1
|
Chick RC, Beane JD, Contreras CM. Adoptive T-Cell Therapy in Melanoma: How This Will Impact Surgical Practice and the Role of Surgeons. Surg Oncol Clin N Am 2025; 34:423-436. [PMID: 40413008 DOI: 10.1016/j.soc.2025.01.002] [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: 05/27/2025]
Abstract
Melanoma is one of a small number of cancers where there is a clear role for surgery in selected patients with metastatic disease. However, the role of surgery for metastatic melanoma in the age of immune checkpoint blockade is not clearly delineated. Adoptive cell therapies, which include tumor-infiltrating lymphocytes and chimeric antigen receptor T cells, often require metastasectomy to obtain the tumor-specific immune cells and/or antigens necessary to create personalized cell-based products. It is, therefore, essential that the surgeon be well-versed in techniques for procuring appropriate tissue and familiar with their delivery to ensure appropriate preoperative planning and postoperative recovery.
Collapse
Affiliation(s)
- R Connor Chick
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 410 West Tenth Avenue, Columbus, OH 43210, USA
| | - Joal D Beane
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 410 West Tenth Avenue, Columbus, OH 43210, USA
| | - Carlo M Contreras
- Division of Surgical Oncology, The Ohio State University Wexner Medical Center, 410 West Tenth Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Paranthaman S, Uthaiah CA, Md S, Alkreathy HM. Comprehensive strategies for constructing efficient CRISPR/Cas based cancer therapy: Target gene selection, sgRNA optimization, delivery methods and evaluation. Adv Colloid Interface Sci 2025; 341:103497. [PMID: 40157335 DOI: 10.1016/j.cis.2025.103497] [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: 09/20/2024] [Revised: 12/17/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cancer is a complicated disease that results from the interplay between specific changes in cellular genetics and diverse microenvironments. The application of high-performance and customizable clustered regularly interspaced palindromic repeats/associated protein (CRISPR/Cas) nuclease systems has significantly enhanced genome editing for accurate cancer modeling and facilitated simultaneous genetic modification for cancer therapy and mutation identification. Achieving an effective CRISPR/Cas platform for cancer treatment depends on the identification, selection, and optimization of specific mutated genes in targeted cancer tissues. However, overcoming the off-target effects, specificity, and immunogenicity are additional challenges that must be addressed while developing a gene editing system for cancer therapy. From this perspective, we briefly covered the pipeline of CRISPR/Cas cancer therapy, identified target genes to optimize gRNAs and sgRNAs, and explored alternative delivery modalities, including viral, non-viral, and extracellular vesicles. In addition, the list of patents and current clinical trials related to this unique cancer therapy method is discussed. In summary, we have discussed comprehensive start-to-end pipeline strategies for CRISPR/Cas development to advance the precision, effectiveness, and safety of clinical applications for cancer therapy.
Collapse
Affiliation(s)
- Sathishbabu Paranthaman
- Department of Cell Biology and Molecular Genetics, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research, Tamaka, Kolar 563103, Karnataka, India.
| | - Chinnappa A Uthaiah
- Genetics Laboratory, Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raipur, Chhattisgarh 492099, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Mohammed Alkreathy
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Li J, Chen L, Chen M, Lin M, Xie Z, Wu H, Zhou Z, Lin W. Dap10 co-stimulation enhances the anti-HCC efficacy of NKp30 chimeric antigen receptor T cells. Transl Oncol 2025; 57:102425. [PMID: 40393250 DOI: 10.1016/j.tranon.2025.102425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/30/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell immunotherapy has made significant breakthroughs in the treatment of relapsed or refractory hematologic malignancies, but its efficacy in solid tumors remains limited. In this study, we developed a chimeric NKp30 (chNKp30) receptor whose ligand, B7H6, is often up-regulated in various tumor cells and sparsely expressed in healthy cells. Introduction of the cytoplasmic structural domain of dnax-activating protein 10 (DAP10) into CAR resulted in chNKp30-Dap10 CAR-T cells that showed superior cell proliferation, activation, and apoptosis inhibition after antigenic stimulation compared with conventional chNKp30-CD28 and chNKp30-Wt CAR-T cells lacking any structural domains, along with inducing a central memory T cell phenotype, whereas chNKp30-CD28 and chNKp30-Wt triggered an effector memory phenotype. In addition, chNKp30-Dap10 T cells secreted higher levels of pro-inflammatory cytokines such as IL-2, IFN-γ, and TNF-α, while chNKp30-CD28 T cells secreted more of the anti-inflammatory cytokine IL-10. In the killing assay, chNKp30-Dap10 T cells demonstrated stronger anti-tumor effects. Similarly, better tumor regression was observed in the hepatocellular carcinoma transplantation tumor model. These findings suggest that B7H6 is an attractive therapeutic target and DAP10 signaling is involved in the functional regulation of CAR-T cells in hepatocellular carcinoma, which may induce preferential cytokine profiling and differentiation for cancer therapy, and that NKp30-Dap10 CAR-T cell therapy offers a potential option for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- JieYu Li
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, Fujian Province, PR China
| | - LiMei Chen
- Department of Clinical Laboratory, Fuzhou 350014, PR China
| | - MingShui Chen
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - Miao Lin
- Fuzhou University College of Chemistry, Fuzhou 350002, PR China
| | - Zineng Xie
- Fuzhou University College of Chemistry, Fuzhou 350002, PR China
| | - HuiLing Wu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China
| | - ZhiFeng Zhou
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, Fujian Province, PR China.
| | - WanSong Lin
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, PR China; Fuzhou University College of Chemistry, Fuzhou 350002, PR China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, PR China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, Fujian Province, PR China.
| |
Collapse
|
4
|
Hinnekens C, Ramon J, Birben M, Germeraad WTV, Harizaj A, De Velder M, De Smedt SC, Vandekerckhove B, Braeckmans K, Fraire JC. Gentle and efficient engineering of primary human NK cells by photoporation with polydopamine nanosensitizers. J Control Release 2025; 382:113742. [PMID: 40250627 DOI: 10.1016/j.jconrel.2025.113742] [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: 09/24/2024] [Revised: 04/08/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
Over the past several years, adoptive T cell therapies have accounted for great success in treating diverse malignancies. More recently, however, NK cells are being investigated as a promising alternative. Due to the innate antiviral properties of NK cells, viral engineering has proven to be challenging, prompting the development of non-viral transfection technologies. In this work, we evaluated photoporation with polydopamine nanosensitizers as a notable upcoming transfection technology for the engineering of NK cells and compared its performance to Nucleofection. Our results demonstrated the successful transfection of NK cells with eGFP mRNA and gene editing with Cas9 ribonucleoproteins (RNPs) for knock-out of the KLRC1 gene, encoding for the inhibitory NK cell receptor NKG2A. Importantly, no alterations to the phenotype of the cells (e.g. expression of surface markers and release of cytokines) could be detected, nor was the proliferation or cytolytic capacity of the cells influenced by either of the treatments. Overall, our findings highlight the potential of polydopamine-sensitized photoporation as a gentle and efficient transfection technology for NK cell engineering.
Collapse
Affiliation(s)
- C Hinnekens
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - J Ramon
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - M Birben
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - W T V Germeraad
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center, Maastricht, the Netherlands
| | | | - M De Velder
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - S C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - B Vandekerckhove
- GMP Unit Cell therapy, Cord blood Bank and Hematopoietic Stem cell Bank, UZ Gent, Belgium
| | - K Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium.
| | - J C Fraire
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| |
Collapse
|
5
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
6
|
Chen J, Bu C, Lu Y, Peng X, Yu J, Ding X, Yuan P, Hong S. Bioresponsive nanoreactor initiates cascade reactions for tumor vascular normalization and lactate depletion to augment immunotherapy. Biomaterials 2025; 317:123100. [PMID: 39799700 DOI: 10.1016/j.biomaterials.2025.123100] [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: 09/12/2024] [Revised: 12/05/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment. However, abnormal tumor vasculature and excess lactate contribute to tumor immunosuppression and confer resistance to ICB therapy, seriously limiting its clinical application. Here, we have developed a bioresponsive nanoreactor, ALMn, which consists of hollow manganese dioxide nanoparticles with encapsulation of lactate oxidase and L-Arginine, to overcome immunosuppression and sensitize ICB therapy. In the tumor microenvironment, lactate oxidase catalyzes lactate to produce hydrogen peroxide, which subsequently oxidizes L-Arginine to generate nitric oxide for vascular normalization. Through cascade reactions, ALMn effectively depletes excess lactate and normalize tumor vasculature, reshaping the immunosuppressive phenotype to an immune-activated one. Transcriptomics and immunological analyses prove that ALMn facilitates the infiltration and activation of effector cells, further potentiating antitumor immunity. Consequently, ALMn sensitizes anti-PD-L1 therapy, significantly suppressing tumor growth with an 83.7 % suppression, and prolonging the survival of mice, with the median survival time increasing from 29.5 days to 54.5 days. Our study demonstrates that ALMn effectively alleviates tumor immunosuppression and synergizes with anti-PD-L1, which shows promise in boosting ICB therapy.
Collapse
Affiliation(s)
- Jiaoyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Changxin Bu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yuting Lu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xinran Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jiayin Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xin Ding
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China.
| | - Sheng Hong
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China.
| |
Collapse
|
7
|
Zheng Y, Gu Z, Shudde CE, Piper TL, Wang X, Aleck GA, Zhou J, King D, Chanda MK, Trinch L, Zou W, Courtney AH. An engineered viral protein activates STAT5 to prevent T cell suppression. Sci Immunol 2025; 10:eadn9633. [PMID: 40408430 DOI: 10.1126/sciimmunol.adn9633] [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: 01/25/2024] [Revised: 01/08/2025] [Accepted: 04/30/2025] [Indexed: 05/25/2025]
Abstract
T cell therapy efficacy can be compromised if cytokine-induced Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling is dysregulated or insufficient to sustain functionality. Here, we demonstrate that LCK kinase activity can be recruited to noncanonical protein substrates to directly activate targeted STAT proteins in T cells. STAT activation was accomplished by engineering the herpesvirus saimiri tyrosine kinase interacting protein (TIP) to provide a platform for the enforced recruitment of LCK to STAT proteins. We determined that a minimal region of TIP that binds to LCK could be combined with STAT binding sites derived from endogenous cytokine receptors. These constructs activated targeted STAT proteins in a cytokine-independent manner. We identified a STAT5 activator that sustained CD8+ T cell survival and cytotoxic function ex vivo in the absence of interleukin-2. Tumor outgrowth was reduced in vivo because of enhanced T cell persistence and functionality. Single-cell transcriptomics revealed that the STAT5 activator prevented the expression of genes associated with an exhausted T cell fate. Our findings demonstrate that signaling pathways can be rewired in T cells to sustain their function in solid tumors.
