for capturing and screening ∼100% UCH-L1(+) and PD-L1(+) exosomes selectively from infected whole-blood samples. Furthermore, we showcase the potential application for the screening of UCH-L1 (+) and PD-L1(+) exosomes simultaneously from whole blood, which indicates that the nanoplatform may be used for monitoring chemotherapeutic and immunotherapeutic resistance of cancer.
Collapse
Affiliation(s)
- Olorunsola Praise Kolawole
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Avijit Pramanik
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Pragathi Kasani-Akula
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Shivangee Rai
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Sanika Janorkar
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Zoe Edorodion
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Kaelin Gates
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Sanchita Kundu
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| | - Paresh Chandra Ray
- Department of Chemistry and Biochemistry, Jackson State University, Jackson, Mississippi 39217, United States
| |
Collapse
|
5
|
Ni C, Sun Q, Yin H. Comprehensive multi-omics analysis of histone acetylation modulators identifies ASH1L as a novel aggressive marker for osteosarcoma. Discov Oncol 2025; 16:1070. [PMID: 40504347 PMCID: PMC12162460 DOI: 10.1007/s12672-025-02920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 06/05/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Osteosarcoma, a highly malignant bone tumor prevalent in children and adolescents, continues to have poor long-term survival rates, particularly in metastatic cases. While histone acetylation dysregulation has been implicated in cancer progression, the role of histone acetylation modification-related proteins (HAMRPs) in osteosarcoma immune infiltration and prognosis remains unclear. METHODS The expression patterns, prognostic implications, and clinical correlations of HAMRPs in osteosarcoma were analyzed using the TARGET, GEO, TISCH, and HPA databases. The effectiveness of HAMRPs in predicting the immune landscape of osteosarcoma was confirmed using CIBERSORT, ssGSEA, and ESTIMATE algorithms. The study employed GSEA analysis, wound healing assay, Transwell, and western blot to explore the role and regulatory mechanism of the key gene ASH1L in osteosarcoma progression. RESULTS Two distinct histone acetylation modification patterns were identified, showing significant differences in survival, clinical features, and immune landscape. Comprehensive clinical correlation analysis and Kaplan-Meier analysis of all HAMRPs used for two subtypes revealed that higher ASH1L expression was found in metastatic osteosarcoma cases and indicated poorer survival outcomes. In vitro experiments confirmed that ASH1L promoted osteosarcoma metastasis and epithelial-mesenchymal transition via the AKT/mTOR pathway. Additionally, an ASH1L-derived risk model was developed to aid personalized clinical decisions. CONCLUSIONS This study elucidates the prognostic and immunological significance of HAMRPs and highlights ASH1L as a novel aggressive marker in osteosarcoma.
Collapse
Affiliation(s)
- Chenlie Ni
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Qiwen Sun
- Haining Yuanhua Township Central Health Hospital, Jiaxing, 314000, China
| | - Haibo Yin
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China.
| |
Collapse
|
6
|
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] [MESH Headings] [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
|
7
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [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: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
8
|
Yang F, Xue H, Fan Y, Zhang T, Wang T, Gu F, Guan L, Zhou L, Guan X, Chen G. Engineered hybrid cell membrane nanovesicles for potentiated cancer immunotherapy through dual immune checkpoint inhibition. Biomater Sci 2025; 13:2642-2650. [PMID: 40202456 DOI: 10.1039/d5bm00298b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated remarkable success in treating various types of solid tumors; however, only a limited number of patients currently benefit from these therapeutic agents. Developing novel ICIs that elicit systemic and durable antitumor immune responses remains a significant challenge in improving immunotherapy outcomes. In this study, we engineered PD-1/LAG-3 receptors onto cell membrane nanovesicles to simultaneously block two immune checkpoints for the treatment of colorectal cancer. This dual-checkpoint blockade strategy led to significantly more potent tumor growth suppression in mice with MC38 xenografts compared to nanovesicles targeting PD-1 or LAG-3 alone. Notably, the hybrid nanovesicles substantially rejuvenated exhausted CD8+ T cells, promoting dendritic cell maturation and depleting regulatory T cells (Tregs). This research highlights the promising potential of cell membrane nanovesicles as an effective platform for delivering multiple immune checkpoints in cancer immunotherapy, offering a novel strategy to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Fuxu Yang
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Han Xue
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Yuxin Fan
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Ting Zhang
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Ting Wang
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Fanlin Gu
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Longxue Guan
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Lisha Zhou
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Xingang Guan
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Guofu Chen
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| |
Collapse
|
9
|
Nahon P. Establishing five-year overall survival as a new standard for trials in advanced HCC. J Hepatol 2025:S0168-8278(25)02199-3. [PMID: 40373978 DOI: 10.1016/j.jhep.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Affiliation(s)
- Pierre Nahon
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, APHP, Liver Unit, Bobigny, Université Sorbonne Paris Nord, F-93000 Bobigny, Inserm, UMR-1138 "Functional Genomics of Solid Tumors", Centre de recherche des Cordeliers, Université de Paris, Paris, France.
| |
Collapse
|
10
|
Nykaza I, Murciano-Goroff YR, Desilets A, Harada G, Postow MA, Callahan MK, Lee CH, Rudin CM, Kelsen DP, Stadler ZK, Wibmer AG, Hechtman JF, Drilon A, Friedman CF. Sarcoid-like reactions in patients treated with checkpoint inhibitors for advanced solid tumors. Oncologist 2025; 30:oyaf017. [PMID: 40349135 PMCID: PMC12065934 DOI: 10.1093/oncolo/oyaf017] [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: 06/17/2024] [Accepted: 12/23/2024] [Indexed: 05/14/2025] Open
Abstract
IMPORTANCE While new intrathoracic adenopathy in a patient with cancer can represent progression of disease, the differential diagnosis is broad. Sarcoid-like reactions (SLR) remain an underreported source of lymphadenopathy in patients treated with immune checkpoint inhibitors (ICI), with limited reports in patients with cancers other than melanoma. OBJECTIVE To characterize SLRs among patients treated with ICI for advanced solid tumors. METHODS Data were collected on the clinical, pathologic, and radiographic presentation of patients treated with ICI who developed clinical or imaging findings suggestive of an SLR, including the presence of hilar or mediastinal lymphadenopathy, cutaneous/subcutaneous involvement, and/or worsening of existing sarcoidosis on ICI. RESULTS Twelve patients were identified as having experienced an SLR. While 6 patients had melanoma, SLRs were also observed among patients with lung, gynecologic, and genitourinary cancers, including high-grade serous ovarian carcinoma, and an angiomyolipoma. Median time from initiation of ICI to diagnosis of an SLR was 3.4 months (range: 1.8-9.1). All but one patient (92%) were deemed to have had a radiographic response to ICI. CONCLUSIONS AND RELEVANCE Clinicians should maintain the awareness of the possibility of SLRs in patients receiving ICI, particularly in patients whose scans show evidence of "mixed" response, with decreases in certain lesions coupled with new/increasing intrathoracic lymphadenopathy and/or other systemic signs of sarcoid.
Collapse
Affiliation(s)
- Ian Nykaza
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Yonina R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Antoine Desilets
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Guilherme Harada
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Margaret K Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - David Paul Kelsen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Zsofia K Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Andreas G Wibmer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Jaclyn F Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| | - Claire F Friedman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, United States
| |
Collapse
|
11
|
Huang K, Bostock IC. Role of Metastasectomy for Tissue Acquisition for Tumor-Infiltrating Lymphocyte Harvest and Biomarker/Genomic Testing. Thorac Surg Clin 2025; 35:169-174. [PMID: 40246406 DOI: 10.1016/j.thorsurg.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Tumor-infiltrating lymphocyte therapy is an emerging paradigm with promising early results for a patient population without many good alternatives. In some cases where the primary tumor or soft tissues metastases cannot be harvested, there may be a role for the thoracic surgeon to harvest tissue from pulmonary metastases. Typically, these can be limited resections with most cases being amenable to a wedge resection as only 1 cm of tissue is usually required. As a result, in appropriately selected patients, there has been shown to be a low risk of pulmonary, cardiovascular, infectious, and wound complications.
Collapse
Affiliation(s)
- Kevin Huang
- Department of Surgery, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Ian C Bostock
- Department of Thoracic Surgery, Miami Cancer Institute, Baptist Health South Florida, 8900 N Kendall Drive, Miami, FL 33176, USA.
| |
Collapse
|
12
|
Liu J, Wu Z, Zhou S, Lv W, Wang Y, Xia P, Zhu L, Hu J. Neoadjuvant immunochemotherapy for locally advanced esophageal squamous cell carcinoma in real-world practice: an analysis of the clinical outcomes and long-term survival, and the feasibility of using major pathological response as a surrogate endpoint. Eur J Med Res 2025; 30:342. [PMID: 40301916 PMCID: PMC12038973 DOI: 10.1186/s40001-025-02599-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/15/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Neoadjuvant immunochemotherapy is expected to become the standard treatment mode for locally advanced esophageal squamous cell carcinoma (ESCC). This study aims to analyze the clinical outcomes and long-term survival of neoadjuvant immunochemotherapy for locally advanced ESCC, and explore the feasibility of using major pathological response (MPR) as a surrogate endpoint. METHODS This real-world retrospective study consecutively included eligible patients with stage II-IVA locally advanced ESCC who received neoadjuvant immunochemotherapy and surgery between 2019 and 2022 at the Department of Thoracic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine. RESULTS This study collected a total of 166 patients, and ultimately included 126 patients after screening. The objective response rate (ORR) was 69.8% (88/126). The incidence of grade 3-4 adverse events (AEs) was 13.5% (17/126). MPR was observed in 49 (38.9%) patients, and 24 (19.0%) patients achieved a complete pathological response (pCR). The median progression-free survival (PFS) was 31.7 months and the 3-year PFS rate was 56.3%. The median overall survival (OS) was not reached and the 3-year OS rate was 70.6%. The median PFS of the non-MPR group was 25.0 months, with the MPR group not achieved (hazard ratio [HR], 2.503; 95% CI 1.359-4.610; P = 0.0022). The median OS in the non-MPR group was 31.7 months and not reached in the MPR group (HR, 3.607; 95% CI 1.576-8.254; P = 0.0012). MPR is an independent prognostic factor affecting OS (HR, 2.522; 95% CI 1.018-6.401; P = 0.046). CONCLUSIONS Neoadjuvant immunochemotherapy is safe and effective for locally advanced ESCC, and can result in certain survival benefits. MPR can serve as a surrogate endpoint for predicting long-term OS.
Collapse
Affiliation(s)
- Jiacong Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Ziheng Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shihong Zhou
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Wang Lv
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Yiqing Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Pinghui Xia
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China
| | - Linhai Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China.
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, China.
