1
|
Sun Q, Li CH, Liu QS, Zhang YB, Hu BS, Feng Q, Lang Y. Research status of biomaterials based on physical signals for bone injury repair. Regen Ther 2025; 28:544-557. [PMID: 40027992 PMCID: PMC11872413 DOI: 10.1016/j.reth.2025.01.025] [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: 10/08/2024] [Revised: 01/02/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025] Open
Abstract
Bone defects repair continues to be a significant challenge facing the world. Biological scaffolds, bioactive molecules, and cells are the three major elements of bone tissue engineering, which have been widely used in bone regeneration therapy, especially with the rise of bioactive molecules in recent years. According to their physical properties, they can be divided into force, magnetic field (MF), electric field (EF), ultrasonic wave, light, heat, etc. However, the transmission of bioactive molecules has obvious shortcomings that hinder the development of the tissue-rearing process. This paper reviews the mechanism of physical signal induction in bone tissue engineering in recent years. It summarizes the application strategies of physical signal in bone tissue engineering, including biomaterial designs, physical signal loading strategies and related pathways. Finally, the ongoing challenges and prospects for the future are discussed.
Collapse
Affiliation(s)
- Qi Sun
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Chao-Hua Li
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Qi-Shun Liu
- Department of Orthopedics, Zhejiang Medical & Health Group Hangzhou Hospital, Hangzhou, 310015, China
| | - Yuan-Bin Zhang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Bai-Song Hu
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Qi Feng
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| | - Yong Lang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou, 311499, China
| |
Collapse
|
2
|
Cao B, Liu M, Xiao Z, Leng D, Zhou Y, Zhang Z, Wang L, Huang X, Ni Q, Cheng W, Assaraf YG, Zhao Q, Shen J, Zhu K. CV1-secreting sCAR-T cells potentiate the abscopal effect of microwave ablation in heterogeneous tumors. Cell Rep Med 2025; 6:101965. [PMID: 39970874 DOI: 10.1016/j.xcrm.2025.101965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/01/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025]
Abstract
Microwave ablation (MWA) triggers a weak systemic immune response that leads to the abscopal regression of distant metastases while killing local tumors, known as the abscopal effect. Combining MWA with chimeric antigen receptor (CAR)-T cells demonstrates promise in enhancing the abscopal effect in antigen-homogeneous tumors. However, the loss of the antigen recognized by CAR or intrinsic antigenic heterogeneity in solid tumors poses a major obstacle. SIRPα variant (CV1)-secreting CAR-T (sCAR-T) cells elicit an abscopal effect on distant tumors with antigen heterogeneity in mice receiving local MWA. Mechanistically, sCAR-T cells can locally eliminate antigen-positive tumors and secrete CV1, whereas the secreted CV1 can activate macrophages that migrate to non-ablated tumor sites in response to post-MWA chemokines, eliciting a macrophage-dependent abscopal effect that enables phagocytosis of antigen-heterogeneous cancer cells. This macrophage-dependent abscopal effect instigated by MWA and sCAR-T cells offers a clinically translatable strategy in metastatic solid tumors with antigen heterogeneity.
Collapse
Affiliation(s)
- Bihui Cao
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China; Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA
| | - Manting Liu
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Zecong Xiao
- Nanomedicine Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Dongliang Leng
- MoE Frontiers Science Center for Precision Oncology, Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yubo Zhou
- Department of Library, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Zhenfeng Zhang
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Lu Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xinkun Huang
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Qianqian Ni
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Wei Cheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Qi Zhao
- MoE Frontiers Science Center for Precision Oncology, Institute of Translational Medicine, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China.
| | - Jia Shen
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA; Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, Department of Radiology, Central Laboratory, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| |
Collapse
|
3
|
Nafie EHO, Pastori C, Acharya R, Kaviani R, Hunter D, Shalaby W, Krimsky WS, Neal RE. Understanding the aliya pulsed electric field dose-response relationship: Implications for ablation size, thermal load, and immune response in an orthotopic murine breast cancer model. PLoS One 2025; 20:e0318440. [PMID: 39946406 PMCID: PMC11824980 DOI: 10.1371/journal.pone.0318440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Aliya®Pulsed Electric Field (PEF) technology is an emerging strategy in the field of cancer treatment, offering a novel approach to ablation therapy that does not rely on thermal mechanisms. By employing a multi-stage experimental setup, including potato tuber, porcine liver, and murine breast cancer models, we explored the dose-response relationship on ablation and immune modulation by varying the pulse packets delivered from 20 to 100. The biologic response observed with 60 packets represented a minimum effective dose yielding reproducible ablation parameters, immune response, and efficacy which could be augmented with immune checkpoint blockade. This pre-clinical analysis provides a first step toward understanding the therapeutic index for PEF technology beyond ablation, a consideration that will require robust clinical validation in well-designed prospective studies.
Collapse
Affiliation(s)
| | - Chiara Pastori
- Galvanize Therapeutics, Redwood City, CA, United States of America
| | - Rupsa Acharya
- Galvanize Therapeutics, Redwood City, CA, United States of America
| | - Rosa Kaviani
- Galvanize Therapeutics, Redwood City, CA, United States of America
| | - David Hunter
- Galvanize Therapeutics, Redwood City, CA, United States of America
| | - Waleed Shalaby
- Galvanize Therapeutics, Redwood City, CA, United States of America
| | | | - Robert E. Neal
- Galvanize Therapeutics, Redwood City, CA, United States of America
| |
Collapse
|
4
|
Liu Q, Zhang R, Shen W. Advancements in locoregional therapy for advanced hepatocellular carcinoma: Emerging perspectives on combined treatment strategies. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109502. [PMID: 39615292 DOI: 10.1016/j.ejso.2024.109502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/17/2024] [Accepted: 11/23/2024] [Indexed: 01/03/2025]
Abstract
Hepatocellular carcinoma (HCC) persists as a leading cause of cancer-related mortality, often diagnosed at advanced stages with limited treatment options. Locoregional therapies (LRTs) are crucial in HCC management, playing significant roles in neoadjuvant and palliative treatments, among others. However, the unique disease background of HCC necessitates multidisciplinary and integrated treatment strategies. The therapeutic landscape for advanced HCC has been significantly broadened by the advent of combined therapies, presenting multiple approaches aimed at improving long-term survival, which remains a critical challenge. This review offers a comprehensive overview of major LRTs for HCC, highlighting recent technological advancements and exploring the challenges and limitations in their application, and presents the latest developments in combination therapies, including combinations between different LRTs and their integration with systemic treatments. Additionally, we outline future directions for the development of integrated treatment modalities for advanced HCC.
Collapse
Affiliation(s)
- Qi Liu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China; The Second Clinical Medical College of Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Renjie Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China; The Second Clinical Medical College of Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Weixi Shen
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
| |
Collapse
|
5
|
Xie GL, Zhong ZH, Ye TW, Xiao ZQ. Radiofrequency ablation combined with immunotherapy to treat hepatocellular carcinoma: a comprehensive review. BMC Surg 2025; 25:47. [PMID: 39875933 PMCID: PMC11776151 DOI: 10.1186/s12893-025-02778-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND AND AIM Hepatocellular carcinoma (HCC) is a highly immunogenic tumor and the third leading cause of cancer-related deaths worldwide with an increasing incidence. Therefore, the combination of immunotherapy with other approaches, such as anti-angiogenic agents and local area therapy, has become a new strategy for HCC treatment. METHODS We searched PubMed and Web of Science and extracted publications relating to the radiofrequency ablation (RFA) and immunotherapy. The search terms were: "radiofrequency ablation", "immunotherapy" and "hepatocellular carcinoma", and manual searches of eligible articles from literature reference lists were performed. We then thoroughly reviewed the literature on ablation combined with immunotherapy for HCC, analyzed the relevant mechanism, and explored the safety and effectiveness of this form of combination therapy. RESULTS RFA combined with immunotherapy in HCC is reported to have good efficacy and controllable safety. On the one hand, RFA can induce the immunogenic substances including Ficolin-3, IL-1 and heat shock protein and regulate the immune cells by mediating the Th1/Th2 ratio, increasing Th17 cells, etc. On the other hand, RFA treatment can lead to tumor immune microenvironment reconstruction, increasing the proportion of functional T cells and upregulate PD-1 in T cells in distant tumors without RFA. This combined strategy has the ability to enhance the anti-tumor immune response through synergies, significantly reduce the risk of recurrence and improve survival. CONCLUSIONS RFA combined with immunotherapy yields a good synergistic effect: it can further strengthen anti-tumor response, delay distant tumor growth, reduce tumor recurrence and metastasis, providing new options for HCC systemic treatment.
Collapse
Affiliation(s)
- Gui-Lin Xie
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, China
| | - Zhi-Han Zhong
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Tai-Wei Ye
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zun-Qiang Xiao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
6
|
Wang M, Sang J, Xu F, Wang S, Liu P, Ma J, Chen Z, Xie Q, Wei Z, Ye X. Microwave Ablation Combined with Flt3L Provokes Tumor-Specific Memory CD8 + T Cells-Mediated Antitumor Immunity in Response to PD-1 Blockade. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413181. [PMID: 39629989 PMCID: PMC11775548 DOI: 10.1002/advs.202413181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Indexed: 01/12/2025]
Abstract
For medically inoperable non-small cell lung cancer, microwave ablation (MWA) represents a super minimally invasive alternative treatment. However, tumor recurrence remains a concern. Here, it is demonstrated that the combination of MWA with Flt3L significantly inhibits tumor recurrence by CD8+ central memory T (TCM)-like cell-dependent antitumor immune responses within the tumor-draining lymph nodes (TdLN). TdLN-TCM-like cells encompassed both tumor-specific memory T (TTSM) and progenitor-exhausted T (TPEX) cells. The expansion of these cells markedly altered the differentiation of exhausted T cells within the tumor microenvironment (TME). TPEX predominantly differentiated into transitory effector-like exhausted T cells (TEX-int). The expansion of TTSM cells elicited by the combined therapy was reliant on conventional dendritic cells (cDCs) and was likely specifically dependent on the migratory cDC1s (Mig cDC1s) within the TdLN. The upregulation of ICOSL on migratory cDC1s was pivotal in initiating TTSM-like cell-mediated antitumor responses. Slc38a2 may be a critical gene responsible for the upregulation of ICOSL in Mig cDC1s following combined treatment. Finally, the combined treatment significantly enhanced the antitumor efficacy of immunotherapy based on PD-1 blockade. The research thereby afforded a novel strategic approach to forestall tumor recurrence after MWA therapy, while also providing the foundational proof-of-concept for impending clinical investigations.
Collapse
Affiliation(s)
- Meixiang Wang
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
| | - Jing Sang
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
- Department of PathologyShandong Provincial Third Hospital11 Wuyingshan Zhonglu RoadJinan250100P. R. China
| | - Fengkuo Xu
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
| | - Shulong Wang
- Shandong Academy of Preventive MedicineShandong Center for Disease Control and Prevention16992 Jingshi RoadJinan250014P. R. China
| | - Peng Liu
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
| | - Ji Ma
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
| | - Zhengtao Chen
- School of Laboratory Animal & Shandong Laboratory Animal CenterShandong First Medical University & Shandong Academy of Medical Sciences6699 Qingdao RoadJinan250014P. R. China
| | - Qi Xie
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
| | - Zhigang Wei
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
- Cheeloo College of MedicineShandong University27 Shanda Nanlu RoadJinan250100P. R. China
| | - Xin Ye
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalShandong Provincial Lab for Clinical Immunology Translational Medicine in UniversitiesShandong Lung Cancer Institute16766 Jingshi RoadJinan250014P. R. China
| |
Collapse
|
7
|
Xie Y, Lin N, Song P, Ni X, Wang Y, Huang P, Han Z, Wang D, Sun N. Identification of immunostimulatory activities and active compounds from sequentially extracted fractions of rhizosphere fungal fermentation broth of Atractylodes macrocephala Koidz. rhizomes. Front Pharmacol 2024; 15:1460614. [PMID: 39759456 PMCID: PMC11695301 DOI: 10.3389/fphar.2024.1460614] [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: 07/09/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Pharmacological studies have shown that the rhizome of Atractylodes macrocephala Koidz. (Compositae), commonly known as atractylodes macrocephala rhizome (AMR), can modulate immunity. Nevertheless, its resources have been largely depleted, and the pharmacological activity of artificial AMR is relatively modest. We hypothesized that the fermented crude extracts of the rhizosphere fungi of AMR would have similar immunomodulatory effects since the metabolites generated by these fungi are similar to those of the host plant given their long-term synergistic evolution. Methods Rhizosphere fungi were isolated from the rhizosphere soil of AMR and cultured to produce the secondary metabolites. These metabolites were then sequentially extracted with four solvents of increasing polarities (petroleum ether, ethyl acetate, n-butanol, and water). The in vitro immunomodulatory activities of the metabolite extracts were evaluated by cell proliferation capacity, cell phagocytosis activity, NO secretion capacity, cell morphology changes, and cytokine (TNF-α, IL-1β and IL-6) secretion capacity in RAW264.7 macrophage cells. The biologically active secondary metabolites produced by the rhizosphere fungi were identified using ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS). Results Three rhizosphere fungi, namely Penicillium (MK-1), Penicillium glaucoroseum (MN-1), and Purpureocillium lilalium (MG-1), were isolated from the rhizosphere soil of AMR. The assays for cell proliferation capacity, cell phagocytosis activity, and NO secretion capacity showed that all metabolite extracts exhibited in vitro immunomodulatory activities. The crude extracts of MG-1 exhibited the highest levels of in vitro immunomodulatory activities compared to the other extracts. Furthermore, it was demonstrated that the fermented extracts of MG-1 could facilitate immunological enhancement in vitro by altering the cellular morphology in the resting state and increasing the secretions of TNF-α, IL-1β, and IL-6. Meanwhile, there was no observable endotoxin contamination. The metabolite profiling of MG-1 by UHPLC-Q-TOFMS revealed the presence of several compounds with established immunoreactive activities, including L-arginine, prostaglandin I2, deoxyguanosine, bestatin, and osthole. Discussion The present study demonstrated that the metabolite extracts of the rhizosphere fungi isolated from the rhizosphere soil of AMR exhibited in vitro immunoreactive activities and that these rhizosphere fungi could produce several bioactive metabolites. The crude extracts of the rhizosphere fungi may hence extend the medicinal utility of AMR and provide a basis for further development of natural plant-based immunomodulators.