Collapse
Affiliation(s)
- Yating Zheng
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zehui Gu
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Claire E Shudde
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Taylor L Piper
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xinyu Wang
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Grace A Aleck
- Cellular and Molecular Biology Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jiajia Zhou
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dana King
- BRCF Bioinformatics Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Monica K Chanda
- Cancer Biology Training Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lilliana Trinch
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Adam H Courtney
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
8
|
Chen LE, Nittayacharn P, Exner AA. Progress and potential of nanobubbles for ultrasound-mediated drug delivery. Expert Opin Drug Deliv 2025:1-24. [PMID: 40353846 DOI: 10.1080/17425247.2025.2505044] [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: 02/13/2025] [Revised: 04/18/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Despite much progress, nanomedicine-based drug therapies in oncology remain limited by systemic toxicity and insufficient particle accumulation in the tumor. To address these barriers, formulations responsive to external physical stimuli have emerged. One most promising system is the ultrasound stimulation of drug-loaded, gas-core particles (bubbles). Ultrasound induces bubble cavitation for cell and tissue permeabilization, triggers on-demand drug release, and provides opportunities for real-time imaging of delivery. AREAS COVERED Here, we focus on shell-stabilized, gas-core nanoparticles (also termed nanobubbles or ultrafine bubbles) and their role in ultrasound-mediated therapeutic delivery to tumors. This review frames the advantages of nanobubbles within the ongoing deficits in nanomedicine, describes mechanisms of ultrasound-mediated therapy, and details formulation techniques for nanobubble delivery systems. It then highlights the past decade of research in nanobubble-facilitated drug delivery for cancer therapy and anticipates new directions in the field. EXPERT OPINION Nanobubble ultrasound contrast agents offer a spatiotemporally triggerable therapeutic coupled with a safe, accessible imaging modality. Nanobubbles can be loaded with diverse therapeutic cargoes to treat disease and overcome numerous barriers limiting delivery to solid tumors. Close attention to formulation, characterization methods, acoustic testing parameters, and the biological mechanisms of nanobubble delivery will facilitate preclinical research toward clinical adoption.
Collapse
Affiliation(s)
- Laura E Chen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pinunta Nittayacharn
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Radiology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
9
|
Chao CJ, Zhang E, Trinh DN, Udofa E, Lin H, Silvers C, Huo J, He S, Zheng J, Cai X, Bao Q, Zhang L, Phan P, Elgendy SM, Shi X, Burdette JE, Lee SSY, Gao Y, Zhang P, Zhao Z. Integrating antigen capturing nanoparticles and type 1 conventional dendritic cell therapy for in situ cancer immunization. Nat Commun 2025; 16:4578. [PMID: 40379691 DOI: 10.1038/s41467-025-59840-w] [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] [Received: 09/17/2024] [Accepted: 05/02/2025] [Indexed: 05/19/2025] Open
Abstract
Eliciting a robust immune response against tumors is often hampered by the inadequate presence of effective antigen presenting cells and their suboptimal ability to present antigens within the immunosuppressive tumor microenvironment. Here, we report a cascade antigen relay strategy integrating antigen capturing nanoparticles (AC-NPs) and migratory type 1 conventional dendritic cells (cDC1s), named Antigen Capturing nanoparticle Transformed Dendritic Cell therapy (ACT-DC), to facilitate in situ immunization. AC-NPs are engineered to capture antigens directly from the tumor and facilitate their delivery to adoptively transferred migratory cDC1s, enhancing antigen presentation to the lymph nodes and reshaping the tumor microenvironment. Our findings suggest that ACT-DC improves in situ antigen collection, triggers a robust systemic immune response without the need for exogenous antigens, and transforms the tumor environment into a more "immune-hot" state. In multiple tumor models including colon cancer, melanoma, and glioma, ACT-DC in combination with immune checkpoint inhibitors eliminates primary tumors in 50-100% of treated mice and effectively rejects two separate tumor rechallenges. Collectively, ACT-DC could provide a broadly effective approach for in situ cancer immunization and tumor microenvironment modulation.
Collapse
Affiliation(s)
- Chih-Jia Chao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Endong Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Duong N Trinh
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Edidiong Udofa
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Hanchen Lin
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Caylee Silvers
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jiawei Huo
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shan He
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Jingtian Zheng
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Xiaoying Cai
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Qing Bao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Luyu Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Philana Phan
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Sara M Elgendy
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Xiangqian Shi
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Steve Seung-Young Lee
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Yu Gao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, Chicago, IL, USA
| | - Peng Zhang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL, USA.
- University of Illinois Cancer Center, Chicago, IL, USA.
| |
Collapse
|
10
|
Li YR, Zhu Y, Halladay T, Yang L. In vivo CAR engineering for immunotherapy. Nat Rev Immunol 2025:10.1038/s41577-025-01174-1. [PMID: 40379910 DOI: 10.1038/s41577-025-01174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 05/19/2025]
Abstract
Chimeric antigen receptor (CAR)-engineered immune cell therapy represents an important advance in cancer treatments. However, the complex ex vivo cell manufacturing process and stringent patient selection criteria curtail its widespread use. In vivo CAR engineering is emerging as a promising off-the-shelf therapy, providing advantages such as streamlined production, elimination of patient-specific manufacturing, reduced costs and simplified logistics. A large set of preclinical findings has inspired further investigation into treatments for hard-to-treat diseases such as solid tumours and has facilitated the development of advanced products to enhance in vivo CAR engineering efficacy, the persistence of the cellular therapeutic and safety. In this Review, we summarize current in vivo CAR engineering strategies, including nanoparticle-based and viral delivery systems as well as bioinstructive implantable scaffolds, and discuss their advantages and disadvantages. Additionally, we provide a systematic comparison between in vivo and conventional ex vivo CAR engineering methods and address the challenges and future prospects of in vivo CAR engineering.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Tang S, Zhang Y, Wang P, Tang Q, Liu Y, Lu F, Han M, Zhou M, Hu Q, Feng M, Liang D. NKG2D-CAR-targeted iPSC-derived MSCs efficiently target solid tumors expressing NKG2D ligand. iScience 2025; 28:112343. [PMID: 40276759 PMCID: PMC12020857 DOI: 10.1016/j.isci.2025.112343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Mesenchymal stem cells (MSCs) hold potential in cancer therapy; however, insufficient tumor homing ability and heterogeneity limit their therapeutic benefits. Obviously, the homogeneous induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) with enhanced ability of tumor targeting could be the solution. In this study, a CAR containing the NKG2D extracellular domain was targeted at the B2M locus of iPSCs to generate NKG2D-CAR-iPSCs, which were subsequently differentiated into NKG2D-CAR-iMSCs. In vitro, NKG2D-CAR significantly enhanced migration and adhesion of iMSCs to a variety of solid tumor cells expressing NKG2D ligands. RNA sequencing (RNA-seq) revealed significant upregulation of genes related to cell adhesion, migration, and binding in NKG2D-CAR-iMSCs. In A549 xenograft model, NKG2D-CAR-iMSCs demonstrated a 57% improvement in tumor-homing ability compared with iMSCs. In conclusion, our findings demonstrate enhanced targeting specificity of NKG2D-CAR-iMSCs to tumor cells expressing NKG2D ligands in vitro and in vivo, facilitating future investigation of iMSCs as an off-the-shelf living carrier for targeted delivery of anti-tumor agents.
Collapse
Affiliation(s)
- Shuqing Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yusang Zhang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Peiyun Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Qiyu Tang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Yating Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Fan Lu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Mengting Han
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Miaojin Zhou
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Qian Hu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| | - Mai Feng
- Hunan Key Laboratory of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha 410078, China
| | - Desheng Liang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
12
|
Chen Y, Song J, Chen X, Chen G. Synthetic Glycopolymers in Tumor Immunotherapy. Macromol Rapid Commun 2025:e2401089. [PMID: 40372066 DOI: 10.1002/marc.202401089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/30/2025] [Indexed: 05/16/2025]
Abstract
Glycopolymers, as an emerging immunomodulatory material, exhibit considerable promise in the field of tumor immunotherapy. Compared to native saccharides, they offer significant advantages, including enhanced immune activity, controllable structure and sequence, elevated stability, and high purity. By mimicking the multivalency of native sugar chains, glycopolymers significantly enhance their interactions with receptors, a phenomenon known as the "glycocluster effect." Glycopolymers are capable of modulating immune cell functions, inhibiting tumor immune evasion, and reconfiguring the tumor microenvironment. This review provides a comprehensive overview of recent advancements in the application of glycopolymers, protein-glycopolymer conjugates, glycopolymer-based micro/nanoparticles, and glycopolymer-engineered cells in tumor immunotherapy. These glycopolymer-based materials enhance antitumor immune responses by specifically interacting with immune cell surface receptors, significantly improving the precision and efficacy of immunotherapy, and providing valuable insights for the development of innovative therapeutic strategies in cancer treatment.
Collapse
Affiliation(s)
- Yuru Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Jiaxin Song
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Xiangqian Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Gaojian Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
13
|
Jusztus V, Szöőr Á, Hajdu P. Role of CAR-T cell K+ channels in tumor infiltration and elimination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf084. [PMID: 40373182 DOI: 10.1093/jimmun/vkaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/02/2025] [Indexed: 05/17/2025]
Abstract
Genetic modification of T cells to express chimeric antigen receptors (CAR, CAR-T cells) enable them to recognize the specific antigen on tumor surface and then eliminate the tumor. T lymphocyte ion channels such as Kv1.3, KCa3.1 and CRAC influence T cell activation and proliferation by regulating Ca2+ signaling, as well as other effector functions such as cytokine release, migration and even target cell killing. Here we established two CAR cell lines (using CEM T cell line and primary T cells) recognizing CD19 antigen on surface of Raji B and human breast cancer MCF-7 expressing CD19 cell lines. First, we exposed that KCa3.1 and Kv1.3 functional expressions of CEM cells were comparable to those in T cells, which demonstrated their suitability for primary T cell mimics. Next, we studied the tumor cell killing efficiency of CAR-T and CEM-CAR cells in monolayer and 3D spheroid tumor models. We could show that CAR expressing cells specifically eliminate tumor cells regardless of tumor models. Furthermore, the application of Kv1.3 (Vm24) and KCa3.1 (TRAM34) inhibitors significantly improved the tumor eradication efficiency for both CEM-CAR and CAR-T cells in spheroids, however, the infiltration rate was not influenced upon addition of antagonists. We could conclude that modification of Kv1.3 and KCa3.1 ion channels could contribute to a more effective immunotherapy of solid tumor.
Collapse
Affiliation(s)
- Vivien Jusztus
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Hungary
| | - Árpád Szöőr
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Hungary
| | - Péter Hajdu
- Faculty of Medicine, Department of Biophysics and Cell Biology, University of Debrecen, Debrecen, Hungary
- Faculty of Dentistry, Division of Dental Biochemistry, Department of Basic Medical Sciences, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
14
|
Zhang X, Fam KT, Dai T, Hang HC. Microbiota mechanisms in cancer progression and therapy. Cell Chem Biol 2025; 32:653-677. [PMID: 40334660 DOI: 10.1016/j.chembiol.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
The composition of the microbiota in patients has been shown to correlate with cancer progression and response to therapy, highlighting unique opportunities to improve patient outcomes. In this review, we discuss the challenges and advancements in understanding the chemical mechanisms of specific microbiota species, pathways, and molecules involved in cancer progression and treatment. We also describe the modulation of cancer and immunotherapy by the microbiota, along with approaches for investigating microbiota enzymes and metabolites. Elucidating these specific microbiota mechanisms and molecules should offer new opportunities for developing enhanced diagnostics and therapeutics to improve outcomes for cancer patients. Nonetheless, many microbiota mechanisms remain to be determined and require innovative chemical genetic approaches.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Kyong Tkhe Fam
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tingting Dai
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
15
|
Zhang K, Zhang Y, Xiang P, Wang Y, Li Y, Jiang S, Zhang Y, Chen M, Su W, Li X, Li S. Advances in T Cell-Based Cancer Immunotherapy: From Fundamental Mechanisms to Clinical Prospects. Mol Pharm 2025. [PMID: 40359327 DOI: 10.1021/acs.molpharmaceut.4c01502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
T cells and their T cell receptors (TCRs) play crucial roles in the adaptive immune system's response against pathogens and tumors. However, immunosenescence, characterized by declining T cell function and quantity with age, significantly impairs antitumor immunity. Recent years have witnessed remarkable progress in T cell-based cancer treatments, driven by a deeper understanding of T cell biology and innovative screening technologies. This review comprehensively examines T cell maturation mechanisms, T cell-mediated antitumor responses, and the implications of thymic involution on T cell diversity and cancer prognosis. We discuss recent advances in adoptive T cell therapies, including tumor-infiltrating lymphocyte (TIL) therapy, engineered T cell receptor (TCR-T) therapy, and chimeric antigen receptor T cell (CAR-T) therapy. Notably, we highlight emerging DNA-encoded library technologies in mammalian cells for high-throughput screening of TCR-antigen interactions, which are revolutionizing the discovery of novel tumor antigens and optimization of TCR affinity. The review also explores strategies to overcome challenges in the solid tumor microenvironment and emerging approaches to enhance the efficacy of T cell therapy. As our understanding of T cell biology deepens and screening technologies advances, T cell-based immunotherapies show increasing promise for delivering durable clinical benefits to a broader patient population.