- Key Laboratory of Clinical Evaluation Technology for Medical Device of Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
13
|
Rimassa L, Chan SL, Sangro B, Lau G, Kudo M, Reig M, Breder V, Ryu MH, Ostapenko Y, Sukeepaisarnjaroen W, Varela M, Tougeron D, Crysler OV, Bouattour M, Van Dao T, Tam VC, Faccio A, Furuse J, Jeng LB, Kang YK, Kelley RK, Paskow MJ, Ran D, Xynos I, Kurland JF, Negro A, Abou-Alfa GK. Five-year overall survival update from the HIMALAYA study of tremelimumab plus durvalumab in unresectable HCC. J Hepatol 2025:S0168-8278(25)00226-0. [PMID: 40222621 DOI: 10.1016/j.jhep.2025.03.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND & AIMS In the phase III HIMALAYA study (NCT03298451), STRIDE (Single Tremelimumab Regular Interval Durvalumab) significantly improved overall survival (OS) versus sorafenib in unresectable HCC (uHCC) and demonstrated long-term survival benefits. We report an updated exploratory analysis of OS with 5 years of follow-up, including survival by multiple tumour response measures. METHODS Participants were randomised to STRIDE (tremelimumab plus durvalumab), durvalumab or sorafenib. OS, depth of response and serious adverse events (AEs) were assessed. Extended long-term survivors (eLTS; ≥48 months beyond randomisation) were described. Updated data cut-off: 01 March 2024. RESULTS Median (95% CI) follow-up durations were 62.49 (59.47-64.79) months (STRIDE) and 59.86 (58.32-61.54) months (sorafenib). The OS HR (95% CI) for STRIDE versus sorafenib was 0.76 (0.65-0.89). OS rates at 60 months for STRIDE versus sorafenib were 19.6% versus 9.4% overall, 28.7% versus 12.7% in participants achieving disease control per RECIST v1.1 and 50.7% versus 26.3% in participants achieving >25% tumour shrinkage. No late-onset treatment-related serious AEs were reported for STRIDE. There were more eLTS with STRIDE (83/393, 21.1%) than sorafenib (45/389, 11.6%), and extended long-term survival occurred across all clinically relevant subgroups. CONCLUSIONS At 5 years, STRIDE sustained an OS benefit versus sorafenib and maintained a manageable safety profile. OS benefit with STRIDE was improved in participants with disease control. Data suggest that any degree of tumour shrinkage with STRIDE can be associated with improved OS, indicating that conventional response measures may not fully capture STRIDE benefits. Nevertheless, participants experiencing deep responses appear to have the greatest benefit. STRIDE continues to set new benchmarks in uHCC with 1 in 5 patients alive at 5 years. IMPACT AND IMPLICATIONS The phase III HIMALAYA study showed that STRIDE (Single Tremelimumab Regular Interval Durvalumab) improved overall survival (OS) versus sorafenib in participants with unresectable HCC (uHCC), including after 4 years of follow-up. Understanding the efficacy and safety of STRIDE over the longer term is important for healthcare providers; here, we demonstrate that STRIDE sustained an OS benefit versus sorafenib and maintained a manageable safety profile after 5 years of follow-up. OS benefit with STRIDE was improved in participants with disease control and any degree of tumour shrinkage, indicating that conventional response measures may not fully capture the benefits of STRIDE. These findings are important as they set new benchmarks in uHCC and may help guide clinical decisions in the future.
Collapse
Affiliation(s)
- Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy.
| | - Stephen L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir Yue-Kong Pao Center for Cancer, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Bruno Sangro
- Liver Unit and HPB Oncology Area, Clínica Universidad de Navarra and CIBEREHD, Pamplona - Madrid, Spain
| | - George Lau
- Humanity and Health Clinical Trial Center, Humanity and Health Medical Group, Hong Kong SAR, China
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Maria Reig
- Barcelona Clinic Liver Cancer (BCLC), Liver Unit, Hospital Clinic de Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain
| | - Valeriy Breder
- Department of Chemotherapy, N. N. Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yuriy Ostapenko
- Department of Minimally Invasive and Endoscopic Surgery, Interventional Radiology, National Cancer Institute, Kyiv, Ukraine
| | | | - Maria Varela
- Liver Unit, Hospital Universitario Central de Asturias, IUOPA, ISPA, FINBA, Universidad de Oviedo, Oviedo, Spain
| | - David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Oxana V Crysler
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Tu Van Dao
- Cancer Research and Clinical Trials Center, Department of Optimal Therapy, National Cancer Hospital, Hanoi, Vietnam
| | - Vincent C Tam
- Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Adilson Faccio
- Department of Oncology, CEON - Centro Especializado em Oncologia, Ribeirao Preto, Brazil
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Long-Bin Jeng
- Department of Surgery, China Medical University and Hospital, Taichung, Taiwan, Republic of China
| | - Yoon Koo Kang
- Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Robin K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Michael J Paskow
- Global Medical Affairs, AstraZeneca, Gaithersburg, Maryland, USA
| | - Di Ran
- Statistics, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ioannis Xynos
- Oncology R&D, Late-Stage Development, AstraZeneca, Cambridge, UK
| | - John F Kurland
- Oncology R&D, Late-Stage Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Alejandra Negro
- Oncology R&D, Late-Stage Development, AstraZeneca, Gaithersburg, Maryland, USA
| | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Cornell University, New York, New York, USA; Weill Medical College, Cornell University, New York, New York, USA; Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Bi C, Patel JS, Liang SH. Development of CD73 Inhibitors in Tumor Immunotherapy and Opportunities in Imaging and Combination Therapy. J Med Chem 2025; 68:6860-6869. [PMID: 40106690 PMCID: PMC11998006 DOI: 10.1021/acs.jmedchem.4c02151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/14/2025] [Accepted: 02/13/2025] [Indexed: 03/22/2025]
Abstract
CD73 is a member of the membrane-bound enucleotidase family, which catalyzes the extracellular hydrolysis of adenosine monophosphate (AMP) to produce anti-inflammatory and immunosuppressive adenosine. As a novel checkpoint protein, CD73 is overexpressed in the immune system of various tumors, where adenosine is abundantly enriched. A large number of monoclonal antibodies (mAbs), nucleotides, and non-nucleotides as potent CD73 inhibitors are being discovered, providing opportunities for novel tumor immunotherapy. Currently, 18 CD73 inhibitors are in clinical trials, showing promising results in combination therapy for various solid tumors. The development of CD73-specific companion positron emission tomography imaging ligands holds potential for facilitating diagnosis, patient selection, and treatment efficacy evaluation throughout the entire process of CD73-targeted therapeutic development.
Collapse
Affiliation(s)
- Chunyang Bi
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- PharmaCenter
Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Jimmy S. Patel
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Department
of Radiation Oncology, Winship Cancer Institute
of Emory University, Atlanta, Georgia 30322, United States
| | - Steven H. Liang
- Department
of Radiology and Imaging Sciences, Emory
University, Atlanta, Georgia 30322, United States
- Department
of Biomedical Engineering, Emory University
and Georgia Institute of Technology, Atlanta, Georgia 30322, United States
| |
Collapse
|
15
|
Wang Q, Xiao C, Hu P, Zhang X, Lian J, Su X, Yu X, Chen J, Zheng Y. Artificial liver support systems bridge severe immune-mediated hepatotoxicity to clinical recovery. Immunopharmacol Immunotoxicol 2025; 47:194-200. [PMID: 39819181 DOI: 10.1080/08923973.2025.2454030] [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: 01/10/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND The incidence of hepatic immune-related adverse events has increased with the wide use of immune checkpoint inhibitors (ICIs), some immune-mediated hepatotoxicity (IMH) cases are severe and lack of clinical recommendations. OBJECTIVE This study aimed to evaluate the efficacy of artificial liver support systems (ALSSs) in the treatment of IMH. METHODS This retrospective case series included six patients with grade 4 hepatotoxicity with high bilirubin induced by ICIs treated between 1 January 2019 and 31 December 2021. All patients received ALSS treatment. RESULTS After treatment and recovery, four of the six patients experienced improvement in hepatotoxicity, with total bilirubin (TBIL) levels reduced to ≤ grade 2, and two patients achieved complete recovery (TBIL grade = 0). CONCLUSION ALSS serve as a therapeutic option for severe IMH.
Collapse
Affiliation(s)
- Qiangfeng Wang
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Xiao
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Peipei Hu
- Department of General Medicine, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiuming Zhang
- Department of Pathology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangshan Lian
- Department of Infectious Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xingyun Su
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiongfei Yu
- Department of Surgical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajia Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yulong Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
16
|
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
|
17
|
Zhang J, Sun Z, Li G, Ding L, Wang Z, Liu M. Discovering biomarkers associated with infiltration of CD8 + T cells and tumor-associated fibrosis in colon adenocarcinoma using single-cell RNA sequencing and gene co-expression network. Front Immunol 2025; 16:1496640. [PMID: 40230854 PMCID: PMC11994618 DOI: 10.3389/fimmu.2025.1496640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/11/2025] [Indexed: 04/16/2025] Open
Abstract
Background Colorectal adenocarcinoma (COAD) is a prevalent malignant tumor associated with a high mortality rate. Within the tumor microenvironment, CD8+ T cells play a pivotal role in the anti-tumor immune response within the human body. Fibrosis directly and indirectly affects the therapeutic response of tumor immunotherapy. However, the significance of regulatory genes associated with tumor-associated fibrosis and CD8+ T cell infiltration remains uncertain. Therefore, it is imperative to identify biomarkers with prognostic value and elucidate the precise role of CD8+ T cells and tumor-associated fibrosis. Methods We performed a single-cell transcriptome analysis of COAD samples from the GEO database. To evaluate immune infiltration in COAD samples, we utilized CIBERSORT and ESTIMATE. Furthermore, we analyzed the correlation between CD8+ T cells and immune infiltration. To analyze COAD expression's quantitative immune cell composition data, we conducted a Weighted Gene Correlation Network Analysis and utilized a deconvolution algorithm. The data for these analyses were obtained from the GEO database. We utilized univariate Cox regression and LASSO analysis to create a prognostic model. The predictive model was assessed through Kaplan-Meier analysis, and a survival prediction nomogram was created. Additionally, we analyzed the correlation between the prognostic model and chemotherapy drug sensitivity. To estimate the expression of hub genes, we employed immunohistochemistry, real-time PCR, and western blot techniques. Results Single-cell transcriptome analysis has indicated a higher prevalence of CD8+ T cells in COAD tumor samples. The connection between COAD and CD8+ T cells was further confirmed by WGCNA and deconvolution analysis using the GEO database. The Protein-Protein Interaction network analysis revealed three hub genes: LARS2, SEZ6L2, and SOX7. A predictive model was subsequently created using LASSO and univariate COX regression, which included these three genes. Two of these hub genes (LARS2 and SEZ6L2) were found to be upregulated in COAD cell lines and tissues, while SOX7 was observed to be downregulated. The prognostic model demonstrated a significant association with CD8+ T cells, suggesting that these genes could serve as potential biomarkers and targets for gene therapy in treating COAD. Conclusion This study has identified three key genes associated with CD8+ T cells and the prognosis of COAD, providing new prognostic biomarkers for diagnosing and treating COAD.
