1
|
Zhang Q, Kuang G, Li W, Zhao Y. Cryo-Inactivated Cancer Cells Derived Magnetic Micromotors for Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04986. [PMID: 40405693 DOI: 10.1002/advs.202504986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/24/2025] [Indexed: 05/24/2025]
Abstract
Immunotherapy represents a highly promising modality in cancer treatment, with substantial advancements in therapeutic strategies. The primary challenge lies in enhancing the efficacy of immunotherapy approaches. Here, novel cryo-inactivated cancer cells (CICC) derived magnetic micromotors (CICC@FeMnP) are reported for tumor synergistic immunotherapy. Through the magnetic control, the CICC@FeMnP micromotors can on-demand target and accumulate at the tumor site. The FeMnP can induce ferroptosis and then trigger immunogenic cell death of tumor cells. The CICC containing the whole cancer antigen can conduct vaccination effects. Together with the Mn2+-mediated cGAS-STING pathway to stimulate the immune response, substantial anti-tumor immune effects can be achieved. Importantly, the CICC@FeMnP micromotors not only facilitate the establishment of a collaborative anti-tumor immune network to enhance effective tumoricidal immunity but also induce long-lasting immune memory effects. These results contribute to the inhibition of tumor progression, recurrence and lung metastasis, thereby prolonging the overall survival of tumor-bearing mice. This work underscores the potential of an engineered biohybrid micromotor system as an alternative therapeutic approach in immunotherapy to enhance efficacy against tumors.
Collapse
Affiliation(s)
- Qingfei Zhang
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Gaizhen Kuang
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Wenzhao Li
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Yuanjin Zhao
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
2
|
Christie JR, Romine P, Eddy K, Chen DL, Daher O, Abdelrazek M, Malthaner RA, Qiabi M, Nayak R, Kinahan P, Nair VS, Mattonen SA. Thorax-encompassing multi-modality PET/CT deep learning model for resected lung cancer prognostication: A retrospective, multicenter study. Med Phys 2025. [PMID: 40317503 DOI: 10.1002/mp.17862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/10/2025] [Accepted: 03/02/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Patients with early-stage non-small cell lung cancer (NSCLC) typically receive surgery as their primary form of treatment. However, studies have shown that a high proportion of these patients will experience a recurrence after their resection, leading to an increased risk of death. Cancer staging is currently the gold standard for establishing a patient's prognosis and can help clinicians determine patients who may benefit from additional therapy. However, medical images which are used to help determine the cancer stage, have been shown to hold unutilized prognostic information that can augment clinical data and better identify high-risk NSCLC patients. There remains an unmet need for models to incorporate clinical, pathological, surgical, and imaging information, and extend beyond the current staging system to assist clinicians in identifying patients who could benefit from additional therapy immediately after surgery. PURPOSE We aimed to determine whether a deep learning model (DLM) integrating FDG PET and CT imaging from the thoracic cavity along with clinical, surgical, and pathological information can predict NSCLC recurrence-free survival (RFS) and stratify patients into risk groups better than conventional staging. MATERIALS AND METHODS Surgically resected NSCLC patients enrolled between 2009 and 2018 were retrospectively analyzed from two academic institutions (local institution: 305 patients; external validation: 195 patients). The thoracic cavity (including the lungs, mediastinum, pleural interfaces, and thoracic vertebrae) was delineated on the preoperative FDG PET and CT images and combined with each patient's clinical, surgical, and pathological information. Using the local cohort of patients, a multi-modal DLM using these features was built in a training cohort (n = 225), tuned on a validation cohort (n = 45), and evaluated on testing (n = 35) and external validation (n = 195) cohorts to predict RFS and stratify patients into risk groups. The area under the curve (AUC), Kaplan-Meier curves, and log-rank test were used to assess the prognostic value of the model. The DLM's stratification performance was compared to the conventional staging stratification. RESULTS The multi-modal DLM incorporating imaging, pathological, surgical, and clinical data predicted RFS in the testing cohort (AUC = 0.78 [95% CI:0.63-0.94]) and external validation cohort (AUC = 0.66 [95% CI:0.58-0.73]). The DLM significantly stratified patients into high, medium, and low-risk groups of RFS in both the testing and external validation cohorts (multivariable log-rank p < 0.001) and outperformed conventional staging. Conventional staging was unable to stratify patients into three distinct risk groups of RFS (testing: p = 0.94; external validation: p = 0.38). Lastly, the DLM displayed the ability to further stratify patients significantly into sub-risk groups within each stage in the testing (stage I: p = 0.02, stage II: p = 0.03) and external validation (stage I: p = 0.05, stage II: p = 0.03) cohorts. CONCLUSION This is the first study to use multi-modality imaging along with clinical, surgical, and pathological data to predict RFS of NSCLC patients after surgery. The multi-modal DLM better stratified patients into risk groups of poor outcomes when compared to conventional staging and further stratified patients within each staging classification. This model has the potential to assist clinicians in better identifying patients that may benefit from additional therapy.
Collapse
Affiliation(s)
- Jaryd R Christie
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Baines Imaging Research Laboratory, London Health Sciences Center, London, Ontario, Canada
| | | | - Karen Eddy
- Baines Imaging Research Laboratory, London Health Sciences Center, London, Ontario, Canada
| | - Delphine L Chen
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Omar Daher
- Department of Medical Imaging, Western University, London, Ontario, Canada
| | - Mohamed Abdelrazek
- Department of Medical Imaging, Western University, London, Ontario, Canada
| | - Richard A Malthaner
- Division of Thoracic Surgery, Department of Surgery, Western University, London, Ontario, Canada
| | - Mehdi Qiabi
- Division of Thoracic Surgery, Department of Surgery, Western University, London, Ontario, Canada
| | - Rahul Nayak
- Division of Thoracic Surgery, Department of Surgery, Western University, London, Ontario, Canada
| | - Paul Kinahan
- Department of Radiology, University of Washington, Seattle, Washington, USA
| | - Viswam S Nair
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Pulmonary, Critical Care & Sleep Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| | - Sarah A Mattonen
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Baines Imaging Research Laboratory, London Health Sciences Center, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
| |
Collapse
|
3
|
Li L, Wu J, Wu X, Li Z, Zhang X, Yan Z, Liang Y, Huang C, Qu S. Carbon Dot-Linked Hydrogel for TAMs Transform: Spatiotemporal Manipulation to Reshape Tumor Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2420068. [PMID: 40181663 PMCID: PMC12087752 DOI: 10.1002/adma.202420068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/20/2025] [Indexed: 04/05/2025]
Abstract
As one of the most crucial immune cells in the tumor microenvironment (TME), regulating tumor-associated macrophages (TAMs) is vital for enhancing antitumor immunity. Here, an injectable carbon dots (CDs)-linked egg white hydrogel was developed, termed TAMs Transform Factory (TTF-L-C), to spatiotemporally manipulate TAMs. The fabricated CDs significantly promoted macrophage migration. Notably, TTF-L-C achieved macrophage spatial enrichment through CDs-induced directional recruitment with molecular Ctnnd1 upregulation. Subsequently, the recruited macrophages were locoregionally reprogrammed within TTF-L-C, as well as blocking the upregulated PD-L1. Finally, through multi-stage regulation at spatial, cellular, and molecular levels, TTF-L-C released immune-activated M1 macrophages to the tumor site as it degraded. Moreover, TTF-L-C promoted dendritic cell (DCs) maturation and further boosted T cell activation, thereby reshaping the tumor-suppressive TME. Through peritumoral injection, TTF-L-C enhanced tumor immunotherapy in both subcutaneous and recurrent 4T1 tumor models with satisfactory biosafety. Therefore, TTF-L-C is proposed to become a safe and powerful platform for various biomedical applications.
Collapse
Affiliation(s)
- Lingyun Li
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Jun Wu
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Xue Wu
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Zhenjian Li
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Xianming Zhang
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Zekun Yan
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Yingqi Liang
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Caishi Huang
- Faculty of Health ScienceUniversity of MacauTaipaMacau SAR999078P. R. China
| | - Songnan Qu
- Joint Key Laboratory of the Ministry of EducationInstitute of Applied Physics and Materials EngineeringUniversity of MacauTaipaMacau SAR999078P. R. China
- Department of Physics and ChemistryFaculty of Science and Technology University of MacauMacao SAR999078P. R. China
- MOE Frontier Science Centre for Precision Oncology University of MacauMacao SAR999078P. R. China
- Zhuhai UM Science and Technology Research InstituteZhuhai519031P. R. China
| |
Collapse
|
4
|
Zhou JJ, Feng YC, Zhao ML, Guo Q, Zhao XB. Nanotechnology-driven strategies in postoperative cancer treatment: innovations in drug delivery systems. Front Pharmacol 2025; 16:1586948. [PMID: 40371327 PMCID: PMC12075547 DOI: 10.3389/fphar.2025.1586948] [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: 03/03/2025] [Accepted: 04/23/2025] [Indexed: 05/16/2025] Open
Abstract
Cancer remains a global health challenge, and this challenge comes with a significant burden. Current treatment modalities, such as surgery, chemotherapy, and radiotherapy, have their limitations. The emergence of nanomedicines presents a new frontier in postoperative cancer treatment, offering potential to inhibit tumor recurrence and manage postoperative complications. This review deeply explores the application and potential of nanomedicines in the treatment of cancer after surgery. In particular, it focuses on local drug delivery systems (LDDS), which consist of in situ injection, implantation, and spraying. LDDS can provide targeted drug delivery and controlled release, which enhancing therapeutic efficacy. At the same time, it minimizes damage to healthy tissues and reduces systemic side effects. The nanostructures of these systems are unique. They facilitate the sustained release of drugs, prolong the effects of treatment, and decrease the frequency of dosing. This is especially beneficial in the postoperative period. Despite their potential, nanomedicines have limitations. These include high production costs, concerns regarding long-term toxicity, and complex regulatory approval processes. This paper aims to analyze several aspects. These include the advantages of nanomedicines, their drug delivery systems, how they combine with multiple treatment methods, and the associated challenges. Future research should focus on certain issues. These issues are stability, tumor specificity, and clinical translation. By addressing these, the delivery methods can be optimized and their therapeutic efficacy enhanced. With the advancements in materials science and biomedical engineering, the future design of LDDS is set to become more intelligent and personalized. It will cater to the diverse needs of clinical treatment and offer hope for better outcomes in cancer patients after surgery.
Collapse
Affiliation(s)
- Jun-Jie Zhou
- The Stomatological Hospital, Anyang Sixth People’s Hospital, Anyang, China
| | | | | | | | | |
Collapse
|
5
|
Luo Z, Li X, Zhu D, Fu W, Liu Y, Zheng L, Chen P, Gong C, Liu X. Implantable Immunostimulant Microneedle Patch for Post-Surgical Prevention of Cancer Recurrence and Distant Tumor Inhibition. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22362-22374. [PMID: 40194999 DOI: 10.1021/acsami.5c01155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cancer recurrence after surgical resection remains a grand challenge in achieving long-term eradication. Here, we develop a biocompatible and implantable immunostimulant microneedle patch designed to suppress local tumor recurrence after surgery. The patch, fabricated using methacrylate-modified hyaluronic acid, incorporates 2'3'-cGAMP, a STING agonist, and IL-2, a cytokine approved for clinical cancer immunotherapy that expands T cells. The patch enables controlled release of cGAMP to induce dendritic cell maturation, antitumor macrophage polarization (M1 macrophage), and T cell priming and activation. Simultaneously, localized IL-2 activates CD8+ T cells and recruits immune cells to the tumor microenvironment. When combined with an anti-CTLA-4 antibody, an immune checkpoint blockade, the hybrid microneedle patch significantly reduces Treg cells at the surgery sites, enhancing immune responses and effectively inhibiting the progression of distant tumors in both prophylactic and therapeutic models. Compared with traditional postsurgical chemotherapy and radiotherapy, this patch-mediated immunotherapy demonstrates superior efficacy in mitigating tumor relapse while offering higher biocompatibility. Our findings suggest that this immunotherapeutic patch has potential as a translational tool to prevent cancer recurrence in patients with resectable tumors.
Collapse
Affiliation(s)
- Zichao Luo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031 China
- NHC Key laboratory of Myopia and Related Eye Diseases; Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, Shanghai 200031 China
- Shanghai Research Center of Ophthalmology and Optometry, Shanghai, 200031 China
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
| | - Xinchao Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dandan Zhu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
| | - Wangxian Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxia Liu
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
| | - Lewen Zheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 637457 Singapore
- Skin Research Institute of Singapore, 308232 Singapore
- Lee Kong Chian School of Medicine, Institute for Digital Molecular Analytics and Science, Nanyang Technological University, 636921 Singapore
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaogang Liu
- Department of Chemistry and the N.1 Institute for Health, National University of Singapore, 117543 Singapore
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research, 138634, Singapore
| |
Collapse
|
6
|
Xie RL, Cai WL, Ouyang YF, Zou YH, Sun R. Effect and safety of immunotherapy among elder patients (age ≥ 65) with recurrent or metastatic nasopharyngeal carcinoma. BMC Cancer 2025; 25:691. [PMID: 40229737 PMCID: PMC11998246 DOI: 10.1186/s12885-025-14108-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The use of immune checkpoint inhibitors (ICIs) combined with chemotherapy constitutes the first-line treatment for recurrent or metastatic nasopharyngeal carcinoma (RM-NPC). However, elderly patients are underrepresented in the majority of related clinical trials. PATIENTS AND METHODS This retrospective study included RM-NPC patients aged 65 years or older who received immunotherapy between January 2015 and February 2022. Cox regression models were utilized to compare the outcomes. Comorbidity assessments (ACE-27, CCI, and ACCI) were used for the geriatric evaluation. RESULTS Among the 95 of 243 patients included in this analysis (71 men), the median follow-up time was 29.3 months. Patients receiving local therapy had longer progression-free survival (PFS) (HR 0.352; 95% CI: 0.145-0.853; p = .021). No significant differences in survival outcomes or toxicity profiles were observed between age groups or among the ICI agent groups. CONCLUSIONS The findings suggest that immunotherapy is efficacious and safe for treating RM-NPC in elderly patients. The combination of ICIs and local therapy significantly prolonged survival and could be an option for this vulnerable population.
