1
|
Sardela de Miranda F, Martinez-Marin D, Babcock RL, Castro M, Boligala GP, Khan SY, Furr KL, Castro-Piedras I, Wagner N, Robison DE, Daniele K, Singh SP, Pruitt K, Melkus MW, Layeequr Rahman R. Cryoablation of primary breast cancer tumors induces a systemic abscopal effect altering TIME (Tumor Immune Microenvironment) in distant tumors. Front Immunol 2024; 15:1498942. [PMID: 39703517 PMCID: PMC11657241 DOI: 10.3389/fimmu.2024.1498942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/04/2024] [Indexed: 12/21/2024] Open
Abstract
Introduction Despite recent advances, triple-negative breast cancer (TNBC) patients remain at high risk for recurrence and metastasis, which creates the need for innovative therapeutic approaches to improve patient outcomes. Cryoablation is a promising, less invasive alternative to surgical resection, capable of inducing tumor necrosis via freeze/thaw cycles. Necrotic cell death results in increased inflammatory signals and release of preserved tumor antigens, which have the potential to boost the local and systemic anti-tumor immune response. Thus, compared to surgery, cryoablation enhances the activation of T cells leading to an improved abscopal effect, defined as the occurrence of a systemic response after local treatment. We previously showed with a bilateral-tumor mouse model of TNBC that cryoablation of the primary tumor leads to increased infiltration of distant (abscopal) tumors by tumor infiltrating lymphocytes (TILs) and decreased rates of recurrence and metastasis. However, the early drivers of the cryoablation generated abscopal effect are still unknown and knowledge of the mechanism could provide insight into improving the anti-tumor immune response through pharmacologic immune modulation in addition to cryoablation. Methods One million 4T1-12B-luciferase expressing cells were transplanted into the mammary fat pad of BALB/c mice. Two weeks later, left (primary) tumors were either resected or cryoablated. A week after the procedure, right (abscopal) and left tumors, along with spleen, tumor-draining lymph node and blood were collected and processed for flow cytometry and/or RNA-sequencing and immunofluorescence. Results Here we show that cryoablation of mouse mammary carcinomas results in smaller abscopal tumors that harbor increased frequencies of anti-tumor cells [such as natural killer (NK) cells], accompanied by a systemic increase in the frequency of migratory conventional type 1 dendritic cells (cDC1; CD103+ XCR1+), compared to resection. The changes in cell frequencies are mirrored by the immune gene signature of the abscopal tumors, with cryoablation inducing genes involved with NK cell activation and leukocyte-mediated toxicity, including IL11ra1 and Pfr1. Conclusions These results better define the early mechanisms through which cryoablation improves tumor elimination, which is mediated by enhanced frequencies of anti-tumoral cells such as NK and cDC1s at the abscopal tumor and in the spleen of mice treated with cryoablation, respectively.
Collapse
Affiliation(s)
- Flávia Sardela de Miranda
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Dalia Martinez-Marin
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rachel L. Babcock
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Cell Biology and Biochemistry, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Maribel Castro
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Geetha P. Boligala
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Sonia Y. Khan
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Surgery, The University of Texas Rio Grande Valley (UTRGV) Rio Grande Valley, Harlingen, TX, United States
| | - Kathryn L. Furr
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Isabel Castro-Piedras
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Nicholas Wagner
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Dakota E. Robison
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Karla Daniele
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sharda P. Singh
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Kevin Pruitt
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael W. Melkus
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Cancer Institute, MetroHealth System, Cleveland, OH, United States
| |
Collapse
|
2
|
Holtermann A, Gislon M, Angele M, Subklewe M, von Bergwelt-Baildon M, Lauber K, Kobold S. Prospects of Synergy: Local Interventions and CAR T Cell Therapy in Solid Tumors. BioDrugs 2024; 38:611-637. [PMID: 39080180 PMCID: PMC11358237 DOI: 10.1007/s40259-024-00669-y] [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] [Accepted: 06/17/2024] [Indexed: 08/30/2024]
Abstract
Chimeric antigen receptor T cell therapy has been established in the treatment of various B cell malignancies. However, translating this therapeutic effect to treat solid tumors has been challenging because of their inter-tumoral as well as intratumoral heterogeneity and immunosuppressive microenvironment. Local interventions, such as surgery, radiotherapy, local ablation, and locoregional drug delivery, can enhance chimeric antigen receptor T cell therapy in solid tumors by improving tumor infiltration and reducing systemic toxicities. Additionally, ablation and radiotherapy have proven to (re-)activate systemic immune responses via abscopal effects and reprogram the tumor microenvironment on a physical, cellular, and chemical level. This review highlights the potential synergy of the combined approaches to overcome barriers of chimeric antigen receptor T cell therapy and summarizes recent studies that may pave the way for new treatment regimens.