Collapse
Affiliation(s)
- Yuxin Xie
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Na Lin
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Pingping Song
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xiangyan Ni
- Beijing Municipal Bureau of Agriculture and Rural Affairs, Beijing Agricultural Product Quality and Safety Center, Beijing, China
| | - Yakun Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Peng Huang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Zhili Han
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Dianlei Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| | - Nianxia Sun
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Research and Development of Chinese Medicine, Hefei, China
| |
Collapse
|
8
|
Alférez MD, Corda A, de Blas I, Gago L, Fernandes T, Rodríguez-Piza I, Balañá B, Pentcheva P, Caruncho J, Barbero-Fernández A, Llinás J, Rivas D, Escudero A, Gómez-Ochoa P. Computed Tomography-Guided Radiofrequency Ablation of Nasal Carcinomas in Dogs. Animals (Basel) 2024; 14:3682. [PMID: 39765586 PMCID: PMC11672759 DOI: 10.3390/ani14243682] [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: 11/26/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Nasal carcinomas in dogs are locally invasive neoplasms with a low metastatic rate that pose significant treatment challenges due to their location and aggressiveness. This study evaluates the safety, feasibility, and therapeutic outcomes of computed tomography-guided radiofrequency ablation (CT-guided RFA) in 15 dogs diagnosed with nasal adenocarcinoma. All patients underwent staging and histopathological diagnosis before treatment. CT-guided RFA achieved a significant tumor volume reduction (82.8%) and improvement in clinical signs such as nasal discharge, epistaxis, and respiratory distress, without complications. Post-RFA CT examinations demonstrated a significant decrease in Hounsfield units and tumor volume. This study has shown that CT-guided RFA is an effective cytoreductive option for minimally invasive management of nasal adenocarcinomas in dogs, particularly when traditional therapies like radiation therapy or surgery are not feasible.
Collapse
Affiliation(s)
| | - Andrea Corda
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | - Ignacio de Blas
- Department of Animal Pathology, University of Zaragoza, 50013 Zaragoza, Spain;
| | - Lucas Gago
- Department of Mathematics and Computer Science, University of Barcelona, 08007 Barcelona, Spain;
| | - Telmo Fernandes
- Imaginologia Veterinaria do Porto, 4490-479 Porto, Portugal;
| | | | - Beatriz Balañá
- Hospital Anicura Aralar Veterinarios, 50410 Zaragoza, Spain; (B.B.); (D.R.); (A.E.)
| | - Plamena Pentcheva
- Department of Veterinary Medicine, University of Sassari, 07100 Sassari, Italy;
| | | | | | - Jorge Llinás
- Hospital Anicura Valencia Sur, 46460 Valencia, Spain;
| | - David Rivas
- Hospital Anicura Aralar Veterinarios, 50410 Zaragoza, Spain; (B.B.); (D.R.); (A.E.)
| | - Amaia Escudero
- Hospital Anicura Aralar Veterinarios, 50410 Zaragoza, Spain; (B.B.); (D.R.); (A.E.)
| | - Pablo Gómez-Ochoa
- VetCorner Unavets, 50012 Zaragoza, Spain; (M.D.A.); (P.G.-O.)
- Department of Animal Pathology, University of Zaragoza, 50013 Zaragoza, Spain;
| |
Collapse
|
9
|
He W, Yang F, Chen K, Zeng Q. Targeted gold nanoparticles for ovarian cancer (Review). Oncol Lett 2024; 28:589. [PMID: 39417039 PMCID: PMC11481100 DOI: 10.3892/ol.2024.14723] [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: 06/07/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Among all malignant gynecological tumors, ovarian cancer (OC) has the highest mortality rate. OC is often diagnosed at advanced and incurable stages; however, early diagnosis can enable the use of optimized and personalized treatments. Intensive research into the synthesis and characterization of gold nanoparticles (AuNPs) has been performed with the aim of developing innovative materials for use in biological and photothermal therapies for OC. AuNPs can be chemically modified and functionalized by binding to a variety of organic compounds and biomolecules, such as peptides, antibodies and therapeutic agents, via simple synthetic processes. They are particularly suitable for use as carriers for drug delivery. In the present review, the synthesis and characteristics of AuNPs are summarized, and their potential in OC therapy are discussed.
Collapse
Affiliation(s)
- Wenjuan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Fuyuan Yang
- School of Basic Medicine, Yangtze University Health Science Center, Jingzhou, Hubei 434000, P.R. China
| | - Keming Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| | - Qingsong Zeng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
10
|
Mekers V, de Visser M, Suijkerbuijk K, Bos C, Moonen C, Deckers R, Adema G. Mechanical HIFU and immune checkpoint inhibition: toward clinical implementation. Int J Hyperthermia 2024; 41:2430333. [PMID: 39566471 DOI: 10.1080/02656736.2024.2430333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/21/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Objective: Immune checkpoint inhibition (ICI) has significantly advanced the field of immuno-oncology, yet not all patients benefit from this therapy. Combining ICI with other therapeutic modalities, including tumor ablation, is currently being explored as a method to enhance ICI efficacy. Mechanical High-Intensity Focused Ultrasound (M-HIFU) represents a promising tumor ablative therapy, inducing cavitation within the tumor, resulting in tumor cell destruction and the release of danger signals and tumor antigens, two key factors contributing to anti-tumor immune responses. Methods/Results: Preclinical studies on the impact of M-HIFU on the anti-tumor immune response are guiding the translational application of this technique in the clinical setting. This review provides a comprehensive overview of the current understanding of the effects of M-HIFU on the immune system. We report on the effect of M-HIFU on soluble immune modulators and immune cells in different preclinical models, and potential contributions to the anti-tumor immune response. We discuss clinical studies applying M-HIFU and studies that have combined ICI with other ablative therapies to draw parallels to clinical implementation of M-HIFU. Further, we will highlight essential questions that should be addressed in future clinical trials exploring the combination of M-HIFU and ICI in the clinical setting. Conclusion: Overall, this review offers guidance for the clinical implementation of combining M-HIFU with ICI and highlights key questions that remain to be addressed in first clinical studies.
Collapse
Affiliation(s)
- Vera Mekers
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mirjam de Visser
- Division of Imaging & Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Karijn Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Clemens Bos
- Division of Imaging & Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Chrit Moonen
- Division of Imaging & Oncology, University Medical Center Utrecht, Utrecht, Netherlands
- Focused Ultrasound Foundation, Charlottesville, VA, USA
| | - Roel Deckers
- Division of Imaging & Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Gosse Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
11
|
He P, Ma L, Xu B, Wang Y, Li X, Chen H, Li Y. Research progress and future directions of immune checkpoint inhibitor combination therapy in advanced gastric cancer. Ther Adv Med Oncol 2024; 16:17588359241266156. [PMID: 39091604 PMCID: PMC11292724 DOI: 10.1177/17588359241266156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
In recent years, with the continuous development of molecular immunology, immune checkpoint inhibitors (ICIs) have also been widely used in the treatment of gastric cancer, but they still face some challenges: The first is that only some people can benefit, the second is the treatment-related adverse events (TRAEs) that occur during treatment, and the third is the emergence of varying degrees of drug resistance with long-term use. How to overcome these challenges, combined therapy based on ICIs has become one of the important strategies. This article summarizes the clinical application of ICIs combined with chemotherapy, targeted therapy, radiotherapy, photodynamic therapy, thermotherapy, immune adjuvant, and dual immunotherapy and discusses the mechanism, and also summarizes the advantages and disadvantages of the current combination modalities and the potential research value. The aim of this study is to provide more and more optimized combination regimen for ICI combined therapy in patients with advanced gastric cancer and to provide reference for clinical and scientific research.
Collapse
Affiliation(s)
- Puyi He
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Long Ma
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Bo Xu
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Yunpeng Wang
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Xiaomei Li
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Hao Chen
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
- No. 82, Cuiyingmen, Chengguan, Lanzhou 730030, China
| | - Yumin Li
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
- No. 82, Cuiyingmen, Chengguan, Lanzhou 730030, China
| |
Collapse
|
12
|
Waidhauser J, Gantner AK, Schifano P, Rippel K, Schiele S, Arndt TT, Müller G, Steinestel J, Rank A, Kröncke T. Influence of cryoablation versus operation on circulating lymphocyte subsets in patients with early-stage renal cell carcinoma. BMC Cancer 2024; 24:825. [PMID: 38987735 PMCID: PMC11238514 DOI: 10.1186/s12885-024-12596-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: 08/07/2023] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Immune response is known to play an important role in local tumor control especially in renal cell carcinoma (RCC), which is considered highly immunogenic. For localized tumors, operative resection or local ablative procedures such as cryoablation are common therapeutical options. For thermal ablative procedures such as cryoablation, additional immunological anti-tumor effects have been described.The purpose of this prospective study was to determine changes in peripheral blood circulating lymphocytes and various of their subsets in RCC patients treated with cryoablation or surgery in a longitudinal approach using extensive flow cytometry. Additionally, lymphocytes of RCC patients were compared to a healthy control group.We included 25 patients with RCC. Eight underwent cryoablation and 17 underwent surgery. Univariate and multivariable analysis revealed significantly lower values of B cells, CD4 and CD8 T cells, and various of their subsets in the treatment groups versus the healthy control group. Comparing the two different therapeutical approaches, a significant decline of various lymphocyte subsets with a consecutive normalization after three months was seen for the surgery group, whereas cryoablation led to increased values of CD69 + CD4 + and CD69 + CD8 + cell counts as well as memory CD8 + cells after three months.Treatment-naïve RCC patients showed lower peripheral blood lymphocyte counts compared to healthy controls. The post-treatment course revealed different developments of lymphocytes in the surgery versus cryoablation group, and only cryoablation seems to induce a sustained immunological response after three months.
Collapse
Affiliation(s)
- Johanna Waidhauser
- Department of Hematology and Oncology, University Medical Center Augsburg, Stenglinstr.2, 86156, Augsburg, Germany.
| | - Anna-Katharina Gantner
- Department of Hematology and Oncology, University Medical Center Augsburg, Stenglinstr.2, 86156, Augsburg, Germany
| | - Paola Schifano
- Department of Urology, University Medical Center Augsburg, Augsburg, Germany
| | - Katharina Rippel
- Department of Diagnostic and Interventional Radiology, University Medical Center Augsburg, Augsburg, Germany
| | - Stefan Schiele
- Institute of Mathematics, Faculty of Mathematics and Natural Sciences, University of Augsburg, Augsburg, Germany
| | - Tim Tobias Arndt
- Institute of Mathematics, Faculty of Mathematics and Natural Sciences, University of Augsburg, Augsburg, Germany
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Gernot Müller
- Institute of Mathematics, Faculty of Mathematics and Natural Sciences, University of Augsburg, Augsburg, Germany
| | - Julie Steinestel
- Department of Urology, University Medical Center Augsburg, Augsburg, Germany
| | - Andreas Rank
- Department of Hematology and Oncology, University Medical Center Augsburg, Stenglinstr.2, 86156, Augsburg, Germany
| | - Thomas Kröncke
- Department of Diagnostic and Interventional Radiology, University Medical Center Augsburg, Augsburg, Germany
| |
Collapse
|
13
|
Xiao YL, Wu SY, Jiang YZ. Thermo-immune synergy: Camrelizumab plus microwave ablation in preoperative early-stage breast cancer. MED 2024; 5:278-280. [PMID: 38614071 DOI: 10.1016/j.medj.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 04/15/2024]
Abstract
Immunotherapy has enhanced breast cancer outcomes, but optimizing combination therapies is crucial. Integrating additional treatment modalities, like physical therapies, holds promise for optimizing efficacy. Pan et al. recently reported that combining preoperative immunotherapy with microwave ablation is safe and feasible in early-stage breast cancer, effectively sensitizing peripheral CD8+ T cells.1.
Collapse
Affiliation(s)
- Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Song-Yang Wu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
14
|
Liu P, Wei Z, Ye X. Immunostimulatory effects of thermal ablation: Challenges and future prospects. J Cancer Res Ther 2024; 20:531-539. [PMID: 38687922 DOI: 10.4103/jcrt.jcrt_2484_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/13/2023] [Indexed: 05/02/2024]
Abstract
ABSTRACT This literature explores the immunostimulatory effects of thermal ablation in the tumor microenvironment, elucidating the mechanisms such as immunogenic cell death, tumor-specific antigens, and damage-associated molecular patterns. Furthermore, it outlines critical issues associated with thermal ablation-induced immunostimulatory challenges and offers insights into future research avenues and potential therapeutic strategies.