Collapse
Affiliation(s)
- Rui-Ling Xie
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Wu-Lin Cai
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yan-Feng Ouyang
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ye-Hao Zou
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Rui Sun
- Department of Nasopharyngeal Carcinoma, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
7
|
Acuña MI, Lamirault C, Larcher T, Brisebard E, Schneider T, Juchaux M, Iglesias-Rey R, Fernández-Rodicio S, Aguiar P, Gómez-Lado N, Martínez-Rovira I, González-Vegas R, Yousef I, Gomez-Caamano A, Pombar M, Luna V, Sanchez M, Prezado Y. Mini-GRID therapy delivers optimised spatially fractionated radiation therapy using a flattening free filter accelerator. COMMUNICATIONS MEDICINE 2025; 5:101. [PMID: 40188304 PMCID: PMC11972377 DOI: 10.1038/s43856-025-00809-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 03/14/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Radioresistant tumours remain a challenge for conventional radiation therapy (RT), and often, only palliative treatment can be offered. Recently developed techniques, such as spatially fractionated radiation therapy (SFRT) could potentially improve treatment. However, current clinical SFRT implementations do not allow the full potential to be exploited. We further optimize SFRT, developing mini-GRID, which uses a flattening free filter accelerator. METHODS The increase in normal tissue tolerances provided by mini-GRID compared to conventional RT and GRID therapy was validated in a rat model of brain irradiation in a longitudinal imaging study, behavioural tests and by histopathological evaluation. RESULTS The implementation optimizes mini-GRID therapy, with beam widths around 2 mm2. The peak-to-valley dose ratios and peak dose rates are around 4 and 7 Gy/min, respectively. Mini-GRID RT allows the use of high peak doses: 42 Gy in one fraction, a factor more than twice higher than the peak doses generally employed in conventional GRID therapy (20 Gy peak dose). This enables the use of more aggressive and potentially curative treatments. Infrared microspectroscopy analysis suggests different early biochemical changes in both modalities, with conventional RT leading to stronger modifications in the secondary protein structure, and higher oxidative damage than mini-GRID RT. CONCLUSIONS The possibility to treat both large and small tumours, and to perform safe and potentially curative dose escalations in previously untreatable cases, makes mini-GRID a promising approach to expand the clinical use of SFRT.
Collapse
Affiliation(s)
- M Isabel Acuña
- New Approaches in Radiotherapy Lab, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, A Coruña, Spain
| | - Charlotte Lamirault
- Translational Research Department, Institut Curie, Experimental Radiotherapy Platform, Université Paris Saclay, Orsay, France
| | | | | | - Tim Schneider
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Marjorie Juchaux
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Ramon Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Pablo Aguiar
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Santiago de Compostela, Spain
| | - Noemi Gómez-Lado
- Molecular Imaging and Pharmacokinetic Modelling Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Spain; Nuclear Medicine and Molecular Imaging Group, Health Research Institute of Santiago de Compostela (IDIS), University Hospital Santiago de Compostela, Santiago de Compostela, Spain
| | - Immaculada Martínez-Rovira
- Physics Department, Universitat Autònoma de Barcelona (UAB), 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Roberto González-Vegas
- Physics Department, Universitat Autònoma de Barcelona (UAB), 08193 Cerdanyola del Vallès, Barcelona, Spain
| | - Ibraheem Yousef
- MIRAS Beamline, ALBA Synchrotron, 08209 Cerdanyola del Vallès, Barcelona, Spain
| | - Antonio Gomez-Caamano
- Department of Radiation Oncology, Hospital Clínico Universitario Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Pombar
- Department of Medical Physics, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Victor Luna
- Department of Medical Physics, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Sanchez
- Department of Medical Physics, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Yolanda Prezado
- New Approaches in Radiotherapy Lab, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, A Coruña, Spain.
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France.
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France.
- Oportunius Program, Galician Agency of Innovation (GAIN), Xunta de Galicia, A Coruña, Spain.
| |
Collapse
|
8
|
Verginadis II, Velalopoulou A, Kim MM, Kim K, Paraskevaidis I, Bell B, Oliaei Motlagh SA, Karaj A, Banerjee E, Finesso G, Assenmacher CA, Radaelli E, Lu J, Lin Y, Putt ME, Diffenderfer ES, Guha C, Qin L, Metz JM, Maity A, Cengel KA, Koumenis C, Busch TM. FLASH proton reirradiation, with or without hypofractionation, reduces chronic toxicity in the normal murine intestine, skin, and bone. Radiother Oncol 2025; 205:110744. [PMID: 39880309 DOI: 10.1016/j.radonc.2025.110744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND AND PURPOSE The normal tissue sparing afforded by FLASH radiotherapy is being intensely investigated for potential clinical translation. Here, we studied the effects of FLASH proton radiotherapy (F-PRT) in the reirradiation setting, with or without hypofractionation. Chronic toxicities in three murine models of normal tissue toxicity including the intestine, skin, and bone were investigated. MATERIALS AND METHODS In studies of the intestine, single-dose irradiation was performed with 12 Gy of standard proton RT (S-PRT), followed by a second dose of 12 Gy of F-PRT or S-PRT. Additionally, a hypofractionation scheme was applied in the reirradiation setting (3 x 6.4 Gy of F-PRT or S-PRT, given every 48 hrs). In studies of skin/bone of the murine leg, 15 Gy of S-PRT was followed by hypofractionated reirradiation with F-PRT or S-PRT (3 x 11 Gy). RESULTS Compared to reirradiation with S-PRT, F-PRT induced less intestinal fibrosis and collagen deposition that was accompanied by significantly increased survival rate, demonstrating its protective effects on intestinal tissues in the reirradiation setting. In previously irradiated leg tissues, reirradiation with hypofractionated F-PRT created transient dermatitis that fully resolved in contrast to reirradiation with hypofractionated S-PRT. Lymphedema was also alleviated after a second course of radiation with F-PRT, along with significant reductions in the accumulation of fibrous connective tissue in the skin, compared to mice reirradiated with S-PRT. The delivery of a second course of fractionated S-PRT induced tibial fractures in 83.3% of the mice, whereas only 20% of mice reirradiated with F-PRT presented with fractures. CONCLUSION These studies provide the first evidence of the sparing effects of F-PRT in the setting of hypofractionated reirradiation. The results support FLASH as highly relevant to the reirradiation regimen where it exhibits significant potential to minimize chronic complications for patients undergoing RT.
Collapse
Affiliation(s)
- Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michele M Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyle Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis Paraskevaidis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brett Bell
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Antoneta Karaj
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Esha Banerjee
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giovanni Finesso
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiawei Lu
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuewei Lin
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary E Putt
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric S Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ling Qin
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Metz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Maity
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Zhang L, Wang S, Hou Y. Magnetic Micro/nanorobots in Cancer Theranostics: From Designed Fabrication to Diverse Applications. ACS NANO 2025; 19:7444-7481. [PMID: 39970007 DOI: 10.1021/acsnano.4c10382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cancer poses a substantial threat and a serious challenge to public human health, driving the promotion of sophisticated technologies for cancer therapy. While conventional chemotherapy has bottlenecks such as low delivery efficiency, strong toxic side effects, and tumor enrichment barriers, magnetic micro/nanorobots (MNRs) emerge as promising therapeutic candidates that provide alternative strategies for cancer therapy. MNR is a kind of human-made machine that is micro- or nanosized, is reasonably designed, and performs command tasks through self-actuated or externally controlled propulsion mechanisms, which can be potentially applied in cancer theranostics. Here, this review first introduces the components that constitute a typical magnetic MNR, including the body part, the driving part, the control part, the function part, and the sensing part. Subsequently, this review elucidates representative fabrication methods to construct magnetic MNRs from top-down approaches to bottom-up approaches, covering injection molding, self-rolling, melt electrospinning writing, deposition, biotemplate method, lithography, assembling, 3D printing, and chemical synthesis. Furthermore, this review focuses on multiple applications of magnetic MNRs facing cancer diagnosis and treatment, encompassing imaging, quantification, drug release, synergy with typical therapies, cell manipulation, and surgical assistance. Then, this review systematically elaborates on the biocompatibility and biosafety of magnetic MNRs. Finally, the challenges faced by magnetic MNRs are discussed alongside future research directions. This review is intended to provide scientific guidance that may improve the comprehension and cognition of cancer theranostics through the platform of magnetic MNRs, promoting and prospering the practical application development of magnetic MNRs.
Collapse
Affiliation(s)
- Lin Zhang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Shuren Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Yanglong Hou
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
- School of Materials, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
10
|
Li L, Zhu C, Yan Q, Li J, Chen Y, Hu X. Effectiveness of Dyadic Interventions on Quality of Life for Cancer Patients and Family Caregivers: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. J Clin Nurs 2025. [PMID: 39972207 DOI: 10.1111/jocn.17700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 01/24/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Increases in cancer survivorship negatively impact patients and family caregivers, decreasing quality of life. Previous dyadic interventions involved them as a unit and focused on their outcomes, but inconsistent results existed in influencing quality of life. OBJECTIVES To assess dyadic intervention effect on quality of life for cancer patients and family caregivers across different cancer types and intervention durations. DESIGN A systematic review and meta-analysis based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). METHODS Six databases were searched from establishment until 14 January 2024. Two authors independently performed the search process, literature screening, and data extraction. The ROB version 2 and GRADE were respectively used to check the methodology and evidence quality. The data were analysed via RStudio, and intervention effects were estimated with 95% CIs and SMDs. The statistical heterogeneity was explored through the I2 statistic, P values, and Egger's test, and differences in overall effects were deemed statistically significant, having a P value < 0.05. Subgroup analysis was also conducted. RESULTS 13 RCTs with 1625 participants, published from 2005 to 2021, were included. The results demonstrated that dyadic interventions enhanced quality of life for both cancer patients and family caregivers. Subgroup analysis suggested that family-centred interventions for patients with specific cancer types, which lasted for a long period (> 6 weeks), enhanced quality of life for cancer patients and family caregivers. The evidence and methodology were of a moderate quality. CONCLUSIONS Nurses are important practitioners of culture-oriented dyadic interventions. Long-term (> 6 weeks) and family-centred dyadic interventions for patients with a specific cancer type can enhance cancer patients' and family caregivers' quality of life, along with digital intelligence approaches to promote mutual communication and strengthen family relationships, thereby optimising oncology clinical nursing and enhancing the quality of life, health, and welfare of the entire family. RELEVANCE TO CLINICAL PRACTICE Dyadic interventions emphasising the involvement of both cancer patients and family caregivers should be considered and tailored by professionals and oncology nurses to establish harmonious family relationships, improve family coping techniques and decision-making to enhance the whole family's quality of life and well-being according to their cultural contexts, and promote more efficient, targeted, and economical oncology care. PATIENT OR PUBLIC CONTRIBUTION No Patient or Public Contribution because all the involved participants were from existing studies, and the design, conduction, analysis, and interpretation of the data were completed by the authors in this article. TRIAL REGISTRATION International Prospective Register of Systematic Reviews: CRD42024519432; https://www.crd.york.ac.uk/PROSPERO/#recordDetails.
Collapse
Affiliation(s)
- Linna Li
- Department of Nursing, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Chuanmei Zhu
- Outpatient Department, West China Hospital, Sichuan University, Chengdu, China
| | - Qianwen Yan
- Department of Nursing, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Juejin Li
- Department of Nursing, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yang Chen
- Department of Nursing, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Xiaolin Hu
- Department of Nursing, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, China
| |
Collapse
|
11
|
Wang C, Li J, Chen J, Wang Z, Zhu G, Song L, Wu J, Li C, Qiu R, Chen X, Zhang L, Li W. Multi-omics analyses reveal biological and clinical insights in recurrent stage I non-small cell lung cancer. Nat Commun 2025; 16:1477. [PMID: 39929832 PMCID: PMC11811181 DOI: 10.1038/s41467-024-55068-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/26/2024] [Indexed: 02/13/2025] Open
Abstract
Post-operative recurrence rates of stage I non-small cell lung cancer (NSCLC) range from 20% to 40%. Nonetheless, the molecular mechanisms underlying recurrence hitherto remain largely elusive. Here, we generate genomic, epigenomic and transcriptomic profiles of paired tumors and adjacent tissues from 122 stage I NSCLC patients, among which 57 patients develop recurrence after surgery during follow-up. Integrated analyses illustrate that the presence of predominantly solid or micropapillary histological subtypes, increased genomic instability, and APOBEC-related signature are associated with recurrence. Furthermore, TP53 missense mutation in DNA-binding domain could contribute to shorter time to recurrence. DNA hypomethylation is pronounced in recurrent NSCLC, and PRAME is the significantly hypomethylated and overexpressed gene in recurrent lung adenocarcinoma (LUAD). Mechanistically, hypomethylation at TEAD1 binding site facilitates the transcriptional activation of PRAME. Inhibition of PRAME restrains the tumor metastasis via downregulation of epithelial-mesenchymal transition-related genes. We also identify that enrichment of AT2 cells with higher copy number variation burden, exhausted CD8 + T cells and Macro_SPP1, along with the reduced interaction between AT2 and immune cells, is essential for the formation of ecosystem in recurrent LUAD. Finally, multi-omics clustering could stratify the NSCLC patients into 4 subclusters with varying recurrence risk and subcluster-specific therapeutic vulnerabilities. Collectively, this study constitutes a promising resource enabling insights into the biological mechanisms and clinical management for post-operative recurrence of stage I NSCLC.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Jingwei Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyao Chen
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhoufeng Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guonian Zhu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lujia Song
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Changshu Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Qiu
- Department of Respiratory and Critical Care Medicine, Suining Central Hospital, Suining, China
| | - Xuelan Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, Sichuan, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Precision Therapeutics, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Wang SF, Pan XB, Huang W, Zhao YN. Postoperative radiotherapy for stage pIIIA-N2 non-small cell lung cancer patients undergoing sublobar resection: A retrospective cohort study. Medicine (Baltimore) 2025; 104:e41462. [PMID: 39928784 PMCID: PMC11813015 DOI: 10.1097/md.0000000000041462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/02/2024] [Accepted: 01/17/2025] [Indexed: 02/12/2025] Open
Abstract
This study assesses the effect of postoperative radiotherapy on survival outcomes in patients diagnosed with stage pIIIA-N2 non-small cell lung cancer (NSCLC) after sublobar resection. Data of patients with stage pIIIA-N2 NSCLC who underwent sublobar resection were extracted from the Surveillance, Epidemiology, and End Results database spanning from 2000 to 2020. Patients were divided into 2 groups: postoperative radiotherapy and observation. Cancer-specific survival (CSS) and overall survival (OS) were analyzed and compared between the 2 groups. A total of 444 patients were included in the study, with 210 (47.3%) receiving postoperative radiotherapy and 234 (52.7%) with observation. The CSS (hazard ratio [HR] = 0.99, 95% confidence interval [CI]: 0.78-1.26; P = .926) and OS (HR = 0.93, 95% CI: 0.75-1.15; P = .512) did not show significant differences between the postoperative radiotherapy and observation groups. Subgroup analysis of patients receiving postoperative chemotherapy revealed comparable CSS (HR = 1.24, 95% CI: 0.89-1.71; P = .203) and OS (HR = 1.12, 95% CI: 0.85-1.49; P = .425) between the 2 groups. Similarly, for patients without postoperative chemotherapy, CSS (HR = 1.11, 95% CI: 0.66-1.84; P = .699) and OS (HR = 1.08, 95% CI: 0.68-1.71; P = .740) were not significantly different between the 2 groups. Postoperative radiotherapy does not improve survival in patients with stage pIIIA-N2 NSCLC following sublobar resection.