Collapse
Affiliation(s)
- Anne Holtermann
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Mila Gislon
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health Neuherberg, Munich, Germany.
| |
Collapse
|
3
|
Zhang X, Shao S, Song N, Yang B, Liu F, Tong Z, Wang F, Li J. Integrated omics characterization reveals reduced cancer indicators and elevated inflammatory factors after thermal ablation in non-small cell lung cancer patients. Respir Res 2024; 25:309. [PMID: 39143582 PMCID: PMC11325606 DOI: 10.1186/s12931-024-02917-9] [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: 04/20/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Thermal ablation is a minimally invasive treatment for non-small cell lung cancer (NSCLC). Aside from causing an immediate direct tumour cell injury, the effects of thermal ablation on the internal microenvironment are unknown. This study aimed to investigate the effects of thermal ablation on the plasma internal environment in patients with NSCLC. METHODS 128 plasma samples were collected from 48 NSCLC (pre [LC] and after thermal ablation [LC-T]) patients and 32 healthy controls (HCs). Olink proteomics and metabolomics were utilized to construct an integrated landscape of the cancer-related immune and inflammatory responses after ablation. RESULTS Compared with HCs, LC patients exhibited 58 differentially expressed proteins (DEPs) and 479 differentially expressed metabolites (DEMs), which might participate in tumour progression and metastasis. Moreover, 75 DEPs were identified among the HC, LC, and LC-T groups. Forty-eight highly expressed DEPs (eg, programmed death-ligand 1 [PD-L1]) in the LC group were found to be downregulated after thermal ablation. These DEPs had significant impacts on pathways such as angiogenesis, immune checkpoint blockade, and pro-tumour chemotaxis. Metabolites involved in tumour cell survival were associated with these proteins at the expression and functional levels. In contrast, 19 elevated proteins (eg, interleukin [IL]-6) were identified after thermal ablation. These proteins were mainly associated with inflammatory response pathways (NF-κB signalling and tumour necrosis factor signalling) and immune cell activation. CONCLUSIONS Thermal ablation-induced changes in the host plasma microenvironment contribute to anti-tumour immunity in NSCLC, offering new insights into tumour ablation combined with immunotherapy. Trial registration This study was registered on the Chinese Clinical Trial Registry ( https://www.chictr.org.cn/index.html ). ID: ChiCTR2300076517. Registration Date: 2023-10-11.
Collapse
Affiliation(s)
- Xinglu Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Beijing, 100020, Chaoyang District, China
| | - Shuai Shao
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Beijing, 100020, Chaoyang District, China
| | - Nan Song
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Baolu Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Beijing, 100020, Chaoyang District, China
| | - Fengjiao Liu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Beijing, 100020, Chaoyang District, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Beijing, 100020, Chaoyang District, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Feng Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, No. 8 Gongti South Road, Beijing, 100020, Chaoyang District, China.