Collapse
Affiliation(s)
- Kaili Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Pan Xiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Wang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yifan Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuze Jiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yuxuan Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Min Chen
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaoling Li
- Cell Biotechnology Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin 300308, China
- National Clinical Research Center for Cancer, Tianjin 300060, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300090, China
| | - Shuai Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
16
|
Hu X, Wang Z, Zhu Y, Li Z, Yan H, Zhao X, Wang Q. Advancements in molecular imaging for the diagnosis and treatment of pancreatic ductal adenocarcinoma. NANOSCALE ADVANCES 2025; 7:2887-2903. [PMID: 40270837 PMCID: PMC12012634 DOI: 10.1039/d4na01080a] [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: 12/30/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant tumor characterized by poor overall patient survival and prognosis, largely due to challenges in early diagnosis, limited surgical options, and a high propensity for therapy resistance. The integration of various imaging modalities through molecular imaging techniques, particularly multimodal molecular imaging, offers the potential to provide more precise and comprehensive information about the lesion. With advances in nanomedicine, new imaging and drug delivery approaches that allow the development of multifunctional theranostic agents offer opportunities for improving pancreatic cancer treatment using precision oncology. Herein, we review the diagnostic and therapeutic applications of molecular imaging for PDAC and discuss the adoption of multimodal imaging approaches that combine the strengths of different imaging techniques to enhance diagnostic accuracy and therapeutic efficacy. We emphasize the significant role of nanomedicine technology in advancing multimodal molecular imaging and theranostics, and their potential impact on PDAC management. This comprehensive review aims to serve as a valuable reference for researchers and clinicians, offering insights into the current state of molecular imaging in PDAC and outlining future directions for improving early diagnosis, combination therapies, and prognostic evaluations.
Collapse
Affiliation(s)
- Xun Hu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zihua Wang
- School of Basic Medical Sciences, Fujian Medical University Fuzhou 350122 Fujian Province China
| | - Yuting Zhu
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Zhangfu Li
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital Shenzhen Guangdong 518036 China
| | - Hao Yan
- Tsinghua Shenzhen International Graduate School/Tsinghua University Shenzhen 518055 China
| | - Xinming Zhao
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| | - Qian Wang
- Department of Diagnostic Imaging, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100021 China
| |
Collapse
|
17
|
Yassin AAK, Ureña Martin C, Le Saux G, Pandey A, Tzadka S, Toledo E, Pandit JJ, Sherf T, Nusbaum I, Bhattachrya B, Banerji R, Greenshpan Y, Abu Ahmad MA, Radinsky O, Sklartz M, Gazit R, Elkabets M, Ghassemi S, Cohen O, Schvartzman M, Porgador A. Mechanostimulatory Platform for Improved CAR T Cell Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2412482. [PMID: 40348587 DOI: 10.1002/adma.202412482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 04/29/2025] [Indexed: 05/14/2025]
Abstract
Chimeric Antigen Receptor T (CAR T) cell immunotherapy has revolutionized cancer treatment, yet it is hindered by rapid T-cell exhaustion caused by uncontrolled activation during CAR generation. Leveraging insights into T-cell mechanosensing, a novel mechanostimulatory platform is engineered for T-cell activation based on an antigen-carrying surface with controlled elasticity and nanotopography. The platform is designed to optimize and balance T-cell exhaustion, proliferation, and CAR expression. It enhances the differentiation of T cells into the central memory subset, which is crucial for the persistence of CAR T cell therapy's anticancer effects. The platform produces CAR T cells with higher antitumor efficacy, as validated through ex vivo experiments, and with higher in vivo persistence and ability to suppress tumor proliferation, as compared to CAR T cells generated by standard protocols. RNA-seq analysis confirmed an increased transcriptional signature of central memory T cells. Furthermore, this platform completely eliminates T-cell toxicity associated with the non-viral transfection process typically observed with standard activation methods. This platform presents a promising pathway for improving the efficiency and safety of CAR T cell therapy.
Collapse
Affiliation(s)
- Abed Al-Kader Yassin
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Carlos Ureña Martin
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Guillaume Le Saux
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Ashish Pandey
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Sivan Tzadka
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Esti Toledo
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Jatin Jawhir Pandit
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Tomer Sherf
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Idan Nusbaum
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Baisali Bhattachrya
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Rajashri Banerji
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Yariv Greenshpan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Muhammad Abu Abu Ahmad
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Olga Radinsky
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Menachem Sklartz
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Roi Gazit
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Sabah Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ofir Cohen
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Department of Software and Information System Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Mark Schvartzman
- Department of Materials Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Ilse Katz Institute for the Nanoscale Science and Technology, Beer-Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| |
Collapse
|
18
|
Cao G, Hu Y, Pan T, Tang E, Asby N, Althaus T, Wan J, Riedell PA, Bishop MR, Kline JP, Huang J. Two-stage CD8 + CAR T-cell differentiation in patients with large B-cell lymphoma. Nat Commun 2025; 16:4205. [PMID: 40328775 PMCID: PMC12055983 DOI: 10.1038/s41467-025-59298-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Advancements in chimeric antigen receptor (CAR) T-cell therapy for treating diffuse large B-cell lymphoma (DLBCL) have been limited by an incomplete understanding of CAR T-cell differentiation in patients. Here, we show via single-cell, multi-modal, and longitudinal analyses, that CD8+ CAR T cells from DLBCL patients successfully treated with axicabtagene ciloleucel undergo two distinct waves of clonal expansion in vivo. The first wave is dominated by an exhausted-like effector memory phenotype during peak expansion (day 8-14). The second wave is dominated by a terminal effector phenotype during the post-peak persistence period (day 21-28). Importantly, the two waves have distinct ontogeny from the infusion product and are biologically uncoupled. Precursors of the first wave exhibit more effector-like signatures, whereas precursors of the second wave exhibit more stem-like signatures. We demonstrate that CAR T-cell expansion and persistence are mediated by clonally, phenotypically, and ontogenically distinct CAR T-cell populations that serve complementary clinical purposes.
Collapse
Affiliation(s)
- Guoshuai Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Yifei Hu
- Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Nicholas Asby
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Thomas Althaus
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Peter A Riedell
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA
| | - Michael R Bishop
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Justin P Kline
- The David and Etta Jonas Center for Cellular Therapy, University of Chicago, Chicago, IL, 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA
| | - Jun Huang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Cancer Biology, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
19
|
Gupta D, Shaz B. Surveying local CAR T-cell manufacturing processes to facilitate standardization and expand accessibility. J Transl Med 2025; 23:507. [PMID: 40329308 PMCID: PMC12057077 DOI: 10.1186/s12967-025-06400-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CAR T-cell) therapies have shown significant promise in treating cancers and other diseases. However, the manufacturing processes for CAR T-cell therapies exhibit considerable variability, which can affect treatment consistency and patient outcomes. While centralized manufacturing models dominate, local decentralized approaches, including point-of-care production, are being explored to address logistical and access challenges. This study aims to evaluate the current landscape of local CAR T-cell manufacturing at academic institutions. METHODS A comprehensive, cross-sectional survey was distributed to 130 FACT and/or JACIE accredited academic institutions globally. The survey, developed from semi-structured interviews with CAR T-cell manufacturing experts, assessed practices in cell modification methods, equipment protocols, and regulatory challenges. Data were analyzed using descriptive statistics, comparing responses across institutions and regions. RESULTS 45 of the 130 institutions (35 from the United States and 10 internationally, from the European Union, the United Kingdom, and Australia) responded to the survey (35% response rate). Of the 45 responding institutions, 40 were actively engaged or planning to engage in CAR T-cell production, while five had no plans to initiate manufacturing. Within the 40 institutions engaged in CAR T-cell production, 63% (25/40) reported active manufacturing, while 37% (15/40) were in the process of developing manufacturing capabilities. The most commonly reported barriers to local manufacturing were cost constraints (70%, 28/40), regulatory complexities (70%, 28/40), and facility requirements (57%, 17/40). Variability in product quality was cited by 73% (29/40) of institutions. Equipment costs and the need for specialized training emerged as major challenges, particularly for international institutions. Institutions also highlighted the need for automated platforms, with 60% (24/40) using the Miltenyi CliniMACS Prodigy and 50% (20/40) using the Lonza Cocoon. CONCLUSIONS This study highlights the widespread adoption of local CAR T-cell manufacturing and the significant variability in production processes across institutions. The findings emphasize the importance of establishing quality control benchmarks and data reporting frameworks to improve product consistency and access to CAR T-cell therapies. Addressing barriers such as cost, infrastructure, and regulatory challenges through standardization efforts and international collaboration could enhance the reproducibility, scalability, and accessibility of CAR T-cell therapies globally.
Collapse
Affiliation(s)
- Deven Gupta
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA.
- Margolis Institute for Health Policy, Duke University, Durham, NC, USA.
| | - Beth Shaz
- Marcus Center for Cellular Cures, Duke University School of Medicine, Durham, NC, USA
- Margolis Institute for Health Policy, Duke University, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
20
|
Ramola A, Shakya AK, Bergman A. Comprehensive Analysis of Advancement in Optical Biosensing Techniques for Early Detection of Cancerous Cells. BIOSENSORS 2025; 15:292. [PMID: 40422031 DOI: 10.3390/bios15050292] [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: 03/28/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025]
Abstract
This investigation presents an overview of various optical biosensors utilized for the detection of cancer cells. It covers a comprehensive range of technologies, including surface plasmon resonance (SPR) sensors, which exploit changes in refractive index (RI) at the sensor surface to detect biomolecular interactions. Localized surface plasmon resonance (LSPR) sensors offer high sensitivity and versatility in detecting cancer biomarkers. Colorimetric sensors, based on color changes induced via specific biochemical reactions, provide a cost-effective and simple approach to cancer detection. Sensors based on fluorescence work using the light emitted from fluorescent molecules detect cancer-specific targets with specificity and high sensitivity. Photonics and waveguide sensors utilize optical waveguides to detect changes in light propagation, offering real-time and label-free detection of cancer biomarkers. Raman spectroscopy-based sensors utilize surface-enhanced Raman scattering (SERS) to provide molecular fingerprint information for cancer diagnosis. Lastly, fiber optic sensors offer flexibility and miniaturization, making them suitable for in vivo and point-of-care applications in cancer detection. This study provides insights into the principles, applications, and advancements of these optical biosensors in cancer diagnostics, highlighting their potential in improving early detection and patient outcomes.
