1
|
Gao Y, He J, Wang J, Xu H, Ma L. Chimeric antigen receptor T cell immunotherapy for gynecological malignancies. Crit Rev Oncol Hematol 2025; 209:104680. [PMID: 40024355 DOI: 10.1016/j.critrevonc.2025.104680] [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: 11/17/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Gynecologic malignancies pose a serious threat to women's health worldwide. Although immunotherapy has significantly revolutionized cancer treatment strategies, effective therapeutic options for recurrent or advanced gynecologic malignancies are still deficient, posing significant challenges to clinical therapy. Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable efficacy in treating hematologic malignancies, marking a significant change in the oncology treatment paradigm. However, despite the gradual increase in CAR T cell therapy used in treating solid tumors in recent years, its efficacy in treating gynecologic malignancies still needs further validation. This review will thoroughly examine CAR-T cell engineering and its mechanism of action on specific antigens associated with gynecologic malignancies, systematically assess the current application of CAR T cell therapy in gynecologic tumors and the advancements in clinical trials, and discuss the significant challenges and corresponding strategies, thereby offering a scientific foundation and guidance for future research in this area.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China.
| | - Jing He
- Department of Emergency, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China
| | - Jing Wang
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China
| | - Haiou Xu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Lin Ma
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China.
| |
Collapse
|
2
|
Sobti A, Skinner H, Wilke CT. Predictors of Radiation Resistance and Novel Radiation Sensitizers in Head and Neck Cancers: Advancing Radiotherapy Efficacy. Semin Radiat Oncol 2025; 35:224-242. [PMID: 40090749 DOI: 10.1016/j.semradonc.2025.02.008] [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: 10/15/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/18/2025]
Abstract
Radiation resistance in head and neck squamous cell carcinoma (HNSCC), driven by intrinsic and extrinsic factors, poses a significant challenge in radiation oncology. The key contributors are tumor hypoxia, cancer stem cells, cell cycle checkpoint activation, and DNA repair processes (homologous recombination and non-homologous end-joining). Genetic modifications such as TP53 mutations, KRAS mutations, EGFR overexpression, and abnormalities in DNA repair proteins like BRCA1/2 additionally affect radiation sensitivity. Novel radiosensitizers targeting these pathways demonstrate the potential to overcome resistance. Hypoxia-activated drugs and gold nanoparticles enhance the efficacy of radiotherapy and facilitate targeted distribution. Integrating immunotherapy, especially immune checkpoint inhibitors, with radiation therapy, enhances anti-tumor responses and reduces resistance. Epigenetic alterations, such as DNA methylation and histone acetylation, significantly influence radiation response, with the potential for sensitization through histone deacetylase inhibitors and non-coding RNA regulators. Metabolic changes linked to glucose, lipid, and glutamine metabolism influence radiosensitivity, uncovering new targets for radiosensitization. Human papillomavirus (HPV)-associated malignancies exhibit increased radiosensitivity relative to other tumors due to impaired DNA repair mechanisms and heightened immunogenicity. Furthermore, understanding the interplay between HPV oncoproteins and p53 functionality can enhance treatment strategies for HPV-related cancers. Using DNA damage response inhibitors (PARP, ATM/ATR), cell cycle checkpoint inhibitors (WEE1, CHK1/2), and hypoxia-targeted agents as radiosensitizing strategies exhibit considerable promise. Immunomodulatory approaches, including PD-1 and CTLA-4 inhibitors in conjunction with radiation, enhance anti-tumor immunity. Future directions emphasize personalized radiation therapy using genetics, sophisticated medication delivery systems, adaptive radiotherapy, and real-time monitoring. These integrated strategies seek to diminish radiation resistance and improve therapeutic efficacy in HNSCC.
Collapse
Affiliation(s)
- Aastha Sobti
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Heath Skinner
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA
| | - Christopher T Wilke
- Department of Radiation Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA..
| |
Collapse
|
3
|
Laverdure E, Mollica L, Ahmad I, Cohen S, Lachance S, Veilleux O, Bernard M, Marchand EL, Delisle JS, Bernard L, Boileau M, Petrella T, Pilon SJ, Bouchard P, Roy DC, Busque L, Fleury I. Enhancing CAR-T Efficacy in Large B-Cell Lymphoma with Radiation Bridging Therapy: A Real-World Single-Center Experience. Curr Oncol 2025; 32:173. [PMID: 40136377 PMCID: PMC11941054 DOI: 10.3390/curroncol32030173] [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: 01/05/2025] [Revised: 02/25/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
One challenge of chimeric antigen receptor T-cell therapy (CAR-T) for relapsed or refractory large B-cell lymphoma (LBCL) is achieving disease control during manufacturing. We report real-word outcomes of 100 patients treated with axicabtagene ciloleucel (axi-cel, n = 50) or tisagenlecleucel (tisa-cel, n = 50) at our center. Most patients received bridging therapy (BT) with 48 undergoing radiation BT (RBT) and 32 receiving systemic BT (SBT). The best overall response rate (ORR) was 84% (78% complete response (CR)) for axi-cel and 60% (42% CR) for tisa-cel. At a median follow-up of 16 months, 12-month progression-free survival (PFS) and overall survival (OS) were 72% and 82% for axi-cel, compared to 35% and 57% for tisa-cel. By the bridging approach, 12-month PFS was 60% with RBT, 59% without BT and 35% with SBT (p = 0.06). Notably, axi-cel patients without lymphoma progression during manufacturing (n = 24) achieved 12-month PFS and OS rates of 91% and 96%, respectively. Axi-cel was associated with more cytokine release syndrome (92% vs. 66%, p = 0.003) and neurotoxicity (all-grade 56% vs. 10%, p < 0.001, grade ≥ 328% vs. 4%, p = 0.002). Multivariate analysis identified RBT as independently associated with improved PFS (HR 0.46, 95% CI 0.22-0.96). Pending prospective validation, RBT shows promise for improving CAR-T outcomes in LBCL.
Collapse
MESH Headings
- Humans
- Middle Aged
- Male
- Female
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/radiotherapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Aged
- Immunotherapy, Adoptive/methods
- Adult
- Biological Products/therapeutic use
- Treatment Outcome
- Aged, 80 and over
- Receptors, Antigen, T-Cell/therapeutic use
- Antigens, CD19/therapeutic use
Collapse
Affiliation(s)
- Eva Laverdure
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Department of Hemato-Oncology, Hôpital Fleurimont, Centre Hospitalier Universitaire de Sherbrooke, CIUSSS de l’Estrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Luigina Mollica
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Imran Ahmad
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Sandra Cohen
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Silvy Lachance
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Olivier Veilleux
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Maryse Bernard
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Radiation Therapy, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Eve-Lyne Marchand
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Radiation Therapy, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Jean-Sébastien Delisle
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Lea Bernard
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Mélissa Boileau
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Tony Petrella
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Pathology, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Sarah-Jeanne Pilon
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
- Department of Pathology, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Philippe Bouchard
- Department of Pharmacy, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, Montréal, QC H1T 2M4, Canada
| | - Denis-Claude Roy
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Lambert Busque
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| | - Isabelle Fleury
- Department of Medicine, Institut Universitaire d’Hémato-Oncologie et de Thérapie Cellulaire, Hôpital Maisonneuve-Rosemont, CIUSSS de l’Est-de-l’Île-de-Montréal, l, Montréal, QC HIT 2M4, Canada; (E.L.); (L.M.); (I.A.); (S.C.); (S.L.); (O.V.); (J.-S.D.); (D.-C.R.)