| | - Jieqiong Li
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Gu J, Yu Z, Tang X, Chen W, Deng X, Zhu X. Cryoablation combined with dual immune checkpoint blockade enhances antitumor efficacy in hepatocellular carcinoma model mice. Int J Hyperthermia 2024; 41:2373319. [PMID: 38955354 DOI: 10.1080/02656736.2024.2373319] [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: 04/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Cryoablation (Cryo) is a minimally invasive treatment for tumors. Cryo can activate the body's immune response, although it is typically weak. The immune response induced by Cryo in hepatocellular carcinoma (HCC) is poorly understood. PD-1 and CTLA-4 monoclonal antibodies are immune checkpoint inhibitors used in immunotherapy for tumors. The combined use of these antibodies with Cryo may enhance the immune effect. METHODS A Balb/c mouse model of HCC was established and treated with Cryo, immune checkpoint blockade (ICB), or Cryo + ICB (combination therapy). The growth trend of right untreated tumors and survival time of mice were determined. The expression of apoptosis-related proteins was detected by Western blot (WB) assay. The percentages of immune cells and immunosuppressive cells were analyzed by flow cytometry. The numbers of infiltrating T lymphocytes were checked by immunohistochemistry, and the levels of T-cell-associated cytokines were detected by Quantitative real-time Polymerase Chain Reaction (qRT-PCR) assays and Enzyme-Linked Immunosorbent Assays (ELISA) assays. RESULTS Cryo + ICB inhibited the growth of right untreated tumors, promoted tumor cell apoptosis, and prolonged the survival time of mice. Local T-cell infiltration in right tumor tissues increased after the combination therapy, while the number of immunosuppressive cells was significantly reduced. In addition, the combination therapy may induce the production of multiple Th1-type cytokines but reduce the production of Th2-type cytokines. CONCLUSIONS Cryo can activate CD8+ and CD4+ T-cell immune responses. Cryo + ICB can relieve the immunosuppressive tumor microenvironment and shift the Th1/Th2 balance toward Th1 dominance, further enhancing the Cryo-induced T-cell immune response and resulting in a stronger antitumor immune response.
Collapse
Affiliation(s)
- Jun Gu
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Zepeng Yu
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xiangxiang Tang
- School of Nursing, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Wenying Chen
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xuedong Deng
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xiaoli Zhu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
5
|
Séguier D, Adams ES, Kotamarti S, D'Anniballe V, Michael ZD, Deivasigamani S, Olivier J, Villers A, Hoimes C, Polascik TJ. Intratumoural immunotherapy plus focal thermal ablation for localized prostate cancer. Nat Rev Urol 2024; 21:290-302. [PMID: 38114768 DOI: 10.1038/s41585-023-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 12/21/2023]
Abstract
Major advances have been made in the use of immunotherapy for the treatment of solid tumours, including the use of intratumourally injected immunotherapy instead of systemically delivered immunotherapy. The success of immunotherapy in prostate cancer treatment has been limited to specific populations with advanced disease, which is thought to be a result of prostate cancer being an immunologically 'cold' cancer. Accordingly, combining intratumoural immunotherapy with other treatments that would increase the immunological heat of prostate cancer is of interest. Thermal ablation therapy is currently one of the main strategies used for the treatment of localized prostate cancer and it causes immunological activation against prostate tissue. The use of intratumoural immunotherapy as an adjunct to thermal ablation offers the potential to elicit a systemic and lasting adaptive immune response to cancer-specific antigens, leading to a synergistic effect of combination therapy. The combination of thermal ablation and immunotherapy is currently in the early stages of investigation for the treatment of multiple solid tumour types, and the potential for this combination therapy to also offer benefit to prostate cancer patients is exciting.
Collapse
Affiliation(s)
- Denis Séguier
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
- Department of Urology, Lille University, Lille, France.