Collapse
Affiliation(s)
- Peng Liu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, No. 16766 Jingshi Road, Jinan, Shandong Province, China
| | | | | |
Collapse
|
15
|
Mustafa AR, Miyasato D, Wehrenberg-Klee E. Synergizing Thermal Ablation Modalities with Immunotherapy: Enough to Induce Systemic Antitumoral Immunity? J Vasc Interv Radiol 2024; 35:185-197. [PMID: 38272639 DOI: 10.1016/j.jvir.2023.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 01/27/2024] Open
Abstract
Thermal ablation modalities (cryoablation, radiofrequency ablation, and microwave ablation) have long been noted to occasionally induce a systemic antitumoral response. With the widespread use of checkpoint inhibitors, there is a significant interest in whether thermal ablation can promote immune system tumor recognition and increase checkpoint inhibitor response rates. In this review, we examine the current state of preclinical and clinical evidence examining the combination of checkpoint inhibitor therapies and thermal ablation modalities as well as discuss remaining the unanswered questions and directions for future research.
Collapse
Affiliation(s)
- Abdul Rehman Mustafa
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | | | - Eric Wehrenberg-Klee
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
16
|
Altun I, Demirlenk YM, Atar D, Cevik E, Gunduz S, Albadawi H, Oklu R. Advances and Challenges in Interventional Immuno-Oncology Locoregional Therapies. J Vasc Interv Radiol 2024; 35:164-172. [PMID: 38272636 DOI: 10.1016/j.jvir.2023.10.009] [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: 08/19/2023] [Accepted: 10/15/2023] [Indexed: 01/27/2024] Open
Abstract
Interventional immuno-oncology is making strides in locoregional therapies to address complex tumor microenvironments. Long-standing interventional radiology cancer therapies, such as tumor ablation and embolization, are being recharacterized in the context of immunotherapy. Intratumoral injections, such as those of genetically engineered or unaltered viruses, and the delivery of immune cells, antibodies, proteins, or cytokines into targeted tumors, along with advancements in delivery techniques, have produced promising results in preliminary studies, indicating their antitumor effectiveness. Emerging strategies using DNA scaffolding, polysaccharides, glycan, chitosan, and natural products are also showing promise in targeted cancer therapy. The future of interventional immuno-oncology lies in personalized immunotherapies that capitalize on individual immune profiles and tumor characteristics, along with the exploration of combination therapies. This study will review various interventional immuno-oncology strategies and emerging technologies to enhance delivery of therapeutics and response to immunotherapy.
Collapse
Affiliation(s)
- Izzet Altun
- Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Yusuf M Demirlenk
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Dila Atar
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Enes Cevik
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Seyda Gunduz
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona; Department of Medical Oncology, Istinye University Bahcesehir Liv Hospital, Istanbul, Turkey
| | - Hassan Albadawi
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona
| | - Rahmi Oklu
- Division of Vascular and Interventional Radiology, Laboratory for Patient Inspired Engineering, Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
17
|
Deng H, Huang J, Gao N, Liu Z, Yi Z, Xiao J, Gao X, Zhang C, Juliet M, Hu J, Chen J, Zu X. Nanotherapeutic System with Effective Microwave Sensitization and Pyroptosis Programming Enable Synergistic Microwave-Immunotherapy in Bladder Cancer. Biomater Res 2024; 28. [DOI: 10.34133/bmr.0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 08/10/2024] [Indexed: 12/01/2024] Open
Abstract
Currently, the prognosis for patients with advanced bladder cancer remains poor, with only a minority being sensitive to immune checkpoint inhibitors. There is a need to develop additional treatment strategies. Microwave therapy, as a promising approach for some inoperable tumors, still faces challenges such as limited efficacy and high recurrence rates. Additionally, the cell damage and necrosis induced by conventional microwave treatment only act as weak immunostimulatory factors for antitumor immunity, failing to activate effective antitumor immune responses. Recent discoveries have shown that inducing pyroptosis can provide a good opportunity for enhancing systemic immune responses and alleviating immune suppression in cancer therapy. Here, we have developed Mn-ZrMOF@DAC, a microwave-sensitized nanoparticle loaded with the DNA methylation inhibitor decitabine. The Mn-ZrMOF@DAC can enhance the effect of microwave thermal therapy and generate reactive oxygen species under microwave irradiation, causing thermal and oxidative damage to cancer cells. Furthermore, there was an important up-regulation of the key pyroptosis protein GSDME, with a marked increase in pyroptotic cell numbers. In vivo experiments demonstrated that mice injected with Mn-ZrMOF@DAC nanoparticles followed by microwave radiation treatment exhibited potent antitumor effects and enhanced the efficacy of anti-PD-1 therapy. This therapy not only enhanced the efficacy of microwave treatment, exhibiting significant antitumor effects, but also activated antitumor immunity by inducing pyroptosis, thus enhancing the efficacy of immunotherapy for bladder cancer. It holds promise for providing new avenues in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Hao Deng
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders,
Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, Southwest Hospital,
Army Medical University, Chongqing, People’s Republic of China
| | - Jinliang Huang
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders,
Xiangya Hospital, Central South University, Changsha, China
| | - Ning Gao
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Zhi Liu
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Zhenglin Yi
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jiatong Xiao
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Xin Gao
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Chunyu Zhang
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Matsika Juliet
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders,
Xiangya Hospital, Central South University, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders,
Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital,
Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders,
Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The First Affiliate Hospital of Hunan Normal,
University (Hunan Provincial People’s Hospital), Changsha, Hunan Province China
| |
Collapse
|
18
|
Luo T, Wang Z, Yu X, Han Z, Cheng Z, Liu F, Yu J, Liang P. More Ultrasound-Guided Percutaneous Microwave Ablation Leads to Higher Immune-Related Gene Expression and Boosts PD-1 Monoclonal Antibodies for Liver Cancer. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:150-157. [PMID: 37867090 DOI: 10.1016/j.ultrasmedbio.2023.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023]
Abstract
OBJECTIVE The aim of the work described here was to investigate the relative contribution of subtotal ultrasound-guided percutaneous microwave ablation (MWA) to amplifying programmed cell death protein-1 (PD-1) inhibition in advanced hepatocellular carcinoma (HCC). METHODS Between April 2019 and December 2021, advanced HCC patient demographics, tumor response, survival data, neutrophil-to-lymphocyte ratio (NLR) and peripheral lymphocyte profiles were retrospectively collected and analyzed. In hepa1-6 tumor-bearing C57BL/6J mice, RNA sequencing, flow cytometry, immunohistochemistry staining and cytokine tests were also performed. RESULTS Twenty-nine HCC patients were enrolled, with a median follow-up duration of 15.1 mo. Compared with the ablation rate (AR) ≤50% group (n = 10), the AR >50% group (n = 19) had a higher disease control rate, a longer time to progression and a longer overall survival. More patients in the AR >50% group had an early decrease in NLR and better immune activation. RNA sequencing of murine tumors subjected to MWA >50% AR showed that immune-related gene expression upregulated. CD8+ T cells, interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) were also increased, indicating that MWA >50% AR boosted the immunomodulatory effect of PD-1 inhibitors. CONCLUSION More MWA could induce superior antitumor immunity by enhancing immune-related gene expression, priming CD8+ T cells and thereby boosting PD-1 inhibition. It is advisable that eradication of tumors to the degree possible should be considered within technical access to obtain a better prognosis.
Collapse
Affiliation(s)
- Ting Luo
- School of Medicine, Nankai University, Tianjin, China; Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhen Wang
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaoling Yu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhiyu Han
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhigang Cheng
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Fangyi Liu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jie Yu
- Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Ping Liang
- School of Medicine, Nankai University, Tianjin, China; Department of Interventional Ultrasound, Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
19
|
Liao C, Zhang G, Huang R, Zeng L, Chen B, Dai H, Tang K, Lin R, Huang Y. Inducing the Abscopal Effect in Liver Cancer Treatment: The Impact of Microwave Ablation Power Levels and PD-1 Antibody Therapy. Pharmaceuticals (Basel) 2023; 16:1672. [PMID: 38139799 PMCID: PMC10747918 DOI: 10.3390/ph16121672] [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: 10/18/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Microwave ablation (MWA) is an effective treatment for liver cancer (LC), but its impact on distant tumors remains to be fully elucidated. This study investigated the abscopal effects triggered by MWA treatment of LC, at different power levels and with or without combined immune checkpoint inhibition (ICI). We established a mouse model with bilateral subcutaneous LC and applied MWA of varied power levels to ablate the right-sided tumor, with or without immunotherapy. Left-sided tumor growth was monitored to assess the abscopal effect. Immune cell infiltration and distant tumor neovascularization were quantified via immunohistochemistry, revealing insights into the tumor microenvironment and neovascularization status. Th1- and Th2-type cytokine concentrations in peripheral blood were measured using ELISA to evaluate systemic immunological changes. It was found that MWA alone, especially at lower power, promoted distant tumor growth. On the contrary, combining high-power MWA with anti-programmed death (PD)-1 therapy promoted CD8+ T-cell infiltration, reduced regulatory T-cell infiltration, upregulated a Th1-type cytokine (TNF-α) in peripheral blood, and inhibited distant tumor growth. In summary, combining high-power MWA with ICI significantly enhances systemic antitumor immune responses and activates the abscopal effect, offering a facile and robust strategy for improving treatment outcomes.
Collapse
Affiliation(s)
- Changli Liao
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
- Department of Interventional Therapy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, No. 55 South Renmin Road, Section 4, Chengdu 610041, China
| | - Guiyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| | - Ruotong Huang
- Department of Metabolism, Digestion, and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK;
| | - Linyuan Zeng
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| | - Bin Chen
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| | - Haitao Dai
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| | - Keyu Tang
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| | - Run Lin
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| | - Yonghui Huang
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-sen University, No.58 Zhongshan 2nd Road, Guangzhou 510080, China; (C.L.); (G.Z.); (L.Z.); (B.C.); (H.D.); (K.T.)
| |
Collapse
|
20
|
Cho SH, Kim DH, Seo DW, Yoo SK, Oh D, Song TJ, Lee SK. Expanded indication for EUS-guided radiofrequency ablation: management of adrenal tumors. Gastrointest Endosc 2023; 98:790-796. [PMID: 37356635 DOI: 10.1016/j.gie.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/27/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023]
Abstract
BACKGROUND AND AIMS EUS-guided radiofrequency ablation (EUS-RFA) has been performed as an alternative to surgery for the treatment of pancreatic tumors. The promising results obtained using this procedure suggest that the indication for EUS-RFA could be expanded to the management of extrapancreatic tumors. Here, we evaluate the feasibility, efficacy, and safety of EUS-RFA in the treatment of left adrenal tumors. METHODS This single-center, prospective study was conducted at Asan Medical Center between January 2016 and November 2021. A total of 11 patients with left adrenal tumors were enrolled in the study. The technical success rate, treatment response, and adverse events were evaluated. RESULTS EUS-RFA was performed successfully in all patients (technical success rate, 100%). The indications for the procedure were an increase in tumor size (n = 8), and adrenal tumor with adrenal hormone excess (n = 3). After a median of 2 EUS-RFA sessions (range, 1-2), 73% of patients had a complete response, and 27% had a partial response. During follow-up, 5 patients experienced self-limiting mild abdominal pain; no moderate or severe adverse events were reported. CONCLUSIONS EUS-RFA showed high technical feasibility, clinical success, and an acceptable safety profile in the treatment of left adrenal tumors. In patients at high surgical risk, EUS-RFA can be considered as an alternative therapeutic modality to surgery for the treatment of left adrenal tumors.
Collapse
Affiliation(s)
- Sung Hyun Cho
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dae Hyeon Kim
- University of Ulsan College of Medicine, Seoul, South Korea
| | - Dong-Wan Seo
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Seul Ki Yoo
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Dongwook Oh
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Tae Jun Song
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung Koo Lee
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Mikhail AS, Morhard R, Mauda-Havakuk M, Kassin M, Arrichiello A, Wood BJ. Hydrogel drug delivery systems for minimally invasive local immunotherapy of cancer. Adv Drug Deliv Rev 2023; 202:115083. [PMID: 37673217 PMCID: PMC11616795 DOI: 10.1016/j.addr.2023.115083] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/27/2023] [Accepted: 09/02/2023] [Indexed: 09/08/2023]
Abstract
Although systemic immunotherapy has achieved durable responses and improved survival for certain patients and cancer types, low response rates and immune system-related systemic toxicities limit its overall impact. Intratumoral (intralesional) delivery of immunotherapy is a promising technique to combat mechanisms of tumor immune suppression within the tumor microenvironment and reduce systemic drug exposure and associated side effects. However, intratumoral injections are prone to variable tumor drug distribution and leakage into surrounding tissues, which can compromise efficacy and contribute to toxicity. Controlled release drug delivery systems such as in situ-forming hydrogels are promising vehicles for addressing these challenges by providing improved spatio-temporal control of locally administered immunotherapies with the goal of promoting systemic tumor-specific immune responses and abscopal effects. In this review we will discuss concepts, applications, and challenges in local delivery of immunotherapy using controlled release drug delivery systems with a focus on intratumorally injected hydrogel-based drug carriers.