Collapse
Affiliation(s)
- Shou-Feng Wang
- Department of Thoracic Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, P.R. China
| | - Xin-Bin Pan
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, P.R. China
| | - Wei Huang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, P.R. China
| | - Yin-Nong Zhao
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, P.R. China
| |
Collapse
|
13
|
Rao Z, Zhu Y, Chen Z, Luo Y, Yang Z, Liu W, Qiao C, Xia Y, Yang P, Ye D, Wang Z. Injectable Autocatalytic Hydrogel Triggers Pyroptosis to Stimulate Anticancer Immune Response for Preventing Postoperative Tumor Recurrence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408415. [PMID: 39465669 PMCID: PMC11714207 DOI: 10.1002/advs.202408415] [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: 07/23/2024] [Revised: 09/14/2024] [Indexed: 10/29/2024]
Abstract
Modulating immunosuppression while eliminating residual microscopic tumors is critical for inhibiting the postoperative recurrence of triple-negative breast cancer (TNBC). Although immunotherapy has shown potential in achieving this goal, due to multiple immunosuppression and poor immunogenicity of apoptosis, a satisfactory anti-recurrence effect still faces the challenge. Herein, an injectable hydrogel-encapsulated autocatalytic copper peroxide (CP@Gel) therapeutic platform is designed and combine it with the clinical-grade DNA methyltransferase inhibitor decitabine (DAC) to effectively inhibit TNBC growth and postoperative recurrence via pyroptosis, killing residual cancer cells that bypass apoptosis resistance while also improving immunogenicity and modulating immunosuppression to achieve an intense anti-tumor immune response. Following injection of the CP@Gel, the sustained release of CP leads to the autocatalytic generation of reactive oxygen species, resulting in caspase-3 activation, and the pre-administered DAC inhibits the methylation of Gsdme to elevate the GSDME protein levels, leading to intense pyroptosis and anti-tumor immune responses. The in vivo results show a 67% elimination of local tumor recurrence via treatment with DAC+CP@Gel, suggesting the successful integration of sustained drug release with autocatalysis and epigenetic modification. The results thus suggest great potential for pyroptosis-based and injectable hydrogel-aided strategies for preventing the postoperative recurrence of TNBC.
Collapse
Affiliation(s)
- Zhiping Rao
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Yutong Zhu
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
- Medical collegeXi'an International UniversityXi'anShaanxi710077P. R. China
| | - Zhuang Chen
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Yi Luo
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Zuo Yang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Weijing Liu
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Chaoqiang Qiao
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Yuqiong Xia
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Peng Yang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| | - Dong‐Man Ye
- Department of Medical ImagingCancer Hospital of China Medical UniversityLiaoning Cancer Hospital & InstituteShenyangLiaoning110042P. R. China
| | - Zhongliang Wang
- Lab of Molecular Imaging and Translational Medicine (MITM)Engineering Research Center of Molecular & NeuroimagingMinistry of EducationSchool of Life Science and TechnologyXidian University & International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and TreatmentXi'anShaanxi710126P. R. China
| |
Collapse
|
14
|
Qadir A, Singh N, Moe AAK, Cahoon G, Lye J, Chao M, Foroudi F, Uribe S. Potential of MRI in Assessing Treatment Response After Neoadjuvant Radiation Therapy Treatment in Breast Cancer Patients: A Scoping Review. Clin Breast Cancer 2025; 25:e1-e9.e2. [PMID: 38906720 DOI: 10.1016/j.clbc.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/07/2024] [Accepted: 05/26/2024] [Indexed: 06/23/2024]
Abstract
The objective of this scoping review is to evaluate the potential of Magnetic Resonance Imaging (MRI) and to determine which of the available MRI techniques reported in the literature are the most promising for assessing treatment response in breast cancer patients following neoadjuvant radiotherapy (NRT). Ovid Medline, Embase, CINAHL, and Cochrane databases were searched to identify relevant studies published from inception until March 13, 2023. After primary selection, 2 reviewers evaluated each study using a standardized data extraction template, guided by set inclusion and exclusion criteria. A total of 5 eligible studies were selected. The positive and negative predictive values for MRI predicting pathological complete response across the studies were 67% to 88% and 76% to 85%, respectively. MRI's potential in assessing postradiotherapy tumor sizes was greater for volume measurements than uni-dimensional longest diameter measurements; however, overestimation in surgical tumor sizes was observed. Apparent diffusion coefficient (ADC) values and Time to Enhance (TTE) was seen to increase post-NRT, with a notable difference between responders and nonresponders at 6 months, indicating a potential role in assessing treatment response. In conclusion, this review highlights tumor volume measurements, ADC, and TTE as promising MRI metrics for assessing treatment response post-NRT in breast cancer. However, further research with larger cohorts is needed to confirm their utility. If MRI can accurately identify responders from nonresponders to NRT, it could enable a more personalized and tailored treatment approach, potentially minimizing radiation therapy related toxicity and enhancing cosmetic outcomes.
Collapse
Affiliation(s)
- Ayyaz Qadir
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Melbourne, Australia.
| | - Nabita Singh
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Melbourne, Australia
| | - Aung Aung Kywe Moe
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Melbourne, Australia
| | - Glenn Cahoon
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Heidelberg, Victoria, Australia
| | - Jessica Lye
- Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Heidelberg, Victoria, Australia
| | - Michael Chao
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Melbourne, Australia; Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Heidelberg, Victoria, Australia
| | - Farshad Foroudi
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Melbourne, Australia; Department of Radiation Oncology, Olivia Newton-John Cancer Wellness and Research Centre, Austin Health, Heidelberg, Victoria, Australia
| | - Sergio Uribe
- Department of Medical Imaging and Radiation Sciences, School of Primary and Allied Health Care, Monash University, Melbourne, Australia
| |
Collapse
|
15
|
Li L, Xing Y, Chen Y, Li K, Wu Y, Cai K, Wang L, Zhang J. Flower-Like Nanosensors for Photoacoustic-Enhanced Lysosomal Escape and Cytoplasmic Marker-Activated Fluorescence: Enabling High-Contrast Identification and Photothermal Ablation of Minimal Residual Disease in Breast Cancer. Adv Healthc Mater 2025; 14:e2403042. [PMID: 39580677 DOI: 10.1002/adhm.202403042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/31/2024] [Indexed: 11/26/2024]
Abstract
The clearance of minimal residual disease (MRD) after breast cancer surgery is crucial for inhibiting metastasis and recurrence. However, the most promising biomarker-activated fluorescence imaging strategies encounter accessibility issues of the delivered sensors to cytoplasmic targets. Herein, a flower-like composite nanosensor with photoacoustic (PA) effect-enhanced lysosomal escape and cytoplasmic marker-activated fluorescence is developed to address this challenge. Specifically, the incorporation of Co2+ into the synthesis of 2D Zn2+-derived metal-organic frameworks enabled rapid dopamine polymerization and deposition. Subsequently, the composite nanoflower (FHN), characterized by an average size of ≈80 nm and petal thickness of ≈6 nm, is formed through the sealing of micropores and simultaneous cross-linking of nanosheets. The pronounced reduction in thermal conductivity of FHN, and superposition of interpetal thermal fields under a pulsed laser (PL), lead to enhanced PA effect and membrane permeability. Thereby, nanosensors efficiently escape from lysosomes resulting in synergistic fluorescence activation by dual-factors (ATP, miRNA-21) and DNA probes installed on FHN. A subsequently high tumor-to-normal tissue signal ratio (TNR) of 17.4 lead to precise guidance of NIR irradiation for efficient MRD eradication and recurrence inhibition. This study provides a new approach for high-contrast identification and precise ablation of MRD based on the synergistic response of endogenous and exogenous factors.
Collapse
Affiliation(s)
- Lin Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Yuxin Xing
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Yuhua Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Kunlin Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Yunyun Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| | - Lu Wang
- Department of Clinical Laboratory, Laboratory Medicine Center, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), No.158 Shangtang Road, Hangzhou, Zhejiang, 310014, China
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, No.8 Yikang Road, Hangzhou, Zhejiang, 311399, China
| | - Jixi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing, 400044, China
| |
Collapse
|
16
|
Kang S, Mansurov A, Kurtanich T, Chun HR, Slezak AJ, Volpatti LR, Chang K, Wang T, Alpar AT, Refvik KC, Hansen OI, Borjas GJ, Berg BTK, Shim HN, Hultgren KT, Gomes S, Wang Y, Solanki A, Ishihara J, Swartz MA, Hubbell JA. Engineered GM-CSF polarizes protumorigenic tumor-associated macrophages to an antitumorigenic phenotype and potently synergizes with IL-12 immunotherapy. J Immunother Cancer 2024; 12:e009541. [PMID: 39794939 PMCID: PMC11667343 DOI: 10.1136/jitc-2024-009541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/18/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND The use of immune checkpoint inhibitors (CPIs) has become a dominant regimen in modern cancer therapy, however immune resistance induced by tumor-associated macrophages (TAMs) with immune suppressive and evasion properties limits responses. Therefore, the rational design of immune modulators that can control the immune suppressive properties of TAMs and polarize them, as well as dendritic cells (DCs), toward a more proinflammatory phenotype is a principal objective in cancer immunotherapy. METHODS Here, using a protein engineering approach to enhance cytokine residence in the tumor microenvironment, we examined combined stimulation of the myeloid compartment via tumor stroma-binding granulocyte-macrophage colony-stimulating factor (GM-CSF) to enhance responses in both DCs and T cells via stroma-binding interleukin-12 (IL-12). We evaluated tumor responses at the levels of outcome, cellular responses, and cytokine responses in both the tumors and the tumor-draining lymph nodes. We further investigated the potentiation of DC response to IL-12 by GM-CSF stimulation ex vivo. RESULTS Engineered GM-CSF restored an antitumorigenic tumor myeloid microenvironment otherwise suppressed by TAMs, while engineered IL-12 provided effector signals to T cells, thereby boosting both tumor-resident antitumor macrophage and CD8+ T cell populations. Furthermore, engineered GM-CSF potentiated DC response to IL-12, upregulating DC expression of IL-12 receptor and enhancing their expression of proinflammatory cytokines and chemokines on IL-12 exposure. This resulted in remarkable synergistic efficacy in multiple solid tumor models treated with the dual cytokine combination. The combination therapy also improved the efficacy of CPI in a CPI-resistant genetically-engineered melanoma model and exhibited synergistic antitumor efficacy in a pulmonary metastasis model. CONCLUSION Our strategy provides a rational design for combination immunotherapy targeting both myeloid and lymphoid compartments through complementary mechanisms.
Collapse
Affiliation(s)
- Seounghun Kang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Trevin Kurtanich
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Hye Rin Chun
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
| | - Anna J Slezak
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Lisa R Volpatti
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Kevin Chang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Medical Scientist Training Program, The University of Chicago, Chicago, Illinois, USA
| | - Thomas Wang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Aaron T Alpar
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Kirsten C Refvik
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - O Isabella Hansen
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Gustavo J Borjas
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Brendan T K Berg
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Medical Scientist Training Program, The University of Chicago, Chicago, Illinois, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Kevin T Hultgren
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Yue Wang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
| | - Ani Solanki
- Animal Resource Center, The University of Chicago, Chicago, Illinois, USA
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, UK
| | - Melody A Swartz
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
- Committee on Cancer Biology, The University of Chicago, Chicago, Illinois, USA
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, New York, New York, USA
| |
Collapse
|
17
|
Cuiffo B, Maxwell M, Yan D, Guemiri R, Boone A, Bellet D, Rivest B, Cardia J, Robert C, Fricker SP. Self-delivering RNAi immunotherapeutic PH-762 silences PD-1 to generate local and abscopal antitumor efficacy. Front Immunol 2024; 15:1501679. [PMID: 39697325 PMCID: PMC11652358 DOI: 10.3389/fimmu.2024.1501679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
Objective Immunotherapeutic inhibition of PD-1 by systemically administered monoclonal antibodies is widely used in cancer treatment, but it may cause severe immune-related adverse events (irSAEs). Neoadjuvant PD-1 inhibition before surgery has shown promise in reducing recurrence by stimulating durable antitumor immunity. Local intratumoral (IT) immunotherapy is a potential strategy to minimize irSAEs, but antibodies have limited tumor penetration, making them less suitable for this approach. Therapeutic self-delivering RNAi (INTASYL) is an emerging modality well-suited for neoadjuvant immunotherapy. This study presents preclinical proof-of-concept for PH-762, an INTASYL designed to silence PD-1, currently in clinical development for advanced cutaneous malignancies (ClinicalTrials.gov#NCT06014086). Methods and analysis PH-762 pharmacology was characterized in vitro, and in vivo antitumor efficacy was evaluated using a murine analogue (mPH-762) in syngeneic tumor models with varying PD-1 responsiveness. Bilateral Hepa1-6 models assessed abscopal effects of local treatment. Ex vivo analyses explored mechanisms of direct and abscopal efficacy. Results PH-762 was rapidly internalized by human T cells, silencing PD-1 mRNA and decreasing PD-1 surface protein, enhancing TCR-stimulated IFN-γ and CXCL10 secretion. In vivo, IT mPH-762 provided robust antitumor efficacy, local and lymphatic biodistribution, and was well tolerated. Ex vivo analyses revealed that IT mPH-762 depleted PD-1 protein, promoted leukocyte and T cell infiltration, and correlated with tumor control. IT mPH-762 also demonstrated efficacy against untreated distal tumors (abscopal effect) by priming systemic antitumor immunity. Conclusion These data support PH-762 as a promising candidate for neoadjuvant immunotherapy in clinical studies.