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Eric S Adams
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Srinath Kotamarti
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Vincent D'Anniballe
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Zoe D Michael
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Sriram Deivasigamani
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Jonathan Olivier
- Department of Urology, Lille University, Lille, France
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Arnauld Villers
- Department of Urology, Lille University, Lille, France
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Christopher Hoimes
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, 27708, USA
| | - Thomas J Polascik
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
6
|
Shen L, Tan H, Nie J, Jiang Y, Nuerhashi G, Qi H, Cao F, Wen C, Chen S, Zhang T, Zheng W, Liu P, Liu Y, Huang T, Li D, Zhang X, Fan W. Size selection of intrahepatic lesions for cryoablation contributes to abscopal effect and long-term survival in patients with liver metastatic melanoma receiving PD-1 blockade therapy. Cancer Immunol Immunother 2024; 73:68. [PMID: 38430269 PMCID: PMC10908608 DOI: 10.1007/s00262-024-03637-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 01/16/2024] [Indexed: 03/03/2024]
Abstract
OBJECTIVES In this study, we aimed to examine parameters of cryoablation, tumor characteristics, and their correlations with distant tumor response and survival of liver metastatic melanoma patients receiving cryoablation and PD-1 blockade (cryo-PD-1) combination treatment. MATERIALS AND METHODS A retrospective study was conducted among 45 melanoma patients who received combined PD-1 blockade therapy and cryoablation for liver metastasis from 2018 to 2022. Cox regression was utilized to determine the associations between factors and overall survival (OS). Changes in cytokines and immune cell compositions in peripheral blood samples following the combined treatment were investigated, along with their correlations with treatment response. RESULTS The mean cycle of cryo-PD-1 combination treatment was 2.2 (range, 1-6), and the 3-month overall response rate (RECIST 1.1 criteria) was 26.7%. Of the 21 patients who failed previous PD-1 blockade therapy after diagnosis of liver metastasis, 4 (19.0%) achieved response within 3 months since combination treatment. The diameter of ablated lesion ≤ 30 mm, metastatic organs ≤ 2, and pre-treatment LDH level ≤ 300 U/L were independent prognostic factors for favorable OS. Further analysis showed patients with intrahepatic tumor size of 15-45 mm, and ablated lesion size of ≤ 30 mm had significantly higher 3-month response rate (42.9% vs 12.5%; P = 0.022) and survival time (30.5 vs 14.2 months; P = 0.045) than their counterparts. The average increase in NLR among patients with ablated tumor size of ≤ 3 cm and > 3 cm were 3.59 ± 5.01 and 7.21 ± 12.57, respectively. The average increase in serum IL-6 levels among patients with ablated tumor size of ≤ 3 cm and > 3 cm were 8.62 ± 7.95 pg/ml and 15.40 ± 11.43 pg/ml, respectively. CONCLUSION Size selection of intrahepatic lesions for cryoablation is important in order to achieve abscopal effect and long-term survival among patients with liver metastatic melanoma receiving PD-1 blockade therapy.
Collapse
Affiliation(s)
- Lujun Shen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Hongtong Tan
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Juan Nie
- Department of Research & Education, Guangzhou Concord Cancer Center, Guangzhou, 510054, People's Republic of China
| | - Yiquan Jiang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Gulijiayina Nuerhashi
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Han Qi
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Fei Cao
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chunyong Wen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Shuanggang Chen
- Department of Oncology, Yuebei People's Hospital, Shaoguan, 511100, People's Republic of China
| | - Tianqi Zhang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Wei Zheng
- Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, 400000, People's Republic of China
| | - Peng Liu
- Department of Oncology, General Hospital of Southern Theater Command, Guangzhou, 510060, People's Republic of China
| | - Ying Liu
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Tao Huang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Dandan Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- Department of Biological Therapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Xiaoshi Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
- Department of Biological Therapy Center, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Weijun Fan
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
7
|
Lucas AP, Lewis AR, Kasi PM, Toskich BB, Paz-Fumagalli R. Abscopal downstaging of intermediate stage hepatocellular via combination cryoablation and immunotherapy with complete pathologic response. Radiol Case Rep 2024; 19:910-914. [PMID: 38188944 PMCID: PMC10770505 DOI: 10.1016/j.radcr.2023.11.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 01/09/2024] Open
Abstract
The abscopal effect is a rare phenomenon characterized by disease regression in distant sites after tumoral locoregional therapy. Locoregional therapy, such as cryoablation, can induce an antitumor immunological response, potentially improving outcomes in cancer patients receiving immunotherapy. This report describes a patient with multifocal hepatocellular carcinoma who progressed through multiple locoregional therapies, was initially unresponsive to immunotherapy, and later achieved rapid and sustained disease regression with a combination cryoablation and immunotherapy. A 5-year sustained complete tumor response successfully bridged to liver transplantation.