- Faculty of Medicine, Université de Montréal, Montréal, QC H2V 0B3, Canada; (M.B.); (E.-L.M.); (T.P.); (S.-J.P.)
| |
Collapse
|
4
|
Zhang X, Shi C, Liu Q, Zhong Y, Zhu L, Zhao Y. Combination of adenosine blockade and ferroptosis for photo-immunotherapy of triple negative breast cancer with aptamer-modified copper sulfide. J Mater Chem B 2025; 13:2504-2519. [PMID: 39834279 DOI: 10.1039/d4tb02125h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Combination of immunotherapy and photothermal therapy (PTT) provides a promising therapeutic performance for tumors. However, it still faces negative feedback from suppressive factors such as adenosine. Herein, we developed a new nanodrug that can combine adenosine blockade and ferroptosis to promote the photoimmunotherapy of triple negative breast cancer (TNBC). The nanodrug, named CuS-PEG@Apt, was constructed via the modification of copper sulfide (CuS) nanoparticles with adenosine aptamer and PEG. CuS-PEG@Apt could be effectively enriched in the tumor site and locally generate a strong photothermal effect, directly ablating tumors and inducing immunogenic death (ICD). On the other hand, the aptamers could block the adenosine pathway to inhibit the immune suppression by adenosine, which further promoted the anti-tumor immunity. Moreover, the CuS nanoparticles could consume GSH and inhibit GPX4 to cause the ferroptosis of tumor cells. Collectively, CuS-PEG@Apt achieved potent efficacy of tumor suppression via the combination of PTT, immune activation and ferroptosis, representing an appealing platform for TNBC treatment.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Chengyu Shi
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Qiao Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuting Zhong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Lipeng Zhu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuetao Zhao
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| |
Collapse
|
5
|
Cao Y, Zhao X, Miao Y, Wang X, Deng D. How the Versatile Self-Assembly in Drug Delivery System to Afford Multimodal Cancer Therapy? Adv Healthc Mater 2025; 14:e2403715. [PMID: 39587000 DOI: 10.1002/adhm.202403715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/04/2024] [Indexed: 11/27/2024]
Abstract
The rapid development of self-assembly technology during the past few decades has effectively addressed plenty of the issues associated with carrier-based drug delivery systems, such as low loading efficiency, complex fabrication processes, and inherent toxicity of carriers. The integration of nanoscale delivery systems with self-assembly techniques has enabled efficient and targeted self-administration of drugs, enhanced bioavailability, prolonged circulation time, and controllable drug release. Concurrently, the limitations of single-mode cancer treatment, including low bioavailability, poor therapeutic outcomes, and significant side effects, have highlighted the urgent need for multimodal combined antitumor therapies. Set against the backdrop of multimodal cancer therapy, this review summarizes the research progress and applications of a large number of self-assembled drug delivery platforms, including natural small molecule self-assembled, carrier-free self-assembled, amphiphilic polymer-based self-assembled, peptide-based self-assembled, and metal-based self-assembled nano drug delivery systems. This review particularly analyzes the latest advances in the application of self-assembled nano drug delivery platforms in combined antitumor therapies mediated by chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, and immunotherapy, providing innovative research insights for further optimization and expansion of self-assembled nano drug delivery systems in the clinical translation and development of antitumor combined therapy.
Collapse
Affiliation(s)
- Yuqi Cao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaomin Zhao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhang Miao
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Xin Wang
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Dawei Deng
- Department of Pharmaceutical Engineering and Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| |
Collapse
|
6
|
Huang Q, Li H, Zhang Y. A bibliometric and knowledge-map study on the treatment of hematological malignancies with CAR-T cells from 2012 to 2023. Hum Vaccin Immunother 2024; 20:2371664. [PMID: 38961667 PMCID: PMC11225924 DOI: 10.1080/21645515.2024.2371664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
Recently, CAR-T cell therapy in hematological malignancies has received extensive attention. The objective of this study is to gain a comprehensive understanding of the current research status, development trends, research hotspots, and emerging topics pertaining to CAR-T cells in the treatment of hematological malignancies. Articles pertaining to CAR-T cell therapy for hematological malignancies from the years 2012 to 2023 were obtained and assessed from the Web of Science Core Collection (WoSCC). A bibliometric approach was employed to conduct a scientific, comprehensive, and objective quantitative analysis, as well as a visual analysis, of this particular research domain. A comprehensive analysis was conducted on a corpus of 3643 articles, which were collaboratively authored by 72 countries and various research institutions. CAR-T cell research in treating hematological malignancies shows an increasing trend each year. Notably, the study identified the countries and institutions displaying the highest level of activity, the journals with the most citations and output, as well as the authors who garnered the highest frequency of citations and co-citations. Furthermore, the analysis successfully identified the research hotspots and highlighted six emerging topics within this domain. This study conducted a comprehensive exploration and analysis of the research status, development trends, research hotspots, and emerging topics about CAR-T cells in the treatment of hematological malignancies from 2012 to 2023. The findings of this study will serve as a valuable reference and guide for researchers seeking to delve deeper into this field and determine the future direction of their research.
Collapse
Affiliation(s)
- Qing Huang
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Huimin Li
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yuan Zhang
- Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
7
|
Aniogo E, Kujawski M, Awuah D, Cha SE, Espinosa R, Hui S, Ghimire H, Yazaki PJ, Brown CE, Wang X, Shively JE. Targeting CEA in metastatic triple negative breast cancer with image-guided radiation followed by Fab-mediated chimeric antigen receptor (CAR) T-cell therapy. Front Immunol 2024; 15:1499471. [PMID: 39759518 PMCID: PMC11695362 DOI: 10.3389/fimmu.2024.1499471] [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: 09/20/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Although CAR-T cell therapy has limited efficacy against solid tumors, it has been hypothesized that prior treatment with Image-Guided Radiation Therapy (IGRT) would increase CAR-T cell tumor infiltration, leading to improved antigen specific expansion of CAR-T cells. Methods To test this hypothesis in a metastatic triple negative breast cancer (TNBC) model, we engineered two anti-CEA single-chain Fab (scFab) CAR-T cells with signaling domains from CD28zeta and 4-1BBzeta, and tested them in vitro and in vivo. Results The anti-CEA scFab CAR-T cells generated from three different human donors demonstrated robust in vitro expression, expansion, and lysis of only CEA-positive TNBC cells, with the CD28z-CAR-T cells showing the highest cytotoxicity. IFN-γ and granzyme B release assays revealed significantly higher IFN-γ production at a 4:1 effector-to-target (E:T) ratio in CD28z-CAR-T cells compared to 4-1BBz-CAR-T cells. Treatment of CEA-positive TNBC MDA-MB231 xenografts in the mammary fat pads of NSG mice, that produced spontaneous lung metastases over time, resulted in significant tumor growth reduction compared to either therapy alone (p<0.01). Immunohistochemical (IHC) analysis revealed that only combined IGRT and CAR-T therapy resulted in the elimination of lung metastases. Discussion These findings demonstrate that the combination of IGRT and anti-CEA scFab CAR-T therapy induces a strong antitumor response, effectively targeting both the primary tumor and distant metastatic lesions in the lungs, thus demonstrating that IGRT enhances CAR-T cell infiltration, persistence, and overall efficacy within both primary and metastatic lesions.