Collapse
Affiliation(s)
- Andrew S Mikhail
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Robert Morhard
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michal Mauda-Havakuk
- Interventional Oncology service, Interventional Radiology, Tel Aviv Sourasky Medical Center, Tel Aviv District, Israel
| | - Michael Kassin
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
22
|
Zhou J, Sun H, Wang Z, Cong W, Zeng M, Zhou W, Bie P, Liu L, Wen T, Kuang M, Han G, Yan Z, Wang M, Liu R, Lu L, Ren Z, Zeng Z, Liang P, Liang C, Chen M, Yan F, Wang W, Hou J, Ji Y, Yun J, Bai X, Cai D, Chen W, Chen Y, Cheng W, Cheng S, Dai C, Guo W, Guo Y, Hua B, Huang X, Jia W, Li Q, Li T, Li X, Li Y, Li Y, Liang J, Ling C, Liu T, Liu X, Lu S, Lv G, Mao Y, Meng Z, Peng T, Ren W, Shi H, Shi G, Shi M, Song T, Tao K, Wang J, Wang K, Wang L, Wang W, Wang X, Wang Z, Xiang B, Xing B, Xu J, Yang J, Yang J, Yang Y, Yang Y, Ye S, Yin Z, Zeng Y, Zhang B, Zhang B, Zhang L, Zhang S, Zhang T, Zhang Y, Zhao M, Zhao Y, Zheng H, Zhou L, Zhu J, Zhu K, Liu R, Shi Y, Xiao Y, Zhang L, Yang C, Wu Z, Dai Z, Chen M, Cai J, Wang W, Cai X, Li Q, Shen F, Qin S, Teng G, Dong J, Fan J. Guidelines for the Diagnosis and Treatment of Primary Liver Cancer (2022 Edition). Liver Cancer 2023; 12:405-444. [PMID: 37901768 PMCID: PMC10601883 DOI: 10.1159/000530495] [Citation(s) in RCA: 112] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/24/2023] [Indexed: 10/31/2023] Open
Abstract
Background Primary liver cancer, of which around 75-85% is hepatocellular carcinoma in China, is the fourth most common malignancy and the second leading cause of tumor-related death, thereby posing a significant threat to the life and health of the Chinese people. Summary Since the publication of Guidelines for Diagnosis and Treatment of Primary Liver Cancer in China in June 2017, which were updated by the National Health Commission in December 2019, additional high-quality evidence has emerged from researchers worldwide regarding the diagnosis, staging, and treatment of liver cancer, that requires the guidelines to be updated again. The new edition (2022 Edition) was written by more than 100 experts in the field of liver cancer in China, which not only reflects the real-world situation in China but also may reshape the nationwide diagnosis and treatment of liver cancer. Key Messages The new guideline aims to encourage the implementation of evidence-based practice and improve the national average 5-year survival rate for patients with liver cancer, as proposed in the "Health China 2030 Blueprint."
Collapse
Affiliation(s)
- Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huichuan Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenming Cong
- Department of Pathology, The Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Ping Bie
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lianxin Liu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianfu Wen
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ming Kuang
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guohong Han
- Department of Liver Diseases and Digestive Interventional Radiology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Zhiping Yan
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Maoqiang Wang
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, China
| | - Ruibao Liu
- Department of Interventional Radiology, The Tumor Hospital of Harbin Medical University, Harbin, China
| | - Ligong Lu
- Department of Interventional Oncology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhenggang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaochong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping Liang
- Department of Interventional Ultrasound, Chinese PLA General Hospital, Beijing, China
| | - Changhong Liang
- Department of Radiology, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Min Chen
- Editorial Department of Chinese Journal of Digestive Surgery, Chongqing, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinlin Hou
- Department of Infectious Diseases, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jingping Yun
- Department of Pathology, Tumor Prevention and Treatment Center, Sun Yat-sen University, Guangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dingfang Cai
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weixia Chen
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, China
| | - Yongjun Chen
- Department of Hematology, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenwu Cheng
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shuqun Cheng
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Chaoliu Dai
- Department of Hepatobiliary and Spleenary Surgery, The Affiliated Shengjing Hospital, China Medical University, Shenyang, China
| | - Wengzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yabing Guo
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Baojin Hua
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowu Huang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weidong Jia
- Department of Hepatic Surgery, Affiliated Provincial Hospital, Anhui Medical University, Hefei, China
| | - Qiu Li
- Department of Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Xun Li
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Yaming Li
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Yexiong Li
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Beijing, China
| | - Changquan Ling
- Changhai Hospital of Traditional Chinese Medicine, Second Military Medical University, Shanghai, China
| | - Tianshu Liu
- Department of Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiufeng Liu
- Department of Medical Oncology, PLA Cancer Center, Nanjing Bayi Hospital, Nanjing, China
| | - Shichun Lu
- Institute and Hospital of Hepatobiliary Surgery of Chinese PLA, Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, China
| | - Guoyue Lv
- Department of General Surgery, The First Hospital of Jilin University, Jilin, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiqiang Meng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weixin Ren
- Department of Interventional Radiology the First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guoming Shi
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Shi
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tianqiang Song
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jianhua Wang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kui Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Wentao Wang
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Xiaoying Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiming Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Bangde Xiang
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Baocai Xing
- Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jianming Xu
- Department of Gastrointestinal Oncology, Affiliated Hospital Cancer Center, Academy of Military Medical Sciences, Beijing, China
| | - Jiamei Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jianyong Yang
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yefa Yang
- Department of Hepatic Surgery and Interventional Radiology, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yunke Yang
- Department of Integrative Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shenglong Ye
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenyu Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Yong Zeng
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Bixiang Zhang
- Department of Surgery, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Boheng Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Leida Zhang
- Department of Hepatobiliary Surgery Institute, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, ZhengZhou, China
| | - Ti Zhang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, The Affiliated Tumor Hospital of Harbin Medical University, Harbin, China
| | - Ming Zhao
- Minimally Invasive Interventional Division, Liver Cancer Group, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yongfu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, ZhengZhou, China
| | - Honggang Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ledu Zhou
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People’s Hospital, Beijing, China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rong Liu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yinghong Shi
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongsheng Xiao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lan Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun Yang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhifeng Wu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhi Dai
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Minshan Chen
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jianqiang Cai
- Department of Abdominal Surgical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiujun Cai
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qiang Li
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Feng Shen
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Shukui Qin
- Department of Medical Oncology, PLA Cancer Center, Nanjing Bayi Hospital, Nanjing, China
| | - Gaojun Teng
- Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Jiahong Dong
- Department of Hepatobiliary and Pancreas Surgery, Beijing Tsinghua Changgung Hospital (BTCH), School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Canakis A, Sharaiha RZ. Radiofrequency ablation for pancreatobiliary disease: an updated review. Ann Gastroenterol 2023; 36:497-503. [PMID: 37664225 PMCID: PMC10433252 DOI: 10.20524/aog.2023.0828] [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: 04/11/2023] [Accepted: 07/05/2023] [Indexed: 09/05/2023] Open
Abstract
Endoscopic radiofrequency ablation (RFA) has emerged as a minimally invasive treatment option in cases of malignant biliary obstruction, pancreatic cancer, and other pancreatic cystic neoplasms. Intraductal biliary RFA is safe, effective, and confers a survival advantage over stenting alone, where it should be used an adjunct to biliary stenting. Endoscopic ultrasound-guided RFA can also provide pancreatic cyst resolution in patients who are not ideal operative candidates. The aim of this review is to describe the endoscopic applications and associated outcomes of RFA.
Collapse
Affiliation(s)
- Andrew Canakis
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland (Andrew Canakis)
| | - Reem Z. Sharaiha
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York (Reem Z. Sharaiha), USA
| |
Collapse
|
24
|
Spyridon P, Antonia S, Dionysia M, Ioanna S, Georgia B, Nikolaos P, Petros G, Ioannis E. Efficacy of Αtezolizumab-Βevacizumab in BCLC-C cirrhotic patients with hepatocellular carcinoma according to the type of disease progression, the type of BCLC-C and liver disease severity. J Cancer Res Clin Oncol 2023; 149:9253-9261. [PMID: 37199835 DOI: 10.1007/s00432-023-04846-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/07/2023] [Indexed: 05/19/2023]
Abstract
PURPOSE The aim of our study was to evaluate, under real-life conditions, survival of patients with advanced HCC (BCLC-C), either initially presenting in that stage or migrating from BCLC-A to BCLC-C within 2 years after curative LR/RFA, treated either with Atezolizumab-Bevacizumab or TKIs. METHODS Sixty-four cirrhotic patients with advanced HCC, who either initially presented as BCLC-C and were treated with Atezo-Bev (group A, N = 23) or TKIs (group B, N = 15) or who migrated from BCLC-A to BCLC-C stage within 2 years after LR/RFA and were either treated with Atezo-Bev (group C, N = 12) or TKIs (group D, N = 14), were retrospectively evaluated. RESULTS The four groups were comparable for all baseline parameters (demographics/platelets/liver disease etiology/diabetes/varices/Child-Pugh stage/ALBI grade) except for CPT score and MELD-Na. Using Cox-regression analysis, we observed that survival of group C after systemic treatment onset was significantly higher compared to group A (HR 3.71, 1.20-11.46, p = 0.02) and presented a trend to statistical significance when compared to group D (HR 3.14, 0.95-10.35, p = 0.06), adjusted for liver disease severity scores. When all BCLC-C patients classified as such due to PS only were excluded from the study, a trend for the same survival benefit in group C was shown, even in the most difficult-to-treat population with extrahepatic disease or macrovascular invasion. CONCLUSION Cirrhotic patients with advanced HCC initially diagnosed in BCLC-C, exhibit the worst survival irrespective of treatment schedule, whereas patients progressing to BCLC-C following disease recurrence after LR/RFA, seem to mostly benefit from Atezo-Bev, even patients with extrahepatic disease and/or macrovascular invasion. Liver disease severity seems to drive survival of these patients.
Collapse
Affiliation(s)
- Pantzios Spyridon
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece.
| | - Syriha Antonia
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| | - Mandilara Dionysia
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| | - Stathopoulou Ioanna
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| | - Barla Georgia
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| | - Ptohis Nikolaos
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| | - Galanis Petros
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| | - Elefsiniotis Ioannis
- Academic Department of Internal Medicine-Hepatogastroenterology Unit, General and Oncology Hospital of Kifisia "Agioi Anargyroi", National and Kapodistrian University of Athens, Timiou Stavrou and Noufaron 14, Kalyftaki, N. Kifisia, 14564, Athens, Greece
| |
Collapse
|
25
|
Domouchtsidou A, Beckmann F, Marenbach B, Mueller SP, Best J, Herrmann K, Horn PA, Barsegian V, Lindemann M. In Patients Treated by Selective Internal Radiotherapy, Cellular In Vitro Immune Function Is Predictive of Survival. Cancers (Basel) 2023; 15:4055. [PMID: 37627082 PMCID: PMC10452121 DOI: 10.3390/cancers15164055] [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: 07/03/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
In patients with liver malignancies, the cellular immune function was impaired in vitro after selective internal radiotherapy (SIRT). Because immunosuppression varied substantially, in the current study, we investigated in 25 SIRT patients followed up for ten years whether the lymphocyte function was correlated with survival. Peripheral blood mononuclear cells were stimulated with four microbial antigens (tuberculin, tetanus toxoid, Candida albicans and CMV) before therapy and at four time points thereafter, and lymphocyte proliferation was determined by H3-thymidine uptake. The median sum of the responses to these four antigens decreased from 39,464 counts per minute (CPM) increment (range 1080-204,512) before therapy to a minimum of 700 CPM increment on day 7 after therapy (0-93,187, p < 0.0001). At all five time points, the median survival in patients with weaker responses was 2- to 3.5-fold shorter (p < 0.05). On day 7, the median survival in patients with responses below and above the cutoff of a 2 CPM increment was 185 and 523 days, respectively (χ2 = 9.4, p = 0.002). In conclusion, lymphocyte function could be a new predictor of treatment outcome after SIRT.
Collapse
Affiliation(s)
- Aglaia Domouchtsidou
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; (A.D.); (F.B.); (B.M.); (P.A.H.)
- Department of Microbiology, General Anticancer Oncological Hospital “Agios Savvas”, 115 22 Athens, Greece
| | - Ferdinand Beckmann
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; (A.D.); (F.B.); (B.M.); (P.A.H.)
| | - Beate Marenbach
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; (A.D.); (F.B.); (B.M.); (P.A.H.)
| | - Stefan P. Mueller
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (S.P.M.); (K.H.); (V.B.)
| | - Jan Best
- Department of Gastroenterology and Hepatology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
- Department of Internal Medicine, University Hospital Knappschaftskrankenhaus Bochum, Ruhr University Bochum, 44892 Bochum, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (S.P.M.); (K.H.); (V.B.)
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; (A.D.); (F.B.); (B.M.); (P.A.H.)
| | - Vahé Barsegian
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany; (S.P.M.); (K.H.); (V.B.)