Collapse
Affiliation(s)
- Ayushman Ramola
- Department of Electrical and Electronics Engineering, Ariel University, Ariel 40700, Israel
| | - Amit Kumar Shakya
- Department of Electrical and Electronics Engineering, Ariel University, Ariel 40700, Israel
| | - Arik Bergman
- Department of Electrical and Electronics Engineering, Ariel University, Ariel 40700, Israel
| |
Collapse
|
21
|
Joly F, Castel H, Compter A, Nicola C, Duivon M, Lange M. Neuropsychological and central neurologic effects of cancer immunotherapy: the start of a new challenge. J Clin Exp Neuropsychol 2025:1-20. [PMID: 40323211 DOI: 10.1080/13803395.2025.2498713] [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: 05/06/2024] [Accepted: 04/22/2025] [Indexed: 05/15/2025]
Abstract
INTRODUCTION Cognitive difficulties are frequently reported after cancer treatments, such as chemotherapy or hormone therapy, and have a negative impact on patients' quality of life. Recently, some studies have shown that new cancer treatments, such as immunotherapy agents, can induce cognitive changes. METHOD This review presents the central neurological immune adverse events of immunotherapy treatments including Immune Checkpoint Inhibitors (ICI) and Chimeric Antigen Receptor (CAR) T-cell therapy. The physiopathological mechanisms and risk factors are developed and clinical studies on immunotherapy agents and cognition (among adult patients, using validated questionnaires and/or cognitive tests), psychological factors and quality of life were presented. RESULTS Neurological toxicities are frequently observed with CAR-T cell therapies at acute stage, such as the immune effector cell-associated neurotoxicity syndrome (ICANS), inducing cognitive disorders such as disorientation and aphasia. However, few studies have accurately assessed the impact of immunotherapy on cognition. The methodology of these studies is heterogeneous and they mainly included nonspecific self-report questionnaires of cognitive complaints. Variable results have been obtained concerning the cognitive impact of ICI and CAR-T cell several months following immunotherapy: overall, while some studies reported cognitive difficulties (mainly processing speed and executive functions), the majority has not. Although anxiety and depression are frequently reported in patients treated with ICI or CAR-T cells, these symptoms tend to decrease after the start of immunotherapy. The current neurobiological investigations are too fragmentary to explain neurological symptoms and potential cognitive alteration, but neuroinflammation, vascular inflammation, brain blood barrier disruption, and immune cell brain infiltration would constitute common mechanisms relayed by CAR-T and to a lesser degree, ICI. CONCLUSIONS Acute neurological toxicities following CAR-T cell therapies are a major issue. Further studies are needed to better assess cognitive difficulties after the initiation of immunotherapy, in particular ICI, to better understand the physiopathology, including imaging studies, and risk factors.
Collapse
Affiliation(s)
- Florence Joly
- ANTICIPE U1086 INSERM-UCN, Equipe Labellisée Ligue Contre le Cancer, Centre François Baclesse, Normandie Université UNICAEN, Caen, France
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
- Clinical Research Department, Centre François Baclesse, Caen, France
- Medical oncology department, CHU de Caen, Caen, France
| | - Hélène Castel
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
- UNIROUEN, INSERM, U1245, Cancer and Brain Genomics, Normandie University, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Annette Compter
- Department of Neuro-Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Celeste Nicola
- UNIROUEN, INSERM, U1245, Cancer and Brain Genomics, Normandie University, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Mylène Duivon
- ANTICIPE U1086 INSERM-UCN, Equipe Labellisée Ligue Contre le Cancer, Centre François Baclesse, Normandie Université UNICAEN, Caen, France
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
| | - Marie Lange
- ANTICIPE U1086 INSERM-UCN, Equipe Labellisée Ligue Contre le Cancer, Centre François Baclesse, Normandie Université UNICAEN, Caen, France
- Services Unit PLATON, Cancer and Cognition Platform, University of Caen Normandy, Caen, France
- Clinical Research Department, Centre François Baclesse, Caen, France
| |
Collapse
|
22
|
Sheikh M, Saiyyad A, Aliunui A, Jirvankar PS. The evolving landscape of oncolytic virus immunotherapy: combinatorial strategies and novel engineering approaches. Med Oncol 2025; 42:190. [PMID: 40314865 DOI: 10.1007/s12032-025-02746-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Oncolytic viruses (OVs) are a promising class of cancer therapy, exploiting their abilities to selectively infect and kill cancer cells while stimulating antitumor immune responses. The current assessment explores the changing horizons of OV immunotherapy, focusing on recent advances in technology plans to improve OV projects and combined approaches to improve curative efficacy. We discuss how OVs induce direct oncolysis and promote the release of tumor-associated antigens, leading to the activation of both innate and adaptive immunity. Special attention shall be given to programs for arm OVs to express curative genes, modify the tumor microenvironment and overcome immunosuppression. Moreover, we assess the synergies of uniting OVs with other immunotherapeutic techniques, such as immune checkpoint inhibitors and cell therapy, to improve tolerant outcomes. The present assessment provides an understanding of the relevant declaration of the OV analysis, highlighting the main obstacles and the future directions for the development of other capable and targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Mujibullah Sheikh
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India.
| | - Arshiya Saiyyad
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| | - Aimé Aliunui
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| | - Pranita S Jirvankar
- Datta Meghe College of Pharmacy DMIHER (Deemed to be University), Wardha, Maharashtra, 442001, India
| |
Collapse
|
23
|
Liang J, Wang P, Lin Y, Jia A, Tong F, Li Z. Advances in Photothermal Therapy for Oral Cancer. Int J Mol Sci 2025; 26:4344. [PMID: 40362580 PMCID: PMC12072920 DOI: 10.3390/ijms26094344] [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: 04/10/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Oral cancer represents a critical global health issue, where traditional treatment modalities are often characterized by considerable adverse effects and suboptimal effectiveness. Photothermal therapy (PTT) offers an innovative method for tumor treatment, leveraging photothermal agents to convert light into hyperthermia, ultimately leading to tumor ablation. PTT offers unique advantages in treating oral cancer due to its superficial anatomical location and consequent accessibility to laser irradiation. PTT's advantage is further enhanced by its capacity to facilitate drug release and promote tissue regeneration. Consequently, the application of PTT for oral cancer has garnered widespread interest and has undergone rapid development. This review outlines advances in PTT for oral cancer, emphasizing strategies to improve efficacy and combination therapy approaches. The key challenges, including temperature control and long-term biosafety, are discussed alongside future directions. The review also encompasses PTT's role in managing oral potentially malignant disorders and postoperative defects, conditions intimately linked with oral cancer. We aim to provide guidance for emerging PTT research in oral cancer and to promote the development of precise and efficient treatment strategies.
Collapse
Affiliation(s)
- Jian Liang
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.L.); (P.W.); (Y.L.); (A.J.)
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang 330006, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Pei Wang
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.L.); (P.W.); (Y.L.); (A.J.)
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang 330006, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Yanfang Lin
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.L.); (P.W.); (Y.L.); (A.J.)
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang 330006, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Ao Jia
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.L.); (P.W.); (Y.L.); (A.J.)
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang 330006, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Fei Tong
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.L.); (P.W.); (Y.L.); (A.J.)
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang 330006, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang 330006, China
| | - Zhihua Li
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (J.L.); (P.W.); (Y.L.); (A.J.)
- Jiangxi Provincial Key Laboratory of Oral Diseases, Nanchang 330006, China
- Jiangxi Provincial Clinical Research Center for Oral Diseases, Nanchang 330006, China
| |
Collapse
|
24
|
Walters AJ, Yang X, Olson SD, Bashor CJ. Enhancing the safety and efficacy of cell therapy with programmed sense-and-respond function. Clin Transl Med 2025; 15:e70328. [PMID: 40292750 PMCID: PMC12035645 DOI: 10.1002/ctm2.70328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Affiliation(s)
- Andrew J. Walters
- Department of BioengineeringRice UniversityHoustonTexasUSA
- Department of Pediatric SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Xiaoyu Yang
- Department of BioengineeringRice UniversityHoustonTexasUSA
| | - Scott D. Olson
- Department of Pediatric SurgeryMcGovern Medical SchoolUniversity of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Caleb J. Bashor
- Department of BioengineeringRice UniversityHoustonTexasUSA
- Department of BiosciencesRice UniversityHoustonTexasUSA
| |
Collapse
|
25
|
Goblirsch TJ, Huang V, Maguire A, Chebolu AP. BILATERAL CHOROIDAL DETACHMENTS FOLLOWING NOVEL CHIMERIC ANTIGEN RECEPTOR T-CELL IMMUNOTHERAPY REGIMEN. Retin Cases Brief Rep 2025; 19:342-345. [PMID: 38437821 DOI: 10.1097/icb.0000000000001558] [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: 10/09/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND/PURPOSE The aim of this study was to describe a vision-threatening adverse event of a novel chimeric antigen receptor T-cell immunotherapy for metastatic prostate cancer. METHODS This was an observational case report. RESULTS A 77-year-old male patient with a history of metastatic prostate cancer and pulmonary embolism enrolled in a clinical trial investigating the use of chimeric antigen receptor T-cell immunotherapy for treatment of metastatic prostate cancer presented with a subjective left temporal visual disturbance. The patient was found to have bilateral extensive choroidal detachments on examination. Optical coherence tomography macula demonstrated no intraretinal fluid or supraretinal fluid in both eyes. B-scan of both eyes redemonstrated choroidal detachments in both eyes and no retinal detachment in either eye. The patient was initiated on a topical and systemic steroid regimen and experienced symptomatic and clinical improvement. CONCLUSION The authors exhibit a case of bilateral choroidal detachments secondary to systemic reaction to a novel immunotherapy for metastatic cancer successfully treated with systemic and topical steroids. A close follow-up may be required for patients receiving similar novel immunotherapeutic regimens.
Collapse
Affiliation(s)
- Timothy J Goblirsch
- Department of Ophthalmology, University of Pennsylvania Scheie Eye Institute, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
26
|
Li YR, Zhou K, Lee D, Zhu Y, Halladay T, Yu J, Zhou Y, Lyu Z, Fang Y, Chen Y, Semaan S, Yang L. Generating allogeneic CAR-NKT cells for off-the-shelf cancer immunotherapy with genetically engineered HSP cells and feeder-free differentiation culture. Nat Protoc 2025; 20:1352-1388. [PMID: 39825143 DOI: 10.1038/s41596-024-01077-w] [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: 06/13/2024] [Accepted: 09/20/2024] [Indexed: 01/20/2025]
Abstract
The clinical potential of current chimeric antigen receptor-engineered T (CAR-T) cell therapy is hampered by its autologous nature that poses considerable challenges in manufacturing, costs and patient selection. This spurs demand for off-the-shelf therapies. Here we introduce an ex vivo feeder-free culture method to differentiate gene-engineered hematopoietic stem and progenitor (HSP) cells into allogeneic invariant natural killer T (AlloNKT) cells and their CAR-armed derivatives (AlloCAR-NKT cells). We include detailed information on lentivirus generation and titration, as well as the five stages of ex vivo culture required to generate AlloCAR-NKT cells, including HSP cell engineering, HSP cell expansion, NKT cell differentiation, NKT cell deep differentiation and NKT cell expansion. In addition, we describe procedures for evaluating the pharmacology, antitumor efficacy and mechanism of action of AlloCAR-NKT cells. It takes ~2 weeks to generate and titrate lentiviruses and ~6 weeks to generate mature AlloCAR-NKT cells. Competence with human stem cell and T cell culture, gene engineering and flow cytometry is required for optimal results.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kuangyi Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Derek Lee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jiaji Yu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yang Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yuning Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sasha Semaan
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Centre of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
27
|
Kret ZS, Sweder RJ, Pollock R, Tinoco G. Potential Mechanisms for Immunotherapy Resistance in Adult Soft-Tissue Sarcoma. Target Oncol 2025; 20:485-502. [PMID: 40289241 DOI: 10.1007/s11523-025-01145-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Soft-tissue sarcomas represent a diverse group of rare malignancies originating from mesenchymal tissue, accounting for less than 1% of adult cancers in the USA. With over 13,000 new cases and around 5350 deaths annually, patients with metastatic soft-tissue sarcomas face limited therapeutic options and an estimated median overall survival of 18 months. While immunotherapy has demonstrated effectiveness in several cancers, its application in soft-tissue sarcomas remains challenging owing to the tumors' largely "cold" immunological environment, characterized by low levels of tumor-infiltrating lymphocytes and a lack of soft-tissue sarcoma-specific biomarkers. This review examines potential mechanisms underlying immunotherapy resistance in soft-tissue sarcomas, including the complex interplay between innate and adaptive immunity, the tumor microenvironment, and the role of immune-related genes. Despite preliminary findings suggesting correlations between immune profiles and histological subtypes, consistent biomarkers for predicting immunotherapeutic responses across soft-tissue sarcoma types are absent. Emerging strategies focus on converting "cold" tumors to "hot" tumors, enhancing their susceptibility to immunologic activation. While research is ongoing, personalized treatment approaches may offer hope for overcoming the inherent heterogeneity and resistance seen in soft-tissue sarcomas, ultimately aiming to improve outcomes for affected patients.