Collapse
Affiliation(s)
- Eric Aniogo
- Department of Immunology and Theranostics, City of Hope, Duarte, CA, United States
| | - Maciej Kujawski
- Department of Immunology and Theranostics, City of Hope, Duarte, CA, United States
| | - Dennis Awuah
- T-Cell Therapeutic Laboratory, City of Hope, Duarte, CA, United States
| | - Seung E. Cha
- Department of Immunology and Theranostics, City of Hope, Duarte, CA, United States
| | - Ruby Espinosa
- T-Cell Therapeutic Laboratory, City of Hope, Duarte, CA, United States
| | - Susanta Hui
- Department of Radiation Oncology, City of Hope, Duarte, CA, United States
| | - Hemendra Ghimire
- Department of Radiation Oncology, City of Hope, Duarte, CA, United States
| | - Paul J. Yazaki
- Department of Immunology and Theranostics, City of Hope, Duarte, CA, United States
| | - Christine E. Brown
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope Beckman Research Institute and Medical Center, Duarte, CA, United States
| | - Xiuli Wang
- T-Cell Therapeutic Laboratory, City of Hope, Duarte, CA, United States
| | - John E. Shively
- Department of Immunology and Theranostics, City of Hope, Duarte, CA, United States
| |
Collapse
|
8
|
Yao J, Cui Z, Zhang F, Li H, Tian L. Biomaterials enhancing localized cancer therapy activated anti-tumor immunity: a review. J Mater Chem B 2024; 13:117-136. [PMID: 39544081 DOI: 10.1039/d4tb01995d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Localized cancer therapies such as radiotherapy, phototherapy, and chemotherapy are precise cancer treatment strategies aimed at minimizing systemic side effects. However, cancer metastasis remains the primary cause of mortality among cancer patients in clinical settings, and localized cancer treatments have limited efficacy against metastatic cancer. Therefore, researchers are exploring strategies that combine localized therapy with immunotherapy to activate robust anti-tumor immune responses, thereby eradicating metastatic cancer. Biomaterials, as novel materials, exhibit great potential in biomedical applications and have achieved great progress in clinic translation. This review introduces biomaterials and their applications in research focused on enhancing localized cancer treatment activated anti-tumor immunity. Additionally, the current challenges and future directions of biomaterials are also discussed, providing insights and references for related research.
Collapse
Affiliation(s)
- Jipeng Yao
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Zhencun Cui
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- Department of Nuclear Medicine, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, 730000, China
| | - Feifei Zhang
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Haidong Li
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Longlong Tian
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China.
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| |
Collapse
|
9
|
Chang Y, Chang M, Bao X, Dong C. Advancements in adoptive CAR immune cell immunotherapy synergistically combined with multimodal approaches for tumor treatment. Bioact Mater 2024; 42:379-403. [PMID: 39308543 PMCID: PMC11415837 DOI: 10.1016/j.bioactmat.2024.08.046] [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: 06/24/2024] [Revised: 08/26/2024] [Accepted: 08/31/2024] [Indexed: 09/25/2024] Open
Abstract
Adoptive immunotherapy, notably involving chimeric antigen receptor (CAR)-T cells, has obtained Food and Drug Administration (FDA) approval as a treatment for various hematological malignancies, demonstrating promising preclinical efficacy against cancers. However, the intricate and resource-intensive autologous cell processing, encompassing collection, expansion, engineering, isolation, and administration, hamper the efficacy of this therapeutic modality. Furthermore, conventional CAR T therapy is presently confined to addressing solid tumors due to impediments posed by physical barriers, the potential for cytokine release syndrome, and cellular exhaustion induced by the immunosuppressive and heterogeneous tumor microenvironment. Consequently, a strategic integration of adoptive immunotherapy with synergistic multimodal treatments, such as chemotherapy, radiotherapy, and vaccine therapy etc., emerges as a pivotal approach to surmount these inherent challenges. This collaborative strategy holds the key to addressing the limitations delineated above, thereby facilitating the realization of more precise personalized therapies characterized by heightened therapeutic efficacy. Such synergistic strategy not only serves to mitigate the constraints associated with adoptive immunotherapy but also fosters enhanced clinical applicability, thereby advancing the frontiers of therapeutic precision and effectiveness.
Collapse
Affiliation(s)
- Yun Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| | - Mingyang Chang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
| | - Xiaoping Bao
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue University Institute for Cancer Research, West Lafayette, IN, 47907, USA
| | - Cheng Dong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, 999077, Hong Kong, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518000, China
| |
Collapse
|
10
|
Yang L, Wu M, Yang H, Sun X, Xing L, Liu D, Xing L, Yu J. Case report: Bridging radiation therapy before chimeric antigen receptor T-cell therapy induces sustained remission in patients with relapsed/refractory double-expressor diffuse large B-cell lymphoma with localized compressive symptoms. Front Immunol 2024; 15:1441404. [PMID: 39290703 PMCID: PMC11405209 DOI: 10.3389/fimmu.2024.1441404] [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: 05/31/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Background High-risk double-expressor diffuse large B-cell lymphoma has an inferior prognosis following standard first-line therapy. After failure of second-line therapy, treatment options are limited if accompanied by localized compressive symptoms. Chimeric Antigen Receptor T cell (CAR-T) therapy preceded by bridging radiotherapy may be an effective emerging therapy. Case presentation We report a 66-year-old female patient diagnosed with stage IV double-expressor diffuse large B-cell lymphoma. The patient achieved progressive disease after two cycles of rituximab, cyclophosphamide, liposomal doxorubicin, vincristine, and prednisone and continued to develop cervical lymph node recurrence after second-line therapy. The patient was infused with CAR-T cells after receiving focal bridging radiotherapy and remained in complete response more than 9 months after treatment. In addition, the patients did not experience serious adverse reactions related to radiotherapy as well as CAR-T cell therapy. Conclusions In this article, we describe a patient with double-expressor diffuse large B-cell lymphoma with localized compression symptoms after second-line treatment failure who benefited from CAR-T combined with focal bridging radiotherapy.
Collapse
Affiliation(s)
- Liying Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengdi Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hao Yang
- Department of Graduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaorong Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lijie Xing
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Dan Liu
- Department of Hematology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
11
|
Cheng Z, Cui X, Li S, Liang Y, Yang W, Ouyang J, Wei M, Yan Z, Yu W. Harnessing cytokines to optimize chimeric antigen receptor-T cell therapy for gastric cancer: Current advances and innovative strategies. Biomed Pharmacother 2024; 178:117229. [PMID: 39096620 DOI: 10.1016/j.biopha.2024.117229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/20/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
Enormous patients with gastric cancer (GC) are insensitive to chemotherapy and targeted therapy without the chance of radical surgery, so immunotherapy may supply a novel choice for them. Chimeric antigen receptor (CAR)-T cell therapy has the advantages of higher specificity, stronger lethality, and longer-lasting efficacy, and it has the potential for GC in the future. However, its application still faces numerous obstacles in terms of accuracy, efficacy, and safety. Cytokines can mediate the migration, proliferation, and survival of immune cells, regulate the duration and strength of immune responses, and are involved in the occurrence of severe side effects in CAR-T cell therapy. The expression levels of specific cytokines are associated with the genesis, invasion, metastasis, and prognosis of GC. Applications of cytokines and their receptors in CAR-T cell therapy have emerged, and various cytokines and their receptors have contributed to improving CAR-T cell anti-tumor capabilities. Large amounts of central cytokines in this therapy include chemokines, interleukins (ILs), transforming growth factor-β (TGF-β), and colony-stimulating factors (CSFs). Meanwhile, researchers have explored the combination therapy in treating GC, and several approaches applied to other malignancies can also be considered as references. Therefore, our review comprehensively outlines the biological functions and clinical significance of cytokines and summarizes current advances and innovative strategies for harnessing cytokines to optimize CAR-T cell therapy for GC.