Collapse
Affiliation(s)
- Jinning Zhang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ziquan Sun
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Province Key Laboratory of Digestive Surgery and Nutrition & Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guodong Li
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Province Key Laboratory of Digestive Surgery and Nutrition & Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixian Ding
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zitong Wang
- Colorectal Cancer Surgery Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Heilongjiang Province Key Laboratory of Digestive Surgery and Nutrition & Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
18
|
Rhodin KE, O'Connor MH, Therien A, Hollander S, Geron V, Nair U, Rakestraw E, Salama AK, Shah R, Tyler DS, Beasley GM. Circulating Tumor DNA in High-Risk Stage II/III Cutaneous Melanoma: A Feasibility Study. Ann Surg Oncol 2025:10.1245/s10434-025-17194-z. [PMID: 40146490 DOI: 10.1245/s10434-025-17194-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/25/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND Adjuvant therapies reduce recurrence in patients with clinical stage IIB/IIC/III melanoma; however, better risk stratification and patient selection are needed. Circulating tumor DNA (ctDNA) as a marker of micrometastatic residual disease is being explored for such purposes in other malignancies. We aimed to explore the feasibility of serial ctDNA monitoring in patients with stage II/III melanoma, as well as the association of ctDNA elevation with disease burden and outcomes. METHODS A single-institution prospective study was conducted on patients with clinical stage IIB/IIC/III melanoma. Primary tumor was sent to Natera for generation of a tumor-informed mPCR-NGS assay (Signatera™). Peripheral blood was collected for analysis at pre-specified timepoints. Patients were stratified by ctDNA elevations both pre- and postoperatively to compare tumor characteristics and recurrence-free survival (RFS). RESULTS Overall, 30 patients were enrolled. The median Breslow depth was 4.4 mm and 70% were ulcerated. Signatera™ assays were successfully created for all 30 patients. Median follow-up from the time of surgery was 16 months and 13 patients recurred with median RFS of 19 months. Eight of these 13 patients (62%) had detectable ctDNA levels predating their clinical or radiographic recurrence. Elevated ctDNA at the first post-operative timepoint was associated with worse RFS. CONCLUSIONS ctDNA monitoring is feasible for patients with high-risk cutaneous melanoma. Our findings suggest that detectable ctDNA post-operatively may be associated with worse outcomes. Elevations during surveillance may predict subsequent clinical recurrence; however, the role of ctDNA in adjuvant therapy decision-making and surveillance is not yet ready for broad application.
Collapse
Affiliation(s)
| | | | - Aaron Therien
- Department of Surgery, Duke University, Durham, NC, USA
| | | | - Viviana Geron
- Department of Surgery, Duke University, Durham, NC, USA
| | - Uma Nair
- Department of Surgery, Duke University, Durham, NC, USA
| | | | - April K Salama
- Department of Medicine, Duke University, Durham, NC, USA
| | | | - Douglas S Tyler
- Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Georgia M Beasley
- Department of Surgery, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| |
Collapse
|
19
|
Cheng B, Li H, Hong Y, Zhou Y, Chen J, Shao C, Kong Z. Research progress in bifunctional small molecules for cancer immunotherapy. Eur J Med Chem 2025; 286:117289. [PMID: 39919914 DOI: 10.1016/j.ejmech.2025.117289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/09/2025]
Abstract
Immunotherapy has become one of the most revolutionary modalities for cancer treatment with the approval of many anti-PD-L1 (programmed cell death-ligand 1)/PD-1 (programmed cell death-1) monoclonal antibodies (mAbs). However, anti-PD-L1/PD-1 mAbs suffer from several drawbacks including limited clinical efficacy (∼20 %), poor pharmacokinetics, and the development of immune resistance. Hence, the search for PD-1/PD-L1-based combination therapies and other PD-L1-based bifunctional small molecule modulators [e.g. PD-L1/HDAC (Histone Deacetylase), PD-L1/CXCL12 (C-X-C chemokine ligand 12), PD-L1/Tubulin, PD-L1/IDO1 (Indoleamine 2,3 dioxygenase 1), PD-L1/PARP (Poly(ADP-ribose) polymerase), PD-L1/STING (Stimulator of interferon genes), and PD-L1/NAMPT (Nicotinamide phosphoribosyltransferase)-targeting dual inhibitors] has been intensified with considerable strides achieved in the past couple of years. Herein, we summarize the latest development of bifunctional small molecules as immunotherapy for tumor treatment, including those PD-L1-based, A2AR (Adenosine 2A receptor)-based, IDO1-based, Toll-like receptor (TLR)-based, SHP2 (Src homology 2 domain-containing phosphatase 2)-based, and HPK1 (Hematopoietic progenitor kinase 1)-based dual-acting compounds. In addition, we also summarize the tumorigenesis and synergy mechanism of various targets. Finally, the challenges and future directions for bifunctional small molecules for cancer immunotherapy are also discussed in detail.
Collapse
Affiliation(s)
- Binbin Cheng
- Hubei Polytechnic University, Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China; Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Hongqiao Li
- The Central Hospital of Huangshi, Huangshi, 435000, China
| | - Yimeng Hong
- Hubei Polytechnic University, Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China
| | - Yingxing Zhou
- Hubei Polytechnic University, Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, 435003, China; Huangshi Key Laboratory of Molecular Diagnosis and Individualized Treatment, Huangshi Love&health Hospital Affiliated of Hubei Polytechnic University, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Chuxiao Shao
- Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang, 323000, China.
| | - Zhihua Kong
- Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, FoShan, 528200, China.
| |
Collapse
|
20
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
21
|
Duan W, Hosea R, Wang L, Ruan C, Zhao F, Liu J, Zhao H, Miyagishi M, Wu S, Kasim V. Chromosome Missegregation Triggers Tumor Cell Pyroptosis and Enhances Anti-Tumor Immunotherapy in Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409769. [PMID: 39903759 PMCID: PMC11948012 DOI: 10.1002/advs.202409769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Immune checkpoint inhibitor (ICI) therapy is a promising anti-tumor therapeutic strategy; however, its efficacy in solid tumors is limited. Chromosome missegregation is common in various solid tumors; however, its role in tumor progression remains poorly understood, and its correlation with ICI is yet to be explored. Here, it is found that increased chromosome missegregation promotes tumor immune microenvironment, and eventually immunotherapeutic efficacy, by triggering pyroptosis. yin yang 2 (YY2) is identified as a mitotic checkpoint regulator, which promotes chromosome missegregation by upregulating BUB1B transcription. Increased chromosome missegregation promoted the formation of micronuclei and release of double-stranded DNA (dsDNA) into the cytosol, triggering an AIM2-mediated cytosolic dsDNA response. The subsequent pyroptosis strengthened the tumor immune microenvironment, thereby enhancing immunoinfiltration and cytotoxicity of CD8+ T cells, while preventing their exhaustion. Finally, through in vitro and in vivo experiments, it is demonstrated that combining YY2 overexpression-induced chromosome missegregation/cytosolic dsDNA response and PD-1 inhibitor significantly enhanced the efficacy of ICI immunotherapy in microsatellite instable and microsatellite stable colorectal cancer cells. Together, these findings provide new insights on the role of chromosome missegregation in triggering cytosolic dsDNA response-mediated pyroptosis and modulating the tumor immune microenvironment, suggesting a novel strategy for improving ICI therapeutic efficacy in colorectal cancer.
Collapse
Affiliation(s)
- Wei Duan
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Rendy Hosea
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Lingxian Wang
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Cao Ruan
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Fuqiang Zhao
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Jingyi Liu
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
| | - Hezhao Zhao
- Department of Gastrointestinal SurgeryChongqing University Cancer HospitalChongqing UniversityChongqing400030China
| | - Makoto Miyagishi
- Life Science InnovationSchool of Integrative and Global MajorsUniversity of TsukubaTsukubaIbaraki305‐0006Japan
| | - Shourong Wu
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized TreatmentChongqing University Cancer HospitalChongqing UniversityChongqing400030China
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and TechnologyMinistry of EducationCollege of BioengineeringChongqing UniversityChongqing400044China
- The 111 Project Laboratory of Biomechanics and Tissue RepairCollege of BioengineeringChongqing UniversityChongqing400044China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized TreatmentChongqing University Cancer HospitalChongqing UniversityChongqing400030China
| |
Collapse
|
22
|
Mukhopadhyay P, Abdullah HA, Opalinska JB, Paka P, Richards E, Weisel K, Trudel S, Mateos MV, Dimopoulos MA, Lonial S. The clinical journey of belantamab mafodotin in relapsed or refractory multiple myeloma: lessons in drug development. Blood Cancer J 2025; 15:15. [PMID: 39920159 PMCID: PMC11806103 DOI: 10.1038/s41408-025-01212-0] [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: 10/25/2024] [Revised: 12/13/2024] [Accepted: 01/14/2025] [Indexed: 02/09/2025] Open
Abstract
Patients with relapsed/refractory multiple myeloma (RRMM) have a poor prognosis and a need remains for novel effective therapies. Belantamab mafodotin, an anti-B-cell maturation antigen antibody-drug conjugate, was granted accelerated/conditional approval for patients with RRMM who have received at least 4 prior lines of therapy, based on response rates observed in DREAMM-1/DREAMM-2. Despite the 41% response rate and durable responses observed with belantamab mafodotin in the Phase III confirmatory DREAMM-3 trial, the marketing license for belantamab mafodotin was later withdrawn from US and European markets when the trial did not meet its primary endpoint of superiority for progression-free survival compared with pomalidomide and dexamethasone. This review reflects on key lessons arising from the clinical journey of belantamab mafodotin in RRMM. It considers how incorporating longer follow-up in DREAMM-3 may have better captured the clinical benefits of belantamab mafodotin, particularly given its multimodal, immune-related mechanism of action with responses deepening over time. A non-inferiority hypothesis may have been more appropriate rather than superiority in the context of a monotherapy versus an active doublet therapy. Further, anticipation of, and planning for, non-proportional hazards arising from response heterogeneity may have mitigated loss of statistical power. With the aim of improving the efficacy of belantamab mafodotin, other Phase III trials in the RRMM development program (DREAMM-7 and DREAMM-8) proceeded to evaluate the synergistic potential of combination regimens in earlier lines of treatment. The aim was to increase the proportion of patients responding to belantamab mafodotin (and thus the likelihood of seeing a clear separation of the progression-free survival curve versus comparator regimens). Protocol amendments reflecting DREAMM-3 learnings could also be implemented prospectively on the combinations trials to optimize the follow-up duration and mitigate risk. The wider implications of the lessons learned for clinical research in RRMM and in earlier treatment settings are discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Katja Weisel
- University Medical Center of Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Sagar Lonial
- Winship Cancer Institute, Emory University Hospital, Atlanta, GA, USA.
| |
Collapse
|
23
|
Shi Y, Yu Y, Zhao J, Huang L, Wang Q, Sun Q, Liu L, Sun C. Development of a prognostic model based on four genes related to exhausted CD8+ T cell in triple-negative breast cancer patients: a comprehensive analysis integrating scRNA-seq and bulk RNA-seq. Discov Oncol 2025; 16:114. [PMID: 39899181 PMCID: PMC11790537 DOI: 10.1007/s12672-025-01812-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Low immune infiltration is closely associated with poor clinical results and an unfavorable response to therapy in triple-negative breast cancer (TNBC). T-cell exhaustion (TEX) is a significant risk factor for tumor immunosuppression and invasion. Although improving TEX and enhancing effector function are promising strategies for strengthening immunotherapy, their role in the pathogenesis of TNBC remains unclear. This study's objective was to develop a prognostic model for TNBC based on exhausted CD8+ T-cell (CD8+ Tex)-related differentially expressed genes (DEGs) and to investigate its clinical and immune relevance. Initially, 398 CD8+ Tex-related genes were screened utilizing single-cell RNA sequencing (scRNA-seq) data from TNBC patients. Pseudotime analysis confirmed that CD8+ Tex mainly clustered at the end of the differentiation pathways, making them a critical subset in TNBC progression. By analyzing the TCGA cohort, ten CD8+ Tex-related DEGs were identified as significantly correlated with overall survival (OS) in TNBC patients, and a prognostic model containing four biomarkers (GBP1, CTSD, ABHD14B, and HLA-A) was constructed. The model demonstrated robust predictive capability in both the TCGA cohort and an external cohort, with the low-risk group exhibiting elevated expression of immunological checkpoint molecules and immune cell infiltration, as well as better responses to immunotherapy and chemotherapy. Furthermore, these four biomarkers were found to be highly expressed on CD8+ Tex and were associated with cellular communication efficiency. Therefore, this model is expected to be a new method for forecasting TNBC patients' prognosis and effectiveness of treatment, providing new insights for clinical decision-making.