Collapse
Affiliation(s)
- Zaina S Kret
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Ryan J Sweder
- The Ohio State University College of Arts and Sciences and College of Medicine, Columbus, OH, USA
| | - Raphael Pollock
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 1800 Cannon Drive, 1240 Lincoln Tower, Columbus, OH, 43210, USA.
| |
Collapse
|
28
|
Safaei S, Yari A, Pourbagherian O, Maleki LA. The role of cytokines in shaping the future of Cancer immunotherapy. Cytokine 2025; 189:156888. [PMID: 40010034 DOI: 10.1016/j.cyto.2025.156888] [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: 10/28/2024] [Revised: 01/13/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025]
Abstract
As essential immune system regulators, cytokines are essential for modulating both innate and adaptive immunological responses. They have become important tools in cancer immunotherapy, improving the immune system's capacity to identify and destroy tumor cells. This article examines the background, workings, and therapeutic uses of cytokines, such as interleukins, interferons, and granulocyte-macropHage colony-stimulating factors, in the management of cancer. It examines the many ways that cytokines affect immune cell activation, signaling pathways, tumor development, metastasis, and prognosis by modifying the tumor microenvironment. Despite the limited effectiveness of cytokine-based monotherapy, recent developments have concentrated on new fusion molecules such as immunocytokines, cytokine delivery improvements, and combination techniques to maximize treatment efficacy while reducing adverse effects. Current FDA-approved cytokine therapeutics and clinical trial results are also included in this study, which offers insights into how cytokines might be used with other therapies including checkpoint inhibitors, chemotherapy, and radiation therapy to address cancer treatment obstacles. This study addresses the intricacies of cytokine interactions in the tumor microenvironment, highlighting the possibility for innovative treatment methods and suggesting fresh techniques for enhancing cytokine-based immunotherapies. PEGylation, viral vector-mediated cytokine gene transfer, antibody-cytokine fusion proteins (immunocytokines), and other innovative cytokine delivery techniques are among the novelties of this work, which focuses on the most recent developments in cytokine-based immunotherapy. Additionally, the study offers a thorough examination of the little-reviewed topic of cytokine usage in conjunction with other treatment techniques. It also discusses the most recent clinical studies and FDA-approved therapies, providing a modern perspective on the developing field of cancer immunotherapy and suggesting creative ways to improve treatment effectiveness while lowering toxicity. BACKGROUND: Cytokines are crucial in cancer immunotherapy for regulating immune responses and modifying the tumor microenvironment (TME). However, challenges with efficacy and safety have driven research into advanced delivery methods and combination therapies to enhance their therapeutic potential.
Collapse
Affiliation(s)
- Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - AmirHossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Omid Pourbagherian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
29
|
De Domenico P, Gagliardi F, Roncelli F, Snider S, Mortini P. Tumor-infiltrating and circulating B cells mediate local and systemic immunomodulatory mechanisms in Glioblastoma. J Neurooncol 2025; 172:527-548. [PMID: 40080248 DOI: 10.1007/s11060-025-04989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Glioblastoma (GBM) demonstrates extensive immunomodulatory mechanisms that challenge effective therapeutic interventions. These phenomena extend well beyond the tumor microenvironment (TME) and are reflected in the circulating immunophenotype. B lymphocytes (B cells) have received limited attention in GBM studies despite their emerging importance in mediating both local and systemic immune responses. Recent findings highlight the complex regulatory interactions between B cells and other immune cell populations, including tumor-infiltrating macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and other infiltrating lymphocytes (TILs). B cells are believed to hinder the efficacy of modern immunotherapy strategies focusing on T cells. METHODS This is a focused review of available evidence regarding B cells in GBM through January 2025. RESULTS Peripheral blood reflects a systemically dampened immune response, with sustained lymphopenia, increased plasma cells, and dysfunctional memory B cells. The tumor immune landscape is enriched in cells of B-lineage. Subsets of poorly characterized B regulatory cells (Bregs) populate the TME, developing their phenotype due to their proximity to MDSCs, TAMs, and tumoral cells. The Bregs inhibit CD8+ T activity and may have potential prognostic significance. CONCLUSION Understanding the role of B cells, how they are recruited, and their differentiation shifted towards an immunomodulatory role could inform better therapeutic strategies and unleash their full antitumoral potential in GBM.
Collapse
Affiliation(s)
- Pierfrancesco De Domenico
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy.
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Roncelli
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Silvia Snider
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
30
|
Kim H, Lee M, Han B, Kim J, Cho D, Doh J, Chung AJ. Advancing Allogeneic NK Cell Immunotherapy through Microfluidic Gene Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412544. [PMID: 40052491 PMCID: PMC12061328 DOI: 10.1002/advs.202412544] [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] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/26/2025] [Indexed: 05/10/2025]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer treatment, yet challenges such as manufacturing complexity, high costs, and safety concerns have spurred the development of alternatives like CAR-natural killer (NK) cell immunotherapies. CAR-NK cell therapies provide innate cytotoxicity with antigen-independent targeting, reducing safety risks while improving therapeutic efficacy. However, efficient genomic engineering and large-scale production of allogeneic NK cells remain significant obstacles. To address these challenges, a novel microfluidic gene delivery platform is developed, the Y-hydroporator, designed for allogeneic NK cell immunotherapy. This platform features a Y-shaped microchannel where NK cells experience rapid hydrodynamic stretching near the stagnation point, creating transient membrane discontinuities that facilitate the uptake of exogenous cargo. The Y-hydroporator achieves high delivery and transfection efficiency, processing ≈2 × 106 cells min-1 while maintaining long-term cell viability (>89%) and functionality. Using this platform, human primary CAR-NK cells and NKG2A-knockout NK cells are successfully generated by delivering anti-CD19 CAR mRNA and CRISPR/Cas9 ribonucleoproteins, respectively. These engineered NK cells demonstrated enhanced cytotoxicity, underscoring the potential of the Y-hydroporator as a transformative tool for advancing allogeneic NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Hyelee Kim
- Department of BioengineeringKorea UniversitySeoul02841Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH)Korea UniversitySeoul02841Republic of Korea
| | - Mujin Lee
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Bohwa Han
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Jinho Kim
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06355Republic of Korea
| | - Duck Cho
- Department of Health Sciences and TechnologySAIHSTSungkyunkwan UniversitySeoul06355Republic of Korea
- Department of Laboratory Medicine and GeneticsSamsung Medical CenterSungkyunkwan University School of MedicineSeoul03063Republic of Korea
| | - Junsang Doh
- Department of Materials Science and EngineeringSeoul National UniversitySeoul08826Republic of Korea
| | - Aram J. Chung
- Department of BioengineeringKorea UniversitySeoul02841Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH)Korea UniversitySeoul02841Republic of Korea
- School of Biomedical EngineeringKorea UniversitySeoul02841Republic of Korea
- MxT BiotechSeoul04785Republic of Korea
| |
Collapse
|
31
|
Ding M, Lu Y, Lei QK, Zheng YW. Advantages and challenges of ex vivo generation and expansion of human hematopoietic stem cells from pluripotent stem cells. Exp Hematol 2025; 145:104752. [PMID: 40086687 DOI: 10.1016/j.exphem.2025.104752] [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: 01/08/2025] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 03/16/2025]
Abstract
Hematopoietic stem cell transplantation (HSCT) is an essential and increasing therapeutic approach for treating conditions such as leukemia, lymphoma, and other blood cancers. However, its widespread use faces significant challenges, including limited donor availability, pathogens, and the risk of immune rejection. The emergence of pluripotent stem cells (PSCs) offers a potential solution to these challenges. By enabling the generation of hematopoietic stem cells (HSCs) and blood cells in vitro, PSCs open pathways to address the limitations of traditional HSC sources. Self-induced or gene-edited PSCs from patients may provide an accessible and personalized option for clinical applications. In this review, we examine the current protocols for differentiating PSCs into HSCs and blood cells, highlighting their benefits and shortcomings. Despite advancements in this field, two primary challenges persist: low differentiation efficiency and difficulties in isolating and enriching functional HSCs. These problems make it difficult to obtain HSCs for long-term survival. Thus, we propose innovative strategies and potential improvements including induction scheme optimization, reprogramming, and cell fate tracking. Future research should prioritize the development of efficient and reliable differentiation protocols for PSCs to obtain more functional HSCs. Additionally, establishing effective methods for enriching functional HSCs and blood cells will be critical for optimizing their use in clinical applications. These efforts hold the promise of overcoming current limitations and advancing the therapeutic potential of PSC-derived blood cells.
Collapse
Affiliation(s)
- Min Ding
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China; Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China; Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
| | - Yu Lu
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China; Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China; Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China
| | - Quan-Kai Lei
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun-Wen Zheng
- Institute of Regenerative Medicine, and Department of Dermatology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu, China; Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Chinese Academy of Medical Sciences, Tianjin, China; Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, and South China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, Guangdong, China; Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan.
| |
Collapse
|
32
|
Wali AF, Talath S, Sridhar SB, El-Tanani M, Rangraze IR. Endosialin-directed CAR-T cell therapy: A promising approach for targeting triple-negative breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167852. [PMID: 40318845 DOI: 10.1016/j.bbadis.2025.167852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/03/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
In triple-negative breast cancer, this review article explores into the utilization of Chimeric antigen receptor T-cell (CAR-T) cell therapy to target cells expressing endosialin. Even with all the new treatments available, breast cancer still kills more women than any other disease. Drug resistance and ineffective cancer cell targeting are two major problems with targeted medications, chemotherapy, and surgery. Among cancer treatments, CAR-T cell therapy stands out. To identify endosialin as a therapeutic target, it is essential to understand its molecular structure and its involvement in tumor angiogenesis and progression. An effective target for CAR-T cells is breast cancer, which overexpresses endosialin. The development of CARs that are specific to endosialin and the results of early trials are covered in relation to CAR-T cell therapy that targets endosialin. Perhaps the most effective cancer treatment is endosialin targeting, since it is expressed only in tumors and plays a crucial role in the course of cancer. This article reviews endosialin-directed CAR-T cell breast cancer treatments' safety and efficacy from current and completed clinical trials. Despite promising results, these trials reveal that clinical translation must overcome significant challenges. The report suggests further research and combination tactics to improve endosialin-targeted CAR-T cell treatment.