Collapse
Affiliation(s)
- Zewei Cheng
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaohan Cui
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Song Li
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yize Liang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenshuo Yang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jun Ouyang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meng Wei
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhibo Yan
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenbin Yu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
12
|
Srivastava S, Tyagi A, Pawar VA, Khan NH, Arora K, Verma C, Kumar V. Revolutionizing Immunotherapy: Unveiling New Horizons, Confronting Challenges, and Navigating Therapeutic Frontiers in CAR-T Cell-Based Gene Therapies. Immunotargets Ther 2024; 13:413-433. [PMID: 39219644 PMCID: PMC11365499 DOI: 10.2147/itt.s474659] [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: 05/11/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
The CAR-T cell therapy has marked the dawn of new era in the cancer therapeutics and cell engineering techniques. The review emphasizes on the challenges that obstruct the therapeutic efficiency caused by cell toxicities, immunosuppressive tumor environment, and decreased T cell infiltration. In the interest of achieving the overall survival (OS) and event-free survival (EFS) of patients, the conceptual background of potential target selection and various CAR-T cell design techniques are described which can minimize the off-target effects, reduce toxicity, and thus increase the resilience of CAR-T cell treatment in the haematological malignancies as well as in solid tumors. Furthermore, it delves into cutting-edge technologies like gene editing and synthetic biology, providing new opportunities to enhance the functionality of CAR-T cells and overcome mechanisms of immune evasion. This review provides a comprehensive understanding of the complex and diverse aspects of CAR-T cell-based gene treatments, including both scientific and clinical aspects. By effectively addressing the obstacles and utilizing the capabilities of cutting-edge technology, CAR-T cell therapy shows potential in fundamentally changing immunotherapy and reshaping the approach to cancer treatment.
Collapse
Affiliation(s)
- Shivani Srivastava
- Department of Pathology, School of Medicine, Yale University, New Haven, CT, USA
| | - Anuradha Tyagi
- Department of cBRN, Institute of Nuclear Medicine and Allied Science, Delhi, India
| | | | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Arora
- Advanced Instrumentation Research Facility, Jawaharlal Nehru University, New Delhi, India
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Chaitenya Verma
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
- Department of Biotechnology, SSET, Sharda University, Greater Noida, 201306, India
| | - Vinay Kumar
- Pennsylvania State University Hershey Medical Center, 500 University Dr, Heshey, PA, USA
| |
Collapse
|
13
|
Li Y, Song Y, Yin J, Pan W, Li N, Tang B. Organelle-based immunotherapy strategies for fighting against cancer. Chem Commun (Camb) 2024; 60:8170-8185. [PMID: 38979965 DOI: 10.1039/d4cc01594k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Destruction of subcellular organelles can cause dysfunction and even death of cells to elicit immune responses. In this review, the characteristics and functions of important organelles are mainly summarized. Then, the intelligent immunotherapeutic strategies and suggestions based on influencing the organelles are further highlighted. This review will provide ideas for developing novel and effective immunotherapy strategies and advance the development of cancer immunotherapy.
Collapse
Affiliation(s)
- Yanhua Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Yingying Song
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Jiaqi Yin
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China.
- Laoshan Laboratory, Qingdao 266237, P. R. China
| |
Collapse
|
14
|
Espelage L, Wagner N, Placke JM, Ugurel S, Tasdogan A. The Interplay between Metabolic Adaptations and Diet in Cancer Immunotherapy. Clin Cancer Res 2024; 30:3117-3127. [PMID: 38771898 DOI: 10.1158/1078-0432.ccr-22-3468] [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: 07/06/2023] [Revised: 11/07/2023] [Accepted: 04/15/2024] [Indexed: 05/23/2024]
Abstract
Over the past decade, cancer immunotherapy has significantly advanced through the introduction of immune checkpoint inhibitors and the augmentation of adoptive cell transfer to enhance the innate cancer defense mechanisms. Despite these remarkable achievements, some cancers exhibit resistance to immunotherapy, with limited patient responsiveness and development of therapy resistance. Metabolic adaptations in both immune cells and cancer cells have emerged as central contributors to immunotherapy resistance. In the last few years, new insights emphasized the critical role of cancer and immune cell metabolism in animal models and patients. During therapy, immune cells undergo important metabolic shifts crucial for their acquired effector function against cancer cells. However, cancer cell metabolic rewiring and nutrient competition within tumor microenvironment (TME) alters many immune functions, affecting their fitness, polarization, recruitment, and survival. These interactions have initiated the development of novel therapies targeting tumor cell metabolism and favoring antitumor immunity within the TME. Furthermore, there has been increasing interest in comprehending how diet impacts the response to immunotherapy, given the demonstrated immunomodulatory and antitumor activity of various nutrients. In conclusion, recent advances in preclinical and clinical studies have highlighted the capacity of immune-based cancer therapies. Therefore, further exploration into the metabolic requirements of immune cells within the TME holds significant promise for the development of innovative therapeutic approaches that can effectively combat cancer in patients.
Collapse
Affiliation(s)
- Lena Espelage
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Natalie Wagner
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Jan-Malte Placke
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen and German Cancer Consortium (DKTK), Essen, Germany
| |
Collapse
|
15
|
Tseng YT, Tsai CC, Chen PC, Lin BY, Hsu SCN, Huang SP, Huang B. Mechanical shear flow regulates the malignancy of colorectal cancer cells. Kaohsiung J Med Sci 2024; 40:650-659. [PMID: 38757734 DOI: 10.1002/kjm2.12844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Colorectal cancer (CRC) is notable for its high mortality and high metastatic characteristics. The shear force generated by bloodstream provides mechanical signals regulating multiple responses of cells, including metastatic cancer cells, dispersing in blood vessels. We, therefore, studied the effect of shear flow on circulating CRC cells in the present study. The CRC cell line SW620 was subjected to shear flow of 12.5 dynes/cm2 for 1 and 2 h separately. Resulting elevated caspase-9 and -3 indicated that shear flow initiated the apoptosis of SW620. Enlarged cell size associated with a higher level of cyclin D1 was coincident with the flow cytometric results indicating that the cell cycle was arrested at the G1 phase. An elevated phosphor-eNOSS1177 increased the production of nitric oxide and led to reactive oxygen species-mediated oxidative stress. Shear flow also regulated epithelial-mesenchymal transition (EMT) by increasing E-cadherin and ZO-1 while decreasing Snail and Twist1. The migration and invasion of sheared SW620 were also substantially decreased. Further investigations showed that mitochondrial membrane potential was significantly decreased, whereas mitochondrial mass and ATP production were not changed. In addition to the shear flow of 12.5 dynes/cm2, the expressions of EMT were compared at lower (6.25 dynes/cm2) and at higher (25 dynes/cm2) shear flow. The results showed that lower shear flow increased mesenchymal characteristics and higher shear flow increased epithelial characteristics. Shear flow reduces the malignancy of CRC in their metastatic dispersal that opens up new ways to improve cancer therapies by applying a mechanical shear flow device.