Collapse
Affiliation(s)
- Yulin Shi
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, and Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, 999078, Macau, China
| | - Jiahan Zhao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Linan Huang
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China
| | - Qingyang Wang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qi Sun
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261000, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261000, China.
| |
Collapse
|
24
|
Li Y, Dong Y, Shen D, Guo Y, Cao Y, Zhang K, Li X, Zhu R, Yi J, Yao X, Dang X, Li R, Zhang Z, Qin Z, Yang W. Personalized Nanovaccine Based on STING-Activating Nanocarrier for Robust Cancer Immunotherapy. ACS NANO 2025; 19:3226-3239. [PMID: 39817337 DOI: 10.1021/acsnano.4c11014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Tumor-specific T cells play a vital role in potent antitumor immunity. However, their efficacy is severely affected by the spatiotemporal orchestration of antigen-presentation as well as the innate immune response in dendritic cells (DCs). Herein, we develop a minimalist nanovaccine that exploits a dual immunofunctional polymeric nanoplatform (DIPNP) to encapsulate ovalbumin (OVA) via electrostatic interaction when the nanocarrier serves as both STING agonist and immune adjuvant in DCs. In vitro results reveal that the nanocarrier induces STING activation via facilitating interferon regulatory factor 3 phosphorylation by block poly 18-crown-6-yl methacrylate (P18C6MA) mediated K+ perturbation cascade with endoplasmic reticulum stress, and stimulates DC maturation via the Toll-like receptor 4 activation by primary amine. In vivo studies indicate that the smart nanovaccine dramatically inhibits tumor growth with a long-term immune memory response in both the B16-OVA and EG7-OVA tumor models. After combination with programmed death ligand-1 antibody (aPD-L1), mice survival rate is notably prolonged. In addition, DIPNP forms a personalized nanovaccine after resected autologous primary tumor cell membranes decoration with a high antitumor activity in a homologous distant tumor model. The rational design provides inspiration for personalized nanovaccine construction via immunofunctional nanocarriers.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ya Dong
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Danyang Shen
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yichen Guo
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kaixin Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Rongrong Zhu
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaohan Yao
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaowei Dang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Rui Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Weijing Yang
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
- School of Pharmaceutical Sciences, Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
25
|
Devasani JR, Guntuku G, Panatula N, Muthyala MKK, Palla MS, Siahaan TJ. Innovative CDR grafting and computational methods for PD-1 specific nanobody design. FRONTIERS IN BIOINFORMATICS 2025; 4:1488331. [PMID: 39897125 PMCID: PMC11782559 DOI: 10.3389/fbinf.2024.1488331] [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: 08/29/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction The development of nanobodies targeting Programmed Cell Death Protein-1 (PD-1) offers a promising approach in cancer immunotherapy. This study aims to design and characterize a PD-1-specific nanobody using an integrated computational and experimental approach. Methods An in silico design strategy was employed, involving Complementarity-Determining Region (CDR) grafting to construct the nanobody sequence. The three-dimensional structure of the nanobody was predicted using AlphaFold2, and molecular docking simulations via ClusPro were conducted to evaluate binding interactions with PD-1. Physicochemical properties, including stability and solubility, were analyzed using web-based tools, while molecular dynamics (MD) simulations assessed stability under physiological conditions. The nanobody was produced and purified using Ni-NTA chromatography, and experimental validation was performed through Western blotting, ELISA, and dot blot analysis. Results Computational findings demonstrated favorable binding interactions, stability, and physicochemical properties of the nanobody. Experimental results confirmed the nanobody's specific binding affinity to PD-1, with ELISA and dot blot analyses providing evidence of robust interaction. Discussion This study highlights the potential of combining computational and experimental approaches for engineering nanobodies. The engineered PD-1 nanobody exhibits promising characteristics, making it a strong candidate for further testing in cancer immunotherapy applications.
Collapse
Affiliation(s)
- Jagadeeswara Reddy Devasani
- Pharmaceutical Biotechnology Division, A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, India
| | - Girijasankar Guntuku
- Pharmaceutical Biotechnology Division, A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, India
| | - Nalini Panatula
- Pharmaceutical Biotechnology Division, A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, India
| | - Murali Krishna Kumar Muthyala
- Pharmaceutical Chemistry Division, A.U. College of Pharmaceutical Sciences, Andhra University, Visakhapatnam, Andhra Pradesh, India
| | - Mary Sulakshana Palla
- GITAM School of Pharmacy, GITAM Deemed to be University, Visakhapatnam, Andhra Pradesh, India
| | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS, United States
| |
Collapse
|
26
|
Dai Y, Dong C, Wang Z, Zhou Y, Wang Y, Hao Y, Chen P, Liang C, Li G. Infiltrating T lymphocytes and tumor microenvironment within cholangiocarcinoma: immune heterogeneity, intercellular communication, immune checkpoints. Front Immunol 2025; 15:1482291. [PMID: 39845973 PMCID: PMC11750830 DOI: 10.3389/fimmu.2024.1482291] [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: 08/18/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Cholangiocarcinoma is the second most common primary liver cancer, and its global incidence has increased in recent years. Radical surgical resection and systemic chemotherapy have traditionally been the standard treatment options. However, the complexity of cholangiocarcinoma subtypes often presents a challenge for early diagnosis. Additionally, high recurrence rates following radical treatment and resistance to late-stage chemotherapy limit the benefits for patients. Immunotherapy has emerged as an effective strategy for treating various types of cancer, and has shown efficacy when combined with chemotherapy for cholangiocarcinoma. Current immunotherapies targeting cholangiocarcinoma have predominantly focused on T lymphocytes within the tumor microenvironment, and new immunotherapies have yielded unsatisfactory results in clinical trials. Therefore, it is essential to achieve a comprehensive understanding of the unique tumor microenvironment of cholangiocarcinoma and the pivotal role of T lymphocytes within it. In this review, we describe the heterogeneous immune landscape and intercellular communication in cholangiocarcinoma and summarize the specific distribution of T lymphocytes. Finally, we review potential immune checkpoints in cholangiocarcinoma.
Collapse
Affiliation(s)
- Yunyan Dai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Chenyang Dong
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhiming Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yunpeng Zhou
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yi Hao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Pinggui Chen
- Department of Nuclear Medicine, Nanyang First People’s Hospital, Nanyang, Henan, China
| | - Chaojie Liang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
- Department of biliary and Pancreatic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Gaopeng Li
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
27
|
Chen Y, Qi F, Sun C, Jiang P, Xue X, Yang X, Li X, He X, Wang Y, Zhang T. Navigating the landscape of neoadjuvant immunotherapy for NSCLC: progress and controversies. Ther Adv Med Oncol 2025; 17:17588359241312501. [PMID: 39781239 PMCID: PMC11707791 DOI: 10.1177/17588359241312501] [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: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Recently, attention has increasingly centered on non-small-cell lung cancer (NSCLC) with immune checkpoint inhibitors application. Numerous clinical studies have underscored the potential of immunotherapy in treating resectable NSCLC, highlighting its role in improving patient outcomes. However, despite these promising results, there is ongoing debate regarding the efficacy of immunological combination therapy strategies, the prevalence of treatment-related side effects, the identification of predictive biomarkers, and various other challenges within the neoadjuvant context. Careful consideration is essential to maximize the benefits of immunotherapy for patients with resectable NSCLC. This article offers a detailed overview of recent advancements in neoadjuvant immunotherapy for resectable NSCLC. By examining these developments, we aim to provide new perspectives and valuable insights into the benefits and challenges of applying neoadjuvant immunotherapy in clinical settings.
Collapse
Affiliation(s)
- Yuzhu Chen
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Fei Qi
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chenhao Sun
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Peng Jiang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiangyu Xue
- Department of Biochemistry and Molecular Biology, Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, Harbin, China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Joint Laboratory for Precision Diagnosis and Treatment Translational Research in Malignant Tumors, Gynecologic Oncology Basic and Clinical Research Laboratory, Capital Medical University, Beijing, China
| | - Xiaomi Li
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xin He
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yishuo Wang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tongmei Zhang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing 101149, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Tian S, Xu M, Geng X, Fang J, Xu H, Xue X, Hu H, Zhang Q, Yu D, Guo M, Zhang H, Lu J, Guo C, Wang Q, Liu S, Zhang W. Network Medicine-Based Strategy Identifies Maprotiline as a Repurposable Drug by Inhibiting PD-L1 Expression via Targeting SPOP in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410285. [PMID: 39499771 PMCID: PMC11714211 DOI: 10.1002/advs.202410285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Indexed: 11/07/2024]
Abstract
Immune checkpoint inhibitors (ICIs) are drugs that inhibit immune checkpoint (ICP) molecules to restore the antitumor activity of immune cells and eliminate tumor cells. Due to the limitations and certain side effects of current ICIs, such as programmed death protein-1, programmed cell death-ligand 1, and cytotoxic T lymphocyte-associated antigen 4 (CTLA4) antibodies, there is an urgent need to find new drugs with ICP inhibitory effects. In this study, a network-based computational framework called multi-network algorithm-driven drug repositioning targeting ICP (Mnet-DRI) is developed to accurately repurpose novel ICIs from ≈3000 Food and Drug Administration-approved or investigational drugs. By applying Mnet-DRI to PD-L1, maprotiline (MAP), an antidepressant drug is repurposed, as a potential PD-L1 modifier for colorectal and lung cancers. Experimental validation revealed that MAP reduced PD-L1 expression by targeting E3 ubiquitin ligase speckle-type zinc finger structural protein (SPOP), and the combination of MAP and anti-CTLA4 in vivo significantly enhanced the antitumor effect, providing a new alternative for the clinical treatment of colorectal and lung cancer.
Collapse
Affiliation(s)
- Saisai Tian
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Mengting Xu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Xiangxin Geng
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Jiansong Fang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhou510006China
| | - Hanchen Xu
- Institute of Digestive DiseasesLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| | - Xinying Xue
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijing100038China
| | - Hongmei Hu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Qing Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Dianping Yu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Mengmeng Guo
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hongwei Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Jinyuan Lu
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Chengyang Guo
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
| | - Qun Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Weidong Zhang
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghai200433China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsInstitute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100193China
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosafetyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
29
|
Oh DY, Rokutanda N, Żotkiewicz M, He P, Stocks J, Johnson ML. Delayed Separation of Kaplan-Meier Curves is Commonly Observed in Studies of Advanced/Metastatic Solid Tumors Treated with Anti-PD-(L)1 Therapy: Systematic Review and Meta-Analysis. Target Oncol 2025; 20:45-56. [PMID: 39522075 PMCID: PMC11762587 DOI: 10.1007/s11523-024-01108-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) Kaplan-Meier (KM) curves often show delayed survival benefit followed by long-term survival in a subgroup of patients. Such outcomes can violate the proportional hazards assumption, leading to a loss of statistical power. OBJECTIVE We aimed to determine common trends in delayed separation to inform future ICI clinical trials. PATIENTS AND METHODS A literature search was performed using Trialtrove® to identify phase III trials of antiprogrammed cell death (ligand)-1 (anti-PD-[L]1) agents in locally advanced/metastatic solid tumors published between January 2010 and September 2021. The frequency of delayed separation of overall survival (OS) and progression-free survival (PFS) KM curves, correlation between duration of delayed separation in OS/PFS KM curves, and correlation between duration of delayed separation in OS/PFS KM curves with corresponding hazard ratios (HRs) were assessed in all-comer and PD-L1 enriched populations. RESULTS Eighty-five studies with OS/PFS KM curves were identified. Most studies showed delayed separation of OS (> 67.9%) and PFS (> 54.5%) KM curves. The correlation between the duration of delayed separation in OS/PFS KM curves was strongest in the PD-L1 enriched population (adjusted R2 = 0.66). No correlation was seen between the duration of delayed separation of OS KM curves and OS HR. A modest correlation was seen between the duration of delayed separation of PFS KM curves and PFS HR in all-comer and PD-L1 enriched populations (adjusted R2 = 0.24 and 0.31, respectively). CONCLUSIONS Delayed separation of KM curves was common in clinical trials of anti-PD-(L)1 agents. Understanding delayed separation is key to clinical study designs and assessing outcomes with ICIs.