Collapse
Affiliation(s)
- Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Sathvik B Sridhar
- Department of Clinical Pharmacy & Pharmacology, RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohamed El-Tanani
- RAK College of Pharmacy, RAK Medical and Health Science University, Ras Al Khaimah 11172, United Arab Emirates
| | - Imran Rashid Rangraze
- Department of Internal Medicine, RAK Medical & Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| |
Collapse
|
33
|
Zhao Z, Cutmore LC, Baleeiro RB, Hartlebury JJ, Brown N, Chard-Dunmall L, Lemoine N, Wang Y, Marshall JF. The Combination of Oncolytic Virus and Antibody Blockade of TGF-β Enhances the Efficacy of αvβ6-Targeting CAR T Cells Against Pancreatic Cancer in an Immunocompetent Model. Cancers (Basel) 2025; 17:1534. [PMID: 40361460 PMCID: PMC12070938 DOI: 10.3390/cancers17091534] [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: 02/19/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES CAR T cell therapy, as a rapidly advancing immuno-oncology modality, has achieved significant success in the treatment of leukaemia and lymphoma. However, its application in solid tumours remains limited. The challenges include the heterogeneity of tumours, local immunosuppression, poor trafficking and infiltration, life-threatening toxicity and the lack of precise representative immunocompetent research models. Considering its typically dense and immunosuppressive tumour microenvironment (TME) and early metastasis, pancreatic ductal adenocarcinoma (PDAC) was employed as a model to address the challenges that hinder CAR T cell therapies against solid tumours and to expand immunotherapeutic options for advanced disease. METHODS A novel murine A20FMDV2 (A20) CAR T cell targeting integrin αvβ6 (mA20CART) was developed, demonstrating efficient and specific on-target cytotoxicity. The mA20CART cell as a monotherapy for orthotopic pancreatic cancer in an immunocompetent model demonstrated modest efficacy. Therefore, a novel triple therapy regimen, combining mA20CART cells with oncolytic vaccinia virus encoding IL-21 and a TGF-β-blocking antibody was evaluated in vivo. RESULTS The triple therapy improved overall survival, improved the safety profile of the CAR T cell therapy, attenuated metastasis and enhanced T cell infiltration. Notably, the potency of mA20CART was dependent on IL-2 supplementation. CONCLUSIONS This study presents an αvβ6-targeting murine CAR T cell, offering a novel approach to developing CAR T cell technologies for solid tumours and a potential adjuvant therapy for pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yaohe Wang
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Z.Z.); (L.C.C.); (R.B.B.); (J.J.H.); (N.B.); (L.C.-D.); (N.L.)
| | - John F. Marshall
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6BQ, UK; (Z.Z.); (L.C.C.); (R.B.B.); (J.J.H.); (N.B.); (L.C.-D.); (N.L.)
| |
Collapse
|
34
|
Xie L, Liu J, Yang Z, Chen H, Wang Y, Du X, Fu Y, Song P, Yu J. Microrobotic Swarms for Cancer Therapy. RESEARCH (WASHINGTON, D.C.) 2025; 8:0686. [PMID: 40302783 PMCID: PMC12038165 DOI: 10.34133/research.0686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Microrobotic swarms hold great promise for the revolution of cancer treatment. The coordination of miniaturized microrobots offers a unique approach to treating cancers at the cellular level with enhanced delivery efficiency and environmental adaptability. Prior studies have summarized the design, functionalization, and biomedical applications of microrobotic swarms. The strategies for actuation and motion control of swarms have also been introduced. In this review, we first give a detailed introduction to microrobot swarming. We then explore the design of microrobots and microrobotic swarms specifically engineered for cancer therapy, with a focus on tumor targeting, infiltration, and therapeutic efficacy. Moreover, the latest developments in active delivery methods and imaging techniques that enhance the precision of these systems are discussed. Finally, we categorize and analyze the various cancer therapies facilitated by functional microrobotic swarms, highlighting their potential to revolutionize treatment strategies for different cancer types.
Collapse
Affiliation(s)
- Leiming Xie
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Jinbo Liu
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Zhen Yang
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Hui Chen
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yibin Wang
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Xingzhou Du
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yongping Fu
- Department of Cardiovascular Medicine,
Affiliated Hospital of Shaoxing University, Shaoxing 312000, China
| | - Peng Song
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Jiangfan Yu
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| |
Collapse
|
35
|
Azmal M, Miah MM, Prima FS, Paul JK, Haque ASNB, Ghosh A. Advances and challenges in cancer immunotherapy: Strategies for personalized treatment. Semin Oncol 2025; 52:152345. [PMID: 40305928 DOI: 10.1016/j.seminoncol.2025.152345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Cancer immunotherapy has transformed oncology by harnessing the immune system to specifically target cancer cells, offering reduced systemic toxicity compared to traditional therapies. This review highlights key strategies, including adoptive cell transfer (ACT), immune checkpoint inhibitors, oncolytic viral (OV) therapy, monoclonal antibodies (mAbs), and mRNA-based vaccines. ACT reinfuses enhanced immune cells like tumor-infiltrating lymphocytes (TILs) to combat refractory cancers, while checkpoint inhibitors (eg, PD-1 and CTLA-4 blockers) restore T-cell activity. OV therapy uses engineered viruses (eg, T-VEC) to selectively lyse cancer cells, and advanced mAbs improve targeting precision. mRNA vaccines introduce tumor-specific antigens to trigger robust immune responses. Despite significant progress, challenges like immune-related side effects, high costs, and immunosuppressive tumor microenvironments persist. This review underscores the need for combination strategies and precision medicine to overcome these barriers and maximize the potential of immunotherapy in personalized cancer treatment.
Collapse
Affiliation(s)
- Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md Munna Miah
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Fatema Sultana Prima
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Anm Shah Newaz Been Haque
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| |
Collapse
|
36
|
Zhao T, You J, Wang C, Li B, Liu Y, Shao M, Zhao W, Zhou C. Cell-based immunotherapies for solid tumors: advances, challenges, and future directions. Front Oncol 2025; 15:1551583. [PMID: 40356763 PMCID: PMC12066282 DOI: 10.3389/fonc.2025.1551583] [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: 12/25/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Cell-based immunotherapies, including CAR-T, CAR-NK, and TCR-T therapies, represent a transformative approach to cancer treatment by offering precise targeting of tumor cells. Despite their success in hematologic malignancies, these therapies encounter significant challenges in treating solid tumors, such as antigen heterogeneity, immunosuppressive tumor microenvironments, limited cellular infiltration, off-target toxicity, and difficulties in manufacturing scalability. CAR-T cells have demonstrated exceptional efficacy in blood cancers but face obstacles in solid tumors, whereas CAR-NK cells offer reduced graft-versus-host disease but encounter similar barriers. TCR-T cells expand the range of treatable cancers by targeting intracellular antigens but require meticulous antigen selection to prevent off-target effects. Alternative therapies like TIL, NK, and CIK cells show promise but require further optimization to enhance persistence and overcome immunosuppressive barriers. Manufacturing complexity, high costs, and ensuring safety and efficacy remain critical challenges. Future advancements in gene editing, multi-antigen targeting, synthetic biology, off-the-shelf products, and personalized medicine hold the potential to address these issues and expand the use of cell-based therapies. Continued research and innovation are essential to improving safety, efficacy, and scalability, ultimately leading to better patient outcomes.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Jinping You
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Congyue Wang
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Bo Li
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Yuhan Liu
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, China
| | - Mingjia Shao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Wuyang Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Chuang Zhou
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| |
Collapse
|
37
|
Mougiakakos D, Meyer EH, Schett G. CAR T cells in autoimmunity: game changer or stepping stone? Blood 2025; 145:1841-1849. [PMID: 39700499 DOI: 10.1182/blood.2024025413] [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: 09/12/2024] [Revised: 11/19/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
ABSTRACT The advent of chimeric antigen receptor (CAR) T cells has revolutionized the treatment landscape for hematologic malignancies, and emerging evidence suggests their potential in autoimmune diseases (AIDs). This article evaluates the early successes and future implications of B-cell-targeting CAR T-cell therapy in AIDs. Initial applications, particularly in refractory systemic lupus erythematosus, have demonstrated significant and durable clinical remissions, with accompanying evaluation of the immune system suggesting a so-called "reset" of innate inflammation and adaptive autoimmunity. This has generated widespread interest in expanding this therapeutic approach. CAR T cells offer unique advantages over other treatment modalities, including very deep B-cell depletion and unique therapeutic activity within inflamed tissues and associated lymphoid structures. However, the field must address key concerns, including long-term toxicity, particularly the risk of secondary malignancies, and future accessibility given the higher prevalence of AIDs compared with malignancies. Technological advances in cell therapy, such as next-generation CAR T cells, allogeneic off-the-shelf products, and alternative cell types, such as regulatory CAR T cells, are being explored in AIDs to improve efficacy and safety. In addition, bispecific antibodies are emerging as potential alternatives or complements to CAR T cells, potentially offering comparable efficacy without the need for complex logistics, lymphodepletion, and the risk of insertional mutagenesis. As the field evolves, cellular therapists will play a critical role in the multidisciplinary teams managing these complex cases. The transformative potential of CAR T cells in AIDs is undeniable, but careful consideration of safety, efficacy, and implementation is essential as this novel therapeutic approach moves forward.
Collapse
Affiliation(s)
- Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Otto von Guericke University, Magdeburg, Germany
| | - Everett H Meyer
- Cellular Immune Tolerance Program, Blood and Marrow Transplantation and Cellular Therapy Division, Stanford School of Medicine, Stanford University, Stanford, CA
| | - Georg Schett
- Department of Medicine 3, Rheumatology and Immunology, Friedrich-Alexander University, Erlangen, Germany
| |
Collapse
|
38
|
Werner J, Lee AG, Zhang C, Abelson S, Xirenayi S, Rivera J, Yousuf K, Shin H, Patiño-Escobar B, Bachl S, Mandal K, Barpanda A, Ramos E, Izgutdina A, Chaudhuri S, Temple WC, Bhatnagar S, Dardis JK, Meyer J, Morales C, Meshinchi S, Loh ML, Braun B, Tasian SK, Wiita AP, Stieglitz E. Cellular immunotherapy targeting CLL-1 for juvenile myelomonocytic leukemia. Nat Commun 2025; 16:3804. [PMID: 40268927 PMCID: PMC12019388 DOI: 10.1038/s41467-025-59040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a myeloproliferative disorder that predominantly affects infants and young children. Hematopoietic stem cell transplantation (HSCT) is standard of care, but post-HSCT relapse is common, highlighting the need for innovative therapies. While adoptive immunotherapy with chimeric antigen receptor (CAR) T cells has improved outcomes for patients with advanced lymphoid malignancies, it has not been comprehensively evaluated in JMML. In the present study, we use bulk and single-cell RNA sequencing, mass spectrometry, and flow cytometry to identify overexpression of CLL-1 (encoded by CLEC12A) on the cell surface of cells from patients with JMML. We develop immunotherapy with CLL-1 CAR T cells (CLL1CART) for preclinical testing and report in vitro and in vivo anti-leukemia activity. Notably, CLL1CART reduce the number of leukemic stem cells and serial transplantability in vivo. These preclinical data support the development and clinical investigation of CLL-1-targeting immunotherapy in children with relapsed/refractory JMML.