Collapse
Affiliation(s)
| | | | - Dingxue Yan
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Ramdane Guemiri
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | - Andrew Boone
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Deborah Bellet
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | | - James Cardia
- Phio Pharmaceuticals, Marlborough, MA, United States
| | - Caroline Robert
- Dermatology Unit, Gustave Roussy Cancer Center, Villejuif, France
| | | |
Collapse
|
18
|
Tang B, Huang R, Ma W. Advances in nanotechnology-based approaches for the treatment of head and neck squamous cell carcinoma. RSC Adv 2024; 14:38668-38688. [PMID: 39654926 PMCID: PMC11626385 DOI: 10.1039/d4ra07193j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), one of the most common types of cancers occurring in the head and neck region, is often associated with high mortality rates due to its invasiveness and morbidity. The mainstream treatment methods in clinical settings, including surgery, chemotherapy, and radiotherapy, may cause poor overall survival rate and prognosis, with issues such as drug resistance, damage to adjacent healthy tissues, and potential recurrences. Other treatment approaches such as immunotherapy, photodynamic therapy (PDT), and photothermal therapy (PPT) also suffer from inefficient tumor targeting and suboptimal therapeutic outcomes. Early detection is vital for HNSCC patients, but it is always limited by insensitivity and confusing clinical manifestations. Hence, it is highly desirable to develop optimized therapeutic and diagnostic strategies. With the boom in nanomaterials, nanotechnology-conducted HNSCC therapy has attracted widespread attention. Nanoparticles (NPs) are distinguished by their unique morphology and superior physicochemical property, and some can exhibit direct antitumor activity, while others serve as promising candidates for drug delivery. In addition, NPs offer the potential for structural modification for drug delivery and tumor targeting, enabling specific delivery to tumor cells through conjugation with biomarker ligands and improving cargo biocompatibility. This work reviews current therapies and diagnosis methods for HNSCC, highlights the characteristics of the major NPs, surveys their uses and advantages in the treatment of HNSCC, and discusses the obstacles and prospects in clinical applications, aiming to enlighten future research directions for nanotechnology-based therapy for HNSCC.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials Chengdu Sichuan 610041 China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Rui Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials Chengdu Sichuan 610041 China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| |
Collapse
|
19
|
Wang L, Ye C, Xue X, Xie M, Zhi Y, Feng X, Zhao P, Zhou J, Mi M, Li J, Gu Q, Zhao Y, Chen J, Zhou Y, Xue Y, Fu Z, Zhou L, Chen L, Pan L, Sun Y, Wang L, Wu S, He Y, Wang J. 3D-Printed Breast Prosthesis that Smartly Senses and Targets Breast Cancer Relapse. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402345. [PMID: 39308160 PMCID: PMC11633491 DOI: 10.1002/advs.202402345] [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: 03/04/2024] [Revised: 09/16/2024] [Indexed: 12/12/2024]
Abstract
Breast reconstruction is essential for improving the appearance of patients after cancer surgery. Traditional breast prostheses are not appropriate for those undergoing partial resections and cannot detect and treat locoregional recurrence. Personalized shape prostheses that can smartly sense tumor relapse and deliver therapeutics are needed. A 3D-printed prosthesis that contains a therapeutic hydrogel is developed. The hydrogel, which is fabricated by crosslinking the polyvinyl alcohol with N1-(4-boronobenzyl)-N3-(4-boronophenyl)-N1, N1, N3, N3-tetramethylpropane-1,3-diaminium, is responsive to reactive oxygen species (ROS) in the tumor microenvironment. Specifically, RSL3, a ferroptosis inducer that is loaded in hydrogels, can trigger tumor ferroptosis. Intriguingly, RSL3 encapsulated in the ROS-responsive hydrogel exerts antitumor effects by increasing the numbers of tumor-infiltrated CD4+ T cells, CD8+ T cells, and M1 macrophages while reducing the number of M2 macrophages. Therefore, this new prosthesis not only allows personalized shape reconstruction, but also detects and inhibits tumor recurrence. This combination of aesthetic appearance and therapeutic function can be very beneficial for breast cancer patients undergoing surgery.
Collapse
Affiliation(s)
- Lu Wang
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Chenyang Ye
- Department of Medical OncologyKey Laboratory of Cancer Prevention and InterventionThe Second Affiliated Hospital of Zhejiang University School of MedicineCancer CenterZhejiang UniversityHangzhou310058China
| | - Xiangjie Xue
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Mingjun Xie
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Yicheng Zhi
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Xiao Feng
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Pengcheng Zhao
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Jichun Zhou
- Department of Surgical OncologyBiomedical Research Center and Key Laboratory of Biotherapy of Zhejiang ProvinceSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Mi Mi
- Department of Medical OncologyKey Laboratory of Cancer Prevention and InterventionThe Second Affiliated Hospital of Zhejiang University School of MedicineCancer CenterZhejiang UniversityHangzhou310058China
| | - Jinrui Li
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Qinhao Gu
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Ye Zhao
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Jiaxin Chen
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Yi Zhou
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Yanan Xue
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Zexin Fu
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Liuyi Zhou
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Lulu Chen
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Lei Pan
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Yi Sun
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Linbo Wang
- Department of Surgical OncologyBiomedical Research Center and Key Laboratory of Biotherapy of Zhejiang ProvinceSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310016China
| | - Sufan Wu
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic SystemsSchool of Mechanical EngineeringZhejiang UniversityHangzhou310058China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang ProvinceSchool of Mechanical Engineering, Zhejiang UniversityHangzhou310058China
| | - Ji Wang
- Center for Plastic & Reconstructive SurgeryDepartment of Plastic & Reconstructive SurgeryZhejiang Provincial People's Hospital (Affiliated People's HospitalHangzhou Medical College)Hangzhou310014China
| |
Collapse
|
20
|
Binks M, Boyages J, Suami H, Ngui N, Meybodi F, Hughes TM, Edirimanne S. Oncoplastic breast surgery - a pictorial classification system for surgeons and radiation oncologists (OPSURGE). ANZ J Surg 2024; 94:2140-2149. [PMID: 39254174 PMCID: PMC11713206 DOI: 10.1111/ans.19212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/10/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024]
Abstract
INTRODUCTION Changes to the tumour bed following oncoplastic breast surgery complicate the administration of adjuvant radiotherapy. Consensus guidelines have called for improved interdisciplinary communication to aid adjuvant boost radiotherapy. We propose a framework of tumour bed classification following oncoplastic surgery to enhance understanding and communication between the multidisciplinary breast cancer team and facilitate effective and more precise delivery of adjuvant boost radiotherapy. METHODS A classification system was devised by grouping oncoplastic procedures based on skin incision, tissue mobilization, tumour bed distortion, seroma formation and flap reconstruction. The system is supplemented by a colour-coded pictorial guide to tumour bed rearrangement with common oncoplastic procedures. RESULTS A 5-tier framework was developed. Representative images were produced to describe tumour bed alterations. CONCLUSION The proposed framework (OPSURGE) improves the identification of the primary tumour bed after initial breast-conserving surgery, which is imperative to both the surgeon in planning re-excision and the radiation oncologist in planning boost radiotherapy.
Collapse
Affiliation(s)
- Matthew Binks
- Division of SurgeryGosford HospitalGosfordNew South WalesAustralia
| | - John Boyages
- School of Medicine and PsychologyAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
- Radiation OncologyIcon Cancer CentreSydneyNew South WalesAustralia
| | - Hiroo Suami
- Department of Health Sciences, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Nicholas Ngui
- School of Medicine and PsychologyAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
- Division of SurgerySydney Adventist HospitalSydneyNew South WalesAustralia
| | - Farid Meybodi
- Division of SurgerySydney Adventist HospitalSydneyNew South WalesAustralia
| | - T. Michael Hughes
- School of Medicine and PsychologyAustralian National UniversityCanberraAustralian Capital TerritoryAustralia
- Division of SurgerySydney Adventist HospitalSydneyNew South WalesAustralia
| | - Senarath Edirimanne
- Division of SurgerySydney Adventist HospitalSydneyNew South WalesAustralia
- Nepean Clinical School, Faculty of Medicine and HealthUniversity of SydneySydneyNew South WalesAustralia
| |
Collapse
|
21
|
Kipnis P, Ramanathan D, Hoehn R, Jethwa AR, Karakla DW, Rohr B, Sutter CM, Mark JR, Khaja SF, Li S, Thuener J, Carroll BT. Tumor seeding across specialties: a systematic review. Front Oncol 2024; 14:1464767. [PMID: 39605885 PMCID: PMC11598697 DOI: 10.3389/fonc.2024.1464767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024] Open
Abstract
Background Understanding shared characteristics underlying reported tumor seeding episodes can reveal when tumor seeding is most likely to occur and guide clinical decision making. Our goal was to systematically review tumor seeding across specialties and determine what types of instrumentation and primary tumor histology are associated with tumor seeding. Methods A systematic review was conducted using PubMed and Web of Science, per PRISMA guidelines. Publications ranged from 1965 to 2022, and studies with five or more reports of seeding were included. Papers were sorted by specialty and assigned a PRISMA Level of Evidence, and data analysis was conducted based on whether each paper supported the clinical significance of seeding. Results 7,165 papers were screened with 156 papers included for analysis. Overall, there were 8,161 cases of tumors seeding across specialties with the majority from general surgery, gastroenterology, and urology. Laparoscopy (n=1,561) and needle biopsy (n=3,448) were most frequently implicated, and carcinomas (n=5,778) and adenocarcinomas (n=1,090) were the most common primary tumor types. Discussion Upon review of the most updated (2023) versions of the NCCN and NICE guidelines across all cancer types, there were identified gaps in the coverage of tumor seeding within these guidelines, with tumor seeding being entirely absent from certain guidelines and partially absent from others. Conclusions Given the high cumulative reports of seeding and the deadly and disseminated nature of secondary disease, it is important to consider seeding risk when manipulating tumors and to modify current cancer care guidelines (NCCN/NICE) to ensure that they appropriately address seeding risk.
Collapse
Affiliation(s)
- Pavel Kipnis
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Diya Ramanathan
- Department of Otolaryngology – Head & Neck Surgery, University of Wisconsin Hospitals and Clinics, Madison, WI, United States
| | - Richard Hoehn
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Surgical Oncology, University Hospitals Medical Center, Cleveland, OH, United States
| | - Ashok R. Jethwa
- Department of Otolaryngology – Head & Neck Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Daniel W. Karakla
- Department of Otolaryngology - Head & Neck Surgery, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Bethany Rohr
- Department of Dermatology, University Hospitals Medical Center, Cleveland, OH, United States
| | - Christopher M. Sutter
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Vascular and Interventional Radiology, University Hospitals Medical Center, Cleveland, OH, United States
| | - Jonathan R. Mark
- Department of Otolaryngology – Head & Neck Surgery, University of Minnesota, Minneapolis, MN, United States
| | - Sobia F. Khaja
- Department of Surgical Oncology, University Hospitals Medical Center, Cleveland, OH, United States
| | - Shawn Li
- Department of Otolaryngology – Head & Neck Surgery, University Hospitals Medical Center, Cleveland, OH, United States
| | - Jason Thuener
- Department of Otolaryngology – Head & Neck Surgery, University Hospitals Medical Center, Cleveland, OH, United States
| | - Bryan T. Carroll
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Dermatology, University Hospitals Medical Center, Cleveland, OH, United States
| |
Collapse
|
22
|
Cortes-Mejia NA, Lillemoe HA, Cata JP. Return to Intended Oncological Therapy: State of the Art and Perspectives. Curr Oncol Rep 2024; 26:1420-1430. [PMID: 39320576 DOI: 10.1007/s11912-024-01594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/26/2024]
Abstract
PURPOSE OF THE REVIEW Despite advances in surgical procedures, cancer recurrence still affects a substantial proportion of patients for whom surgery is considered a curative therapy. This review aims to provide a comprehensive overview of RIOT, addressing its definition, influencing factors, and clinical implications. FINDINGS RIOT can be defined as a continuous variable as the time from surgery to initiation of adjuvant therapies or categorically as whether patients can successfully receive adjuvant therapies or not. Factors influencing RIOT are age, sex, socioeconomic status, access to healthcare, physical performance and comorbidities, and quality of anesthesia and surgical care. Adjuvant therapies such as chemotherapy, radiotherapy, and immunotherapy are often administered to reduce the risk of recurrence after surgery and improve survival. Return to intended oncologic therapy (RIOT) has emerged as a promising outcome metric reflecting patients' functional recovery after surgery and their ability to receive adjuvant therapies.
Collapse
Affiliation(s)
- Nicolas A Cortes-Mejia
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Heather A Lillemoe
- Department of Breast Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA.
- Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA.
- Department of Pain Medicine, The University of Texas - MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
23
|
Prasanna PGS. Harnessing Senescence for Antitumor Immunity to Advance Cancer Treatment. Radiat Res 2024; 202:727-733. [PMID: 39191430 PMCID: PMC11620177 DOI: 10.1667/rade-24-00098.1] [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: 04/01/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Considering the limitations and complexities of the cell-killing-based cancer treatment approaches, one could aim to integrate symbiotic advances in many energy delivery technologies and transformational pieces of evidence in research on senescence and immunomodulators to advance cancer treatment. Although senescent cells contribute to drug tolerance, resistance to therapy, tumorigenesis, maladapting cancer phenotypes, tumor relapse, recurrence, and metastasis, emerging pieces of evidence also demonstrate that acutely induced senescent cells in tumors can elicit a strong and lasting antitumor immune response juxtaposed to the immunologically silent apoptotic cells. This commentary is to help develop an unconventional conceptual framework to advance cancer treatment. Accordingly, it will involve transiently inducing senescent cells in tumors at optimal levels to prime the immune system with radiation, then eliminating senescent cells with senolytics (drugs that specifically eliminate senescent cells) to disrupt their positive feedback accumulation (to prevent tumor maladaptation and adverse effects in healthy cells) and unleash long-lasting antitumor immunity with immunomodulators. The approach is reasonably speculative and will require scientifically rigorous "fit-for-purpose," well-controlled preclinical research and development involving dose and schedule optimization of radiation and drugs, using representative in vitro and in vivo cancer models to obtain high-quality data to proceed to clinical studies.