Collapse
Affiliation(s)
- Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, South Korea.
| | | | | | - Philip He
- Biostatistics and Data Management, Daiichi Sankyo, Basking Ridge, NJ 07920, USA
| | | | - Melissa L Johnson
- Sarah Cannon Research Institute-Tennessee Oncology, Nashville, TN 37203, USA
| |
Collapse
|
30
|
Wang J, Hu X, Wang Y, A R, Li X, Sun Y, Guan Z, Li X, Wu Y, Wang J, Zhao F, Liu Y, Wang H, Yu H, Wang T, Zhu M, Li X, Zhang D, Chen W, Han Z, Sun X. Development and characterisation of [ 18F]TTDP, a novel T cell immunoglobulin and ITIM domain tracer, in humanised mice and non-human primates. Eur J Nucl Med Mol Imaging 2025; 52:416-426. [PMID: 39297961 DOI: 10.1007/s00259-024-06911-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024]
Abstract
PURPOSE The T cell immunoglobulin and ITIM domain (TIGIT) blockade immunotherapy response is directly associated with individual differences of TIGIT expression on tumour-infiltrating lymphocytes (TILs) in tumour immune microenvironment (TIME) of non-small cell lung cancer (NSCLC). Here, we developed a TIGIT-targeted PET tracer to evaluate its feasibility in predicting immunotherapy efficacy, aiming to manage NSCLC patients accurately. METHODS We synthesised a 18F-labeled TIGIT-targeted D-peptide, [18F]TTDP, and investigated the specificity of [18F]TTDP both to murine TIGIT and human TIGIT by a series of in vitro and in vivo assays. [18F]TTDP PET imaging was performed in humanised immune system (HIS) mice models bearing NSCLC patient-derived xenografts (PDXs) to evaluate the predictive value of FDA-approved combination immunotherapy of atezolizumab plus tiragolumab. Lastly, rhesus macaque was applied for [18F] TTDP PET to explore the tracer's in vivo distribution and translational potential in non-human primates. RESULTS [18F]TTDP showed high specificity for both murine TIGIT and human TIGIT in vitro and in vivo. The HIS NSCLC PDX platform was successfully established for [18F]TTDP PET imaging, and tumour uptake of [18F]TTDP was significantly correlated with the TIGIT expression of TILs in the TIME. [18F]TTDP PET imaging, in predicting treatment response to the combination immunotherapy in NSCLC HIS-PDX models, showed a sensitivity of 83.33% and a specificity of 100%. In addition, [18F]TTDP PET also showed cross-species consistency of the tracer biodistribution between non-human primate and murine animals, and no adverse events were observed. CONCLUSION The combined implementation of the [18F]TTDP and HIS-PDX model creates a state-of-the-art preclinical platform that will impact the identification and validation of TIGIT-targeted PET image-guided diagnosis, treatment response prediction, beneficial patient screening, novel immunotherapies, and ultimately the outcome of NSCLC patients. We first provided in vivo biodistribution of [18F]TTDP PET imaging in rhesus macaque, indicating its excellent translational potential in the clinic.
Collapse
Affiliation(s)
- Jing Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Hu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yueqi Wang
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong A
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoqian Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Sun
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhengqi Guan
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaona Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongyi Wu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiannan Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fangyu Zhao
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Liu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongbin Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hong Yu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianyi Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mengyuan Zhu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinyu Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Duoyi Zhang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoguo Han
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xilin Sun
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
31
|
Ren B, Yan S, Li Z, Huang Y, Cai H, Yang J, Fan Q, Chen C, Que F, Wu G, Huang L, Zhou R, Zhu J, Yan C, Liu G, Shen Z, Ning S. A Turbo-Charging System-Like Contrast Agent for MRI-Guided STING Pathway-Activated Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410432. [PMID: 39488791 PMCID: PMC11714149 DOI: 10.1002/advs.202410432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/25/2024] [Indexed: 11/04/2024]
Abstract
To overcome the problems of Gd-based contrast agents (GBCAs) (nephrotoxicity and brain deposition) and stimulator of interferon genes (STING) agonists (poor stability, low delivery efficiency, and potential toxicity), in this study, a Turbo-charging system-like GBCA is designed and constructed for magnetic resonance imaging (MRI) guided STING pathway-activated cancer immunotherapy. Poly(acrylic acid) (PAA) is used to coordinate with Gd3+, forming a Gd/PAA macrochelate. Both Gd/PAA macrochelate and SR717 are conjugated to cystamine (CA) to obtain SR717-CA@Gd/PAA self-assembled nanoparticles (SAN), which are termed as Turbo S because of its similarity with the Turbo-charging system of cars. After accumulation in tumors and internalization in tumor cells, the disulfide linkage in Turbo S undergoes a cleavage process catalyzed by glutathione (GSH), leading to the release of Gd/PAA and SR717. The released Gd/PAA gain a high r1 value (17.11 mM-1 s-1 at 7.0 T; 57.81 mM-1 s-1 at 3.0 T), indicating its strong T1 imaging capability. Turbo S with a low dosage of SR717 (8.9 mg kg-1) achieved a higher tumor immunotherapeutic efficacy than free SR717 with a high dosage (30 mg kg-1). The excellent delivery efficiency, high tumor treatment efficacy, and superior biosafety demonstrate that the Turbo S can be used as a promising candidate for tumor immunotherapy.
Collapse
Affiliation(s)
- Bin Ren
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Sihua Yan
- Department of Breast SurgeryThe Second Affiliated Hospital of Guangxi Medical UniversityNanning530000China
| | - Zongheng Li
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Ya Huang
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Haobin Cai
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Jing Yang
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Qingdeng Fan
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Chunmei Chen
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Fanchao Que
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Guochao Wu
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Lin Huang
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Ruilong Zhou
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Jiaoyang Zhu
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Chenggong Yan
- Medical Imaging CenterNanfang HospitalSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Gang Liu
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsCenter for Molecular Imaging and Translational MedicineSchool of Public HealthXiamen UniversityXiamenFujian361102China
| | - Zheyu Shen
- School of Biomedical EngineeringSouthern Medical University1023 Shatai South RoadGuangzhouGuangdong510515China
| | - Shipeng Ning
- Department of Breast SurgeryThe Second Affiliated Hospital of Guangxi Medical UniversityNanning530000China
| |
Collapse
|
32
|
Yang G, Li H, Yin J, Yao L, Yang J, Tang J, Wu Y, Zhou M, Luo T, Zhang Y, Zhang J, Yang X, Dong X, Liu Z, Li N. Alleviating Tumor Hypoxia and Immunosuppression via Sononeoperfusion: A New Ally for potentiating anti-PD-L1 blockade of solid Tumor. ULTRASONICS SONOCHEMISTRY 2025; 112:107115. [PMID: 39482116 PMCID: PMC11635779 DOI: 10.1016/j.ultsonch.2024.107115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024]
Abstract
The hypoxic and immunosuppressive tumor microenvironment (TME) remains a major obstacle to impede cancer immunotherapy. Here, we found that sononeoperfusion-a new effect of tumor perfusion enhancement induced by low mechanical index ultrasound stimulated microbubble cavitation (USMC)-ameliorated tumor tissue oxygenation and induced tumor vascular normalization (TVN). This TVN might be associated with the down-regulation of hypoxia-inducible factor 1-alpha (HIF-1α) and vascular endothelial growth factor (VEGF) within tumors. Moreover, the sononeoperfusion effect reduced the accumulation of immunosuppressive cells, such as regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs) and M2-like tumor-associated macrophages (M2-TAMs), and decreased the production of immune inhibitory factors like transforming growth factor-β1 (TGF-β1), interleukin 10 (IL-10), chemoattractant chemokines CC-chemokine ligand 22 (CCL22), CCL28, adenosine and lactate within tumors. Notably, flow cytometry analysis revealed that sononeoperfusion not only increased the percentage of tumor infiltrating-CD8+ T cells, but also promoted the generation of interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) by these cells. Furthermore, the improved immune TME by sononeoperfusion effect sensitized anti-PD-L1 treatment both in MC38 colon cancer and Lewis lung carcinoma mice, resulting in tumor regression and prolonged survival. Mechanically, the enhanced efficacy of combination therapy was mainly based on promoting the infiltration and function of CD8+ T cells within tumors. Together, sononeoperfusion could ameliorate hypoxia and immunosuppression in the TME, thereby potentiating anti-PD-L1 therapy for solid tumors. This novel method of USMC generating sononeoperfusion effect may provide a new therapeutic modality for facilitating cancer immunotherapy.
Collapse
Affiliation(s)
- Guoliang Yang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Hui Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Jiabei Yin
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Lei Yao
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Jun Yang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Jiawei Tang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - You Wu
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Meng Zhou
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - TingTing Luo
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Yi Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Jing Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Xuezhi Yang
- Institute of Cancer, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - XiaoXiao Dong
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China
| | - Zheng Liu
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China.
| | - Ningshan Li
- Department of Ultrasound, The Second Affiliated Hospital of Army Medical University, Chongqing 400037, China.
| |
Collapse
|
33
|
Wei J, Li W, Zhang P, Guo F, Liu M. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer 2024; 23:279. [PMID: 39725966 DOI: 10.1186/s12943-024-02179-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed the treatment landscape for various malignancies, achieving notable clinical outcomes across a wide range of indications. Despite these advances, resistance to immune checkpoint blockade (ICB) remains a critical clinical challenge, characterized by variable response rates and non-durable benefits. However, growing research into the complex intrinsic and extrinsic characteristics of tumors has advanced our understanding of the mechanisms behind ICI resistance, potentially improving treatment outcomes. Additionally, robust predictive biomarkers are crucial for optimizing patient selection and maximizing the efficacy of ICBs. Recent studies have emphasized that multiple rational combination strategies can overcome immune checkpoint resistance and enhance susceptibility to ICIs. These findings not only deepen our understanding of tumor biology but also reveal the unique mechanisms of action of sensitizing agents, extending clinical benefits in cancer immunotherapy. In this review, we will explore the underlying biology of ICIs, discuss the significance of the tumor immune microenvironment (TIME) and clinical predictive biomarkers, analyze the current mechanisms of resistance, and outline alternative combination strategies to enhance the effectiveness of ICIs, including personalized strategies for sensitizing tumors to ICIs.