Collapse
Affiliation(s)
- Yu-Ting Tseng
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Chung Tsai
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Pediatrics, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Ping-Chen Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Bo-Yan Lin
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shu-Ping Huang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Bin Huang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
16
|
Szlasa W, Sztuder A, Kaczmar-Dybko A, Maciejczyk A, Dybko J. Efficient combination of radiotherapy and CAR-T - A systematic review. Biomed Pharmacother 2024; 174:116532. [PMID: 38574625 DOI: 10.1016/j.biopha.2024.116532] [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: 01/09/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, a groundbreaking immunotherapy. However, it faces formidable challenges in treating solid tumors, grappling with issues like poor trafficking, limited penetration, and insufficient persistence within the tumor microenvironment (TME). CAR-T cells are engineered to express receptors that target specific cancer antigens, enhancing their ability to recognize and eliminate cancer cells. This review paper explores the intricate interplay between CAR-T therapy and radiotherapy (RT), investigating their synergistic potential. Radiotherapy, a standard cancer treatment, involves using high doses of radiation to target and damage cancer cells, disrupting their ability to grow and divide. We highlight that RT modulates the TME, augments antigen presentation, and promotes immune cell infiltration, bolstering CAR-T cell-mediated tumor eradication. Molecular insights shed light on RT-induced alterations in tumor stroma, T cell recruitment promotion, and induction of immunogenic cell death. Noteworthy, strategies, such as combining hypofractionated radiotherapy with myeloid-derived suppressor cell blockade, underscore innovative approaches to enhance CAR-T cell therapy in solid tumors. Bridging indications for RT and CAR-T cells in hematological malignancies are discussed, emphasizing scenarios where RT strategically enhances CAR-T cell efficacy. The paper critically evaluates the RT as a bridge compared to traditional chemotherapy, highlighting timing and dosage considerations crucial for optimizing CAR-T therapy outcomes. In summary, the paper provides valuable insights into the intricate molecular mechanisms activated by RT and innovative strategies to improve CAR-T cell therapy, fostering a deeper understanding of their combined potential in cancer treatment.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Medical University Hospital, Borowska 213, Wrocław 50-556, Poland.
| | - Aleksandra Sztuder
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | | | - Adam Maciejczyk
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Jarosław Dybko
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology and Hematology, Wroclaw University of Science and Technology, Wrocław 50-370, Poland
| |
Collapse
|
17
|
Zhou S, Zhu M, Wei X, Mu P, Shen L, Wang Y, Wan J, Zhang H, Xia F, Zhang Z. Low-dose radiotherapy synergizes with iRGD-antiCD3-modified T cells by facilitating T cell infiltration. Radiother Oncol 2024; 194:110213. [PMID: 38458258 DOI: 10.1016/j.radonc.2024.110213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND AND PURPOSE Poor penetration of transferred T cells represents a critical factor impeding the development of adoptive cell therapy in solid tumors. We demonstrated that iRGD-antiCD3 modification promoted both T cell infiltration and activation in our previous work. Interest in low-dose radiotherapy has recently been renewed due to its immuno-stimulatory effects including T cell recruitment. This study aims to explore the synergistic effects between low-dose radiotherapy and iRGD-antiCD3-modified T cells. MATERIALS AND METHODS Flow cytometry was performed to assess the expression of iRGD receptors and chemokines. T cell infiltration was evaluated by immunohistofluorescence and in vivo real-time fluorescence imaging and antitumor effects were investigated by in vivo bioluminescence imaging in the gastric cancer peritoneal metastasis mouse model. RESULTS We found that 2 Gy irradiation upregulated the expression of all three iRGD receptors and T-cell chemokines. The addition of 2 Gy low-dose irradiation boosted the accumulation and penetration of iRGD-antiCD3-modified T cells in peritoneal tumor nodules. Combining 2 Gy low-dose irradiation with iRGD-antiCD3-modified T cells significantly inhibited tumor growth and prolonged survival in the peritoneal metastasis mouse model with a favorable safety profile. CONCLUSION Altogether, we demonstrated that low-dose radiotherapy could improve the antitumor potency of iRGD-antiCD3-modified T cells by promoting T cell infiltration, providing a rationale for exploring low-dose radiotherapy in combination of other adoptive T cell therapies in solid tumors.
Collapse
Affiliation(s)
- Shujuan Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Mei Zhu
- Department of Oncology, Xuzhou Cancer Hospital, Xuzhou 221005, China
| | - Xiao Wei
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Peiyuan Mu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Yan Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Juefeng Wan
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Hui Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China
| | - Fan Xia
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China.
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Clinical Research Center for Radiation Oncology, Shanghai 200032, China; Shanghai Key Laboratory of Radiation Oncology, Shanghai 200032, China.
| |
Collapse
|
18
|
Eigendorff F, Filimonova I, Scholl S, Sayer-Klink A, Rummler S, Kunert C, Pietschmann K, Wittig A, Hochhaus A, Schnetzke U. Effective bridging strategies prior to infusion with tisagenlecleucel results in high response rates and long-term remission in relapsed/refractory large B-cell lymphoma: findings from a German monocentric study. J Cancer Res Clin Oncol 2024; 150:224. [PMID: 38693452 PMCID: PMC11062962 DOI: 10.1007/s00432-024-05765-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Incorporating chimeric antigen receptor (CAR)-T cell therapy into relapsed or refractory large B-cell lymphoma (rr LBCL) treatment algorithms has yielded remarkable response rates and durable remissions, yet a substantial portion of patients experience progression or relapse. Variations in outcomes across treatment centers may be attributed to different bridging strategies and remission statuses preceding CAR-T cell therapy. PATIENTS Twenty-nine consecutive adult patients receiving tisagenlecleucel (tisa-cel) for rr LBCL from December 2019 to February 2023 at Jena University Hospital were analyzed. RESULTS The median age was 63, with a median of 3 prior treatments. Twenty patients (69%) were refractory to any systemic therapy before CAR-T cell treatment. Following leukapheresis, 25 patients (86%) received bridging therapy with the majority undergoing chemotherapy (52%) or combined modality therapy (32%). Radiotherapy (RT) was part of the bridging strategy in 44%, with moderately hypofractionated involved site RT (30.0 Gy/2.5 Gy) being applied most frequently (64%). Post-CAR-T infusion, the objective response rate at 30 days was 83%, with 55% achieving complete response. Twelve-month progression-free (PFS) and overall survival (OS) were 60% and 74%, respectively, with a median follow up of 11.1 months for PFS and 17.9 months for OS. Factors significantly associated with PFS were chemotherapy sensitivity pre-leukapheresis and response to bridging. CONCLUSION The study underscores the importance of minimal tumor burden at CAR-T initiation, emphasizing the need for suitable bridging regimens. The findings advocate for clinical trials and further real-world analyses to optimize CAR-T cell therapy outcomes by identifying the most effective bridging strategies.
Collapse
MESH Headings
- Humans
- Male
- Middle Aged
- Female
- Aged
- Immunotherapy, Adoptive/methods
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Adult
- Remission Induction
- Neoplasm Recurrence, Local/therapy
- Neoplasm Recurrence, Local/pathology
- Germany
- Receptors, Antigen, T-Cell/therapeutic use
- Retrospective Studies
- Combined Modality Therapy
Collapse
Affiliation(s)
- Farina Eigendorff
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Irina Filimonova
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Sebastian Scholl
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Anne Sayer-Klink
- Institut für Transfusionsmedizin, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Silke Rummler
- Institut für Transfusionsmedizin, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Christa Kunert
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Klaus Pietschmann
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Jena, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Andrea Wittig
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Jena, Jena, Germany
- Klinik und Poliklinik für Strahlentherapie und Radioonkologie, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Andreas Hochhaus
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany
| | - Ulf Schnetzke
- Klinik Für Innere Medizin II, Abteilung für Hämatologie und Internistische Onkologie, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Germany.