- Institute of Nuclear Medicine, Helios Kliniken, 19049 Schwerin, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; (A.D.); (F.B.); (B.M.); (P.A.H.)
| |
Collapse
|
26
|
Huang H, Liu R, Yang J, Dai J, Fan S, Pi J, Wei Y, Guo X. Gold Nanoparticles: Construction for Drug Delivery and Application in Cancer Immunotherapy. Pharmaceutics 2023; 15:1868. [PMID: 37514054 PMCID: PMC10383270 DOI: 10.3390/pharmaceutics15071868] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer immunotherapy is an innovative treatment strategy to enhance the ability of the immune system to recognize and eliminate cancer cells. However, dose limitations, low response rates, and adverse immune events pose significant challenges. To address these limitations, gold nanoparticles (AuNPs) have been explored as immunotherapeutic drug carriers owing to their stability, surface versatility, and excellent optical properties. This review provides an overview of the advanced synthesis routes for AuNPs and their utilization as drug carriers to improve precision therapies. The review also emphasises various aspects of AuNP-based immunotherapy, including drug loading, targeting strategies, and drug release mechanisms. The application of AuNPs combined with cancer immunotherapy and their therapeutic efficacy are briefly discussed. Overall, we aimed to provide a recent understanding of the advances, challenges, and prospects of AuNPs for anticancer applications.
Collapse
Affiliation(s)
- Huiqun Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Ronghui Liu
- School of Microelectronic, Southern University of Science and Technology, Shenzhen 518000, China
| | - Jie Yang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Jing Dai
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Shuhao Fan
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Jiang Pi
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China
| | - Yubo Wei
- Yunnan Key Laboratory of Pharmacology for Natural Products, School of Pharmaceutical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xinrong Guo
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
27
|
Wang Y, Wang GZ, Chen C, Huang HZ, Wang YH, He XH, Xu LX, Xu LC, Li WT. Exploration of the impact of multimode thermal therapy versus radiofrequency ablation on CD8 + T effector cells of liver malignancies based on single cell transcriptomics. Front Immunol 2023; 14:1172362. [PMID: 37334386 PMCID: PMC10272448 DOI: 10.3389/fimmu.2023.1172362] [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: 02/23/2023] [Accepted: 05/03/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Multimode thermal therapy (MTT) is an innovative interventional therapy developed for the treatment of liver malignancies. When compared to the conventional radiofrequency ablation (RFA), MTT typically offers improved prognosis for patients. However, the effect of MTT on the peripheral immune environment and the mechanisms underlying the enhanced prognosis have yet to be explored. The aim of this study was to further investigate the mechanisms responsible for the difference in prognosis between the two therapies. Methods In this study, peripheral blood samples were collected from four patients treated with MTT and two patients treated with RFA for liver malignancies at different time points before and after the treatment. Single cell sequencing was performed on the blood samples to compare and analyze the activation pathways of peripheral immune cells following the MTT and RFA treatment. Results There was no significant effect of either therapy on the composition of immune cells in peripheral blood. However, the differential gene expression and pathway enrichment analysis demonstrated enhanced activation of T cells in the MTT group compared to the RFA group. In particular, there was a remarkable increase in TNF-α signaling via NF-κB, as well as the expression of IFN-α and IFN-γ in the CD8+ effector T (CD8+ Teff) cells subpopulation, when compared to the RFA group. This may be related to the upregulation of PI3KR1 expression after MTT, which promotes the activation of PI3K-AKT-mTOR pathway. Conclusion This study confirmed that MTT could more effectively activate peripheral CD8+ Teff cells in patients compared with RFA and promote the effector function, thus resulting in a better prognosis. These results provide a theoretical basis for the clinical application of MTT therapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guang-Zhi Wang
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Department of Medical Imaging Center, Affiliated Hospital, Weifang Medical University, Weifang, Shandong, China
| | - Chao Chen
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao-Zhe Huang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yao-Hui Wang
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xin-Hong He
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lisa X. Xu
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Chao Xu
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wen-Tao Li
- Department of Interventional Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
28
|
Jiang M, Fiering S, Shao Q. Combining energy-based focal ablation and immune checkpoint inhibitors: preclinical research and clinical trials. Front Oncol 2023; 13:1153066. [PMID: 37251920 PMCID: PMC10211342 DOI: 10.3389/fonc.2023.1153066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Energy-based focal therapy (FT) uses targeted, minimally invasive procedures to destroy tumors while preserving normal tissue and function. There is strong emerging interest in understanding how systemic immunity against the tumor can occur with cancer immunotherapy, most notably immune checkpoint inhibitors (ICI). The motivation for combining FT and ICI in cancer management relies on the synergy between the two different therapies: FT complements ICI by reducing tumor burden, increasing objective response rate, and reducing side effects of ICI; ICI supplements FT by reducing local recurrence, controlling distal metastases, and providing long-term protection. This combinatorial strategy has shown promising results in preclinical study (since 2004) and the clinical trials (since 2011). Understanding the synergy calls for understanding the physics and biology behind the two different therapies with distinctive mechanisms of action. In this review, we introduce different types of energy-based FT by covering the biophysics of tissue-energy interaction and present the immunomodulatory properties of FT. We discuss the basis of cancer immunotherapy with the emphasis on ICI. We examine the approaches researchers have been using and the results from both preclinical models and clinical trials from our exhaustive literature research. Finally, the challenges of the combinatory strategy and opportunities of future research is discussed extensively.
Collapse
Affiliation(s)
- Minhan Jiang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth Health, Lebanon, NH, United States
| | - Qi Shao
- Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
29
|
Posa A, Contegiacomo A, Ponziani FR, Punzi E, Mazza G, Scrofani A, Pompili M, Goldberg SN, Natale L, Gasbarrini A, Sala E, Iezzi R. Interventional Oncology and Immuno-Oncology: Current Challenges and Future Trends. Int J Mol Sci 2023; 24:ijms24087344. [PMID: 37108507 PMCID: PMC10138371 DOI: 10.3390/ijms24087344] [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: 03/27/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Personalized cancer treatments help to deliver tailored and biologically driven therapies for cancer patients. Interventional oncology techniques are able to treat malignancies in a locoregional fashion, with a variety of mechanisms of action leading to tumor necrosis. Tumor destruction determines a great availability of tumor antigens that can be recognized by the immune system, potentially triggering an immune response. The advent of immunotherapy in cancer care, with the introduction of specific immune checkpoint inhibitors, has led to the investigation of the synergy of these drugs when used in combination with interventional oncology treatments. The aim of this paper is to review the most recent advances in the field of interventional oncology locoregional treatments and their interactions with immunotherapy.
Collapse
Affiliation(s)
- Alessandro Posa
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Andrea Contegiacomo
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Ernesto Punzi
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Giulia Mazza
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Annarita Scrofani
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Maurizio Pompili
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Shraga Nahum Goldberg
- Division of Image-Guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem 12000, Israel
| | - Luigi Natale
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Evis Sala
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Roberto Iezzi
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
30
|
Yao C, Dong J, Ren K, Sun L, Wang H, Zhang J, Wang H, Xu X, Yao B, Zhou H, Zhao L, Peng R. Accumulative Effects of Multifrequency Microwave Exposure with 1.5 GHz and 2.8 GHz on the Structures and Functions of the Immune System. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4988. [PMID: 36981897 PMCID: PMC10049199 DOI: 10.3390/ijerph20064988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 06/18/2023]
Abstract
Microwave ablation can produce immune activation due to thermal effects. However, the nonthermal effects of microwaves on the immune system are still largely unexplored. In this study, we sequentially exposed rats to 1.5 GHz microwave for 6 min and 2.8 GHz microwave for 6 min at an average power density of 5, 10, and 30 mW/cm2. The structure of the thymus, spleen, and mesenteric lymph node were observed, and we showed that multifrequency microwave exposure caused tissue injuries, such as congestion and nuclear fragmentation in lymphocytes. Ultrastructural injuries, including mitochondrial swelling, mitochondrial cristae rupture, and mitochondrial cavitation, were observed, especially in the 30 mW/cm2 microwave-exposed group. Generally, multifrequency microwaves decreased white blood cells, as well as lymphocytes, monocytes, and neutrophils, in peripheral blood, from 7 d to 28 d after exposure. Microwaves with an average density of 30 mW/cm2 produced much more significant inhibitory effects on immune cells. Moreover, multifrequency microwaves at 10 and 30 mW/cm2, but not 5 mW/cm2, reduced the serum levels of several cytokines, such as interleukin-1 alpha (IL-1α), IL-1β, interferon γ (IFN-γ) and tumor necrosis factor α (TNF-α), at 7 d and 14 d after exposure. We also found similar alterations in immunoglobulins (Igs), IgG, and IgM in serum. However, no obvious changes in complement proteins were detected. In conclusion, multifrequency microwave exposure of 1.5 GHz and 2.8 GHz caused both structural injuries of immune tissues and functional impairment in immune cells. Therefore, it will be necessary to develop an effective strategy to protect people from multifrequency microwave-induced immune suppression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Li Zhao
- Correspondence: (L.Z.); (R.P.)
| | | |
Collapse
|
31
|
Kim NJ, Yoon JH, Tuomi AC, Lee J, Kim D. In-situ tumor vaccination by percutaneous ablative therapy and its synergy with immunotherapeutics: An update on combination therapy. Front Immunol 2023; 14:1118845. [PMID: 36969248 PMCID: PMC10030508 DOI: 10.3389/fimmu.2023.1118845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
Percutaneous tumor ablation is now a widely accepted minimally invasive local treatment option offered by interventional radiology and applied to various organs and tumor histology types. It utilizes extreme temperatures to achieve irreversible cellular injury, where ablated tumor interacts with surrounding tissue and host via tissue remodeling and inflammation, clinically manifesting as post-ablation syndrome. During this process, in-situ tumor vaccination occurs, in which tumor neoantigens are released from ablated tissue and can prime one’s immune system which would favorably affect both local and remote site disease control. Although successful in priming the immune system, this rarely turns into clinical benefits for local and systemic tumor control due to intrinsic negative immune modulation of the tumor microenvironment. A combination of ablation and immunotherapy has been employed to overcome these and has shown promising preliminary results of synergistic effect without significantly increased risk profiles. The aim of this article is to review the evidence on post-ablation immune response and its synergy with systemic immunotherapies.
Collapse
Affiliation(s)
- Nicole J. Kim
- Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Diagnostic Imaging, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Jessica H. Yoon
- Department of Diagnostic Imaging, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Adam C. Tuomi
- Department of Diagnostic Imaging, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - John Lee
- Department of Diagnostic Imaging, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Daehee Kim
- Department of Diagnostic Imaging, Warren Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Daehee Kim,
| |
Collapse
|
32
|
Santana JG, Petukhova-Greenstein A, Gross M, Hyder F, Pekurovsky V, Gottwald LA, Boustani A, Walsh JJ, Kucukkaya AS, Malpani R, Madoff DC, Goldberg SN, Ahmed M, Joshi N, Coman D, Chapiro J. MR Imaging-Based In Vivo Macrophage Imaging to Monitor Immune Response after Radiofrequency Ablation of the Liver. J Vasc Interv Radiol 2023; 34:395-403.e5. [PMID: 36423815 PMCID: PMC11042914 DOI: 10.1016/j.jvir.2022.11.013] [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: 04/11/2022] [Revised: 10/27/2022] [Accepted: 11/13/2022] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To establish molecular magnetic resonance (MR) imaging instruments for in vivo characterization of the immune response to hepatic radiofrequency (RF) ablation using cell-specific immunoprobes. MATERIALS AND METHODS Seventy-two C57BL/6 wild-type mice underwent standardized hepatic RF ablation (70 °C for 5 minutes) to generate a coagulation area measuring 6-7 mm in diameter. CD68+ macrophage periablational infiltration was characterized with immunohistochemistry 24 hours, 72 hours, 7 days, and 14 days after ablation (n = 24). Twenty-one mice were subjected to a dose-escalation study with either 10, 15, 30, or 60 mg/kg of rhodamine-labeled superparamagnetic iron oxide nanoparticles (SPIONs) or 2.4, 1.2, or 0.6 mg/kg of gadolinium-160 (160Gd)-labeled CD68 antibody for assessment of the optimal in vivo dose of contrast agent. MR imaging experiments included 9 mice, each receiving 10-mg/kg SPIONs to visualize phagocytes using T2∗-weighted imaging in a horizontal-bore 9.4-T MR imaging scanner, 160Gd-CD68 for T1-weighted MR imaging of macrophages, or 0.1-mmol/kg intravenous gadoterate (control group). Radiological-pathological correlation included Prussian blue staining, rhodamine immunofluorescence, imaging mass cytometry, and immunohistochemistry. RESULTS RF ablation-induced periablational infiltration (206.92 μm ± 12.2) of CD68+ macrophages peaked at 7 days after ablation (P < .01) compared with the untreated lobe. T2∗-weighted MR imaging with SPION contrast demonstrated curvilinear T2∗ signal in the transitional zone (TZ) (186 μm ± 16.9), corresponsing to Iron Prussian blue staining. T1-weighted MR imaging with 160Gd-CD68 antibody showed curvilinear signal in the TZ (164 μm ± 3.6) corresponding to imaging mass cytometry. CONCLUSIONS Both SPION-enhanced T2∗-weighted and 160Gd-enhanced T1-weighted MR imaging allow for in vivo monitoring of macrophages after RF ablation, demonstrating the feasibility of this model to investigate local immune responses.