Collapse
MESH Headings
- Humans
- Leukemia, Myelomonocytic, Juvenile/therapy
- Leukemia, Myelomonocytic, Juvenile/immunology
- Leukemia, Myelomonocytic, Juvenile/genetics
- Leukemia, Myelomonocytic, Juvenile/pathology
- Animals
- Mice
- Immunotherapy, Adoptive/methods
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Receptors, Mitogen/genetics
- Receptors, Mitogen/immunology
- Receptors, Mitogen/metabolism
- Female
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Male
- Xenograft Model Antitumor Assays
- Child
- Mice, SCID
- Neoplastic Stem Cells/immunology
- Infant
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Juwita Werner
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
- Department of Pediatric Hematology and Oncology and Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Alex G Lee
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Chujing Zhang
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Sydney Abelson
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Sherin Xirenayi
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Jose Rivera
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Khadija Yousuf
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Hanna Shin
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | | | - Stefanie Bachl
- Department of Medicine, University of California, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Kamal Mandal
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Department of Animal Biotechnology, Gujarat Biotechnology University, Gandhinagar, India
| | - Abhilash Barpanda
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Emilio Ramos
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Adila Izgutdina
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Sibapriya Chaudhuri
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - William C Temple
- Division of Pediatric Allergy, Immunology, and Bone Marrow Transplant, University of California, San Francisco, CA, USA
- Division of Pediatric Oncology, University of California, San Francisco, CA, USA
| | - Shubhmita Bhatnagar
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jackson K Dardis
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julia Meyer
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Carolina Morales
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Soheil Meshinchi
- Clinical Research Division, Department of Pediatrics, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mignon L Loh
- Seattle Children's Hospital, The Ben Towne Center for Childhood Cancer Research, University of Washington, Seattle, WA, USA
| | - Benjamin Braun
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA
| | - Sarah K Tasian
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Arun P Wiita
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Elliot Stieglitz
- Department of Pediatrics, Benioff Children's Hospitals, University of California, San Francisco, CA, USA.
| |
Collapse
|
39
|
Zhong W, Zhao Z, Fang X, Sun J, Wei Y, Li F, Han B, Jin C. Constructing a neural network model based on tumor-infiltrating lymphocytes (TILs) to predict the survival of hepatocellular carcinoma patients. PeerJ 2025; 13:e19351. [PMID: 40292102 PMCID: PMC12032962 DOI: 10.7717/peerj.19351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary liver cancer worldwide, and early pathological diagnosis is crucial for formulating treatment plans. Despite the widespread attention to pathology in the treatment of HCC patients, a large amount of information contained in pathological images is often overlooked. Methods We retrospectively collected clinical data and pathological slide images from (a) 331 HCC patients at Qingdao University Affiliated Hospital between January 2013 and December 2016 and (b) 180 HCC patients from The Cancer Genome Atlas (TCGA). After data screening, precise quantification of various cell types was achieved using QuPath software. Key factors related to the survival prognosis of pathologically confirmed HCC patients were identified through Cox regression and neural network models, and potential therapeutic targets were screened. Results Our study showed that tumour-infiltrating lymphocytes (TILs) had a protective effect. We quantified the TILs index by machine learning and built a neural network model to predict the prognostic risk of patients (ROC = 0.836 for training set ROC validation set). 95% CI [0.7688-0.896], and there was a significant difference in prognosis in the high-low risk group predicted by the model (p = 2.6e-18, HR = 0.18, 95% CI [0.12-0.27], and TNFSF4 was identified as a possible immunotherapy target. Conclusion This study included a total of 511 patients, divided into a training cohort of 331 cases (from Qingdao University Hospital between January 2013 and December 2016) and a validation cohort of 180 cases (TCGA). The results revealed that tumor-infiltrating lymphocytes (TILs) have a protective effect and successfully predicted the survival risk of liver cancer patients using machine learning and neural network technology. The discovery of TNFSF4 provides a new potential target for immunotherapy.
Collapse
Affiliation(s)
- Wenqing Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ziyin Zhao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xin Fang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Jingyi Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanbing Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fengda Li
- Department of Hepatobiliary Surgery, Gao mi People’s Hospital, Weifang, Shandong, China
| | - Bing Han
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng Jin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
40
|
Pati NB, Jin Y, Kumar S, Kyte JA, Ciosk R. Thermal potentiation improves IFN-γ production but not cytotoxicity in human CAR-T cells. BMC Res Notes 2025; 18:192. [PMID: 40270044 PMCID: PMC12020131 DOI: 10.1186/s13104-025-07249-5] [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: 08/19/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE Body temperature plays an important role in cancer, with febrile temperature generally associated with improved cancer resistance. In murine models, this resistance has been linked to the cytotoxic T cells, whose differentiation into cancer-killing effector cells is poor at lower but robust at elevated temperatures. If conserved, temperature-mediated potentiation of patient-derived T cells could be implemented to improve the existing cancer treatments, including the chimeric antigen receptor T-cell therapy (CAR T-cell therapy). Here, we tested this possibility using human STEAP1 CAR-T cells developed to target prostate cancer. RESULTS In mice, transient exposure to febrile temperature (39-40 ºC) increases the production of IFN-γ and the cancer-killing ability of CD8 + T cells. Using a similar temperature treatment, we observed elevated levels of IFN-γ also in the human CAR-T cells. However, these cells displayed no improvement in their ability to kill cancer cells. Although we cannot discount the possibility that alternative protocols might lead to other outcomes, our findings suggest that incorporating thermal potentiation into existing protocols of CAR-T cell therapy may be more complicated than anticipated.
Collapse
Affiliation(s)
- Niladri Bhusan Pati
- Department of Biosciences, University of Oslo, Oslo, Norway
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Yixin Jin
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Suman Kumar
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| | - Rafal Ciosk
- Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
41
|
Esmaeilzadeh A, Hadiloo K, Yaghoubi S, Makoui MH, Mostanadi P. State of the art in CAR-based therapy: In vivo CAR production as a revolution in cell-based cancer treatment. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01056-7. [PMID: 40261561 DOI: 10.1007/s13402-025-01056-7] [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: 01/24/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Chimeric antigen receptor (CAR) therapy has successfully treated relapsed/refractory hematological cancers. This strategy can effectively target tumor cells. However, despite positive outcomes in clinical applications, challenges remain to overcome. These hurdles pertain to the production of the drugs, solid tumor resistance, and side effects related to the treatment. Some cases have been missed during the drug preparation due to manufacturing issues, prolonged production times, and high costs. These challenges mainly arise from the in vitro manufacturing process, so reevaluating this process could minimize the number of missed patients. The immune cells are traditionally collected and sent to the laboratory; after several steps, the cells are modified to express the CAR gene before being injected back into the patient's body. During the in vivo method, the CAR gene is introduced to the immune cells inside the body. This allows for treatment to begin sooner, avoiding potential failures in drug preparation and the associated high costs. In this review, we will elaborate on the production and treatment process using in vivo CAR, examine the benefits and challenges of this approach, and ultimately present the available solutions for incorporating this treatment into clinical practice.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Pficell R&D Canadian Institution & Corporation, Profound Future Focused Innovative Cell and Gene Therapy, Pficell Canadian Institution and Corporation, Ontario, Canada.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Kaveh Hadiloo
- Pficell R&D Canadian Institution & Corporation, Profound Future Focused Innovative Cell and Gene Therapy, Pficell Canadian Institution and Corporation, Ontario, Canada
- Department of Surgery, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin, Iran
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
| | - Sara Yaghoubi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Parsa Mostanadi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan, Iran
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
42
|
Ma S, Yu J, Caligiuri MA. Natural killer cell-based immunotherapy for cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf036. [PMID: 40246292 DOI: 10.1093/jimmun/vkaf036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 04/19/2025]
Abstract
Natural killer (NK) cells are emerging as a promising tool for cancer immunotherapy due to their innate ability to selectively recognize and eliminate cancer cells. Over the past 3 decades, strategies to harness NK cells have included cytokines, small molecules, antibodies, and the adoptive transfer of autologous or allogeneic NK cells, both unmodified and genetically engineered. Despite favorable safety profiles in clinical trials, challenges such as limited in vivo persistence, exhaustion, and the suppressive tumor microenvironment continue to hinder their efficacy and durability. This review categorizes NK cell-based therapies into 3 major approaches: (i) cellular therapies, including unmodified and chimeric antigen receptor-engineered NK cells; (ii) cytokine-based strategies such as interleukin-2 and interleukin-15 derivatives; and (iii) antibody-based therapies, including immune checkpoint inhibitors and NK cell engagers. We highlight these advancements, discuss current limitations, and propose strategies to optimize NK cell-based therapies for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| | - Jianhua Yu
- Division of Hematology and Oncology, Department of Medicine, School of Medicine, University of California, Irvine, CA, United States
- Institute for Precision Cancer Therapeutics and Immuno-Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA, United States
- Clemons Family Center for Transformative Cancer Research, University of California, Irvine, Irvine, CA, United States
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, United States
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, United States
- City of Hope Comprehensive Cancer Center, Los Angeles, CA, United States
| |
Collapse
|
43
|
Ebrahimiyan H, Sayadmanesh A, Hesaraki M, Ebrahimi M, Baharand H, Basiri M. Engineering CD3 subunits with endoplasmic reticulum retention signal facilitates allogeneic CAR T cell production. Int Immunopharmacol 2025; 152:114412. [PMID: 40056516 DOI: 10.1016/j.intimp.2025.114412] [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: 12/19/2024] [Revised: 02/18/2025] [Accepted: 03/02/2025] [Indexed: 03/10/2025]
Abstract
The success of autologous CAR T cell therapies has driven interest in developing off-the-shelf allogeneic CAR T cells as a scalable and readily available option for broader patient access. Most of the current approaches involve the knockout of T cell receptor (TCR) subunits via genome editing for preventing graft-versus-host disease (GvHD). However, clinical translation of these methods faces challenges due to manufacturing complexities and emerging safety concerns like unintended long deletions and chromosomal loss. In this study, we explored an alternative approach by engineering synthetic CD3 subunits containing an endoplasmic reticulum retention (ERR) signal to suppress TCR surface expression by disrupting its trafficking to the plasma membrane. We screened multiple CD3-ERR candidate designs to identify the construct with the highest efficacy in TCR downregulation. The selected candidate, CD3ζ-ERR, was further characterized, demonstrating its ability to minimize TCR-mediated activation and alloreactivity without affecting T cell phenotype, cell cycle and cytokine-induced expansion. Subsequent assays revealed that CD3ζ-ERR CD19 CAR T cells retained their CAR-mediated cytotoxic function against CD19+ malignant cells. This study presents an alternative approach for TCR downregulation that circumvents genome editing. By using a transgene compatible with conventional viral vector delivery, this approach holds promise for scalable clinical-grade manufacturing of allogeneic CAR T cell therapies.
Collapse
MESH Headings
- Humans
- CD3 Complex/genetics
- CD3 Complex/immunology
- CD3 Complex/metabolism
- Endoplasmic Reticulum/metabolism
- Immunotherapy, Adoptive/methods
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Antigens, CD19/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Hamidreza Ebrahimiyan
- Department of Applied Cell Sciences, Faculty of Basic Sciences and Advanced Medical Technologies, Royan Institute, ACECR, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Sayadmanesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of regenerative medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
44
|
Blanchard T, Faridi P, Xu C, Bear AS, Rasool RU, Huang G, Lim TCC, Ayala R, Gabunia K, Ji M, Posey AD, Scholler J, Asangani IA, Purcell AW, Linette GP, June CH, Carreno BM. LOXHD1 is an oncofusion-regulated antigen of ewing sarcoma. Sci Rep 2025; 15:13007. [PMID: 40234527 PMCID: PMC12000433 DOI: 10.1038/s41598-025-96877-9] [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: 11/22/2024] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Ewing Sarcoma (EwS) is a rare pediatric malignancy characterized by a unique t(11:22) (q24;q12) translocation resulting in the pathognomonic EWSR1::FLI1 fusion. Recent reports indicate that the EWSR1::FLI1 oncofusion drives aberrant expression of numerous transcripts, including Lipoxygenase Homology Domains 1 (LOXHD1). Given its highly restricted protein expression pattern and role in EwS tumorigenesis and metastasis, LOXHD1 may serve as a novel immunotherapeutic target in this malignancy. LOXHD1 immunogenic epitopes restricted to HLA-A*02:01 allowed for the isolation of a high avidity αβTCR. LOXHD1-specific TCR engineered CD8+ T cells conferred cytotoxic activity against a panel of HLA-A*02:01+ EwS tumor cell lines and adoptive transfer led to tumor eradication in a mouse xenograft model of EwS. This study nominates LOXHD1 as an oncofusion regulated, non-mutated tumor associated antigen (TAA) with expression limited to inner hair cells of the cochlea, adult testis, and EwS.