Collapse
Affiliation(s)
- Pataje G. S. Prasanna
- The National Cancer Institute, Division of Cancer Treatment and Diagnosis, Radiation Research Program, Bethesda, Maryland 20892
| |
Collapse
|
24
|
Pizzo A, Leisenring WM, Stratton KL, Lamoureux É, Flynn JS, Alschuler K, Krull KR, Jibb LA, Nathan PC, Olgin JE, Stinson JN, Armstrong GT, Alberts NM. Fear of Cancer Recurrence in Adult Survivors of Childhood Cancer. JAMA Netw Open 2024; 7:e2436144. [PMID: 39361286 PMCID: PMC11450519 DOI: 10.1001/jamanetworkopen.2024.36144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 08/04/2024] [Indexed: 10/05/2024] Open
Abstract
Importance Fear of cancer recurrence is common among survivors of adult-onset cancer and associated with increased distress, functional impairment, and health care utilization. However, little is known about the prevalence and risk factors of fear of cancer recurrence among adult survivors of childhood cancer who are also at high risk for subsequent malignant neoplasms. Objective To characterize the prevalence of and risk factors for clinically significant fear of cancer recurrence in adult survivors of childhood cancer. Design, Setting, and Participants This cross-sectional investigation included participants recruited from the Childhood Cancer Survivor Study, a retrospective cohort study of long-term childhood cancer survivors treated at 31 institutions between 1970 and 1999 across North America. Participants were recruited and completed psychosocial measures via online survey between October 2018 and April 2019. Cancer and treatment-related variables were abstracted from medical records. Data were analyzed from May 2023 to July 2024. Main Outcomes and Measures Clinically significant fear of cancer recurrence was assessed via the Fear of Cancer Recurrence Inventory-Short Form. Poisson regression models estimated prevalence ratios (PRs) with 95% CIs adjusted for age and sex to examine the associations of demographic, disease, treatment, and psychosocial variables with fear of cancer recurrence. Results The final sample included 229 adult survivors of childhood cancer (115 female [50.2%]; mean [SD] age, 39.6 [9.9] years; mean [SD] time since diagnosis, 31.7 [8.4] years). Among survivors, 38 (16.6%; 95% CI, 11.6%-21.6%) reported clinically significant fear of cancer recurrence, and an additional 36 (15.7%) reported high fear of cancer recurrence. Clinically significant fear of cancer recurrence was associated with unemployment (PR, 2.5; 95% CI, 1.3-4.8), presence of neurologic chronic health conditions (PR, 3.3; 95% CI, 1.8-6.1), treatment with pelvic radiation (PR, 2.9; 95% CI, 1.5-5.6), and amputation or limb sparing surgery (PR, 2.4; 95% CI, 1.2-4.9). Higher risk of clinically significant fear of cancer recurrence was also associated with having either elevated anxiety or depression (PR, 2.6; 95% CI, 1.2-5.9), having both elevated (PR, 3.2; 95% CI, 1.2-8.4), and perceived poor health status (PR, 3.0; 95% CI, 3.1-9.7). Conclusions and Relevance Decades following treatment, one-third of childhood cancer survivors in this study reported elevated fear their cancer will recur or a subsequent malignant neoplasm will develop. Findings suggest that fear of cancer recurrence should be routinely screened, and clinically significant symptoms intervened upon as a part of survivorship care.
Collapse
Affiliation(s)
- Alex Pizzo
- Department of Psychology, Concordia University, Montréal, Québec, Canada
| | - Wendy M. Leisenring
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Kayla L. Stratton
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | | | - Jessica S. Flynn
- Department of Psychology and Biobehavioral Sciences, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Kevin Alschuler
- Department of Rehabilitation Medicine, University of Washington, Seattle
| | - Kevin R. Krull
- Department of Psychology and Biobehavioral Sciences, St Jude Children’s Research Hospital, Memphis, Tennessee
| | | | - Paul C. Nathan
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | - Gregory T. Armstrong
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Nicole M. Alberts
- Department of Psychology, Concordia University, Montréal, Québec, Canada
| |
Collapse
|
25
|
Ratnakaram K, Yendamuri S, Groman A, Kalvapudi S. Sex-Based Differences in Lung Cancer Incidence: A Retrospective Analysis of Two Large US-Based Cancer Databases. Cancers (Basel) 2024; 16:3244. [PMID: 39409866 PMCID: PMC11476236 DOI: 10.3390/cancers16193244] [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: 07/16/2024] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Non-small cell lung cancer (NSCLC) has seen a relative rise in incidence among females versus males in recent years, although males still have a higher overall incidence. However, it is unclear whether this trend is consistent across all populations. Therefore, we retrospectively examined this relationship in two large high-risk clinical cohorts. Methods: First, we analyzed lung cancer incidence among individuals with a smoking history of over 40 pack-years in the National Lung Screening Trial (NLST). Then, we investigated the incidence of second primary NSCLC in patients who underwent lobectomy for previous stage I lung cancer using the Surveillance, Epidemiology, and End Results (SEER) database. We performed both univariate and multivariable time-to-event analyses to investigate the relationship between sex and lung cancer incidence. Results: In the NLST cohort (n = 37,627), females had a higher risk of developing primary NSCLC than males (HR = 1.11 [1.007-1.222], p = 0.035) after adjusting for age and pack-year history. In the SEER cohort (n = 19,327), females again exhibited an increased risk of developing a second primary lung cancer (HR = 1.138 [1.02-1.269], p = 0.021), after adjusting for age, race, grade, and histology. Conclusions: Our analysis reveals that females have a modestly higher lung cancer incidence than males in high-risk populations. These findings underscore the importance of further researching the underlying cellular processes that may cause sex-specific differences in lung cancer incidence.
Collapse
Affiliation(s)
- Kalyan Ratnakaram
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (K.R.); (S.K.)
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (K.R.); (S.K.)
| | - Adrienne Groman
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA;
| | - Sukumar Kalvapudi
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; (K.R.); (S.K.)
| |
Collapse
|
26
|
Xu X, Jin K, Xu X, Yang Y, Zhou B. Expression and prognostic value of cell-cycle-associated genes in lung squamous cell carcinoma. J Gene Med 2024; 26:e3735. [PMID: 39171952 DOI: 10.1002/jgm.3735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Lung cancer continues to be a prevalent cause of cancer-related deaths worldwide, with lung squamous carcinoma (LUSC) being a significant subtype characterized by comparatively low survival rates. Extensive molecular studies on LUSC have been conducted; however, the clinical importance of cell-cycle-associated genes has rarely been examined. This study aimed to investigate the relationship between these genes and LUSC. METHODS The expression trends of genes related to the cell cycle in a group of patients with LUSC were analyzed. Clinical information and mRNA expression data were obtained from The Cancer Genome Atlas via cBioportal. Multiple analyses have been performed to investigate the association between these genes and LUSC. RESULTS Three clusters were identified based on the mRNA expression of 124 cell cycle-associated genes. Cluster 3 exhibited the worst prognosis. A comparative analysis showed that nine expressed genes differed distinctly among all the clusters. Among these nine genes, elevated expression of CDK4 was strongly associated with positive prognosis. Furthermore, the expression of ANAPC11, ANAPC5, and ORC4 correlated with the advancement of LUSC pathological stages. CONCLUSIONS Gene expression profiles associated with the cell cycle across various LUSC subtypes were identified, highlighting that specific genes are related to prognosis and disease stages. Based on these results, new prognostic strategies, patient stratification, and targeted therapy trials have been conducted for LUSC.
Collapse
Affiliation(s)
- Xinnan Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kaiqi Jin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoxiong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bin Zhou
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Kang S, Gil YG, Chae SY, Jang H, Min DH. Pt-Te-Nanorod-Based Photothermal Chemokine Immunotherapy for All Stages of Cancer via Adaptive and Innate Immunity. ACS APPLIED MATERIALS & INTERFACES 2024; 16:37555-37568. [PMID: 39007297 DOI: 10.1021/acsami.4c06048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The chemokine (C-X-C) motif ligand 9 (CXCL9) is one of the lymphocyte-traffic-involved chemokines. Despite the immunotherapeutic potential of CXCL9 for recruiting effector T cells (cluster of differentiation 4+ (CD4+) and CD8+ T cells) and natural killer cells (NK cells) around the tumors, practical applications of CXCL9 have been limited because of its immune toxicity and lack of stability in vivo. To overcome these limitations, we designed and synthesized Pt-Te nanorods (PtTeNRs), which exhibited excellent photothermal conversion efficiency with stable CXCL9 payload characteristics under the physiological conditions of in vivo environments. We developed a CXCL9-based immunotherapy strategy by utilizing the unique physicochemical properties of developed PtTeNRs. The investigation revealed that the PtTeNR-loaded CXCL9 was effectively accumulated in the tumor, subsequently released in a sustained manner, and successfully recruited effector T cells for immunotherapy of the designated tumor tissue. In addition, a synergistic effect was observed between the photothermal (PT) therapy and antiprogrammed cell death protein 1 (aPD-1) antibody. In this study, we demonstrated that PtTeNR-based CXCL9, PT, and aPD-1 antibody trimodal therapy delivers an outstanding tumor suppression effect in all stages of cancer, including phases 1-4 and tumor recurrence.
Collapse
Affiliation(s)
- Seounghun Kang
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Yeong-Gyu Gil
- Department of Chemistry, Kwangwoon University, 20 Gwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Se-Youl Chae
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Hongje Jang
- Department of Chemistry, Kwangwoon University, 20 Gwangwoon-ro, Nowon-gu, Seoul 01897, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Biotherapeutics Convergence Technology, Lemonex Inc., Seoul 06683, Republic of Korea
| |
Collapse
|
28
|
Mastronikolis NS, Kyrodimos E, Piperigkou Z, Spyropoulou D, Delides A, Giotakis E, Alexopoulou M, Bakalis NA, Karamanos NK. Matrix-based molecular mechanisms, targeting and diagnostics in oral squamous cell carcinoma. IUBMB Life 2024; 76:368-382. [PMID: 38168122 DOI: 10.1002/iub.2803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a head and neck cancer (HNC) with a high mortality rate. OSCC is developed in the oral cavity and it is triggered by many etiologic factors and can metastasize both regionally and distantly. Recent research advances in OSCC improved our understanding on the molecular mechanisms involved in and the initiation of OSCC metastasis. The key roles of the extracellular matrix (ECM) in OSCC are an emerging area of intensive research as the ECM macromolecular network is actively involved in events that regulate cellular morphological and functional properties, transcription and cell signaling mechanisms in invasion and metastasis. The provisional matrix that is formed by cancer cells is profoundly different in composition and functions as compared with the matrix of normal tissue. Fibroblasts are mainly responsible for matrix production and remodeling, but in cancer, the tumor matrix in the tumor microenvironment (TME) also originates from cancer cells. Even though extensive research has been conducted on the role of ECM in regulating cancer pathogenesis, its role in modulating OSCC is less elucidated since there are several issues yet to be fully understood. This critical review is focused on recent research as to present and discuss on the involvement of ECM macromolecular effectors (i.e., proteoglycans, integrins, matrix metalloproteinases) in OSCC development and progression.
Collapse
Affiliation(s)
- Nicholas S Mastronikolis
- Department of Otorhinolaryngology - Head and Neck Surgery, School of Medicine, University of Patras, Patras, Greece
| | - Efthymios Kyrodimos
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology - Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Despoina Spyropoulou
- Department of Radiation Oncology, School of Medicine, University of Patras, Patras, Greece
| | - Alexander Delides
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Athens, Greece
| | - Evangelos Giotakis
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
- Department of Radiation Oncology, School of Medicine, University of Patras, Patras, Greece
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Athens, Greece
| | - Miranda Alexopoulou
- Department of Maxillofacial Surgery, University Hospital of Patras, Patras, Greece
| | - Nick A Bakalis
- Department of Nursing, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology - Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
29
|
Adams SC, Nambiar AK, Bressler EM, Raut CP, Colson YL, Wong WW, Grinstaff MW. Immunotherapies for locally aggressive cancers. Adv Drug Deliv Rev 2024; 210:115331. [PMID: 38729264 PMCID: PMC11228555 DOI: 10.1016/j.addr.2024.115331] [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: 02/06/2024] [Revised: 03/31/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Improving surgical resection outcomes for locally aggressive tumors is key to inducing durable locoregional disease control and preventing progression to metastatic disease. Macroscopically complete resection of the tumor is the standard of care for many cancers, including breast, ovarian, lung, sarcoma, and mesothelioma. Advancements in cancer diagnostics are increasing the number of surgically eligible cases through early detection. Thus, a unique opportunity arises to improve patient outcomes with decreased recurrence rates via intraoperative delivery treatments using local drug delivery strategies after the tumor has been resected. Of the current systemic treatments (e.g., chemotherapy, targeted therapies, and immunotherapies), immunotherapies are the latest approach to offer significant benefits. Intraoperative strategies benefit from direct access to the tumor microenvironment which improves drug uptake to the tumor and simultaneously minimizes the risk of drug entering healthy tissues thereby resulting in fewer or less toxic adverse events. We review the current state of immunotherapy development and discuss the opportunities that intraoperative treatment provides. We conclude by summarizing progress in current research, identifying areas for exploration, and discussing future prospects in sustained remission.
Collapse
Affiliation(s)
- Sarah C Adams
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Arun K Nambiar
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Eric M Bressler
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Chandrajit P Raut
- Department of Surgery, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yolonda L Colson
- Massachusetts General Hospital, Department of Surgery, Boston, MA 02114, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston MA 02215, USA.
| |
Collapse
|
30
|
Dong C, Wang Y, Chen Z, Yan C, Zhang J, Song C, Wang L. Deformable Smart DNA Nanomachine for Synergistic Intracellular Cancer-Related miRNAs Imaging and Chemo-Gene Therapy of Drug-Resistant Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308562. [PMID: 38441369 DOI: 10.1002/smll.202308562] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 02/19/2024] [Indexed: 07/26/2024]
Abstract
Diagnosis and treatment of tumor especially drug-resistant tumor remains a huge challenge, which requires intelligent nanomedicines with low toxic side effects and high efficacy. Herein, deformable smart DNA nanomachines are developed for synergistic intracellular cancer-related miRNAs imaging and chemo-gene therapy of drug-resistant tumors. The tetrahedral DNA framework (MA-TDNA) with fluorescence quenched component and five antennas is self-assembled first, and then DOX molecules are loaded on the MA-TDNAs followed by linking MUC1-aptamer and Mcl-1 siRNA to the antennas of MA-TDNA, so that the apt-MA-TDNA@DOX-siRNA (DNA nanomachines) is constructed. The DNA nanomachine can respond to two tumor-related miRNAs in vitro and in vivo, which can undergo intelligent miRNA-triggered opening of the framework, resulting in the "turn on" of the fluorescence for sensitively and specifically sensing intracellular miRNAs. Meanwhile, both miRNA-responded rapid release and pH-responded release of DOX are achieved for chemotherapy of tumor. In addition, the gene therapy of the DNA nanomachines is achieved due to the miRNA-specific capture and the RNase H triggered release of Mcl-1 siRNA. The DNA nanomachines intergrading both tumor imaging and chemo-gene therapy in single nanostructures realized efficient tumor-targeted, image-guided, and microenvironment-responsive tumor diagnosis and treatment, which provides a synergetic antitumor effect on drug-resistant tumor.