Collapse
Affiliation(s)
- Jessica G Santana
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Alexandra Petukhova-Greenstein
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Moritz Gross
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Vasily Pekurovsky
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Luzie A Gottwald
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Annemarie Boustani
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - John J Walsh
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Ahmet S Kucukkaya
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, Berlin, Germany
| | - Rohil Malpani
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - David C Madoff
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - S Nahum Goldberg
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard University, Boston, Massachusetts; Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Muneeb Ahmed
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard University, Boston, Massachusetts
| | - Nikhil Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
33
|
Kimura Y, Fujimori M, Rajagopalan NR, Poudel K, Kim K, Nagar K, Vroomen LGPH, Reis H, Al-Ahmadie H, Coleman JA, Srimathveeravalli G. Macrophage activity at the site of tumor ablation can promote murine urothelial cancer via transforming growth factor-β1. Front Immunol 2023; 14:1070196. [PMID: 36761730 PMCID: PMC9902765 DOI: 10.3389/fimmu.2023.1070196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/02/2023] [Indexed: 01/26/2023] Open
Abstract
Cell death and injury at the site of tumor ablation attracts macrophages. We sought to understand the status and activity of these cells while focusing on transforming growth factor-β1 (TGF-β1), a potent immunosuppressive and tumorigenic cytokine. Patients with urothelial cancer who underwent ablation using electrocautery or laser demonstrated increased infiltration and numbers of CD8+ T cells, along with FoxP3+ regulatory T cells, CD68+ macrophages and elevated levels of TGF-β1 in recurrent tumors. Similar findings were reproduced in a mouse model of urothelial cancer (MB49) by partial tumor ablation with irreversible electroporation (IRE). Stimulation of bone marrow derived macrophages with MB49 cell debris produced using IRE elicited strong M2 polarization, with exuberant secretion of TGF-β1. The motility, phenotypic markers and cytokine secretion by macrophages could be muted by treatment with Pirfenidone (PFD), a clinically approved drug targeting TGF-β1 signaling. MB49 cancer cells exposed to TGF-β1 exhibited increased migration, invasiveness and upregulation of epithelial-mesenchymal transition markers α-Smooth Muscle Actin and Vimentin. Such changes in MB49 cells were reduced by treatment with PFD even during stimulation with TGF-β1. IRE alone yielded better local tumor control when compared with control or PFD alone, while also reducing the overall number of lung metastases. Adjuvant PFD treatment did not provide additional benefit under in vivo conditions.
Collapse
Affiliation(s)
- Yasushi Kimura
- Department of Diagnosis and Interventional Radiology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Dept. of Mechanical and Industrial Engineering, University of Massachusetts Amherst,
Amherst, MA, United States
| | | | | | - Krish Poudel
- Dept. of Mechanical and Industrial Engineering, University of Massachusetts Amherst,
Amherst, MA, United States
| | - Kwanghee Kim
- Division of Urology, Department of Surgery, Memorial Sloan Kettering Cancer Center,
New York, NY, United States
| | - Karan Nagar
- Division of Urology, Department of Surgery, Memorial Sloan Kettering Cancer Center,
New York, NY, United States
| | - Laurien GPH. Vroomen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
| | - Henning Reis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jonathan A. Coleman
- Division of Urology, Department of Surgery, Memorial Sloan Kettering Cancer Center,
New York, NY, United States
| | - Govindarajan Srimathveeravalli
- Dept. of Mechanical and Industrial Engineering, University of Massachusetts Amherst,
Amherst, MA, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
34
|
Yu L, Cheng M, Liu J, Ye X, Wei Z, Xu J, Xie Q, Liang J. Crosstalk between microwave ablation and ferroptosis: The next hot topic? Front Oncol 2023; 13:1099731. [PMID: 36712497 PMCID: PMC9880492 DOI: 10.3389/fonc.2023.1099731] [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: 11/16/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Microwave ablation has been one form of thermal ablation in treatments for many tumors, which can locally control unresectable tumors. Ferroptosis is iron-dependent cell death caused by the cumulative reactive oxygen species and lipid peroxidation products. Recently, increasing evidence has shown that ferroptosis might play a vital role in MWA-induced tumor suppression. In this article, we briefly illustrate the concept of ferroptosis, the related signal pathways and inducers, the basic principle of microwave ablation in killing tumors, and the key molecules released after microwave ablation. Then, we describe the cross-talking molecules between microwave ablation and ferroptosis, and discussed the potential mechanism of microwave ablation-induced ferroptosis. This review explores the therapeutic target of ferroptosis in enhancing the systemic antitumor effect after microwave ablation, providing theoretical support in combinational microwave ablation with pro-ferroptosis therapy.
Collapse
Affiliation(s)
- Lu Yu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Min Cheng
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jie Liu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China,School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xin Ye
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Zhigang Wei
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiamei Xu
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi Xie
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China,*Correspondence: Qi Xie, ; Jing Liang,
| | - Jing Liang
- Department of Oncology, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China,*Correspondence: Qi Xie, ; Jing Liang,
| |
Collapse
|
35
|
Wan L, Cao Y, Cheng C, Tang R, Wu N, Zhou Y, Xiong X, He H, Lin X, Jiang Q, Wang X, Guo X, Wang D, Ran H, Ren J, Zhou Y, Hu Z, Li P. Biomimetic, pH-Responsive Nanoplatforms for Cancer Multimodal Imaging and Photothermal Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:1784-1797. [PMID: 36580421 DOI: 10.1021/acsami.2c16667] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Photothermal therapy (PTT), by converting light to thermal energy, has become a novel and noninvasive technique for tumor thermal ablation in clinical practice. However, as a result of phagocytosis of reticuloendothelial cells, current photothermal agents (PTAs) derived from exogenous materials suffer from incompetent tumor targeting and brief internal circulation time. The resulting poor accumulation of PTAs in the target area severely reduces the efficacy of PTT. In addition, the potential toxicity of PTAs, excessive laser exposure, and possibilities of tumor recurrence and metastasis following PTT are still intractable problems that severely influence patients' quality of life. Herein, a biomimetic pH-responsive nanoprobe was prepared via cancer cell membrane coating polydopamine (PDA)-CaCO3 nanoparticles (CPCaNPs) for photoacoustic (PA)/ultrasonic (US)/thermal imaging-guided PTT. When CPCaNPs targeted and infiltrated into the tumor's acidic microenvironment, the decomposed CO2 bubbles from homologous targeting CPCaNPs enhanced ultrasonic (US) signals obviously. At the same time, the PDA of CPCaNPs not only performed efficient PTT of primary tumors but also generated photoacoustic (PA) signals. In addition, an immune checkpoint pathway blockade was combined, which inhibited tumor recurrence and metastasis significantly and improved the immunosuppressive microenvironment after PTT to a large extent. Thus, these proposed biomimetic pH-responsive CPCaNPs provide a promising strategy for precise PTT immunotherapy under the intelligent guidance of PA/US/thermal imaging and show great potential for clinical translation.
Collapse
Affiliation(s)
- Li Wan
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Health Management (Physical Examination) Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Yuting Cao
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Chen Cheng
- Department of Ultrasound, Bishan Hospital of Chongqing, Bishan Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Rui Tang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Nianhong Wu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Ying Zhou
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xialin Xiong
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Hongye He
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xiaohong Lin
- Department of Ultrasound, Chongqing General Hospital, Chongqing 401147, P. R. China
| | - Qinqin Jiang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xiaoting Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xun Guo
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Jianli Ren
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Yang Zhou
- Department of Ultrasound, The Third People's Hospital of Chengdu City, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Zhongqian Hu
- Department of Ultrasound, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P. R. China
| | - Pan Li
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
- Institute of Ultrasound Imaging of Chongqing Medical University, Chongqing 400010, P. R. China
| |
Collapse
|
36
|
Guo RQ, Peng JZ, Li YM, Li XG. Microwave ablation combined with anti-PD-1/CTLA-4 therapy induces an antitumor immune response to renal cell carcinoma in a murine model. Cell Cycle 2023; 22:242-254. [PMID: 35980140 PMCID: PMC9815248 DOI: 10.1080/15384101.2022.2112007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 01/11/2023] Open
Abstract
The study was designed to evaluate the efficiency of microwave ablation (MWA) in combination with anti-programmed death receptor 1 (anti-PD-1)/cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) in renal cell carcinoma (RCC) treatment. After tumors were established on C57/BL6 mice, MWA treatment and/or immune checkpoint inhibitor (ICI) treatment to the mice were performed. Tumor volume was recorded every 7 days. A rechallenge test was conducted on mice with tumors in the left kidney to explore the systemic establishment of antitumor immunity on day 7. In this study, during the 21-day observation period, tumors were continued to grow in all groups. However, compared with the tumor growth rate in MWA or control group, the rate in the ICI or MWA+ICI groups was decreased. Moreover, the population of CD8+T-cells was increased only in the MWA+ICI group, while that of regulatory T cells was decreased in the MWA, ICI, and MWA+ICI groups. Additionally, the MWA+ICI group had the highest interferon-γ level among all groups. Furthermore, histopathological examination revealed that CTLA-4 expression in distant tumors was reduced in the ICI and MWA + ICI groups. MWA treatment increased PD-L1/PD-1 expression; however, after the combination treatment with ICI, PD-L1/PD-1 expression was decreased. According to the rechallenge test, mice (16.7%) in the MWA group, ICI group (50%), and MWA+ICI group (66.7%) exhibited successful tumor rejection, whereas no mice in the control group exhibited the capability of tumor rejection. Overall, the systemic antitumor immunity induced by MWA was boosted when combined with anti-PD-1/CTLA-4 treatment in an RCC murine model.
Collapse
Affiliation(s)
- Run-Qi Guo
- Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R.China
| | - Jin-Zhao Peng
- Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R.China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, P.R.China
| | - Yuan-Ming Li
- Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R.China
| | - Xiao-Guang Li
- Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, P.R.China
| |
Collapse
|
37
|
Marc B, Monino L, Rimbas M. EUS-guided intra-tumoral therapies. Best Pract Res Clin Gastroenterol 2022; 60-61:101817. [PMID: 36577536 DOI: 10.1016/j.bpg.2022.101817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
EUS-guided treatments for focal tumor lesions has been developed since 20 years using at onset of the technique mainly local and guided alcohol injection [1-4]. Pancreatic tumors are the most assessed targeted lesions for EUS treatment because of their accessibility and because EUS management could be a safe alternative to surgery. More and more pancreatic tumors are discovered mainly fortuitously due to the advances in conventional imaging (abdominal ultrasound, CT, MRI) resulting in the question of surgical management of an asymptomatic pancreatic lesion ("incidentaloma") [5-8]. The lesions detected include mostly pancreatic cystic neoplasms (PCN) and neuroendocrine tumors (NET) mainly well differentiated. Clinically, NET are mostly non-functional and do not induce secretory disorders [5-8]. Once their nature is yielded by diagnostic tests like EUS-FNA, incidental nonfunctional NET currently lead to difficult management when their largest diameter is less than 2 cm [2,4,9,10]. EUS-guided treatment for pancreatic adenocarcinoma have also been developed with recent prospective observational study and randomized control study [11,12]. Thus, therapeutic surgical choices could be challenged by EUS- guided treatment [2,4,9].
Collapse
Affiliation(s)
- Barthet Marc
- Aix Marseille Université, Service de Gastro-entérologie, Hôpital Nord, Chemin des Bourrely, 13915, Marseille, cedex 20, France.
| | | | - Mihai Rimbas
- Gastroenterology Department, Colentina Clinical Hospital, Bucharest, Romania
| |
Collapse
|
38
|
Confino H, Dirbas FM, Goldshtein M, Yarkoni S, Kalaora R, Hatan M, Puyesky S, Levi Y, Malka L, Johnson M, Chaisson S, Monson JM, Avniel A, Lisi S, Greenberg D, Wolf I. Gaseous nitric oxide tumor ablation induces an anti-tumor abscopal effect. Cancer Cell Int 2022; 22:405. [PMID: 36514083 PMCID: PMC9745717 DOI: 10.1186/s12935-022-02828-z] [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: 07/04/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In-situ tumor ablation provides the immune system with the appropriate antigens to induce anti-tumor immunity. Here, we present an innovative technique for generating anti-tumor immunity by delivering exogenous ultra-high concentration (> 10,000 ppm) gaseous nitric oxide (UHCgNO) intratumorally. METHODS The capability of UHCgNO to induce apoptosis was tested in vitro in mouse colon (CT26), breast (4T1) and Lewis lung carcinoma (LLC-1) cancer cell lines. In vivo, UHCgNO was studied by treating CT26 tumor-bearing mice in-situ and assessing the immune response using a Challenge assay. RESULTS Exposing CT26, 4T1 and LLC-1 cell lines to UHCgNO for 10 s-2.5 min induced cellular apoptosis 24 h after exposure. Treating CT26 tumors in-situ with UHCgNO followed by surgical resection 14 days later resulted in a significant secondary anti-tumor effect in vivo. 100% of tumor-bearing mice treated with 50,000 ppm UHCgNO and 64% of mice treated with 20,000 ppm UHCgNO rejected a second tumor inoculation, compared to 0% in the naive control for 70 days. Additionally, more dendrocytes infiltrated the tumor 14 days post UHCgNO treatment versus the nitrogen control. Moreover, T-cell penetration into the primary tumor was observed in a dose-dependent manner. Systemic increases in T- and B-cells were seen in UHCgNO-treated mice compared to nitrogen control. Furthermore, polymorphonuclear-myeloid-derived suppressor cells were downregulated in the spleen in the UHCgNO-treated groups. CONCLUSIONS Taken together, our data demonstrate that UHCgNO followed by the surgical removal of the primary tumor 14 days later induces a strong and potent anti-tumor response.