- Comprehensive Cancer Center Central Germany (CCCG) Jena/Leipzig, Campus Jena, Jena, Germany.
| |
Collapse
|
19
|
Amorós-Pérez B, Rivas-Pardo B, Gómez del Moral M, Subiza JL, Martínez-Naves E. State of the Art in CAR-T Cell Therapy for Solid Tumors: Is There a Sweeter Future? Cells 2024; 13:725. [PMID: 38727261 PMCID: PMC11083689 DOI: 10.3390/cells13090725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has proven to be a powerful treatment for hematological malignancies. The situation is very different in the case of solid tumors, for which no CAR-T-based therapy has yet been approved. There are many factors contributing to the absence of response in solid tumors to CAR-T cells, such as the immunosuppressive tumor microenvironment (TME), T cell exhaustion, or the lack of suitable antigen targets, which should have a stable and specific expression on tumor cells. Strategies being developed to improve CAR-T-based therapy for solid tumors include the use of new-generation CARs such as TRUCKs or bi-specific CARs, the combination of CAR therapy with chemo- or radiotherapy, the use of checkpoint inhibitors, and the use of oncolytic viruses. Furthermore, despite the scarcity of targets, a growing number of phase I/II clinical trials are exploring new solid-tumor-associated antigens. Most of these antigens are of a protein nature; however, there is a clear potential in identifying carbohydrate-type antigens associated with tumors, or carbohydrate and proteoglycan antigens that emerge because of aberrant glycosylations occurring in the context of tumor transformation.
Collapse
Affiliation(s)
- Beatriz Amorós-Pérez
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain; (B.A.-P.); (B.R.-P.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Inmunotek S.L., 28805 Madrid, Spain;
| | - Benigno Rivas-Pardo
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain; (B.A.-P.); (B.R.-P.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Manuel Gómez del Moral
- Department of Cellular Biology, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain;
| | | | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ORL, School of Medicine, Universidad Complutense of Madrid (UCM), 28040 Madrid, Spain; (B.A.-P.); (B.R.-P.)
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
20
|
Zhang T, Zhang Y, Wei J. Overcoming the challenges encountered in CAR-T therapy: latest updates from the 2023 ASH annual conference. Front Immunol 2024; 15:1389324. [PMID: 38660304 PMCID: PMC11039902 DOI: 10.3389/fimmu.2024.1389324] [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: 02/21/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024] Open
Abstract
Chimeric antigen receptor (CAR) -T cell therapy has entered the breakthrough era, characterized by a blend of therapeutic opportunities and challenges. With the integration of genome-editing technology, CAR-T cells will be empowered to become super warriors in eradicating tumor cells and attacking various tumors, including T-cell malignancies and acute myeloid leukemia. Notably, the optimization of CAR-T cells, including efficacy, safety, and manufacturing speed, coupled with other therapeutic strategies such as radiotherapy, hematopoietic stem cell transplantation, small-molecule inhibitors, and bispecific antibodies, could revolutionize the therapeutic landscape of tumors. Consequently, next-generation cellular immunotherapy, including universal CAR-NK cells and synergistic combination approaches, are anticipated to significantly impact cancer treatment in the coming decade. Nevertheless, the failure rates of CAR-T therapy continue to be significant. The challenge lies in determining the optimal combination strategy and identifying reliable and robust biomarkers to effectively select the patients who will derive the greatest benefit from CAR-T therapy. Herein, we highlight recent innovations in CAR-T products, combination strategies and predictive biomarkers of response presented at the 2023 ASH Annual Meeting.
Collapse
Affiliation(s)
- Tingting Zhang
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| |
Collapse
|
21
|
Tojjari A, Yu J, Saeed A. Immunotherapy and Radiation Therapy Combinatorial Approaches in Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:1058. [PMID: 38473415 DOI: 10.3390/cancers16051058] [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: 01/27/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
Hepatocellular carcinoma (HCC), a prevalent and often fatal liver cancer, presents significant treatment challenges, especially in its advanced stages. This article delves into the promising approach of combining immunotherapy, particularly immune checkpoint inhibitors, with radiation therapy, a cornerstone of HCC management. Our review synthesizes current preclinical and clinical research, highlighting the potential synergistic effects of this combinational treatment. Emerging evidence suggests that this synergy enhances tumor control and improves patient survival rates. The combination leverages the localized, tumor-targeting ability of radiation therapy and the systemic, immune-boosting effects of immunotherapy, potentially overcoming the limitations inherent in each treatment modality when used separately. This integrative approach is especially promising in addressing the complex tumor microenvironment of HCC. However, the treatment landscape is nuanced, with challenges such as patient-specific response variability and potential resistance to therapies. Future research directions should focus on refining these combination strategies, tailoring them to individual patient profiles, and understanding the underlying mechanisms that govern the interaction between immunotherapy and radiation therapy. Such advancements could significantly improve HCC management, setting new standards for patient care and treatment efficacy.
Collapse
Affiliation(s)
- Alireza Tojjari
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA
| | - James Yu
- Division of Hematology and Medical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Anwaar Saeed
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA
| |
Collapse
|
22
|
Long Y, Yang B, Lei Q, Gao F, Chen L, Chen W, Chen S, Ren W, Cao Y, Xu L, Wu D, Qu J, Li H, Yu Y, Zhang A, Wang S, Chen W, Wang H, Chen T, Chen Z, Li Q. Targeting Senescent Alveolar Epithelial Cells Using Engineered Mesenchymal Stem Cell-Derived Extracellular Vesicles To Treat Pulmonary Fibrosis. ACS NANO 2024; 18:7046-7063. [PMID: 38381372 PMCID: PMC10919282 DOI: 10.1021/acsnano.3c10547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Type 2 alveolar epithelial cell (AEC2) senescence is crucial to the pathogenesis of pulmonary fibrosis (PF). The nicotinamide adenine dinucleotide (NAD+)-consuming enzyme cluster of differentiation 38 (CD38) is a marker of senescent cells and is highly expressed in AEC2s of patients with PF, thus rendering it a potential treatment target. Umbilical cord mesenchymal stem cell (MSC)-derived extracellular vesicles (MSC-EVs) have emerged as a cell-free treatment with clinical application prospects in antiaging and antifibrosis treatments. Herein, we constructed CD38 antigen receptor membrane-modified MSC-EVs (CD38-ARM-MSC-EVs) by transfecting MSCs with a lentivirus loaded with a CD38 antigen receptor-CD8 transmembrane fragment fusion plasmid to target AEC2s and alleviate PF. Compared with MSC-EVs, the CD38-ARM-MSC-EVs engineered in this study showed a higher expression of the CD38 antigen receptor and antifibrotic miRNAs and targeted senescent AEC2s cells highly expressing CD38 in vitro and in naturally aged mouse models after intraperitoneal administration. CD38-ARM-MSC-EVs effectively restored the NAD+ levels, reversed the epithelial-mesenchymal transition phenotype, and rejuvenated senescent A549 cells in vitro, thereby mitigating multiple age-associated phenotypes and alleviating PF in aged mice. Thus, this study provides a technology to engineer MSC-EVs and support our CD38-ARM-MSC-EVs to be developed as promising agents with high clinical potential against PF.