Collapse
Affiliation(s)
- Ana P. Lucas
- Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Andrew R. Lewis
- Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | - Pashtoon M. Kasi
- Weill Cornell Medicine/New York-Presbyterian, 525 E 68th St, New York, NY 10065, USA
| | - Beau B. Toskich
- Mayo Clinic Florida, 4500 San Pablo Rd, Jacksonville, FL, 32224, USA
| | | |
Collapse
|
8
|
Liu Q, Zhang C, Chen X, Han Z. Modern cancer therapy: cryoablation meets immune checkpoint blockade. Front Oncol 2024; 14:1323070. [PMID: 38384806 PMCID: PMC10881233 DOI: 10.3389/fonc.2024.1323070] [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: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Cryoablation, as a minimally invasive technology for the treatment of tumors, destroys target tumors with lethal low temperatures. It simultaneously releases a large number of tumor-specific antigens, pro-inflammatory cytokines, and nucleoproteins, known as "danger signals", activating the body's innate and adaptive immune responses. However, tumor cells can promote the inactivation of immune effector cells by reprogramming immune checkpoints, leading to the insufficiency of these antigens to induce an immune response capable of eradicating the tumor. Immune checkpoint blockers rejuvenate exhausted T cells by blocking immune checkpoints that induce programmed death of T cells, and are therefore considered a promising therapeutic strategy to enhance the immune effects of cryoablation. In this review, we provide a detailed explanation of the immunological mechanisms of cryoablation and articulate the theoretical basis and research progress of the treatment of cancer with cryoablation combined with immune checkpoint blockers. Preliminary data indicates that this combined treatment strategy exhibits good synergy and has been proven to be safe and effective.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
9
|
Yu ZP, Sun XW, He YP, Gu J, Jin Y. PD-1 monoclonal antibodies enhance the cryoablation-induced antitumor immune response: a breast cancer murine model research. Int J Hyperthermia 2023; 40:2164625. [PMID: 36966808 DOI: 10.1080/02656736.2022.2164625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
Abstract
BACKGROUND It has been demonstrated that cryoablation (Cryo) causes specific T-cell immune responses in the body; however, it is not sufficient to prevent tumor recurrence and metastasis. In this report, we evaluated changes in the tumor immune microenvironment (TIME) in distant tumor tissues after Cryo and investigated the immunosuppressive mechanisms that limit the efficacy of Cryo. METHODS Bilateral mammary tumor models were established in mice, and we first observed the dynamic changes in immune cells and cytokines at different time points after Cryo. Then, we confirmed that the upregulation of PD-1 and PD-L1 signaling in the contralateral tumor tissue was closely related to the immunosuppressive state in the TIME at the later stage after Cryo. Finally, we also evaluated the synergistic antitumor effects of Cryo combined with PD-1 monoclonal antibody (mAb) in the treatment of breast cancer (BC) mouse. RESULTS We found that Cryo can stimulate the body's immune response, but it also induces immunosuppression. The elevated PD-1/PD-L1 expression in distant tumor tissues at the later stage after Cryo was closely related to the immunosuppressive state in the TIME but also created the conditions for Cryo combined with PD-1 mAb for BC mouse treatment. Cryo + PD-1 mAb could improve the immunosuppressive state of tumors and enhance the Cryo-induced immune response, thus exerting a synergistic antitumor effect. CONCLUSIONS The PD-1/PD-L1 axis plays an important role in suppressing Cryo-induced antitumor immune responses. This study provides a theoretical basis for Cryo combined with PD-1 mAb therapy in clinical BC patients.