Collapse
Affiliation(s)
| | - Frederick M. Dirbas
- grid.168010.e0000000419368956Department of General Surgery, Stanford University, Stanford, CA USA
| | | | | | | | | | | | - Yakir Levi
- Beyond Cancer Ltd., 7608801 Rehovot, Israel
| | | | | | | | - Jedidiah M. Monson
- Beyond Cancer Ltd., Atlanta, GA USA ,grid.476982.6California Cancer Associates for Research and Excellence, Fresno, CA USA
| | - Amir Avniel
- Beyond Air Ltd., 7608801 Rehovot, Israel ,Beyond Air Inc, Garden City, NY 11530 USA
| | - Steve Lisi
- Beyond Air Inc, Garden City, NY 11530 USA
| | - David Greenberg
- Beyond Air Ltd., 7608801 Rehovot, Israel ,Beyond Air Inc, Garden City, NY 11530 USA
| | - Ido Wolf
- grid.413449.f0000 0001 0518 6922Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel ,grid.12136.370000 0004 1937 0546Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
Cheng X, Wei Y, Jiang X, Wang C, Liu M, Yan J, Zhang L, Zhou Y. Insight into the Prospects for Tumor Therapy Based on Photodynamic Immunotherapy. Pharmaceuticals (Basel) 2022; 15:1359. [PMID: 36355531 PMCID: PMC9693017 DOI: 10.3390/ph15111359] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 10/29/2024] Open
Abstract
Malignancy is one of the common diseases with high mortality worldwide and the most important obstacle to improving the overall life expectancy of the population in the 21st century. Currently, single or combined treatments, including surgery, chemotherapy, and radiotherapy, are still the mainstream regimens for tumor treatment, but they all present significant side effects on normal tissues and organs, such as organ hypofunction, energy metabolism disorders, and various concurrent diseases. Based on this, theranostic measures for the highly selective killing of tumor cells have always been a hot area in cancer-related fields, among which photodynamic therapy (PDT) is expected to be an ideal candidate for practical clinical application due to its precise targeting and excellent safety performance, so-called PDT refers to a therapeutic method mainly composed of photosensitizers (PSs), laser light, and reactive oxygen species (ROS). Photoimmunotherapy (PIT), a combination of PDT and immunotherapy, can induce systemic antitumor immune responses and inhibit continuing growth and distant metastasis of residual tumor cells, demonstrating a promising application prospect. This article reviews the types of immune responses that occur in the host after PDT treatment, including innate and adaptive immunity. To further help PIT-related drugs improve their pharmacokinetic properties and bioavailability, we highlight the potential improvement of photodynamic immunotherapy from three aspects: immunostimulatory agents, tumor-associated antigens (TAAs) as well as different immune cells. Finally, we focus on recent advances in various strategies and shed light on their corresponding mechanisms of immune activation and possible clinical applications such as cancer vaccines. Having discovered the inherent potential of PDT and the mechanisms that PDT triggers host immune responses, a variety of immunotherapeutic strategies have been investigated in parallel with approaches to improve PDT efficiency. However, it remains to be further elucidated under what conditions the immune effect induced by PDT can achieve tumor immunosuppression and to what extent PDT-induced antitumor immunity will lead to complete tumor rejection. Currently, PIT presents several outstanding intractable challenges, such as the aggregation ability of PSs locally in tumors, deep tissue penetration ability of laser light, immune escape, and biological toxicity, and it is hoped that these issues raised will help to point out the direction of preclinical research on PIT and accelerate its transition to clinical practice.
Collapse
Affiliation(s)
- Xiaoxia Cheng
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yiqu Wei
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Xiaomei Jiang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Chunli Wang
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Mengyu Liu
- School of Clinical Medicine, Henan University, Kaifeng 475004, China
| | - Jiaxin Yan
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Lei Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yaqi Zhou
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
- Pathology Department, Jiaozuo Second People’s Hospital, Jiaozuo 454001, China
| |
Collapse
|
40
|
Shou J, Mo F, Zhang S, Lu L, Han N, Liu L, Qiu M, Li H, Han W, Ma D, Guo X, Guo Q, Huang Q, Zhang X, Ye S, Pan H, Chen S, Fang Y. Combination treatment of radiofrequency ablation and peptide neoantigen vaccination: Promising modality for future cancer immunotherapy. Front Immunol 2022; 13:1000681. [PMID: 36248865 PMCID: PMC9559398 DOI: 10.3389/fimmu.2022.1000681] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022] Open
Abstract
Background The safety and immunogenicity of a personalized neoantigen-based peptide vaccine, iNeo-Vac-P01, was reported previously in patients with a variety of cancer types. The current study investigated the synergistic effects of radiofrequency ablation (RFA) and neoantigen vaccination in cancer patients and tumor-bearing mice. Methods Twenty-eight cancer patients were enrolled in this study, including 10 patients who had received RFA treatment within 6 months before vaccination (Cohort 1), and 18 patients who had not (Cohort 2). Individualized neoantigen peptide vaccines were designed, manufactured, and subcutaneously administrated with GM-CSF as an adjuvant for all patients. Mouse models were employed to validate the synergistic efficacy of combination treatment of RFA and neoantigen vaccination. Results Longer median progression free survival (mPFS) and median overall survival (mOS) were observed in patients in Cohort 1 compared to patients in Cohort 2 (4.42 and 20.18 months vs. 2.82 and 10.94 months). The results of ex vivo IFN-γ ELISpot assay showed that patients in Cohort 1 had stronger neoantigen-specific immune responses at baseline and post vaccination. Mice receiving combination treatment of RFA and neoantigen vaccines displayed higher antitumor immune responses than mice receiving single modality. The combination of PD-1 blockage with RFA and neoantigen vaccines further enhanced the antitumor response in mice. Conclusion Neoantigen vaccination after local RFA treatment could improve the clinical and immune response among patients of different cancer types. The synergistic antitumor potentials of these two modalities were also validated in mice, and might be further enhanced by immune checkpoint inhibition. The mechanisms of their synergies require further investigation. Clinical trial registration https://clinicaltrials.gov/, identifier NCT03662815.
Collapse
Affiliation(s)
- Jiawei Shou
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fan Mo
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Hangzhou AI-Force Therapeutics Co., Ltd., Hangzhou, China
| | - Shanshan Zhang
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
- Zhejiang California International Nanosystems Institute, Zhejiang University, Hangzhou, China
| | - Lantian Lu
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Ning Han
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
- Hangzhou AI-Nano Therapeutics Co., Ltd., Hangzhou, China
| | - Liang Liu
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Min Qiu
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Hongseng Li
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weidong Han
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongying Ma
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Xiaojie Guo
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Qianpeng Guo
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Qinxue Huang
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Xiaomeng Zhang
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
| | - Shengli Ye
- Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Hongming Pan
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Hongming Pan, ; Shuqing Chen, ; Yong Fang,
| | - Shuqing Chen
- Hangzhou Neoantigen Therapeutics Co., Ltd., Hangzhou, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Zhejiang California International Nanosystems Institute, Zhejiang University, Hangzhou, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
- *Correspondence: Hongming Pan, ; Shuqing Chen, ; Yong Fang,
| | - Yong Fang
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Hongming Pan, ; Shuqing Chen, ; Yong Fang,
| |
Collapse
|
41
|
Valery M, Cervantes B, Samaha R, Gelli M, Smolenschi C, Fuerea A, Tselikas L, Klotz-Prieux C, Hollebecque A, Boige V, Ducreux M. Immunotherapy and Hepatocellular Cancer: Where Are We Now? Cancers (Basel) 2022; 14:cancers14184523. [PMID: 36139683 PMCID: PMC9497386 DOI: 10.3390/cancers14184523] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy has demonstrated its effectiveness in many cancers. In hepatocellular carcinoma (HCC), promising results shown in the first phase II studies evaluating anti-PD-1 or anti-PD-L1 monotherapies resulted in their approval in the United States. Approval was not obtained in Europe; subsequent randomized studies in first- or second-line treatment did not confirm these initial results. However, first data with immunotherapy plus antiangiogenic treatments or dual immunotherapy combinations were positive. In this context, the combination of bevacizumab and atezolizumab took the lead. The IMbrave150 trial revealed an improved objective response rate (ORR), progression-free survival, and overall survival with this combination versus the previous standard, sorafenib. Subsequent results of dual immunotherapy with the anti-CTLA-4 and anti-PD-1 monotherapies tremelimumab and durvalumab (also superior to sorafenib monotherapy) confirmed the value of using a combination in first-line treatment. These significant therapeutic advances, and the increase in ORR, raise two main questions. Whereas response was very limited with previous treatments, the ORR reported with these new combinations are between 20% and 30%. This raises the question of whether immunotherapy (ICI single agent, combination of ICI with antiangiogenic agent or other antitumoral treatment) can be used in patients beyond those in BCLC group C, the traditional candidate group for systemic therapy. We have thus seen an increasing number of patients previously treated with trans-arterial chemoembolization (BCLC group B) receiving these new treatments, and we develop the results of several studies combining loco-regional therapies and immunotherapy-based systemic treatments. The other major question is that of how and when to use these medical treatments as "adjuvants" to interventional radiology or surgery; the results of several works are discussed for this purpose. In this review, we cover all of these points in a fairly comprehensive manner.
Collapse
Affiliation(s)
- Marine Valery
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
- Correspondence:
| | - Baptiste Cervantes
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
| | - Ramy Samaha
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
| | - Maximiliano Gelli
- Département d’Anesthésie, Chirurgie et Interventionnel, Gustave Roussy, F-94805 Villejuif, France
| | - Cristina Smolenschi
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Alina Fuerea
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
| | - Lambros Tselikas
- Département d’Anesthésie, Chirurgie et Interventionnel, Gustave Roussy, F-94805 Villejuif, France
| | | | - Antoine Hollebecque
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Valérie Boige
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
| | - Michel Ducreux
- Département de Médecine Oncologique, Gustave Roussy, F-94805 Villejuif, France
- Inserm Unité Dynamique des Cellules Tumorales, Gustave Roussy, Université Paris-Saclay, F-94805 Villejuif, France
| |
Collapse
|
42
|
Testoni SGG, Petrone MC, Reni M, Di Serio C, Rancoita PM, Rossi G, Balzano G, Linzenbold W, Enderle M, Della-Torre E, De Cobelli F, Falconi M, Capurso G, Arcidiacono PG. EUS-guided ablation with the HybridTherm Probe as second-line treatment in patients with locally advanced pancreatic ductal adenocarcinoma: A case-control study. Endosc Ultrasound 2022; 11:383-392. [PMID: 36255026 PMCID: PMC9688129 DOI: 10.4103/eus-d-21-00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background and Objectives Data on the clinical efficacy of EUS-guided ablation using the HybridTherm-Probe (EUS-HTP) in locally advanced pancreatic ductal adenocarcinoma (LA-PDAC) are lacking. The aim of the study was to assess the impact of EUS-HTP added to chemotherapy (CT) on overall survival (OS) and progression-free survival (PFS) of LA-PDAC patients with local disease progression (DP) after first-line therapy, compared to CT alone in controls. Methods LA-PDAC cases, prospectively treated by EUS-HTP, were retrospectively compared to matched controls (1:2) receiving standard treatment. Study endpoints were the OS and PFS from local DP after first-line therapy, compared through log-rank test calculating hazard ratios and differences in restricted mean OS/PFS time (RMOST/RMPFST) within prespecified time points (4, 6, and 12 months). Results Thirteen cases and 26 controls were included. Clinical, tumor, and therapy features before and after first-line therapy were case-control balanced. The median OS and PFS were not significantly improved in cases over controls (months: 7 vs. 5 and 5 vs. 3, respectively). At 4 and 6 months, the RMPFST difference was in favor of cases (P = 0.0001 and P = 0.003, respectively). In cases and controls not candidate to further CT (N = 5 and N = 9), the median OS and PFS were not significantly improved in cases over controls (months: 6 vs. 3 and 4 vs. 2, respectively), but the RMPFST difference was in favor of cases at 4 months (P = 0.002). Conclusions In locally progressive PDAC patients experiencing failure of first-line therapy, EUS-HTP achieves a significantly better RMPFST up to 6 months compared to standard treatment, although without a significant impact on OS.
Collapse
Affiliation(s)
- Sabrina Gloria Giulia Testoni
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Maria Chiara Petrone
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Michele Reni
- Department of Oncology, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Clelia Di Serio
- Vita-Salute San Raffaele University, CUSSB, University Centre for Statistics in the Biomedical Sciences, Milan, Italy
| | - Paola Maria Rancoita
- Vita-Salute San Raffaele University, CUSSB, University Centre for Statistics in the Biomedical Sciences, Milan, Italy
| | - Gemma Rossi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Gianpaolo Balzano
- Department of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | | | | | - Emanuel Della-Torre
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Francesco De Cobelli
- Department of Radiology and Center for Experimental Imaging, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| | - Massimo Falconi
- Department of Pancreatic Surgery, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, Vita-Salute San Raffaele University, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
43
|
Chorniak E, Liu Y, Odion R, Etienne W, Canning A, Nair SK, Maccarini P, Palmer GM, Inman BA, Vo-Dinh T. Intravital optical imaging for immune cell tracking after photoimmunotherapy with plasmonic gold nanostars. NANOTECHNOLOGY 2022; 33:10.1088/1361-6528/ac893a. [PMID: 35961291 PMCID: PMC9725032 DOI: 10.1088/1361-6528/ac893a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/11/2022] [Indexed: 06/15/2023]
Abstract
Bladder cancer has been ranked as one of the most commonly occurring cancers in men and women with approximately half of the diagnoses being the late stage and/or metastatic diseases. We have developed a novel cancer treatment by combining gold nanostar-mediated photothermal therapy with checkpoint inhibitor immunotherapy to treat bladder cancer. Experiment results with a murine animal model demonstrated that our developed photoimmunotherapy therapy is more efficacious than any individual studied treatment. In addition, we used intravital optical imaging with a dorsal skinfold window chamber animal model to study immune responses and immune cell accumulation in a distant tumor following our photoimmunotherapy. The mice used have the CX3CR1-GFP receptor on monocytes, natural killer cells, and dendritic cells allowing us to dynamically track their presence by fluorescence imaging. Our proof-of-principle study results showed that the photoimmunotherapy triggered anti-cancer immune responses to generate anti-cancer immune cells which accumulate in metastatic tumors. Our study results illustrate that intravital optical imaging is an efficient and versatile tool to investigate immune responses and mechanisms of photoimmunotherapy in future studies.