Collapse
Affiliation(s)
- Yaoying Long
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bianlei Yang
- Department
of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qian Lei
- West
China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fei Gao
- Department
of Hematology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Li Chen
- Department
of Hematology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Wenlan Chen
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyi Chen
- Department
of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenxiang Ren
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yulin Cao
- Department
of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liuyue Xu
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Wu
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiao Qu
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - He Li
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yali Yu
- Department
of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anyuan Zhang
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shan Wang
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weiqun Chen
- Key
Laboratory for Molecular Diagnosis of Hubei Province, The Central
Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Hongxiang Wang
- Department
of Hematology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ting Chen
- Hubei Engineering
Research Center for Application of Extracellular Vesicle, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhichao Chen
- Department
of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qiubai Li
- Department
of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering
Research Center for Application of Extracellular Vesicle, Hubei University of Science and Technology, Xianning 437100, China
| |
Collapse
|
23
|
Liao YM, Hsu SH, Chiou SS. Harnessing the Transcriptional Signatures of CAR-T-Cells and Leukemia/Lymphoma Using Single-Cell Sequencing Technologies. Int J Mol Sci 2024; 25:2416. [PMID: 38397092 PMCID: PMC10889174 DOI: 10.3390/ijms25042416] [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: 12/22/2023] [Revised: 02/02/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T-cell therapy has greatly improved outcomes for patients with relapsed or refractory hematological malignancies. However, challenges such as treatment resistance, relapse, and severe toxicity still hinder its widespread clinical application. Traditional transcriptome analysis has provided limited insights into the complex transcriptional landscape of both leukemia cells and engineered CAR-T-cells, as well as their interactions within the tumor microenvironment. However, with the advent of single-cell sequencing techniques, a paradigm shift has occurred, providing robust tools to unravel the complexities of these factors. These techniques enable an unbiased analysis of cellular heterogeneity and molecular patterns. These insights are invaluable for precise receptor design, guiding gene-based T-cell modification, and optimizing manufacturing conditions. Consequently, this review utilizes modern single-cell sequencing techniques to clarify the transcriptional intricacies of leukemia cells and CAR-Ts. The aim of this manuscript is to discuss the potential mechanisms that contribute to the clinical failures of CAR-T immunotherapy. We examine the biological characteristics of CAR-Ts, the mechanisms that govern clinical responses, and the intricacies of adverse events. By exploring these aspects, we hope to gain a deeper understanding of CAR-T therapy, which will ultimately lead to improved clinical outcomes and broader therapeutic applications.
Collapse
Affiliation(s)
- Yu-Mei Liao
- Division of Hematology-Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Hsien Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shyh-Shin Chiou
- Division of Hematology-Oncology, Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center of Applied Genomics, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
24
|
Yu X, Cao W, Yang X, Yu C, Jiang W, Guo H, He X, Mei C, Ou C. Prognostic value and therapeutic potential of IAP family in head and neck squamous cell carcinoma. Aging (Albany NY) 2024; 16:3674-3693. [PMID: 38364254 PMCID: PMC10929838 DOI: 10.18632/aging.205551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/17/2024] [Indexed: 02/18/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) ranks as the eighth most prevalent malignancy globally and has the eighth greatest fatality rate when compared to all other forms of cancer. The inhibitor of apoptosis protein (IAP) family comprises a collection of apoptosis-negative modulators characterized by at least one single baculovirus IAP repeat (BIR) domain in its N-terminal region. While the involvement of the IAP family is associated with the initiation and progression of numerous tumours, its specific role in HNSCC remains poorly understood. Thus, this study aimed to comprehensively examine changes in gene expression, immunomodulatory effects, prognosis, and functional enrichment of HNSCC utilising bioinformatics analysis. Elevated levels of distinct IAP family members were observed to varying degrees in HNSCC, with high BIRC2 expression indicating a worse prognosis. Additionally, Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to probe the enrichment of gene expression and biological processes related to the IAP family in HNSCC. The infiltration levels of immune cells were shown to be strongly associated with the IAP gene expression, as determined by subsequent analysis. Hence, BIRC2 could be an effective immunotherapy target for HNSCC. Collectively, novel knowledge of the biological roles and prognostic implications of IAP family members in HNSCC is presented in this study.
Collapse
Affiliation(s)
- Xiaoqian Yu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Weiwei Cao
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha 410008, Hunan, China
| | - Xuejie Yang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Canping Yu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Wenying Jiang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hongbin Guo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiaoyun He
- Departments of Ultrasound Imaging, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Cheng Mei
- Department of Blood Transfusion, Xiangya Hospital, Clinical Transfusion Research Center, Central South University, Changsha 410008, Hunan, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
25
|
Deshwal A, Saxena K, Sharma G, Rajesh, Sheikh FA, Seth CS, Tripathi RM. Nanozymes: A comprehensive review on emerging applications in cancer diagnosis and therapeutics. Int J Biol Macromol 2024; 256:128272. [PMID: 38000568 DOI: 10.1016/j.ijbiomac.2023.128272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023]
Abstract
Nanozymes, a new class of nanomaterials-based artificial enzymes, have gained huge attraction due to their high operational stability, working efficiency in extreme conditions, and resistance towards protease digestion. Nowadays, they are effectively substituted for natural enzymes for catalysis by closely resembling the active sites found in natural enzymes. Nanozymes can compensate for natural enzymes' drawbacks, such as high cost, poor stability, low yield, and storage challenges. Due to their transforming nature, nanozymes are of utmost importance in the detection and treatment of cancer. They enable precise cancer detection, tailored drug delivery, and catalytic therapy. Through enhanced diagnosis, personalized therapies, and reduced side effects, their adaptability and biocompatibility can transform the management of cancer. The review focuses on metal and metal oxide-based nanozymes, highlighting their catalytic processes, and their applications in the prevention and treatment of cancer. It emphasizes their potential to alter diagnosis and therapy, particularly when it comes to controlling reactive oxygen species (ROS). The article reveals the game-changing importance of nanozymes in the future of cancer care and describes future research objectives, making it a useful resource for researchers, and scientists. Lastly, outlooks for future perspective areas in this rapidly emerging field have been provided in detail.
Collapse
Affiliation(s)
- Akanksha Deshwal
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh (AUUP), Noida 201313, India
| | - Kirti Saxena
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh (AUUP), Noida 201313, India
| | - Garima Sharma
- Department of Biomedical Science & Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rajesh
- CSIR-National Physical Laboratory, New Delhi, India
| | - Faheem A Sheikh
- Nanostructured and Biomimetic Lab, Department of Nanotechnology, University of Kashmir Hazratbal, Srinagar, Jammu and Kashmir 190006, India
| | | | - Ravi Mani Tripathi
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh (AUUP), Noida 201313, India.
| |
Collapse
|
26
|
Li X, You J, Hong L, Liu W, Guo P, Hao X. Neoantigen cancer vaccines: a new star on the horizon. Cancer Biol Med 2023; 21:j.issn.2095-3941.2023.0395. [PMID: 38164734 PMCID: PMC11033713 DOI: 10.20892/j.issn.2095-3941.2023.0395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024] Open
Abstract
Immunotherapy represents a promising strategy for cancer treatment that utilizes immune cells or drugs to activate the patient's own immune system and eliminate cancer cells. One of the most exciting advances within this field is the targeting of neoantigens, which are peptides derived from non-synonymous somatic mutations that are found exclusively within cancer cells and absent in normal cells. Although neoantigen-based therapeutic vaccines have not received approval for standard cancer treatment, early clinical trials have yielded encouraging outcomes as standalone monotherapy or when combined with checkpoint inhibitors. Progress made in high-throughput sequencing and bioinformatics have greatly facilitated the precise and efficient identification of neoantigens. Consequently, personalized neoantigen-based vaccines tailored to each patient have been developed that are capable of eliciting a robust and long-lasting immune response which effectively eliminates tumors and prevents recurrences. This review provides a concise overview consolidating the latest clinical advances in neoantigen-based therapeutic vaccines, and also discusses challenges and future perspectives for this innovative approach, particularly emphasizing the potential of neoantigen-based therapeutic vaccines to enhance clinical efficacy against advanced solid tumors.