Collapse
|
10
|
Dronov O, Kozachuk Y, Bakunets Y, Bakunets P, Prytkov F. THE CRYOGENIC TECHNOLOGIES APPLICATION IN THE COMPLEX TREATMENT OF GALLBLADDER ADENOCARCINOMA WITH INVASION INTO THE PORTAL VEIN BRANCHES: CASE REPORT. Exp Oncol 2023; 45:379-385. [PMID: 38186017 DOI: 10.15407/exp-oncology.2023.03.379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Indexed: 01/09/2024]
Abstract
The right trisectionectomy is the main treatment modality for locally advanced gallbladder cancer with invasion of the intraparenchymal portal vein branches because it allows the achievement of negative resection margins (R0). However, only 10%-25% of such patients are eligible for surgery. The cryosurgical method has been successfully used in the complex treatment of hepatopancreatobiliary malignant neoplasms for many years. The possibility of its application close to major blood vessels is one of its advantages. In the presented case, the cryodestruction of the residual tumor with invasion into the anterior wall of the left branch of the portal vein was used as a debulking option during liver resection (R2) due to locally advanced gallbladder cancer. The cryodestruction was performed with application method with a double cryocycle and spontaneous thawing using a Cryo-Pulse device and liquid nitrogen as a cryoagent. No postoperative complications related to cryodestruction were noted. The cryogenic technologies application in the debulking surgery of gallbladder cancer can be a safe treatment modality for residual tumors with invasion into the intraparenchymal branches of the portal vein.
Collapse
Affiliation(s)
- O Dronov
- Bogomolets National Medical University, Kyiv, Ukraine
| | - Y Kozachuk
- Bogomolets National Medical University, Kyiv, Ukraine
| | - Y Bakunets
- Bogomolets National Medical University, Kyiv, Ukraine
| | - P Bakunets
- Bogomolets National Medical University, Kyiv, Ukraine
| | - F Prytkov
- Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
11
|
Hong H, Shen L, Tan H, Wu Y, Liu Y, Fan W. Local treatment of liver metastasis in a patient with advanced malignant melanoma: A case report. J Cancer Res Ther 2023; 19:1675-1679. [PMID: 38156937 DOI: 10.4103/jcrt.jcrt_394_22] [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: 02/16/2022] [Accepted: 08/23/2023] [Indexed: 01/03/2024]
Abstract
Melanoma is a fatal malignant tumor with a high rate of metastasis. Liver metastasis of melanoma is always associated with insensitivity to immunotherapy and a poor prognosis. However, the combination of cryoablation, which is believed to stimulate the antitumor immune response in the body, with immunotherapy can improve the therapeutic response to this condition. Herein, we present the case of a 79-year-old woman with BRAF (B-Raf proto-oncogene) wild-type melanoma who later developed liver metastasis. The patient received intravenous antiprogrammed cell death 1 antibody, which showed poor efficacy, and subsequent treatment with immunotherapy combined with cryoablation yielded a partial response. However, after the second cryoablation, the patient refused further treatment due to a fear of bleeding. Therefore, only immunotherapy was provided, which resulted in disease progression. This report demonstrates the need to consider immunotherapy plus cryoablation for the treatment of liver metastases in patients with BRAF wild-type melanoma.