Collapse
Affiliation(s)
- Chen Dong
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Yeran Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Zhilong Chen
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Chenlong Yan
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Jingjing Zhang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Chunyuan Song
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| |
Collapse
|
31
|
Wyle Y, Lu N, Hepfer J, Sayal R, Martinez T, Wang A. The Role of Biophysical Factors in Organ Development: Insights from Current Organoid Models. Bioengineering (Basel) 2024; 11:619. [PMID: 38927855 PMCID: PMC11200479 DOI: 10.3390/bioengineering11060619] [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: 04/17/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Biophysical factors play a fundamental role in human embryonic development. Traditional in vitro models of organogenesis focused on the biochemical environment and did not consider the effects of mechanical forces on developing tissue. While most human tissue has a Young's modulus in the low kilopascal range, the standard cell culture substrate, plasma-treated polystyrene, has a Young's modulus of 3 gigapascals, making it 10,000-100,000 times stiffer than native tissues. Modern in vitro approaches attempt to recapitulate the biophysical niche of native organs and have yielded more clinically relevant models of human tissues. Since Clevers' conception of intestinal organoids in 2009, the field has expanded rapidly, generating stem-cell derived structures, which are transcriptionally similar to fetal tissues, for nearly every organ system in the human body. For this reason, we conjecture that organoids will make their first clinical impact in fetal regenerative medicine as the structures generated ex vivo will better match native fetal tissues. Moreover, autologously sourced transplanted tissues would be able to grow with the developing embryo in a dynamic, fetal environment. As organoid technologies evolve, the resultant tissues will approach the structure and function of adult human organs and may help bridge the gap between preclinical drug candidates and clinically approved therapeutics. In this review, we discuss roles of tissue stiffness, viscoelasticity, and shear forces in organ formation and disease development, suggesting that these physical parameters should be further integrated into organoid models to improve their physiological relevance and therapeutic applicability. It also points to the mechanotransductive Hippo-YAP/TAZ signaling pathway as a key player in the interplay between extracellular matrix stiffness, cellular mechanics, and biochemical pathways. We conclude by highlighting how frontiers in physics can be applied to biology, for example, how quantum entanglement may be applied to better predict spontaneous DNA mutations. In the future, contemporary physical theories may be leveraged to better understand seemingly stochastic events during organogenesis.
Collapse
Affiliation(s)
- Yofiel Wyle
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
| | - Nathan Lu
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Jason Hepfer
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Rahul Sayal
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Taylor Martinez
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817, USA
| |
Collapse
|
32
|
Li J, Li L, Wang J, Liu N, Liu H, Xu F, Li M, Yuan S. Prognostic biomarker tumor-infiltrating lymphocytes failed to serve as a predictive biomarker for postoperative radiotherapy in completely resected pN2 non-small cell lung cancer: a retrospective analysis. Respir Res 2024; 25:244. [PMID: 38886760 PMCID: PMC11184716 DOI: 10.1186/s12931-024-02863-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Evidence suggests that radiotherapy is a potent immunomodulator in non-small cell lung cancer (NSCLC). Conversely, it has rarely been demonstrated if immune infiltration can influence radiotherapy efficacy. Herein, we explored the effect of tumor-infiltrating lymphocytes (TILs) on the response to postoperative radiotherapy (PORT) in completely resected stage III-pN2 NSCLC. METHODS This retrospective study included 244 patients with pathologically confirmed stage III-N2 NSCLC who underwent complete resection at our institution between 2014 and 2020. TILs were assessed with permanent full-face hematoxylin and eosin (H&E) sections and the evaluation of TILs was based on a published guideline. Patients were stratified into the TILlow or TILhigh group with a cutoff value of 50%. Kaplan-Meier method and Log-rank test were utilized to assess disease-free survival (DFS) and overall survival (OS). Univariate and multivariate Cox regression analysis were conducted to determine prognostic indicators. RESULTS Among 244 patients, a total of 121 patients received PORT whereas 123 did not. TILs level in patients with PORT was significantly higher than that in patients without PORT (p < 0.001). High TILs level was significantly associated with an improved DFS and OS in all the entire chort (DFS, p < 0.001; OS, p = 0.001), PORT chort (DFS, p = 0.003; OS, p = 0.011) and non-PORT chort (DFS, p < 0.001; OS, p = 0.034). There were no significant survival differences between different treatment modalities in the low TILs infiltration (DFS, p = 0.244; OS, p = 0.404) and high TILs infiltration (DFS, p = 0.167; OS, p = 0.958) groups. CONCLUSIONS TILs evaluated with H&E sections could represent a prognostic biomarker in patients with completely resected pN2 NSCLC, and high TILs infiltration was associated with favorable survival outcomes.The predictive value of TILs for PORT still need to be further explored in the future.
Collapse
Affiliation(s)
- Jiaran Li
- Shandong University Cancer Center, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Li Li
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jingjing Wang
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ning Liu
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Haixin Liu
- Shandong University Cancer Center, Jinan, Shandong, China
- Department of Oncology, Qingdao Municipal Hospital (Group), Qingdao, Shandong, China
| | - Fuhao Xu
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengke Li
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Department of Radiation Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, No.17 Lujiang Road, Hefei, Anhui, 230001, China.
- Department of Radiation Oncology, Anhui Provincial Cancer Hospital, Hefei, Anhui, China.
| |
Collapse
|
33
|
Roider HG, Hoff S, Tseng SY, Berndt S, Trautwein M, Filarsky K, Gritzan U, Camps J, Nadler WM, Grudzinska-Goebel J, Ellinger P, Pesch T, Soon CF, Geyer M, Gluske K, Stelte-Ludwig B, Gorjánácz M. Selective depletion of tumor-infiltrating regulatory T cells with BAY 3375968, a novel Fc-optimized anti-CCR8 antibody. Clin Exp Med 2024; 24:122. [PMID: 38856863 PMCID: PMC11164760 DOI: 10.1007/s10238-024-01362-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/28/2024] [Indexed: 06/11/2024]
Abstract
Regulatory T cells (Tregs) are known to facilitate tumor progression by suppressing CD8+ T cells within the tumor microenvironment (TME), thereby also hampering the effectiveness of immune checkpoint inhibitors (ICIs). While systemic depletion of Tregs can enhance antitumor immunity, it also triggers undesirable autoimmune responses. Therefore, there is a need for therapeutic agents that selectively target Tregs within the TME without affecting systemic Tregs. In this study, as shown also by others, the chemokine (C-C motif) receptor 8 (CCR8) was found to be predominantly expressed on Tregs within the TME of both humans and mice, representing a unique target for selective depletion of tumor-residing Tregs. Based on this, we developed BAY 3375968, a novel anti-human CCR8 antibody, along with respective surrogate anti-mouse CCR8 antibodies, and demonstrated their in vitro mode-of-action through induction of potent antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) activities. In vivo, anti-mouse CCR8 antibodies effectively depleted Tregs within the TME primarily via ADCP, leading to increased CD8+ T cell infiltration and subsequent tumor growth inhibition across various cancer models. This monotherapeutic efficacy was significantly enhanced in combination with ICIs. Collectively, these findings suggest that CCR8 targeting represents a promising strategy for Treg depletion in cancer therapies. BAY 3375968 is currently under investigation in a Phase I clinical trial (NCT05537740).
Collapse
MESH Headings
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- Receptors, CCR8/immunology
- Receptors, CCR8/antagonists & inhibitors
- Animals
- Mice
- Humans
- Tumor Microenvironment/immunology
- Tumor Microenvironment/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Female
- Antibody-Dependent Cell Cytotoxicity
- Lymphocyte Depletion
- Cell Line, Tumor
- Phagocytosis/drug effects
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
Collapse
Affiliation(s)
| | | | - Su-Yi Tseng
- Bayer AG, Pharmaceuticals, San Francisco, USA
| | | | | | - Katharina Filarsky
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Roche Diagnostics GmbH, Penzberg, Germany
| | - Uwe Gritzan
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Memorial Sloan Kettering Cancer Center, New York, USA
| | | | | | | | | | | | | | | | | | - Beatrix Stelte-Ludwig
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Vincerx Pharma, Monheim am Rhein, Germany
| | | |
Collapse
|
34
|
Pandiyan S, Wang L. In-silico design of novel potential HDAC inhibitors from indazole derivatives targeting breast cancer through QSAR, molecular docking and pharmacokinetics studies. Comput Biol Chem 2024; 110:108035. [PMID: 38460437 DOI: 10.1016/j.compbiolchem.2024.108035] [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: 12/27/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/11/2024]
Abstract
Latest studies confirmed that abnormal function of histone deacetylase (HDAC) plays a pivotal role in formation of tumors and is a potential therapeutic target for treating breast cancer. In this research, in-silico drug discovery approaches via quantitative structure activity relationship (QSAR) and molecular docking simulations were adapted to 43 compounds of indazole derivatives with HDAC inhibition for anticancer activity against breast cancer. The QSAR models were built from multiple linear regression (MLR), and models predictability was cross-validated by leave-one-out (LOO) method. Based on these results, compounds C32, C26 and C31 from model 3 showed superior inhibitory activity with pIC50 of 9.30103, 9.1549 and 9.1549. We designed 10 novel compounds with molecular docking scores ranging from -7.9 to -9.3 kcal/mol. The molecular docking simulation results reveal that amino acid residues ILE1122 and PRO1123 play a significant role in bonding with 6CE6 protein. Furthermore, newly designed compounds P5, P2 and P7 with high docking scores of -9.3 kcal/mol, -8.9 kcal/mol and -8.8 kcal/mol than FDA-approved drug Raloxifene (-8.5 kcal/mol) and aid in establishment of potential drug candidate for HDAC inhibitors. The in-silico ADME functionality is used in the final phase to evaluate newly designed inhibitors as potential drug candidates. The results suggest that newly designed compounds P5, P2 and P7 can be used as a potential anti-breast cancer drug candidate.
Collapse
Affiliation(s)
- Sanjeevi Pandiyan
- Research Center for Intelligent Information Technology, Nantong University, Nantong, China; School of Information Science and Technology, Nantong University, Nantong, China; Nantong Research Institute for Advanced Communication Technologies, Nantong, China.
| | - Li Wang
- Research Center for Intelligent Information Technology, Nantong University, Nantong, China; School of Information Science and Technology, Nantong University, Nantong, China; Nantong Research Institute for Advanced Communication Technologies, Nantong, China
| |
Collapse
|
35
|
Li M, Zhao D, Yan J, Fu X, Li F, Liu G, Fan Y, Liang Y, Zhang X. A Redox-Triggered Autophagy-Induced Nanoplatform with PD-L1 Inhibition for Enhancing Combined Chemo-Immunotherapy. ACS NANO 2024; 18:12870-12884. [PMID: 38727063 DOI: 10.1021/acsnano.4c00227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Epirubicin (EPI) alone can trigger mildly protective autophagy in residual tumor cells, resulting in an immunosuppressive microenvironment. This accelerates the recurrence of residual tumors and leads to antiprogrammed death ligand 1 (anti-PD-1)/PD-L1 therapy resistance, posing a significant clinical challenge in tumor immunotherapy. The combination of checkpoint inhibitors targeting the PD-1/PD-L1 pathway and amplifying autophagy presents an innovative approach to tumor treatment, which can prevent tumor immune escape and enhance therapeutic recognition. Herein, we aimed to synthesize a redox-triggered autophagy-induced nanoplatform with SA&EA-induced PD-L1 inhibition. The hyaluronic acid (HA) skeleton and arginine segment promoted active nanoplatform targeting, cell uptake, and penetration. The PLGLAG peptide was cleaved by overexpressing matrix metalloproteinase-2 (MMP-2) in the tumor microenvironment, and the PD-L1 inhibitor D-PPA was released to inhibit tumor immune escape. The intense autophagy inducers, STF-62247 and EPI, were released owing to the cleavage of disulfide bonds influenced by the high glutathione (GSH) concentration in tumor cells. The combination of EPI and STF induced apoptosis and autophagic cell death, effectively eliminating a majority of tumor cells. This indicated that the SA&EA nanoplatform has better therapeutic efficacy than the single STF@AHMPP and EPI@AHMPTP groups. This research provided a way to set up a redox-triggered autophagy-induced nanoplatform with PD-L1 inhibition to enhance chemo-immunotherapy.
Collapse
Affiliation(s)
- Ming Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
- Sichuan Kelun Pharmaceutical Research Institute Co. Ltd., Chengdu 611130, China
| | - Dong Zhao
- Sichuan Kelun Pharmaceutical Research Institute Co. Ltd., Chengdu 611130, China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Xiaoheng Fu
- Department of Clinical Laboratory, No.971 Hospital of the People's Liberation Army Navy, Qingdao 266021, China
| | - Fashun Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Gecen Liu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
- College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| |
Collapse
|
36
|
Prezado Y, Grams M, Jouglar E, Martínez-Rovira I, Ortiz R, Seco J, Chang S. Spatially fractionated radiation therapy: a critical review on current status of clinical and preclinical studies and knowledge gaps. Phys Med Biol 2024; 69:10TR02. [PMID: 38648789 DOI: 10.1088/1361-6560/ad4192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/22/2024] [Indexed: 04/25/2024]
Abstract
Spatially fractionated radiation therapy (SFRT) is a therapeutic approach with the potential to disrupt the classical paradigms of conventional radiation therapy. The high spatial dose modulation in SFRT activates distinct radiobiological mechanisms which lead to a remarkable increase in normal tissue tolerances. Several decades of clinical use and numerous preclinical experiments suggest that SFRT has the potential to increase the therapeutic index, especially in bulky and radioresistant tumors. To unleash the full potential of SFRT a deeper understanding of the underlying biology and its relationship with the complex dosimetry of SFRT is needed. This review provides a critical analysis of the field, discussing not only the main clinical and preclinical findings but also analyzing the main knowledge gaps in a holistic way.