Collapse
Affiliation(s)
- 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
| | - Jian You
- Department of Thoracic Oncology, Tianjin Cancer Hospital Airport Hospital, Tianjin 300308, China
- Department of Thoracic Oncology Surgery, Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| | - Liping Hong
- 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
| | - Weijiang Liu
- 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
| | - Peng Guo
- 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
| | - Xishan Hao
- 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
- Tianjin Medical University Cancer Institute & Hospital, Tianjin 300060, China
| |
Collapse
|
27
|
Wang Y, Sun SK, Liu Y, Zhang Z. Advanced hitchhiking nanomaterials for biomedical applications. Theranostics 2023; 13:4781-4801. [PMID: 37771786 PMCID: PMC10526662 DOI: 10.7150/thno.88002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023] Open
Abstract
Hitchhiking, a recently developed bio-inspired cargo delivery system, has been harnessed for diverse applications. By leveraging the interactions between nanoparticles and circulatory cells or proteins, hitchhiking enables efficient navigation through the vasculature while evading immune system clearance. Moreover, it allows for targeted delivery of nutrients to tissues, surveillance of the immune system, and pathogen elimination. Various synthetic nanomaterials have been developed to facilitate hitchhiking with circulatory cells or proteins. By combining the advantages of synthetic nanomaterials and circulatory cells or proteins, hitchhiking nanomaterials demonstrate several advantages over conventional vectors, including enhanced circulatory stability and optimized therapeutic efficacy. This review provides an overview of general strategies for hitchhiking, choices of cells and proteins, and recent advances of hitchhiking nanomaterials for biomedical applications.
Collapse
Affiliation(s)
- Ying Wang
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Yang Liu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhanzhan Zhang
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| |
Collapse
|
28
|
Jia M, Dong T, Cheng Y, Rong F, Zhang J, Lv W, Zhen S, Jia X, Cong B, Wu Y, Cui H, Hao P. Ceruloplasmin is associated with the infiltration of immune cells and acts as a prognostic biomarker in patients suffering from glioma. Front Pharmacol 2023; 14:1249650. [PMID: 37637428 PMCID: PMC10450624 DOI: 10.3389/fphar.2023.1249650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/01/2023] [Indexed: 08/29/2023] Open
Abstract
Glioma is regarded as a prevalent form of cancer that affects the Central Nervous System (CNS), with an aggressive growth pattern and a low clinical cure rate. Despite the advancement of the treatment strategy of surgical resection, chemoradiotherapy and immunotherapy in the last decade, the clinical outcome is still grim, which is ascribed to the low immunogenicity and tumor microenvironment (TME) of glioma. The multifunctional molecule, called ceruloplasmin (CP) is involved in iron metabolism. Its expression pattern, prognostic significance, and association with the immune cells in gliomas have not been thoroughly investigated. Studies using a variety of databases, including Chinese Glioma Genome Atlas (CGGA), The Cancer Genome Atlas (TCGA), and Gliovis, showed that the mRNA and protein expression levels of CP in patients suffering from glioma increased significantly with an increasing glioma grade. Kaplan-Meier (KM) curves and statistical tests highlighted a significant reduction in survival time of patients with elevated CP expression levels. According to Cox regression analysis, CP can be utilized as a stand-alone predictive biomarker in patients suffering from glioma. A significant association between CP expression and numerous immune-related pathways was found after analyzing the data using the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA). Tumor Immune Estimation Resource (TIMER) and CIBERSORT analyses indicated a substantial correlation between the CP expression and infiltration of immunocytes in the TME. Additionally, immune checkpoints and CP expression in gliomas showed a favorable correlation. According to these results, patients with glioma have better prognoses and levels of tumor immune cell infiltration when their CP expression is low. As a result, CP could be used as a probable therapeutic target for gliomas and potentially anticipate the effectiveness of immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Jia
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, China
- Postdoctoral Mobile Station of Biology, Hebei Medical University, Shijiazhuang, Hebei, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Tianyu Dong
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, China
| | - Yangyang Cheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fanghao Rong
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, China
| | - Jiamin Zhang
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, China
| | - Wei Lv
- Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shuman Zhen
- Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xianxian Jia
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Bin Cong
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuming Wu
- Hebei Collaborative Innovation Center for Cardio Cerebrovascular Disease, Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Huixian Cui
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| | - Peipei Hao
- Department of Human Anatomy, Hebei Medical University, Shijiazhuang, Hebei, China
- International Cooperation Laboratory of Stem Cell Research, Shijiazhuang, China
- Hebei Key Laboratory of Neurodegenerative Disease Mechanism, Shijiazhuang, China
| |
Collapse
|
29
|
Nan Y, Ju D, Zhang X. Editorial: Combinational immunotherapy of cancer: novel targets, mechanisms, and strategies. Front Immunol 2023; 14:1250975. [PMID: 37638039 PMCID: PMC10454968 DOI: 10.3389/fimmu.2023.1250975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/03/2023] [Indexed: 08/29/2023] Open
Affiliation(s)
| | | | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
30
|
He M, Zhang D, Cao Y, Chi C, Zeng Z, Yang X, Yang G, Sharma K, Hu K, Enikeev M. Chimeric antigen receptor-modified T cells therapy in prostate cancer: A comprehensive review on the current state and prospects. Heliyon 2023; 9:e19147. [DOI: https:/doi.org/10.1016/j.heliyon.2023.e19147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
|
31
|
He M, Zhang D, Cao Y, Chi C, Zeng Z, Yang X, Yang G, Sharma K, Hu K, Enikeev M. Chimeric antigen receptor-modified T cells therapy in prostate cancer: A comprehensive review on the current state and prospects. Heliyon 2023; 9:e19147. [PMID: 37664750 PMCID: PMC10469587 DOI: 10.1016/j.heliyon.2023.e19147] [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: 05/02/2023] [Revised: 07/31/2023] [Accepted: 08/14/2023] [Indexed: 09/05/2023] Open
Abstract
Recent immunotherapy research has focused on chimeric antigen receptor-modified T cells (CAR-Ts). CAR-T therapies have been clinically applied to manage hematologic malignancies with satisfactory effectiveness. However, the application of CAR-T immunotherapy in solid tumors remains challenging. Even so, current CAR-T immunotherapies for prostate cancer (PCa) have shown some promise, giving hope to patients with advanced metastatic PCa. This review aimed to elucidate different types of prostate tumor-associated antigen targets, such as prostate-specific membrane antigen and prostate stem cell antigen, and their effects. The current status of the corresponding targets in clinical research through their applications was also discussed. To improve the efficacy of CAR-T immunotherapy, we addressed the possible applications of multimodal immunotherapy, chemotherapy, and CAR-T combined therapies. The obstacles of solid tumors were concisely elaborated. Further studies should aim to discover novel potential targets and establish new models by overcoming the inherent barriers of solid tumors, such as tumor heterogeneity and the immunosuppressive nature of the tumor microenvironment.
Collapse
Affiliation(s)
- Mingze He
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| | - Dongqi Zhang
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), 130000, Changchun, China
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Changliang Chi
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), 130000, Changchun, China
| | - Zitong Zeng
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Guodong Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Kritika Sharma
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Russia
| | - Kebang Hu
- Department of Urology, The First Hospital of Jilin University (Lequn Branch), 130000, Changchun, China
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435, Moscow, Russia
| |
Collapse
|