Collapse
Affiliation(s)
- Hongxi Hong
- Department of Oncology, Hospital of Traditional Chinese Medicine of Guangdong Province, Guangzhou, People's Republic of China
| | - Lujun Shen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
- The State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Hongtong Tan
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
- The State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ying Wu
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
- The State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ying Liu
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
- The State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Weijun Fan
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
- The State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| |
Collapse
|
12
|
Khan SY, Cole J, Habrawi Z, Melkus MW, Layeequr Rahman R. Cryoablation Allows the Ultimate De-escalation of Surgical Therapy for Select Breast Cancer Patients. Ann Surg Oncol 2023; 30:8398-8403. [PMID: 37770723 PMCID: PMC10625946 DOI: 10.1245/s10434-023-14332-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/09/2023] [Indexed: 09/30/2023]
Abstract
BACKGROUND Widespread use of screening mammography has allowed breast cancer to be detected at earlier stages. This allows for increased customization of treatment and less aggressive management. De-escalation of therapy plays an important role in decreasing treatment burden and improving patient quality of life. This report examines cryoablation as the next step in the surgical de-escalation of breast cancer. METHODS Women with a diagnosis of clinically node-negative, estrogen receptor-positive (ER +), progesterone receptor-positive (PR +), human epidermal growth factor receptor 2-negative (HER2 -) infiltrating ductal carcinomas 1.5 cm or smaller underwent ultrasound-guided cryoablation. Either the Visica 2 treatment system (before 2020) or the ProSense treatment system (since 2020) was used to perform the cryoablation. Patients received mammograms and ultrasounds at a 6 months follow-up visit, and magnetic resonance images at baseline, then at 1 year follow-up intervals. Adjuvant therapy decisions and disease status were recorded. RESULTS This study enrolled 32 patients who underwent 33 cryoablation procedures (1 patient had bilateral cancer). One patient had a sentinel node biopsy in addition to clinical staging of the axilla. For all the patients, adjuvant endocrine therapy was recommended, and six patients (18.75%) received adjuvant radiation. Of the 32 patients, 20 (60.6%) have been followed up for 2 years or longer, with no residual or recurrent disease at the site of ablation. CONCLUSION Cryoablation of the primary tumor foregoing sentinel node biopsy offers an oncologically safe and feasible minimally invasive office-based procedure option in lieu of surgery for patients with early-stage, low-risk breast cancer.
Collapse
Affiliation(s)
- Sonia Y Khan
- Breast Center of Excellence and Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jaclyn Cole
- Breast Center of Excellence and Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Zaina Habrawi
- Breast Center of Excellence and Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Michael W Melkus
- Breast Center of Excellence and Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Rakhshanda Layeequr Rahman
- Breast Center of Excellence and Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
13
|
Sardela de Miranda F, Castro M, Remmert N, Singh SP, Layeequr Rahman R, Melkus MW. Leveraging cryoablation and checkpoint inhibitors for high-risk triple negative breast cancer. Front Immunol 2023; 14:1258873. [PMID: 37860001 PMCID: PMC10582696 DOI: 10.3389/fimmu.2023.1258873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Breast cancer is the second most common cancer among women in the United States in which the standard of care treatment is surgery with adjunctive therapy. Cryoablation, which destroys the tumor using extremely cold temperatures while preserving the potential tumor antigens, is a promising alternative to surgical resection. It is less invasive, cosmetically appeasing, cost-effective, and capable of contributing to the abscopal effect - the immune response targeting potential distant metastasis. However, to maximize the immunologic benefit of cryoablation in biologically high-risk breast cancers, combination with therapies that enhance immune activation, such as immune checkpoint inhibitors (ICIs) may be necessary. This mini review describes the fundamentals of cryoablation and treatment with ICIs, as well as discuss the caveats in both strategies and current clinical trials aimed to improve this approach to benefit patients.
Collapse
Affiliation(s)
- Flávia Sardela de Miranda
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Maribel Castro
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Nicole Remmert
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Sharda P. Singh
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Internal Medicine, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Rakhshanda Layeequr Rahman
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Michael W. Melkus
- Department of Surgery, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Breast Center of Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
14
|
Liao Y, Chen Y, Liu S, Wang W, Fu S, Wu J. Low-dose total body irradiation enhances systemic anti-tumor immunity induced by local cryotherapy. J Cancer Res Clin Oncol 2023; 149:10053-10063. [PMID: 37261526 DOI: 10.1007/s00432-023-04928-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Strategies that restore the immune system's ability to recognize malignant cells have yielded clinical benefits but only in some patients. Tumor cells survive cryotherapy and produce a vast amount of antigens to trigger innate and adaptive responses. However, because tumor cells have developed immune escape mechanisms, cryotherapy alone may not be enough to induce a significant immune response. METHODS The mice were randomly divided into four groups: Group A: low-dose total body irradiation combined with cryotherapy (L-TBI+cryo); Group B: cryotherapy (cryo); Group C: low-dose total body irradiation(L-TBI); Group D: control group (Control). The tumor growth, recurrence, and survival time of mice in each group were compared and the effects of different treatments on systemic anti-tumor immunity were explored. RESULTS L-TBI in conjunction with cryotherapy can effectively control tumor regrowth, inhibit tumor lung metastasis, extend the survival time of mice, and stimulate a long-term protective anti-tumor immune response to resist the re-challenge of tumor cells. The anti-tumor mechanism of this combination therapy may be related to the stimulation of inflammatory factors IFN-γ and IL-2, as well as an increase in immune effector cells (CD8+ T cells) and a decrease in immunosuppressive cells (MDSC, Treg cells) in the spleen or tumor tissue. CONCLUSIONS We present unique treatment options for enhancing the immune response caused by cryotherapy, pointing to the way forward for cancer treatment.