Collapse
MESH Headings
- Sarcoma, Ewing/immunology
- Sarcoma, Ewing/genetics
- Sarcoma, Ewing/pathology
- Sarcoma, Ewing/therapy
- Sarcoma, Ewing/metabolism
- Humans
- Animals
- Mice
- Cell Line, Tumor
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Oncogene Proteins, Fusion/genetics
- CD8-Positive T-Lymphocytes/immunology
- RNA-Binding Protein EWS/genetics
- Proto-Oncogene Protein c-fli-1/genetics
Collapse
Affiliation(s)
- Tatiana Blanchard
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pouya Faridi
- Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3168, Australia
- Monash Proteomics and Metabolomics Platform, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Chong Xu
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adham S Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Reyaz Ur Rasool
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, BRBII/III, 421 Curie Boulevard, Philadelphia, PA, 19104, USA
| | - Grace Huang
- Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3168, Australia
| | - Terry C C Lim
- Centre for Cancer Research, Hudson Institute of Medical Research, Monash University, Clayton, VIC, 3168, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, 3168, Australia
- Monash Proteomics and Metabolomics Platform, Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Rochelle Ayala
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Khatuna Gabunia
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mei Ji
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Avery D Posey
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - John Scholler
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Irfan A Asangani
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, BRBII/III, 421 Curie Boulevard, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Gerald P Linette
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Nyong E, Kurebayashi Y, Asiedu KO, Choyke PL, Sato N. Intracellular Protein Binding of Zr-89 Oxine Cell Labeling for PET Cell Tracking Studies. Pharmaceutics 2025; 17:518. [PMID: 40284513 PMCID: PMC12030610 DOI: 10.3390/pharmaceutics17040518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/12/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: 89Zr-oxine is an ex vivo cell labeling agent that enables cells to be tracked in vivo by positron emission tomography (PET) over a period of up to two weeks. To better understand where 89Zr-oxine binds within cellular components, factors affecting labeling and intracellular distribution of 89Zr were examined. Methods: Mouse primary T cells, natural killer cells, dendritic cells, and monocytes, and cell lines EL4 (mouse lymphoma), DC2.4 (mouse dendritic cell), Kit225K6 (human T cell leukemia) and MC38 (mouse colon adenocarcinoma) were labeled with 89Zr-oxine or 111In-oxine and protein binding within the cellular compartments, the labeling thresholds, and radioactivity retention were subsequently determined. Results: Cell incorporation of 89Zr-oxine (27.8-71.8 kBq/106 cells) positively correlated with cellular size and protein mass. Most (>97%) 89Zr was protein-bound and primarily localized in the cytoplasm, membrane, and nuclear fractions (>81%) with distribution patterns varying by cell type. By contrast, 111In-oxine showed lower protein-binding activity of approximately 59-65%, with 62-65% of 111In localized in the cytoplasm. Autoradiography of electrophoresed subcellular fractionated cell samples indicated stable binding by 89Zr-oxine to proteins in all subcellular fractions but unstable protein binding by 111In. Saturation studies showed that 89Zr-oxine labeling was saturable, and further labeling reduced cellular retention. Biodistribution of dendritic cells labeled with either 89Zr-oxine or 111In-oxine indicated greater retention of 89Zr in the labeled cells in vivo than 111In. Conclusions: 89Zr-oxine stably binds many intracellular proteins and shows much higher and more stable protein binding than 111In-oxine. Intracellular protein binding of 89Zr accounts for the ability of 89Zr-oxine labeling to successfully track cells in vivo long-term on PET.
Collapse
Affiliation(s)
- Emmanuel Nyong
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.N.); (Y.K.); (K.O.A.); (P.L.C.)
- Department of Surgery, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Yutaka Kurebayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.N.); (Y.K.); (K.O.A.); (P.L.C.)
- Department of Pathology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kingsley O. Asiedu
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.N.); (Y.K.); (K.O.A.); (P.L.C.)
- Department of Radiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.N.); (Y.K.); (K.O.A.); (P.L.C.)
| | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.N.); (Y.K.); (K.O.A.); (P.L.C.)
| |
Collapse
|
46
|
Gao P, Zhang Y, Ma J, Zhang Y. Immunotherapy in chronic lymphocytic leukemia: advances and challenges. Exp Hematol Oncol 2025; 14:53. [PMID: 40211406 PMCID: PMC11984025 DOI: 10.1186/s40164-025-00644-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized as a clonal proliferation of mature B lymphocytes with distinct immunophenotypic traits, predominantly affecting the middle-aged and elderly population. This condition is marked by an accumulation of lymphocytes within the peripheral blood, bone marrow, spleen, and lymph nodes. The associated immune dysregulation predisposes CLL patients to a higher risk of secondary malignancies and infections, which significantly contribute to morbidity and mortality rates. The advent of immunotherapy has revolutionized the prognosis of CLL, advancing treatment modalities and offering substantial benefits to patient outcomes. This review endeavors to synthesize and scrutinize the efficacy, merits, and limitations of the current immunotherapeutic strategies for CLL. The aim is to inform the selection of optimal treatment regimens tailored to individual patient needs. Furthermore, the review juxtaposes various therapeutic combinations to elucidate the comparative advantages of each approach, with the ultimate objective of enhancing patient prognosis and quality of life.
Collapse
Affiliation(s)
- Pan Gao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Yang Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Jun Ma
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
| |
Collapse
|
47
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [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: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
48
|
Limsakul P, Srifa P, Huang Z, Zhu L, Wu Y, Charupanit K. Immunomodulatory Effects of Curcumin on CAR T-Cell Therapy. Antioxidants (Basel) 2025; 14:454. [PMID: 40298832 PMCID: PMC12024323 DOI: 10.3390/antiox14040454] [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: 03/06/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized the treatment of hematological malignancies, demonstrating high efficacy in targeting and eliminating cancer cells. However, its clinical application can be associated with the risk of acute adverse effects, including cytokine release syndrome (CRS), a severe inflammatory response caused by excessive cytokine production. While anti-cytokine therapies are available to manage CRS, additional strategies are needed to optimize CAR T-cell efficacy with reduced toxicities. Curcumin, a bioactive polyphenol known for its anti-inflammatory and antioxidant properties, represents a promising adjunct for CAR T-cell therapy. In this study, we investigated the effects of curcumin on anti-CD19 CAR T-cells in vitro. Our results show that curcumin enhances the cytotoxic activity of CAR T-cells against Nalm-6, a B-cell acute lymphoblastic leukemia model, while reducing the production of pro-inflammatory cytokines, including IL-2 and IFN-γ. To explore its underlying mechanisms, network pharmacology and molecular docking analyses were performed, which revealed that curcumin interacts with key signaling pathways involved in T-cell activation and cytokine regulation. These findings support the potential of curcumin as a therapeutic adjunct to improve CAR T-cell efficacy while mitigating inflammatory toxicity.
Collapse
Affiliation(s)
- Praopim Limsakul
- Division of Physical Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand;
- Center of Excellence for Trace Analysis and Biosensor (TAB-CoE), Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Pemikar Srifa
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| | - Ziliang Huang
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; (Z.H.); (L.Z.)
| | - Linshan Zhu
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA; (Z.H.); (L.Z.)
| | - Yiqian Wu
- National Biomedical Imaging Center, College of Future Technology, Peking University, Beijing 100871, China;
| | - Krit Charupanit
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand;
| |
Collapse
|
49
|
Xie D, Liu Y, Xu F, Dang Z, Li M, Zhang Q, Dang Z. Immune microenvironment and immunotherapy in hepatocellular carcinoma: mechanisms and advances. Front Immunol 2025; 16:1581098. [PMID: 40242773 PMCID: PMC12000014 DOI: 10.3389/fimmu.2025.1581098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality globally. The tumor microenvironment (TME) plays a pivotal role in HCC progression, characterized by dynamic interactions between stromal components, immune cells, and tumor cells. Key immune players, including tumor-associated macrophages (TAMs), tumor-infiltrating lymphocytes (TILs), cytotoxic T lymphocytes (CTLs), regulatory T cells (Tregs), MDSCs, dendritic cells (DCs), and natural killer (NK) cells, contribute to immune evasion and tumor progression. Recent advances in immunotherapy, such as immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell therapy (ACT), and combination therapies, have shown promise in enhancing anti-tumor responses. Dual ICI combinations, ICIs with molecular targeted drugs, and integration with local treatments or radiotherapy have demonstrated improved outcomes in HCC patients. This review highlights the evolving understanding of the immune microenvironment and the therapeutic potential of immunotherapeutic strategies in HCC management.
Collapse
Affiliation(s)
- Dong Xie
- Diagnosis and Treatment Center for Digestive Diseases of Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Yang Liu
- College of Traditional Chinese Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Fangbiao Xu
- Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhibo Dang
- Diagnosis and Treatment Center for Digestive Diseases of Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Mengge Li
- Diagnosis and Treatment Center for Digestive Diseases of Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Qinsheng Zhang
- Diagnosis and Treatment Center for Digestive Diseases of Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, China
| | - Zhongqin Dang
- Diagnosis and Treatment Center for Digestive Diseases of Henan Province Hospital of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
50
|
Rezazadeh‐Gavgani E, Majidazar R, Lotfinejad P, Kazemi T, Shamekh A. Immune Checkpoint Molecules: A Review on Pathways and Immunotherapy Implications. Immun Inflamm Dis 2025; 13:e70196. [PMID: 40243372 PMCID: PMC12004596 DOI: 10.1002/iid3.70196] [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: 05/07/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Today, treating cancer patients with monoclonal antibodies (mAbs), by targeting immune checkpoints, is one of the most outstanding immunotherapeutic methods. Immune checkpoints are special molecules having regulatory role in immune system responses. Once these molecules are presented on cancer cells, these cells will be capable of evading the immune system through their own specific pathways. This Evasion can be prevented by counterbalancing immune system responses with immune checkpoints related antibodies. AIMS The current study aimed to highlight immunotherapy and its methods, describe the immune checkpoints pathways, outline the immune checkpoint inhibitors (ICIs), and recent advances in this field, and sketch an outlook on the best treatment options for the most prevalent cancers. MATERIALS & METHODS This research implemented a narrative review method. A comprehensive literature review on the history, molecular and cellular biology, and the clinical aspects of immune checkpoint molecules was performed to illustrate the pathways involved in various cancers. Also, currently-available and future potential immunotherapies targeting these pathways were extracted from the searched studies. RESULTS The immune checkpoint family consists of many molecules, including CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and TIGIT. Attempts to modify these molecules in cancer treatment led to the development of therapeutic monoclonal antibodies. Most of these antibodies have entered clinical studies and some of them have been approved by the Food and Drug Administration (FDA) to be used in cancer patients' treatment plans. DISCUSSION With these novel treatments and the combination therapies they offer, there is also hope for better treatment outcomes for the previously untreatable metastatic cancers. In spite of the beneficial aspects of immune checkpoint therapy, similar to other treatments, they may cause side effects in some patients. Therefore, more studies are needed to reduce the probable side effects and uncover their underlying mechanism. CONCLUSION Based on the data shown in this review, there is still a lack of knowledge about the complete properties of ICIs and the possible combination therapies that we may be able to implement to achieve a better treatment response in cancer patients.
Collapse
Affiliation(s)
| | - Reza Majidazar
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Parisa Lotfinejad
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Tohid Kazemi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Ali Shamekh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| |
Collapse
|