Collapse
Affiliation(s)
- Yolanda Prezado
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, F-91400, Orsay, France
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, F-91400, Orsay, France
- New Approaches in Radiotherapy Lab, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, E-15706, Spain
- Oportunius Program, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain
| | - Michael Grams
- Department of Radiation Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America
| | - Emmanuel Jouglar
- Institut Curie, PSL Research University, Department of Radiation Oncology, F-75005, Paris and Orsay Protontherapy Center, F-91400, Orsay, France
| | - Immaculada Martínez-Rovira
- Physics Department, Universitat Auto`noma de Barcelona, E-08193, Cerdanyola del Valle`s (Barcelona), Spain
| | - Ramon Ortiz
- University of California San Francisco, Department of Radiation Oncology, 1600 Divisadero Street, San Francisco, CA 94143, United States of America
| | - Joao Seco
- Division of Biomedical physics in Radiation Oncology, DKFZ-German Cancer Research Center, Heidelberg, Germany
- Department of Physics and Astronomy, Heidelberg University, Heidelberg, Germany
| | - Sha Chang
- Dept of Radiation Oncology and Department of Biomedical Engineering, University of North Carolina School of Medicine, United States of America
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolin State University, United States of America
| |
Collapse
|
37
|
Xu M, Hou Y, Li N, Yu W, Chen L. Targeting histone deacetylases in head and neck squamous cell carcinoma: molecular mechanisms and therapeutic targets. J Transl Med 2024; 22:418. [PMID: 38702756 PMCID: PMC11067317 DOI: 10.1186/s12967-024-05169-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/05/2024] [Indexed: 05/06/2024] Open
Abstract
The onerous health and economic burden associated with head and neck squamous cell carcinoma (HNSCC) is a global predicament. Despite the advent of novel surgical techniques and therapeutic protocols, there is an incessant need for efficacious diagnostic and therapeutic targets to monitor the invasion, metastasis and recurrence of HNSCC due to its substantial morbidity and mortality. The differential expression patterns of histone deacetylases (HDACs), a group of enzymes responsible for modifying histones and regulating gene expression, have been demonstrated in neoplastic tissues. However, there is limited knowledge regarding the role of HDACs in HNSCC. Consequently, this review aims to summarize the existing research findings and explore the potential association between HDACs and HNSCC, offering fresh perspectives on therapeutic approaches targeting HDACs that could potentially enhance the efficacy of HNSCC treatment. Additionally, the Cancer Genome Atlas (TCGA) dataset, CPTAC, HPA, OmicShare, GeneMANIA and STRING databases are utilized to provide supplementary evidence on the differential expression of HDACs, their prognostic significance and predicting functions in HNSCC patients.
Collapse
Affiliation(s)
- Mengchen Xu
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Shandong Provincial Clinical Research Center for Oral Diseases, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Yiming Hou
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Shandong Provincial Clinical Research Center for Oral Diseases, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Na Li
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250022, Shandong, China
- Center of Clinical Laboratory, Shandong Second Provincial General Hospital, Jinan, 250022, Shandong, China
| | - Wenqian Yu
- Research Center of Translational Medicine, Department of Cardiac Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, Shandong, People's Republic of China
| | - Lei Chen
- Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Department of Orthodontics, School and Hospital of Stomatology, Shandong Provincial Clinical Research Center for Oral Diseases, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
38
|
Wang S, Qian Z, Xiao H, Yang G, Zhu Z, Gu Y, Song J, Zhang X, Huang X, Weng L, Gao Y, Yang W, Wang L. A photo-responsive self-healing hydrogel loaded with immunoadjuvants and MoS 2 nanosheets for combating post-resection breast cancer recurrence. NANOSCALE 2024; 16:8417-8426. [PMID: 38591110 DOI: 10.1039/d4nr00372a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Tumor recurrence after surgical resection remains a significant challenge in breast cancer treatment. Immune checkpoint blockade therapy, as a promising alternative therapy, faces limitations in combating tumor recurrence due to the low immune response rate. In this study, we developed an implantable photo-responsive self-healing hydrogel loaded with MoS2 nanosheets and the immunoadjuvant R837 (PVA-MoS2-R837, PMR hydrogel) for in situ generation of tumor-associated antigens at the post-surgical site of the primary tumor, enabling sustained and effective activation of the immune response. This PMR hydrogel exhibited potential for near-infrared (NIR) light response, tissue adhesion, self-healing, and sustained adjuvant release. When implanted at the site after tumor resection, NIR irradiation triggered a photothermal effect, resulting in the ablation of residual cancer cells. The in situ-generated tumor-associated antigens promoted dendritic cell (DC) maturation. In a mouse model, PMR hydrogel-mediated photothermal therapy combined with immune checkpoint blockade effectively inhibited the recurrence of resected tumors, providing new insights for combating post-resection breast cancer recurrence.
Collapse
Affiliation(s)
- Siyu Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Zhuoping Qian
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Huaxin Xiao
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Guangwen Yang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Ziyi Zhu
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Yubin Gu
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Junjie Song
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Xin Zhang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Xinxuan Huang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Lixing Weng
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Yu Gao
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Wenjing Yang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Lianhui Wang
- State Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| |
Collapse
|
39
|
Liu F, Yang T, Chang X, Chen L, Cheng C, Peng X, Liu H, Zhang Y, Chen X. Intelligent gold nanocluster for effective treatment of malignant tumor via tumor-specific photothermal-chemodynamic therapy with AIE guidance. Natl Sci Rev 2024; 11:nwae113. [PMID: 38698903 PMCID: PMC11065357 DOI: 10.1093/nsr/nwae113] [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: 11/01/2023] [Revised: 03/04/2024] [Accepted: 03/21/2024] [Indexed: 05/05/2024] Open
Abstract
Precise and efficient therapy of malignant tumors is always a challenge. Herein, gold nanoclusters co-modified by aggregation-induced-emission (AIE) molecules, copper ion chelator (acylthiourea) and tumor-targeting agent (folic acid) were fabricated to perform AIE-guided and tumor-specific synergistic therapy with great spatio-temporal controllability for the targeted elimination and metastasis inhibition of malignant tumors. During therapy, the functional gold nanoclusters (AuNTF) would rapidly accumulate in the tumor tissue due to the enhanced permeability and retention effect as well as folic acid-mediated tumor targeting, which was followed by endocytosis by tumor cells. After that, the overexpressed copper ions in the tumor cells would trigger the aggregation of these intracellular AuNTF via a chelation process that not only generated the photothermal agent in situ to perform the tumor-specific photothermal therapy damaging the primary tumor, but also led to the copper deficiency of tumor cells to inhibit its metastasis. Moreover, the copper ions were reduced to cuprous ions along with the chelation, which further catalysed the excess H2O2 in the tumor cells to produce cytotoxic reactive oxygen species, resulting in additional chemodynamic therapy for enhanced antitumor efficiency. The aggregation of AuNTF also activated the AIE molecules to present fluorescence, which not only imaged the therapeutic area for real-time monitoring of this tumor-specific synergistic therapy, but also allowed us to perform near-infrared radiation at the correct time point and location to achieve optimal photothermal therapy. Both in vitro and in vivo results revealed the strong tumor elimination, effective metastasis inhibition and high survival rate of tumor-bearing mice after treatment using the AuNTF nanoclusters, indicating that this AIE-guided and tumor-specific synergistic strategy could offer a promising approach for tumor therapy.
Collapse
Affiliation(s)
- Feng Liu
- School of Energy and Power Engineering, Xi'an Jiaotong University, Xi'an 710049, China
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Tianfeng Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaowei Chang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Cheng Cheng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiuhong Peng
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Haihu Liu
- School of Energy and Power Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
40
|
Liu R, Zhao Y, Su S, Kwabil A, Njoku PC, Yu H, Li X. Unveiling cancer dormancy: Intrinsic mechanisms and extrinsic forces. Cancer Lett 2024; 591:216899. [PMID: 38649107 DOI: 10.1016/j.canlet.2024.216899] [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: 03/19/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Tumor cells disseminate in various distant organs at early stages of cancer progression. These disseminated tumor cells (DTCs) can stay dormant/quiescent without causing patient symptoms for years or decades. These dormant tumor cells survive despite curative treatments by entering growth arrest, escaping immune surveillance, and/or developing drug resistance. However, these dormant cells can reactivate to proliferate, causing metastatic progression and/or relapse, posing a threat to patients' survival. It's unclear how cancer cells maintain dormancy and what triggers their reactivation. What are better approaches to prevent metastatic progression and relapse through harnessing cancer dormancy? To answer these remaining questions, we reviewed the studies of tumor dormancy and reactivation in various types of cancer using different model systems, including the brief history of dormancy studies, the intrinsic characteristics of dormant cells, and the external cues at the cellular and molecular levels. Furthermore, we discussed future directions in the field and the strategies for manipulating dormancy to prevent metastatic progression and recurrence.
Collapse
Affiliation(s)
- Ruihua Liu
- School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010070, China; Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Yawei Zhao
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Shang Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Augustine Kwabil
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Prisca Chinonso Njoku
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Haiquan Yu
- School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010070, China.
| | - Xiaohong Li
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
41
|
Voznyuk AA, Makarets YA, Advakhova DY, Khafizov KA, Lugovoi ME, Zakharova VA, Senatov FS, Koudan EV. Biodegradable Local Chemotherapy Platform with Prolonged and Controlled Release of Doxorubicin for the Prevention of Local Tumor Recurrence. ACS APPLIED BIO MATERIALS 2024; 7:2472-2487. [PMID: 38480461 DOI: 10.1021/acsabm.4c00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Local recurrence after surgical and therapeutic treatment remains a significant clinical problem in oncology. Recurrence may be due to imperfections in existing therapies, particularly chemotherapy. To improve antitumor activity and prevent local cancer recurrence while keeping toxicity at acceptable levels, we have developed and demonstrated a biodegradable local chemotherapy platform that provides controlled and prolonged drug release. The platform consists of a polycaprolactone (PCL) substrate, which provides the structural integrity of the platform and the predominant unidirectional drug release, and a thin multilayer coating (∼200 nm) containing doxorubicin (DOX). The coating is an electrostatic complex obtained by the layer-by-layer (LbL) assembly and consists of natural polyelectrolytes [poly-γ-glutamic acid (γ-PGA) and chitosan (CS) or poly-l-lysine (PLL)]. To improve the release stability, an ionic conjugate of DOX and γ-PGA was prepared and incorporated into the multilayer coating. By varying the structure of the coating by adding empty (without DOX) bilayers, we were able to control the kinetics of drug release. The resulting platforms contained equal numbers of empty bilayers and DOX-loaded bilayers (15 + 15 or 30 + 30 bilayers) with a maximum loading of 566 ng/cm2. The platforms demonstrated prolonged and fairly uniform drug release for more than 5 months while retaining antitumor activity in vitro on ovarian cancer cells (SKOV-3). The empty platforms (without DOX) showed good cytocompatibility and no cytotoxicity to human fibroblasts and SKOV-3 cells. This study presents the development of a local chemotherapy platform consisting of a PCL-based substrate which provides structural stability and a biodegradable polyelectrolyte layered coating which combines layers containing a polyanion ionic complex with DOX with empty bilayers to ensure prolonged and controlled drug release. Our results may provide a basis for improving the efficacy of chemotherapy using drug delivery systems.
Collapse
Affiliation(s)
- Amina A Voznyuk
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Yulia A Makarets
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Darya Yu Advakhova
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Krestina A Khafizov
- Haute École de la Province de Namur, Henri Blès st. 192, Namur 5000, Belgium
| | - Maksim E Lugovoi
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Vasilina A Zakharova
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Fedor S Senatov
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| | - Elizaveta V Koudan
- National University of Science and Technology MISIS, Leninskiy pr. 4, Moscow 119049, Russian Federation
| |
Collapse
|
42
|
Fujita K, Urano Y. Activity-Based Fluorescence Diagnostics for Cancer. Chem Rev 2024; 124:4021-4078. [PMID: 38518254 DOI: 10.1021/acs.chemrev.3c00612] [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: 03/24/2024]
Abstract
Fluorescence imaging is one of the most promising approaches to achieve intraoperative assessment of the tumor/normal tissue margins during cancer surgery. This is critical to improve the patients' prognosis, and therefore various molecular fluorescence imaging probes have been developed for the identification of cancer lesions during surgery. Among them, "activatable" fluorescence probes that react with cancer-specific biomarker enzymes to generate fluorescence signals have great potential for high-contrast cancer imaging due to their low background fluorescence and high signal amplification by enzymatic turnover. Over the past two decades, activatable fluorescence probes employing various fluorescence control mechanisms have been developed worldwide for this purpose. Furthermore, new biomarker enzymatic activities for specific types of cancers have been identified, enabling visualization of various types of cancers with high sensitivity and specificity. This Review focuses on recent advances in the design, function and characteristics of activatable fluorescence probes that target cancer-specific enzymatic activities for cancer imaging and also discusses future prospects in the field of activity-based diagnostics for cancer.
Collapse
|
43
|
Zhang C, Han ZY, Chen KW, Wang YZ, Bao P, Ji P, Yan X, Rao ZY, Zeng X, Zhang XZ. In Situ Formed Microalgae-Integrated Living Hydrogel for Enhanced Tumor Starvation Therapy and Immunotherapy through Photosynthetic Oxygenation. NANO LETTERS 2024; 24:3801-3810. [PMID: 38477714 DOI: 10.1021/acs.nanolett.4c00471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
The effectiveness of various cancer therapies for solid tumors is substantially limited by the highly hypoxic tumor microenvironment (TME). Here, a microalgae-integrated living hydrogel (ACG gel) is developed to concurrently enhance hypoxia-constrained tumor starvation therapy and immunotherapy. The ACG gel is formed in situ following intratumoral injection of a biohybrid fluid composed of alginate, Chlorella sorokiniana, and glucose oxidase, facilitated by the crossing-linking between divalent ions within tumors and alginate. The microalgae Chlorella sorokiniana embedded in ACG gel generate abundant oxygen through photosynthesis, enhancing glucose oxidase-catalyzed glucose consumption and shifting the TME from immunosuppressive to immunopermissive status, thus reducing the tumor cell energy supply and boosting antitumor immunity. In murine 4T1 tumor models, the ACG gel significantly suppresses tumor growth and effectively prevents postoperative tumor recurrence. This study, leveraging microalgae as natural oxygenerators, provides a versatile and universal strategy for the development of oxygen-dependent tumor therapies.
Collapse
Affiliation(s)
- Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zi-Yi Han
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Ke-Wei Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Yu-Zhang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Peng Bao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Ping Ji
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xiao Yan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Zhi-Yong Rao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, P. R. China
| |
Collapse
|
44
|
Zhang P, Li B, Wang Z, Li J, Wang F, Kong J, Zhou Z, Huang Y, Li L. Durable Attenuation of Tumor pH-Platelet Linkage Reinstates Bioorthogonal Targeting of Residual Tumors Post-Debulking. ACS NANO 2024; 18:4520-4538. [PMID: 38270077 DOI: 10.1021/acsnano.3c11536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
There are circumstances where tumors can only be partially resected. Therefore, multimodality therapy targeting post-operative residuals is important. Here, we show that bioorthogonal click chemistry enables targeted delivery to heterogeneous tumors, but its utility against tumor post-debulking is ineffective due to platelet cloaks that shield tumor cells from bioorthogonal pairing. We further discover tumor-infiltrating platelet levels respond to local pH changes. Elucidating this pH-platelet linkage, we design an injectable hydrogel for resection cavity implantation that simultaneously azido-tags tumor cells and inhibit their catalysis to acidify surrounding milieu. Unlike transient buffering, tumor acidification blockade sustains pH normalization, leading to durable platelet reduction. This reinstates bioorthogonal targeting of dibenzyl cyclooctyne-modified nanoparticles, thereby enhancing photodynamic ablation of residuals while amplifying systemic antitumor immunity. Concurrently, platelet/pH normalization interrupts metastasis cascade from invasion to circulation to colonization. Overall, attenuating tumor pH-platelet linkage unlocks bioorthogonal chemistry as a potential option for adjuvant therapy after tumor debulking.