Collapse
Grants
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Wenke Li
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Pengfei Zhang
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Ming Liu
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
34
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
35
|
Wang K, Zhao J, Duan J, Feng C, Li Y, Li L, Yuan S. Radiomic and dosimetric parameter-based nomogram predicts radiation esophagitis in patients with non-small cell lung cancer undergoing combined immunotherapy and radiotherapy. Front Oncol 2024; 14:1490348. [PMID: 39744008 PMCID: PMC11688372 DOI: 10.3389/fonc.2024.1490348] [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: 09/03/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Background The combination of immune checkpoint inhibitors (ICIs) and radiotherapy (RT) may increase the risk of radiation esophagitis (RE). This study aimed to establish and validate a new nomogram to predict RE in patients with non-small cell lung cancer (NSCLC) undergoing immunochemotherapy followed by RT (ICI-RT). Methods The 102 eligible patients with NSCLC treated with ICI-RT were divided into training (n = 71) and validation (n = 31) cohorts. Clinicopathologic features, dosimetric parameters, inflammatory markers, and radiomic score (Rad-score) were included in the univariate logistic regression analysis, and factors with p < 0.05 in the univariate analysis were included in the multivariate logistic regression analysis. Factors with significant predictive values were obtained and used for developing the nomogram. The area under the receiver operating characteristic curve (AUC), calibration curve, and decision curve were used to validate the model. Results A total of 38 (37.3%) patients developed RE. Univariate and multivariate analyses identified the following independent predictors of RE: a maximum dose delivered to the esophagus >58.4 Gy, a mean esophagus dose >13.3 Gy, and the Rad-score. The AUCs of the nomogram in the training and validation cohorts were 0.918 (95% confidence interval [CI]: 0.824-1.000) and 0.833 (95% CI: 0.697-0.969), respectively, indicating good discrimination. The calibration curves showed good agreement between the predicted occurrence of RE and the actual observations. The decision curve showed a satisfactory positive net benefit at most threshold probabilities, suggesting a good clinical effect. Conclusions We developed and validated a nomogram based on imaging histological features and RT dosimetric parameters. This model can effectively predict the occurrence of RE in patients with NSCLC treated using ICI-RT.
Collapse
Affiliation(s)
- Kang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junfeng Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinghao Duan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Changxing Feng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ying Li
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Li Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China
| |
Collapse
|
36
|
Shahzadi M, Rafique H, Waheed A, Naz H, Waheed A, Zokirova FR, Khan H. Artificial intelligence for chimeric antigen receptor-based therapies: a comprehensive review of current applications and future perspectives. Ther Adv Vaccines Immunother 2024; 12:25151355241305856. [PMID: 39691280 PMCID: PMC11650588 DOI: 10.1177/25151355241305856] [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: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Using artificial intelligence (AI) to enhance chimeric antigen receptor (CAR)-based therapies' design, production, and delivery is a novel and promising approach. This review provides an overview of the current applications and challenges of AI for CAR-based therapies and suggests some directions for future research and development. This paper examines some of the recent advances of AI for CAR-based therapies, for example, using deep learning (DL) to design CARs that target multiple antigens and avoid antigen escape; using natural language processing to extract relevant information from clinical reports and literature; using computer vision to analyze the morphology and phenotype of CAR cells; using reinforcement learning to optimize the dose and schedule of CAR infusion; and using AI to predict the efficacy and toxicity of CAR-based therapies. These applications demonstrate the potential of AI to improve the quality and efficiency of CAR-based therapies and to provide personalized and precise treatments for cancer patients. However, there are also some challenges and limitations of using AI for CAR-based therapies, for example, the lack of high-quality and standardized data; the need for validation and verification of AI models; the risk of bias and error in AI outputs; the ethical, legal, and social issues of using AI for health care; and the possible impact of AI on the human role and responsibility in cancer immunotherapy. It is important to establish a multidisciplinary collaboration among researchers, clinicians, regulators, and patients to address these challenges and to ensure the safe and responsible use of AI for CAR-based therapies.
Collapse
Affiliation(s)
- Muqadas Shahzadi
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Ahmad Waheed
- Department of Zoology, Faculty of Life Sciences, University of Okara, 2 KM Lahore Road, Renala Khurd, Okara 56130, Punjab, Pakistan
| | - Hina Naz
- Department of Zoology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | - Atifa Waheed
- Department of Biology, Faculty of Life Sciences, University of Okara, Okara, Pakistan
| | | | - Humera Khan
- Department of Biochemistry, Sahiwal Medical College, Sahiwal, Pakistan
| |
Collapse
|
37
|
Li B, Xu L, Chen C, Ye J. Mapping the Binding Hotspots and Transient Binding Pockets on V-Domain Immunoglobulin Suppressor of T Cell Activation Protein Surface. ACS OMEGA 2024; 9:48657-48669. [PMID: 39676951 PMCID: PMC11635502 DOI: 10.1021/acsomega.4c07757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/02/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024]
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA), an inhibitory immune checkpoint present on both immune and tumor cells, has emerged as a highly promising target for cancer therapy due to its potential to overcome resistance encountered with existing immune checkpoint treatments. VSIG-3 is determined as an inhibitory ligand for VISTA, leading to the suppression of T cell proliferation. However, hotspots between VISTA/VSIG-3 protein-protein interaction remain ambiguous, mainly attributed to the lack of the structure of the VISTA/VSIG-3 complex. Therefore, in this study, in order to determine the energetic contributions of the interfacial residues on VISTA, we first constructed VISTA/VSIG-3 complex models by the protein docking method, followed by molecular dynamics simulations, binding free-energy decomposition, and alanine scanning. Results suggested that the putative hotspots in VISTA comprise residues His32, Tyr37, Thr35, Glu47, Val48, Gln49, Glu53, Arg54, Gln73, His122, and His126. Moreover, the distribution of the hotspots was clustered into two regions (hot regions I and II), and by using the TRAPP tool, transient subpockets within the hot regions were identified. Furthermore, conformational states of the binding pockets exhibiting druggability scores higher than those observed in the crystal structure were found. Overall, we hope that the findings outlined in this study can be used to facilitate the development of inhibitors targeting the VISTA/VSIG-3 immune checkpoint pathway in the future.
Collapse
Affiliation(s)
- Bingjie Li
- School of Pharmacy, Inflammation and
Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Lixiu Xu
- School of Pharmacy, Inflammation and
Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Chu Chen
- School of Pharmacy, Inflammation and
Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| | - Jiqing Ye
- School of Pharmacy, Inflammation and
Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
38
|
Liu B, Liang BB, Cao WD, Su XX, Cao Q, Mao ZW. Platinum-Metformin Conjugates Acting as Promising PD-L1 Inhibitors through the AMP-Activated Protein Kinase Mediated Lysosomal Degradation Pathway. Angew Chem Int Ed Engl 2024; 63:e202410586. [PMID: 39206686 DOI: 10.1002/anie.202410586] [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: 06/05/2024] [Revised: 07/31/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
With the development of metalloimmunology, the potential of platinum drugs in cancer immunotherapy has attracted extensive attention. Although immunochemotherapy combining PD-1/PD-L1 antibodies with platinum drugs has achieved great success in the clinic, combination therapy commonly brings new problems. Herein, we have developed a platinum-metformin conjugate as a promising alternative to antibody-based PD-L1 inhibitors, not only disrupting PD-1/PD-L1 axis on cell surface but also down-regulating the total PD-L1 levels in non-small cell lung cancer (NSCLC) cells comprehensively, thus achieving highly efficient immunochemotherapy by a single small molecule. Mechanism studies demonstrate that Pt-metformin conjugate can selectively accumulate in lysosomes, promote lysosomal-dependent PD-L1 degradation via the AMPK-TFEB pathway, and modulate the upstream regulatory proteins related to PD-L1 expression (e.g. HIF-1α and NF-κB), eventually decreasing the total abundance of PD-L1 in NSCLC, overcoming tumor hypoxia, and activating anti-tumor immunity in vivo. This work suggests an AMPK-mediated lysosomal degradation pathway of PD-L1 for the first time and provides a unique design perspective for the development of novel platinum drugs for immunochemotherapy.
Collapse
Affiliation(s)
- Bin Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Bing-Bing Liang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Wan-Di Cao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Xu-Xian Su
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Qian Cao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry Guangdong Basic Research Center of Excellence for Functional Molecular Engineering GBRCE for Functional Molecular Engineering School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, People's Republic of China
| |
Collapse
|
39
|
Luo Y, He X, Du Q, Xu L, Xu J, Wang J, Zhang W, Zhong Y, Guo D, Liu Y, Chen X. Metal-based smart nanosystems in cancer immunotherapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230134. [PMID: 39713201 PMCID: PMC11655314 DOI: 10.1002/exp.20230134] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 01/12/2024] [Indexed: 12/24/2024]
Abstract
Metals are an emerging topic in cancer immunotherapy that have shown great potential in modulating cancer immunity cycle and promoting antitumor immunity by activating the intrinsic immunostimulatory mechanisms which have been identified in recent years. The main challenge of metal-assisted immunotherapy lies in the fact that the free metals as ion forms are easily cleared during circulation, and even cause systemic metal toxicity due to the off-target effects. With the rapid development of nanomedicine, metal-based smart nanosystems (MSNs) with unique controllable structure become one of the most promising delivery carriers to solve the issue, owing to their various endogenous/external stimuli-responsiveness to release free metal ions for metalloimmunotherapy. In this review, the state-of-the-art research progress in metal-related immunotherapy is comprehensively summarized. First, the mainstream mechanisms of MSNs-assisted immunotherapy will be delineated. The immunological effects of certain metals and categorization of MSNs with different characters and compositions are then provided, followed by the representative exemplar applications of MSNs in cancer treatment, and synergistic combination immunotherapy. Finally, we conclude this review with a summary of the remaining challenges associated with MSNs and provide the authors' perspective on their further advances.
Collapse
Affiliation(s)
- Ying Luo
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xiaojing He
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
- Clinical Imaging Research CentreCentre for Translational MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Qianying Du
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lian Xu
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Jie Xu
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Junrui Wang
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Wenli Zhang
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yixin Zhong
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Dajing Guo
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yun Liu
- Department of RadiologySecond Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Clinical Imaging Research CentreCentre for Translational MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research ProgramNUS Center for NanomedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of SurgeryChemical and Biomolecular Engineeringand Biomedical EngineeringYong Loo Lin School of Medicine and College of Design and EngineeringNational University of SingaporeSingaporeSingapore
- Institute of Molecular and Cell BiologyAgency for Science, Technology, and Research (A*STAR)SingaporeSingapore
| |
Collapse
|
40
|
Vaishampayan UN, Muzaffar J, Winer I, Rosen SD, Hoimes CJ, Chauhan A, Spreafico A, Lewis KD, Bruno DS, Dumas O, McDermott DF, Strauss JF, Chu QS, Gilbert L, Chaudhry A, Calvo E, Dalal R, Boni V, Ernstoff MS, Velcheti V. Nemvaleukin alfa, a modified interleukin-2 cytokine, as monotherapy and with pembrolizumab in patients with advanced solid tumors (ARTISTRY-1). J Immunother Cancer 2024; 12:e010143. [PMID: 39567211 PMCID: PMC11580269 DOI: 10.1136/jitc-2024-010143] [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: 07/19/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Nemvaleukin alfa (nemvaleukin, ALKS 4230) is a novel, engineered cytokine that selectively binds to the intermediate-affinity interleukin-2 receptor, preferentially activating CD8+ T cells and natural killer cells, with minimal expansion of regulatory T cells, thereby mitigating the risk of toxicities associated with high-affinity interleukin-2 receptor activation. Clinical outcomes with nemvaleukin are unknown. ARTISTRY-1 investigated the safety, recommended phase 2 dose (RP2D), and antitumor activity of nemvaleukin in patients with advanced solid tumors. METHODS This was a three-part, open-label, phase 1/2 study: part A, dose-escalation monotherapy, part B, dose-expansion monotherapy, and part C, combination therapy with pembrolizumab. The study was conducted at 32 sites in 7 countries. Adult patients with advanced solid tumors were enrolled and received intravenous nemvaleukin once daily on days 1-5 (21-day cycle) at 0.1-10 µg/kg/day (part A), or at the RP2D (part B), or with pembrolizumab (part C). Primary endpoints were RP2D selection and dose-limiting toxicities (part A), and overall response rate (ORR) and safety (parts B and C). RESULTS From July 2016 to March 2023, 243 patients were enrolled and treated (46, 74, and 166 in parts A, B, and C, respectively). The maximum tolerated dose was not reached. RP2D was determined as 6 µg/kg/day. ORR with nemvaleukin monotherapy was 10% (7/68; 95% CI 4 to 20), with seven partial responses (melanoma, n=4; renal cell carcinoma, n=3). Robust CD8+ T and natural killer cell expansion, and minimal regulatory T cell expansion were observed following nemvaleukin treatment. ORR with nemvaleukin plus pembrolizumab was 13% (19/144; 95% CI 8 to 20), with 5 complete and 14 partial responses; 6 responses were in PD-(L)1 inhibitor-approved and five in PD-(L)1 inhibitor-unapproved tumor types. Three responses were in patients with platinum-resistant ovarian cancer. The most common grade 3-4 treatment-related adverse events (TRAEs) in parts B and C, respectively, were neutropenia (49%, 21%) and anemia (10%, 11%); 4% of patients in each part discontinued due to TRAEs. CONCLUSIONS Nemvaleukin was well tolerated and demonstrated promising antitumor activity across heavily pretreated advanced solid tumors. Phase 2/3 studies of nemvaleukin are ongoing. TRIAL REGISTRATION NUMBER NCT02799095.