Collapse
Affiliation(s)
- Ericka Chorniak
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yang Liu
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Chemistry, Duke University, Durham, NC 27708, USA
- Fitzpatrick Institute of Photonics, Duke University, Durham, NC 27708, USA
| | - Ren Odion
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Wiguins Etienne
- Division of Urology, Duke University Medical Center, Durham, NC 27710, USA
| | - Aidan Canning
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Smita K. Nair
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Paolo Maccarini
- Fitzpatrick Institute of Photonics, Duke University, Durham, NC 27708, USA
- Department of Electrical and Computer Engineering, Duke University, Durham, NC 27708, USA
| | - Gregory M. Palmer
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC 27710, USA
- Fitzpatrick Institute of Photonics, Duke University, Durham, NC 27708, USA
| | - Brant A. Inman
- Fitzpatrick Institute of Photonics, Duke University, Durham, NC 27708, USA
- Division of Urology, Duke University Medical Center, Durham, NC 27710, USA
| | - Tuan Vo-Dinh
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Chemistry, Duke University, Durham, NC 27708, USA
- Fitzpatrick Institute of Photonics, Duke University, Durham, NC 27708, USA
| |
Collapse
|
44
|
Yin Z, Chen D, Liang S, Li X. Neoadjuvant Therapy for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2022; 9:929-946. [PMID: 36068876 PMCID: PMC9441170 DOI: 10.2147/jhc.s357313] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 06/15/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by low resection and high postoperative recurrence rates, and conventional treatment strategies have failed to meet clinical needs. Neoadjuvant therapy (NAT) is widely employed in the routine management of several solid tumors because it increases resectability and reduces the rate of postoperative recurrence. However, a consensus has not been reached regarding the effects of NAT on HCC. As systemic therapy, particularly targeted therapy and immunotherapy, is given for HCC treatment, accumulating evidence shows that the "spring" of NAT for HCC is imminent. In the future, HCC researchers should focus on identifying biomarkers for treatment response, explore the mechanisms of resistance, and standardize the endpoints of NAT.
Collapse
Affiliation(s)
- Zongyi Yin
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, People’s Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases & Carson International Cancer, Shenzhen University, Shenzhen, 518055, People’s Republic of China
- Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518055, People’s Republic of China
| | - Dongying Chen
- Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518055, People’s Republic of China
- Department of Anesthesiology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, People’s Republic of China
| | - Shuang Liang
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, People’s Republic of China
| | - Xiaowu Li
- Department of Hepatobiliary Surgery, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, People’s Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases & Carson International Cancer, Shenzhen University, Shenzhen, 518055, People’s Republic of China
- Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518055, People’s Republic of China
| |
Collapse
|
45
|
Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23158479. [PMID: 35955613 PMCID: PMC9368760 DOI: 10.3390/ijms23158479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 01/27/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an immunologically heterogenous disease that lacks clinically actionable targets and is more likely to progress to metastatic disease than other types of breast cancer. Tumor ablation has been used to increase response rates to checkpoint inhibitors, which remain low for TNBC patients. We hypothesized that tumor ablation could produce an anti-tumor response without using checkpoint inhibitors if immunosuppression (i.e., Tregs, tumor acidosis) was subdued. Tumors were primed with sodium bicarbonate (200 mM p.o.) to reduce tumor acidosis and low-dose cyclophosphamide (100–200 mg/kg i.p.) to deplete regulatory T cells, as has been shown independently in previous studies. A novel injectable ablative was then used to necrose the tumor, release tumor antigens, and initiate an immune event that could create an abscopal effect. This combination of bicarbonate, cyclophosphamide, and ablation, called “BiCyclA”, was tested in three syngeneic models of TNBC: E0771 (C57BL/6), 67NR (BALB/c), and 4T1-Luc (BALB/c). In E0771 and 67NR, BiCyclA therapy significantly reduced tumor growth and cured 5/7 and 6/10 mice 50 days after treatment respectively. In the metastatic 4T1-Luc tumors, for which surgery and checkpoint inhibitors fail, BiCyclA cured 5/10 mice of primary tumors and lung metastases. Notably, CD4+ and CD8+ T cells were found to be crucial for the anti-metastatic response, and cured mice were able to resist tumor rechallenge, suggesting production of immune memory. Reduction of tumor acidity and regulatory T cells with ablation is a simple yet effective therapy for local and systemic tumor control with broad applicability as it is not limited by expensive supplies.
Collapse
|
46
|
Fahmawi Y, Mehta A, Abdalhadi H, Merritt L, Mizrahi M. Efficacy and safety of endoscopic ultrasound-guided radiofrequency ablation for management of pancreatic lesions: a systematic review and meta-analysis. Transl Gastroenterol Hepatol 2022; 7:30. [PMID: 35892058 PMCID: PMC9257535 DOI: 10.21037/tgh-20-84] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/16/2020] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND Radiofrequency ablation (RFA) has been used to treat various abdominal tumors including pancreatic tumors. Multiple approaches such as laparoscopic, open, and percutaneous have been used for pancreatic tissue ablation. More recently, endoscopic ultrasound (EUS)-guided RFA has emerged as a new technique for pancreatic tissue ablation. The role of EUS-RFA in management of pancreatic lesions is still not well-established. In this study, our aim is to assess efficacy and safety of EUS-RFA for management of pancreatic lesions. METHODS MEDLINE, Scopus, and Cochrane Library databases were searched to identify studies reporting EUS-RFA of pancreatic lesions with outcomes of interest. Studies with <5 patients were excluded. Clinical success was defined as symptom resolution, decrease in tumor size, and/or evidence of necrosis on radiologic imaging. Efficacy was assessed by the pooled clinical response rate whereas safety was assessed by the pooled adverse events rate. Heterogeneity was assessed using I2. Pooled estimates and the 95% CI were calculated using random-effect model. RESULTS Ten studies (5 retrospective and 5 prospective) involving 115 patients with 125 pancreatic lesions were included. 152 EUS-RFA procedures were performed. The lesions comprised of 37.6% non-functional neuroendocrine tumors (NFNETs), 15.4% were insulinomas, 26.5% were pancreatic cystic neoplasms (PCNs), and 19.7% were pancreatic adenocarcinomas. The majority were present in the pancreatic head (40.2%), 38.3% in the body, 11.2% in the tail, and 10.3% in the uncinate process. Pooled overall clinical response rate was 88.9% (95% CI: 82.4-93.7, I2=38.1%). Pooled overall adverse events rate was 6.7% (95% CI: 3.4-11.7, I2=34.0%). The most common complication was acute pancreatitis (3.3%) followed by pancreatic duct stenosis, peripancreatic fluid collection, and ascites (2.8%) each. Only one case of perforation was reported with pooled rate of (2.1%). DISCUSSION This study demonstrates that EUS-RFA is an effective treatment modality for pancreatic lesions, especially functional neuroendocrine tumors such as insulinomas.
Collapse
Affiliation(s)
- Yazan Fahmawi
- Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
| | - Ansh Mehta
- Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
| | - Haneen Abdalhadi
- Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
| | - Lindsey Merritt
- Department of Gastroenterology and Hepatology, Advanced Endoscopy Unit, University of South Alabama, Mobile, AL, USA
| | - Meir Mizrahi
- Department of Gastroenterology and Hepatology, Advanced Endoscopy Unit, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
47
|
Tian Z, Hong B, Chen J, Tang Z. Combination of Radiofrequency Ablation With Resiquimod to Treat Hepatocellular Carcinoma Via Inflammation of Tumor Immune Microenvironment and Suppression of Angiogenesis. Front Oncol 2022; 12:891724. [PMID: 35719978 PMCID: PMC9201999 DOI: 10.3389/fonc.2022.891724] [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: 03/08/2022] [Accepted: 04/22/2022] [Indexed: 12/07/2022] Open
Abstract
Background Radiofrequency ablation (RFA) destroys tumors through hyperthermic injury, which induces the release of immunogenic intracellular substrates and damages associated molecular patterns (DAMPs) to evoke a systemic immune response, but its therapeutic effect is limited. This study aimed to combine RFA with an immunomodulator, resiquimod (R848), to enhance the RFA-induced antitumor immunity. Methods We performed RFA on subcutaneous tumors in immunocompetent mice and intraperitoneally injected R848 to observe the efficacy of the combination therapy. Our research investigated changes in the composition of tumor-infiltrating immune cells in primary and distant tumors by flow cytometry. Natural killer (NK) cell depletion experiment was applied to confirm the role of NK cell in the combination therapy. The expression levels of cytokines and chemokines were detected by real-time quantitative PCR. Immunohistochemical test was conducted to reveal tumor angiogenesis, tumor proliferation, and apoptosis after the different treatments. Results and Conclusion Compared with RFA or R848 monotherapy, the combination therapy significantly slowed the tumor growth, prolonged the survival time, and shrank the tumor-draining lymph nodes of tumor-bearing mice. The flow cytometry results showed that tumor-infiltrating immune cells, total T cells, the ratio of CD8+ T and NK cells to CD45+ cells, and functional NK cells were obviously increased after the combined treatment. Distal tumor growth was also suppressed, and the profile of tumor-infiltrating immune cells was remodeled, too. In addition, the additive effect of the combination therapy disappeared after NK cell depletion. Furthermore, immunohistochemical results verified that R848 inhibited tumor angiogenesis in murine liver cancer, and the combination therapy promoted tumor cell apoptosis. In conclusion, our data suggest that RFA combined with R848 stimulated a stronger antitumor immune response and effectively inhibited liver cancer progression in a NK cell-dependent manner. Meanwhile, we confirmed that R848 inhibited tumor angiogenesis and promoted apoptosis in murine liver cancer. Overall, this is a promising therapeutic strategy to improve the efficacy of RFA in the treatment of liver cancer and provides a novel option for combined thermal ablation and immunotherapy.
Collapse
Affiliation(s)
- Zhou Tian
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China
| | - Baojian Hong
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Jianzhong Chen
- Institute of Immunology School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, China.,Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Abstract
Andrew Canakis.
Collapse
Affiliation(s)
- Andrew Canakis
- Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Linda S Lee
- Division of Gastroenterology Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA, 02115, USA.
| |
Collapse
|
49
|
[Current Status and Progress of Thermal Ablation Combined with Immunotherapy for Lung Tumors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:266-271. [PMID: 35477191 PMCID: PMC9051299 DOI: 10.3779/j.issn.1009-3419.2022.102.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Recent studies have shown that tumor immune microenvironment is closely related to tumor progression, metastasis, recurrence and response to treatment. Some immunotherapies also offer hope for cancer patients. However, the efficacy of tumor immunotherapy is uncertain and has some side effects. In order to enhance its efficacy, tumor immunotherapy combined with tumor thermal ablation has been studied. Thermal ablation has the advantages of minimally invasive, rapid recovery, safety, fewer complications, conformation, reliable effect, repeatable, low cost, and has become the fourth tumor treatment measure after surgery, radiotherapy, and drug therapy. It can directly kill tumor cells and modulate the immune system through a variety of mechanisms, although the corresponding mechanisms are not well understood, but combined tumor immunotherapy has been proposed to treat several solid malignancies. In this review, the current status and progress of thermal ablation combined with immunotherapy for lung tumor were reviewed, and further studies on the efficacy and safety of thermal ablation combined with immunotherapy were expected.
.
Collapse
|
50
|
Senders ZJ, Martin RCG. Intratumoral Immunotherapy and Tumor Ablation: A Local Approach with Broad Potential. Cancers (Basel) 2022; 14:cancers14071754. [PMID: 35406525 PMCID: PMC8996835 DOI: 10.3390/cancers14071754] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/11/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
Several intratumoral immunotherapeutic agents have shown efficacy in controlling local disease; however, their ability to induce a durable systemic immune response is limited. Likewise, tumor ablation is well-established due to its role in local disease control but generally produces only a modest immunogenic effect. It has recently been recognized, however, that there is potential synergy between these two modalities and their distinct mechanisms of immune modulation. The aim of this review is to evaluate the existing data regarding multimodality therapy with intratumoral immunotherapy and tumor ablation. We discuss the rationale for this therapeutic approach, highlight novel combinations, and address the challenges to their clinical utility. There is substantial evidence that combination therapy with intratumoral immunotherapy and tumor ablation can potentiate durable systemic immune responses and should be further evaluated in the clinical setting.
Collapse
|