Collapse
Affiliation(s)
- Ping Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Bo Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ziyan Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Junlin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Fengju Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jinxia Kong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhou Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
45
|
Shakhova M, Elagin V, Plekhanov A, Khilov A, Kurakina D, Kamensky V, Kirillin M. Post-Operational Photodynamic Therapy of the Tumor Bed: Comparative Analysis for Cold Knife and Laser Scalpel Resection. Biomedicines 2024; 12:291. [PMID: 38397893 PMCID: PMC11154242 DOI: 10.3390/biomedicines12020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
In this paper, we report on a study regarding the efficiency of the post-operational phototherapy of the tumor bed after resection with both a cold knife and a laser scalpel in laboratory mice with CT-26 tumors. Post-operational processing included photodynamic therapy (PDT) with a topically applied chlorin-based photosensitizer (PS), performed at wavelengths of 405 or 660 nm, with a total dose of 150 J/cm2. The selected design of the tumor model yielded zero recurrence in the laser scalpel group and 92% recurrence in the cold knife group without post-processing, confirming the efficiency of the laser scalpel in oncology against the cold knife. The application of PDT after the cold knife resection decreased the recurrence rate to 70% and 42% for the 405 nm and 660 nm procedures, respectively. On the other hand, the application of PDT after the laser scalpel resection induced recurrence rates of 18% and 30%, respectively, for the considered PDT performance wavelengths. The control of the penetration of PS into the tumor bed by fluorescence confocal microscopy indicated the deeper penetration of PS in the case of the cold knife, which presumably provided deeper PDT action, while the low-dose light exposure of deeper tissues without PS, presumably, stimulated tumor recurrence, which was also confirmed by the differences in the recurrence rate in the 405 and 660 nm groups. Irradiation-only light exposures, in all cases, demonstrated higher recurrence rates compared to the corresponding PDT cases. Thus, the PDT processing of the tumor bed after resection could only be recommended for the cold knife treatment and not for the laser scalpel resection, where it could induce tumor recurrence.
Collapse
Affiliation(s)
- Maria Shakhova
- Department of Ear, Nose and Throat Diseases, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia;
| | - Vadim Elagin
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
| | - Anton Plekhanov
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
| | - Aleksandr Khilov
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Daria Kurakina
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Vladislav Kamensky
- Institute of Experimental Oncology and Biomedical Technologies, FSBEI HE «Privolzhsky Research Medical University» MOH Russia, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603005, Russia; (A.P.); (V.K.)
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| | - Mikhail Kirillin
- A.V. Gaponov-Grekhov Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanov St., Nizhny Novgorod 603155, Russia; (A.K.); (D.K.); (M.K.)
| |
Collapse
|
46
|
Lian Y, Li X, Lan Y, Li Z, Lin X, Huang J, Zhang B, Feng Z. Bibliometric and visual analysis in the field of tea in cancer from 2013 to 2023. Front Oncol 2024; 13:1296511. [PMID: 38273848 PMCID: PMC10808711 DOI: 10.3389/fonc.2023.1296511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Objective Tea has been utilized in cancer research and is progressively gaining wider recognition, with its roles in cancer prevention and treatment being increasingly affirmed. The objective of this study is to investigate the current state and research hotspots in the field of tea's involvement in cancer research from 2013 to 2023, aiming to offer reference and direction for future studies. Methods We analyzed 4,789 articles published between 2013 and 2022 from the Web of Science database using VOSviewer, R software, and CiteSpace software. Result Tea-related cancer research showed an overall upward trend, with China leading in publications, followed by the United States, India, Japan, and Italy. China also had significant international collaborations, notably with Harvard University and the Egyptian Knowledge Bank. The 'Journal of Agricultural and Food Chemistry' was the most cited journal. Key topics included 'green tea,' 'cancer,' 'in vitro,' 'oxidative stress,' and 'apoptosis.' Research focused on tea's pharmacological effects, anticancer properties, mechanisms of natural compounds (e.g., polyphenols and EGCG), antioxidant and antimicrobial properties, and molecular mechanisms in cancer treatment. Conclusion Tea's potential as an anti-cancer medication is gaining global recognition. Our study provides a comprehensive analysis of tea-related cancer research from 2013 to 2023, guiding future investigations in this field.
Collapse
Affiliation(s)
- Yuanchu Lian
- Scientific Research Center, Guilin Medical University, Guilin, China
- Lingui Clinical College, Guilin Medical University, Guilin, China
| | - Xiating Li
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ying Lan
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Zonghuai Li
- Scientific Research Center, Guilin Medical University, Guilin, China
- Lingui Clinical College, Guilin Medical University, Guilin, China
| | - Xiaoxin Lin
- Scientific Research Center, Guilin Medical University, Guilin, China
- Lingui Clinical College, Guilin Medical University, Guilin, China
| | - Jing’an Huang
- Scientific Research Center, Guilin Medical University, Guilin, China
- Lingui Clinical College, Guilin Medical University, Guilin, China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Zhongwen Feng
- Department of Pharmacy, Guangxi Academy of Medical Sciences and the People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
47
|
He S, Gou X, Zhang S, Zhang X, Huang H, Wang W, Yi L, Zhang R, Duan Z, Zhou P, Qian Z, Gao X. Nanodelivery Systems as a Novel Strategy to Overcome Treatment Failure of Cancer. SMALL METHODS 2024; 8:e2301127. [PMID: 37849248 DOI: 10.1002/smtd.202301127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Despite the tremendous progress in cancer treatment in recent decades, cancers often become resistant due to multiple mechanisms, such as intrinsic or acquired multidrug resistance, which leads to unsatisfactory treatment effects or accompanying metastasis and recurrence, ultimately to treatment failure. With a deeper understanding of the molecular mechanisms of tumors, researchers have realized that treatment designs targeting tumor resistance mechanisms would be a promising strategy to break the therapeutic deadlock. Nanodelivery systems have excellent physicochemical properties, including highly efficient tissue-specific delivery, substantial specific surface area, and controllable surface chemistry, which endow nanodelivery systems with capabilities such as precise targeting, deep penetration, responsive drug release, multidrug codelivery, and multimodal synergy, which are currently widely used in biomedical researches and bring a new dawn for overcoming cancer resistance. Based on the mechanisms of tumor therapeutic resistance, this review summarizes the research progress of nanodelivery systems for overcoming tumor resistance to improve therapeutic efficacy in recent years and offers prospects and challenges of the application of nanodelivery systems for overcoming cancer resistance.
Collapse
Affiliation(s)
- Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xinyu Gou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Shuheng Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Hongyi Huang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Rui Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhongxin Duan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Peizhi Zhou
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| |
Collapse
|
48
|
Huang YY, Deng YS, Liu Y, Qiang MY, Qiu WZ, Xia WX, Jing BZ, Feng CY, Chen HH, Cao X, Zhou JY, Huang HY, Zhan ZJ, Deng Y, Tang LQ, Mai HQ, Sun Y, Xie CM, Guo X, Ke LR, Lv X, Li CF. A deep learning-based semiautomated workflow for triaging follow-up MR scans in treated nasopharyngeal carcinoma. iScience 2023; 26:108347. [PMID: 38125021 PMCID: PMC10730347 DOI: 10.1016/j.isci.2023.108347] [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: 06/27/2023] [Revised: 08/08/2023] [Accepted: 10/24/2023] [Indexed: 12/23/2023] Open
Abstract
It is imperative to optimally utilize virtues and obviate defects of fully automated analysis and expert knowledge in new paradigms of healthcare. We present a deep learning-based semiautomated workflow (RAINMAN) with 12,809 follow-up scans among 2,172 patients with treated nasopharyngeal carcinoma from three centers (ChiCTR.org.cn, Chi-CTR2200056595). A boost of diagnostic performance and reduced workload was observed in RAINMAN compared with the original manual interpretations (internal vs. external: sensitivity, 2.5% [p = 0.500] vs. 3.2% [p = 0.031]; specificity, 2.9% [p < 0.001] vs. 0.3% [p = 0.302]; workload reduction, 79.3% vs. 76.2%). The workflow also yielded a triaging performance of 83.6%, with increases of 1.5% in sensitivity (p = 1.000) and 0.6%-1.3% (all p < 0.05) in specificity compared to three radiologists in the reader study. The semiautomated workflow shows its unique superiority in reducing radiologist's workload by eliminating negative scans while retaining the diagnostic performance of radiologists.
Collapse
Affiliation(s)
- Ying-Ying Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yi-Shu Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Information, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- School of Electronics and Information Technology, Sun Yat-sen University, Guangzhou 510006, China
| | - Yang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Meng-Yun Qiang
- Department of Radiation Oncology, Cancer Hospital of The University of Chinese Academy of Sciences, Hangzhou 310005, China
| | - Wen-Ze Qiu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou 510095, China
| | - Wei-Xiong Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bing-Zhong Jing
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Information, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chen-Yang Feng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Information, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hao-Hua Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Information, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xun Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Critical Care Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jia-Yu Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hao-Yang Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ze-Jiang Zhan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Lin-Quan Tang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hai-Qiang Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan-Miao Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiang Guo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Liang-Ru Ke
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Radiology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xing Lv
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chao-Feng Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou 510060, China
- Department of Information, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
49
|
Amarah A, Elsabagh AA, Ouda A, Karen O, Ferih K, Elmakaty I, Malki MI. Emerging roles of activating transcription factor 2 in the development of breast cancer: a comprehensive review. PRECISION CLINICAL MEDICINE 2023; 6:pbad028. [PMID: 37955015 PMCID: PMC10639104 DOI: 10.1093/pcmedi/pbad028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Activating transcription factor 2 (ATF2) is a member of the leucine zipper family of DNA binding proteins that are responsible for regulating various genes that play an essential role in major biological and cellular functions. Since ATF2 plays a vital role in cellular proliferation and apoptosis, it is believed that it greatly affects the development of breast cancers. However, its exact role in breast cancer is incompletely understood. It remains a subject of debate, ambiguity, and continuous research. Several studies have suggested the role of ATF2 as an oncogene, promoting cellular proliferation and worsening the outcome of cancers. In contrast, other studies have postulated that ATF2 plays a tumor suppressive role in estrogen receptor-positive breast cancer. The ambiguity surrounding its role in breast cancer is the reason why there is an influx of recent studies and research in this area. In this narrative review, we investigate several studies that have been published about the role of ATF2 in breast cancer. We also explore studies that have examined the association between ATF2 and endocrine therapy resistance. ATF2 has been suggested to modulate estrogen receptor (ER) expression and activity, potentially affecting tamoxifen sensitivity in breast cancer cells. Therefore, the role of ATF2 in DNA repair mechanisms and drug resistance has been deeply explored in this review. Additionally, there are numerous ongoing clinical trials exploring the effect of targeting ATF2 pathways and mechanisms on the outcome of breast cancers, some of which we have discussed. The studies and clinical trials that are being conducted to understand the multifaceted role of ATF2 and its signaling pathways may provide valuable insight for developing efficient targeted therapeutic solutions to enhance the outcomes of breast cancer and overcome endocrine resistance. We suggest further research to elucidate the dual roles of ATF2 in breast cancer and potential therapeutic therapies for its treatment.
Collapse
Affiliation(s)
- Ahmed Amarah
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Ahmed Adel Elsabagh
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Amr Ouda
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Omar Karen
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Khaled Ferih
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Ibrahim Elmakaty
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Mohammed Imad Malki
- College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
50
|
You K, Hwang JA, Sohn DK, Lee DW, Park SS, Han KS, Hong CW, Kim B, Kim BC, Park SC, Oh JH. Exfoliate cancer cell analysis in rectal cancer surgery: comparison of laparoscopic and transanal total mesorectal excision, a pilot study. Ann Coloproctol 2023; 39:502-512. [PMID: 38145898 PMCID: PMC10781597 DOI: 10.3393/ac.2023.00479.0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/26/2023] [Accepted: 11/05/2023] [Indexed: 12/27/2023] Open
Abstract
PURPOSE Minimally invasive surgery (MIS) is currently the standard treatment for rectal cancer. However, its limitations include complications and incomplete total mesorectal resection (TME) due to anatomical features and technical difficulties. Transanal TME (TaTME) has been practiced since 2010 to improve this, but there is a risk of local recurrence and intra-abdominal contamination. We aimed to analyze samples obtained through lavage to compare laparoscopic TME (LapTME) and TaTME. METHODS From June 2020 to January 2021, 20 patients with rectal cancer undergoing MIS were consecutively and prospectively recruited. Samples were collected at the start of surgery, immediately after TME, and after irrigation. The samples were analyzed for carcinoembryonic antigen (CEA) and cytokeratin 20 (CK20) through a quantitative real-time polymerase chain reaction. The primary outcome was to compare the detected amounts of CEA and CK20 immediately after TME between the surgical methods. RESULTS Among the 20 patients, 13 underwent LapTME and 7 underwent TaTME. Tumor location was lower in TaTME (7.3 cm vs. 4.6 cm, P=0.012), and negative mesorectal fascia (MRF) was more in LapTME (76.9% vs. 28.6%, P=0.044). CEA and CK20 levels were high in 3 patients (42.9%) only in TaTME. There was 1 case of T4 with incomplete purse-string suture and 1 case of positive MRF with dissection failure. All patients were followed up for an average of 32.5 months without local recurrence. CONCLUSION CEA and CK20 levels were high only in TaTME and were related to tumor factors or intraoperative events. However, whether the detection amount is clinically related to local recurrence remains unclear.
Collapse
Affiliation(s)
- Kiho You
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Jung-Ah Hwang
- Genomics Core Facility, Research Core Center, National Cancer Center, Goyang, Korea
| | - Dae Kyung Sohn
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Dong Woon Lee
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Sung Sil Park
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Kyung Su Han
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Chang Won Hong
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Bun Kim
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Byung Chang Kim
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Sung Chan Park
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| | - Jae Hwan Oh
- Center for Colorectal Cancer, National Cancer Center, Goyang, Korea
| |
Collapse
|