Collapse
Affiliation(s)
- Ulka N Vaishampayan
- Divison of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jameel Muzaffar
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ira Winer
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Seth D Rosen
- Hematology Oncology Association of the Treasure Coast, Port St. Lucie, Florida, USA
| | - Christoper J Hoimes
- Phase I Program, Case Comprehensive Cancer Center, University Hospitals, Cleveland, Ohio, USA
- Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Aman Chauhan
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, USA
| | - Anna Spreafico
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Karl D Lewis
- University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Debora S Bruno
- Department of Medicine, University Hospitals Cleveland Medical Center, Seidman Cancer Center, Cleveland, Ohio, USA
- Case Western School of Medicine, Cleveland, Ohio, USA
| | - Olivier Dumas
- CHU de Québec-Université Laval, Quebec City, Quebec, Canada
| | | | | | - Quincy S Chu
- University of Alberta/Alberta Health Services, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Lucy Gilbert
- Division of Gynecologic Oncology, The Gerald Bronfman Department of Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | | | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Rita Dalal
- Mural Oncology, Inc, Waltham, Massachusetts, USA
| | - Valentina Boni
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Marc S Ernstoff
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, New York University, New York, New York, USA
| |
Collapse
|
41
|
Liu Y, Yang F, Li Z, Wang T, Mu Y, Fan Y, Xue H, Hu X, Guan X, Feng H. Concurrent immune checkpoint blockade for enhanced cancer immunotherapy utilizing engineered hybrid nanovesicles. Front Pharmacol 2024; 15:1487940. [PMID: 39588148 PMCID: PMC11586202 DOI: 10.3389/fphar.2024.1487940] [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: 08/29/2024] [Accepted: 10/30/2024] [Indexed: 11/27/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, demonstrating unprecedented efficacy against advanced cancers. However, their clinical applications are significantly hampered by low overall response rates. Dual blockade of two immune checkpoints represents a promising strategy to enhance immunotherapeutic efficacy. In this study, we developed hybrid cell membrane nanovesicles adorned with PD-1 and SIRPα receptors for combination immunotherapy in melanoma. Our hybrid nanovesicles (PD-1/SIRPα NVs) demonstrated high specificity to PD-L1 and CD47 ligands, facilitating the phagocytosis of melanoma cells by macrophages. In a melanoma mouse model, PD-1/SIRPα NVs significantly suppressed 77% of tumor growth and elicited a robust antitumor immune response for immunotherapy. In conclusion, our findings highlight the promising potential of PD-1/SIRPα NVs as novel and effective ICIs for cancer immunotherapy.
Collapse
Affiliation(s)
- Yuxuan Liu
- Department of Dermatology, The Affiliated Wenling Hospital of Taizhou University, Taizhou, China
- College of Medical Technology, Beihua University, Jilin, China
| | - Fuxu Yang
- College of Medical Technology, Beihua University, Jilin, China
| | - Zhimin Li
- College of Medical Technology, Beihua University, Jilin, China
| | - Ting Wang
- Medical School, Taizhou University, Taizhou, China
| | - Yeteng Mu
- College of Medical Technology, Beihua University, Jilin, China
| | - Yuxin Fan
- College of Medical Technology, Beihua University, Jilin, China
| | - Han Xue
- College of Medical Technology, Beihua University, Jilin, China
| | - Xiuli Hu
- Institute of Polymer Science and Engineering, School of Chemical Engineering, Hebei University of Technology, Tianjin, China
| | - Xingang Guan
- Medical School, Taizhou University, Taizhou, China
| | - Hongxia Feng
- Department of Dermatology, The Affiliated Wenling Hospital of Taizhou University, Taizhou, China
| |
Collapse
|
42
|
Zhang A, Fan T, Liu Y, Yu G, Li C, Jiang Z. Regulatory T cells in immune checkpoint blockade antitumor therapy. Mol Cancer 2024; 23:251. [PMID: 39516941 PMCID: PMC11545879 DOI: 10.1186/s12943-024-02156-y] [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: 07/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Regulatory T cells (Tregs), an essential component of the human immune system, are a heterogeneous group of T lymphocytes with the ability to suppress immune responses and maintain immune homeostasis. Recent evidence indicates that Tregs may impair antitumor immunity and facilitate cancer progression by weakening functions of effector T cells (Teffs). Consequently, targeting Tregs to eliminate them from tumor microenvironments to improve Teffs' activity could emerge as an effective strategy for cancer immunotherapy. This review outlines the biology of Tregs, detailing their origins, classification, and crucial markers. Our focus lies on the complex role of Tregs in cancer's development, progression and treatment, particularly on their suppressive role upon antitumor responses via multiple mechanisms. We delve into Tregs' involvement in immune checkpoint blockade (ICB) therapy, their dual effect on cancer immunotherapy and their potential biomarkers for ICB therapy effectiveness. We also summarize advances in the therapies that adjust Tregs to optimize ICB therapy, which may be crucial for devising innovative cancer treatment strategies.
Collapse
Affiliation(s)
- An Zhang
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guanhua Yu
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
43
|
Chen X, Zhang J, Li B, Yan F. Determining doses for backfill cohorts based on patient-reported outcome. BMC Med Res Methodol 2024; 24:270. [PMID: 39516724 PMCID: PMC11546322 DOI: 10.1186/s12874-024-02398-w] [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: 06/11/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Incorporating backfill cohorts in phase I oncology trials is a recently developed strategy for dose optimization. However, the efficacy assessment window is long in general, causing a lag in identifying ineffective doses and more patients being backfilled to those doses. There is necessity to investigate how to use patient-reported outcomes (PRO) to determine doses for backfill cohorts. METHODS We propose a unified Bayesian design framework, called 'Backfill-QoL', to utilize patient-reported quality of life (QoL) data into phase I oncology trials with backfill cohorts, including methods for trial monitoring, algorithm for dose-finding, and criteria for dose selection. Simulation studies and sensitivity analyses are conducted to evaluate the proposed Backfill-QoL design. RESULTS The simulation studies demonstrate that the Backfill-QoL design is more efficient than traditional dose-expansion strategy, and fewer patients would be allocated to doses with unacceptable QoL profiles. A user-friendly Windows desktop application is developed and freely available for implementing the proposed design. CONCLUSIONS The Backfill-QoL design enables continuous monitoring of safety, efficacy and QoL outcomes, and the recommended phase II dose (RP2D) can be identified in a more patient-centered perspective.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Jingyi Zhang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Bosheng Li
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Department of Biostatistics, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
44
|
Xie F, Luo S, Liu D, Lu X, Wang M, Liu X, Jia F, Pang Y, Shen Y, Zeng C, Ma X, Tang D, Tu L, Yang L, Cheng Y, Luo Y, Xie F, Hou H, Huang T, Ni B, Zhuang C, Zhao W, Li K, Zheng X, Bi W, Jia X, He Y, Wang S, Cao H, Wu K, Wang Y. Genomic and transcriptomic landscape of human gastrointestinal stromal tumors. Nat Commun 2024; 15:9495. [PMID: 39489749 PMCID: PMC11532483 DOI: 10.1038/s41467-024-53821-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
Gastrointestinal stromal tumor (GISTs) are clinically heterogenous exhibiting varying degrees of disease aggressiveness in individual patients. We comprehensively describe the genomic and transcriptomic landscape of a cohort of 117 GISTs including 31 low-risk, 18 intermediate-risk, 29 high-risk, 34 metastatic and 5 neoadjuvant GISTs from 105 patients. GISTs have notably low tumor mutation burden but widespread copy number variations. Aggressive GISTs harbor remarkably more genomic aberrations than low-/intermediate-risk GISTs. Complex genomic alterations, chromothripsis and kataegis, occur selectively in aggressive GISTs. Despite the paucity of mutations, recurrent inactivating YLPM1 mutations are identified (10.3%, 7 of 68 patients), enriched in high-risk/metastatic GIST and functional study further demonstrates YLPM1 inactivation promotes GIST proliferation, growth and oxidative phosphorylation. Spatially and temporally separated GISTs from individual patients demonstrate complex tumor heterogeneity in metastatic GISTs. Finally, four prominent subtypes are proposed with different genomic features, expression profiles, immune characteristics, clinical characteristics and subtype-specific treatment strategies. This large-scale analysis depicts the landscape and provides further insights into GIST pathogenesis and precise treatment.
Collapse
Affiliation(s)
- Feifei Xie
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Shuzhen Luo
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Dongbing Liu
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Xiaojing Lu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Embryo Original Disease, 200030, Shanghai, China
| | - Ming Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Xiaoxiao Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fujian Jia
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Yuzhi Pang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yanying Shen
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chunling Zeng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xinli Ma
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Daoqiang Tang
- Department of Pathology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Lin Tu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Linxi Yang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Yumei Cheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yuxiang Luo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fanfan Xie
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
| | - Hao Hou
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Tao Huang
- Bioinformatics Core, Shanghai Institute of Nutrition and Health, 200031, Shanghai, China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Chun Zhuang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Wenyi Zhao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Ke Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xufen Zheng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Wenbo Bi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Xiaona Jia
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yi He
- Department of Urology, No.1 Hospital of Jiaxing, 314000, Jiaxing, China
| | - Simin Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| | - Hui Cao
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| | - Kui Wu
- Institute of Intelligent Medical Research (IIMR), BGI Genomics, 518083, Shenzhen, China.