Collapse
Affiliation(s)
- Yin Liao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Yao Chen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shuya Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Weizhou Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Shaozhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jingbo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China.
| |
Collapse
|
15
|
Pal K, Sheth RA. Engineering the Tumor Immune Microenvironment through Minimally Invasive Interventions. Cancers (Basel) 2022; 15:196. [PMID: 36612192 PMCID: PMC9818918 DOI: 10.3390/cancers15010196] [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: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is a unique landscape that poses several physical, biochemical, and immune barriers to anti-cancer therapies. The rapidly evolving field of immuno-engineering provides new opportunities to dismantle the tumor immune microenvironment by efficient tumor destruction. Systemic delivery of such treatments can often have limited local effects, leading to unwanted offsite effects such as systemic toxicity and tumor resistance. Interventional radiologists use contemporary image-guided techniques to locally deliver these therapies to modulate the immunosuppressive TME, further accelerating tumor death and invoking a better anti-tumor response. These involve local therapies such as intratumoral drug delivery, nanorobots, nanoparticles, and implantable microdevices. Physical therapies such as photodynamic therapy, electroporation, hyperthermia, hypothermia, ultrasound therapy, histotripsy, and radiotherapy are also available for local tumor destruction. While the interventional radiologist can only locally manipulate the TME, there are systemic offsite recruitments of the immune response. This is known as the abscopal effect, which leads to more significant anti-tumoral downstream effects. Local delivery of modern immunoengineering methods such as locoregional CAR-T therapy combined with immune checkpoint inhibitors efficaciously modulates the immunosuppressive TME. This review highlights the various advances and technologies available now to change the TME and revolutionize oncology from a minimally invasive viewpoint.
Collapse
Affiliation(s)
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
16
|
Olagunju A, Forsman T, Ward RC. An update on the use of cryoablation and immunotherapy for breast cancer. Front Immunol 2022; 13:1026475. [PMID: 36389815 PMCID: PMC9647043 DOI: 10.3389/fimmu.2022.1026475] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/13/2022] [Indexed: 09/10/2023] Open
Abstract
The use of cryoablation, a minimally-invasive image-guided technique to target and kill cancer cells, continues to gain traction within the medical field and with patients. This includes the use of cryoablation for the treatment of small breast cancers and focal sites of metastatic disease. In comparison to open surgical approaches, length of hospital stay and recovery time are decreased with the use of cryoablation. Research studies have also found that cryoablation may actually enhance tumor susceptibility to immunotherapy agents. Immunotherapy enhances a person's own immune system to identify and attack cancer cells. It is proposed that after cryoablation there is increased expression of tumor specific antigens which the body can recognize as foreign invaders and with the combination of immunotherapy, result in an even more robust and efficient attack on the cancer cells. In this review we aim to highlight some of the recent advances in cryoablation which support the potential for cryoablation to induce these tumor-specific immune responses and thus supporting the use of combining cryoablation and immunotherapy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Akindele Olagunju
- Department of Diagnostic Imaging, Rhode Island Hospital, Women & Infants Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Tia Forsman
- Department of Diagnostic Imaging, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Robert C. Ward
- Department of Diagnostic Imaging, Rhode Island Hospital, Women & Infants Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|