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, 518083, Shenzhen, China.
| | - Yuexiang Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
45
|
Feng D, Pu D, Ren J, Liu M, Zhang Z, Liu Z, Li J. CD8 + T-cell exhaustion: Impediment to triple-negative breast cancer (TNBC) immunotherapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189193. [PMID: 39413858 DOI: 10.1016/j.bbcan.2024.189193] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/16/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
CD8+ T-cell exhaustion has been identified as a significant contributor to immunosuppression and immune escape in triple-negative breast cancer (TNBC). Dysfunction due to cell exhaustion is characterized by reduced effector capacity and sustained expression of inhibitory receptors (IRs). The factors contributing to CD8+ T-cell exhaustion are multifaceted, encompassing external influences such as the upregulation of IRs, reduction of effector cytokines, and internal changes within the immune cell, including transcriptomic alterations, epigenetic landscape remodeling, and metabolomic shifts. The impact of the altered TNBC tumor microenvironment (TME) on Tex is also a critical consideration. The production of exhausted CD8+ T-cells (CD8+ Tex) is positively correlated with poor prognosis and reduced response rates to immunotherapy in TNBC patients, underscoring the urgent need for the development of novel TNBC immunotherapeutic strategies that target the mechanisms of CD8+ T-cell exhaustion. This review delineates the dynamic trajectory of CD8+ T-cell exhaustion development in TNBC, provides an update on the latest research advancements in understanding its pathogenesis, and offers insights into potential immunotherapeutic strategies.
Collapse
Affiliation(s)
- Dandan Feng
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dongqing Pu
- Department of Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China
| | - Jinlu Ren
- Shandong Xiandai University, Jinan 250104, China
| | - Ming Liu
- Department of Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhiyong Liu
- Central Laboratory, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China; Shandong Key Laboratory of Dominant Diseases of Traditional Chinese Medicine, Jinan 250014, China.
| | - Jingwei Li
- Department of Breast and Thyroid Surgery, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan 250014, China.
| |
Collapse
|
46
|
Wang Y, Qian M, Xie Y, Zhang X, Qin Y, Huang R. Biodegradable nanoparticles-mediated targeted drug delivery achieves trans-spatial immunotherapy. FUNDAMENTAL RESEARCH 2024; 4:1639-1649. [PMID: 39734540 PMCID: PMC11670710 DOI: 10.1016/j.fmre.2022.11.003] [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: 06/09/2022] [Revised: 10/24/2022] [Accepted: 11/10/2022] [Indexed: 11/18/2022] Open
Abstract
Immunotherapy has been seriously retarded due to inadequate antigen presentation and a tumor cell-dominated immunosuppressive microenvironment (TME). Herein, biodegradable multifunctional mesoporous silica nanoparticles, with dispersed carbon nanodots incorporated into the frameworks, active TKD peptide modification on the surfaces and hydrophobic drug loading in the pores, were prepared for targeted chemotherapy synergized with trans-spatial immunotherapy. The nanoparticles were biodegradable due to nanodot-induced framework swelling, which would (1) kill the in situ tumor cells and promote antigen release by targeted chemotherapy and (2) trigger biodegraded debris involving TKD and CDs to largely adsorb the tumor antigens via π-π conjugation synergized hydrophobic interactions and then massively transport these antigens from the tumor cell-dominated TME to the immune cell-dominated spleen via TKD-mediated small size effects. Thereafter, these antigens can be processed into antigen peptides via TKD-mediated lysosome endocytosis and then activate T cells in the spleen via MHC complex construction and dendritic cell cytomembrane presentation. Therefore, improved immunotherapy with trans-spatial antigen presentation avoided TME immunosuppression, which when synergized with targeted chemotherapy, markedly enhanced the therapeutic outcomes of triple-negative breast cancer.
Collapse
Affiliation(s)
- Yi Wang
- Center for Advanced Low-dimension Materials, State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201600, China
| | - Min Qian
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Yibo Xie
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Xiaoyi Zhang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Yanhui Qin
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Rongqin Huang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| |
Collapse
|
47
|
Duan X, Hu J, Zhang Y, Zhao X, Yang M, Sun T, Liu S, Chen X, Feng J, Li W, Yang Z, Zhang Y, Lin X, Liu D, Meng Y, Yang G, Lin Q, Zhang G, Lei H, Yi Z, Liu Y, Liang X, Wu Y, Diao W, Li Z, Liang H, Zhan M, Sun HW, Li XY, Lu L. RIG-I is an intracellular checkpoint that limits CD8 + T-cell antitumour immunity. EMBO Mol Med 2024; 16:3005-3025. [PMID: 39322862 PMCID: PMC11555380 DOI: 10.1038/s44321-024-00136-9] [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: 11/22/2023] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
Retinoic acid-inducible gene I (RIG-I) is a pattern recognition receptor involved in innate immunity, but its role in adaptive immunity, specifically in the context of CD8+ T-cell antitumour immunity, remains unclear. Here, we demonstrate that RIG-I is upregulated in tumour-infiltrating CD8+ T cells, where it functions as an intracellular checkpoint to negatively regulate CD8+ T-cell function and limit antitumour immunity. Mechanistically, the upregulation of RIG-I in CD8+ T cells is induced by activated T cells, and directly inhibits the AKT/glycolysis signalling pathway. In addition, knocking out RIG-I enhances the efficacy of adoptively transferred T cells against solid tumours, and inhibiting RIG-I enhances the response to PD-1 blockade. Overall, our study identifies RIG-I as an intracellular checkpoint and a potential target for alleviating inhibitory constraints on T cells in cancer immunotherapy, either alone or in combination with an immune checkpoint inhibitor.
Collapse
Affiliation(s)
- Xiaobing Duan
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China.
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China.
| | - Jiali Hu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Yuncong Zhang
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Xiaoguang Zhao
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Mingqi Yang
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Taoping Sun
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Siya Liu
- The Third People's Hospital of Zhuhai, Zhuhai, 519000, China
| | - Xin Chen
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Juan Feng
- Gene Editing Technology Center of Guangdong Province, School of Medicine, Foshan University, Foshan, 528225, China
| | - Wenting Li
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Ze Yang
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Yitian Zhang
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Xiaowen Lin
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Dingjie Liu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Ya Meng
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Guang Yang
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Qiuping Lin
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Guihai Zhang
- Department of Oncology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Haihong Lei
- Department of Radiation Oncology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Zhengsheng Yi
- Department of Radiation Oncology, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Yanyan Liu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
| | - Xiaobing Liang
- Guangdong Huixin Life Science Co., Ltd., Zhuhai, 519000, China
| | - Yujuan Wu
- Zhuhai Central Blood Station, Zhuhai, 519000, China
| | - Wenqing Diao
- Zhuhai Central Blood Station, Zhuhai, 519000, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumours, Shenzhen Key Laboratory of Genitourinary Tumour, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Haihai Liang
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China
- Guangzhou First Pepople's Hospital, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Hong-Wei Sun
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China.
| | - Xian-Yang Li
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China.
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, China.
- Guangzhou First Pepople's Hospital, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
48
|
Abdel-Rahman SA, Gabr MT. Small molecules from antibody pharmacophores (SMAbPs) as a hit identification workflow for immune checkpoints. SCIENCE ADVANCES 2024; 10:eadq5540. [PMID: 39413175 PMCID: PMC11482313 DOI: 10.1126/sciadv.adq5540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
Small-molecule modulators of immune checkpoints are poised to revolutionize cancer immunotherapy. However, efficient strategies for hit identification are lacking. We introduce small molecules from antibody pharmacophores (SMAbPs), a workflow leveraging cocrystal structures of checkpoints with antibodies to create pharmacophore maps for virtual screening. Applying SMAbPs to five immune checkpoints yielded hits with submicromolar potency in both cell-free and cellular assays. Notably, SMAbPs identified the most potent T cell immunoglobulin and mucin-domain containing-3 and V-domain immunoglobulin suppressor of T cell activation (VISTA) inhibitors reported to date and first-in-class modulators of B and T lymphocyte attenuator, 4-IBB, and CD27. Targeting inhibitory and costimulatory checkpoints with hits identified through SMAbPs demonstrated remarkable in vivo antitumor activity, exemplified by MG-V-53 (VISTA inhibitor) and MG-C-30 (CD27 agonist), which significantly reduced tumor volumes in MC38 and EG7-OVA mouse models, respectively.
Collapse
Affiliation(s)
- Somaya A. Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Moustafa T. Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY 10065, USA
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
49
|
Wang L, Yin M, Zhang Z, Liu S, Liu Y, Geng X, Zheng G. Methylation and transcriptome analyses construct a prognostic model and reveal the suppressor role of VMO1 in lung adenocarcinoma. Cell Signal 2024; 122:111313. [PMID: 39053673 DOI: 10.1016/j.cellsig.2024.111313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND DNA methylation is an important epigenetic mechanism of gene regulation. The aberrant DNA methylation has been found to play an important role in the initiation and progression of tumors. RESULTS Transcriptome and DNA methylation data of lung adenocarcinoma (LUAD) patients were co-analyzed and 95 methylation-driven genes (MDGs) was found in relation to LUAD. A prognostic model based on 3 MDGs (GMNN, SPINK2 and VMO1) was constructed by Univariate and Multivariate cox regression analyses. The risk score generated from the prognostic model could be used to classify LUAD patients into high and low risk groups. Furthermore, it was found that the risk score was associated with tumor microenvironment (TME) and clinical characteristics (survival status and T stage) of patients. Interestingly, we identified and validated that the patients in the low-risk group responded better to immunotherapy treatment. Then, a nomogram model based on the risk score and clinical characteristics was established which showed significant prediction value. The down-regulation and hypermethylation levels of vitelline membrane outer layer protein 1 homolog (VMO1) were verified in paired LUAD tumor and non-tumor tissues by pyrosequencing assay and RT-qPCR. Furthermore, MTT, migration and wound healing assays were performed with lentivector-mediated ectopic over-expression and 5-Aza-dC demethylation followed by siRNA rescue experiments to investigate the role of VMO1 in LUAD cells. Our results indicated that VMO1 could inhibit proliferation and migration of A549 and NCI-H1299 cells. CONCLUSIONS In summary, our experiments constructed a prognostic model with high capacity for risk prediction in LUAD patients. VMO1 had a malignant suppressor role in LUAD cells. The correlation between risk score and TME might elucidate a potential mechanism of oncogenesis and provide an avenue for further therapeutic targets.
Collapse
Affiliation(s)
- Lishui Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China
| | - Maopeng Yin
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China; Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, PR China
| | - Zeyu Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China; Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, PR China
| | - Shichao Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China; Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, PR China
| | - Yingjie Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China; Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, PR China
| | - Xueyan Geng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China; Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong Province, PR China
| | - Guixi Zheng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, Shandong Province, PR China.
| |
Collapse
|
50
|
Hu X, Enbar T, Tang L. Delivery approaches of immunomodulatory nucleic acids for cancer therapy. Curr Opin Biotechnol 2024; 89:103182. [PMID: 39178725 DOI: 10.1016/j.copbio.2024.103182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
Messenger RNA (mRNA) vaccines have made remarkable public health contributions during the pandemic and initiated a new era for nucleic acid-based therapeutics. With the unique strength of nucleic acids, including not only mRNA but also DNA, microRNA, small interfering RNA (siRNA), and other nucleic acids, either in tuning off genes or introducing function, nucleic acid therapeutics have been regarded as potential candidates for the treatment of many different diseases, especially for the immunomodulation in cancer. However, the scope of the applications was limited by the challenges in delivery due to intrinsic properties of nucleic acids including low stability, immunogenicity, and toxicity. Bioengineering approaches toward efficient and targeted delivery of therapeutic nucleic acids have gained momentum in clinical applications in the past few decades. Recent advances in the biotechnological approaches for the delivery of mRNA, siRNA, and clustered regularly interspaced short palindromic repeats (CRISPR)/Cas for immunomodulatory are promising alternatives in designing future cancer immunotherapy.
Collapse
Affiliation(s)
- Xiaomeng Hu
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Tom Enbar
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland; Institute of Materials Science & Engineering, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|