1
|
Santana JG, Shewarega A, Nam D, Duncan J, Madoff DC, Hyder F, Coman D, Chapiro J. Molecular MRI of T-cell immune response to cryoablation in immunologically hot vs. cold hepatocellular carcinoma. JHEP Rep 2025; 7:101294. [PMID: 40028344 PMCID: PMC11870164 DOI: 10.1016/j.jhepr.2024.101294] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025] Open
Abstract
Background & Aims Increasing enthusiasm around integrating locoregional therapy with systemic immunotherapy in primary liver cancer underscores the need for non-invasive imaging biomarkers. In this study, we aimed to establish advanced molecular MRI tools for monitoring T-cell responses to cryoablation in murine models, distinguishing between immunologically "hot" and "cold" hepatocellular carcinoma (HCC). Methods Immunocompetent 7-10-week-old C57BL/6J and BALB/cJ mice (n = 18 each) received carbon tetrachloride for 12 weeks to induce cirrhosis. Intrinsically immunogenic Hepa1-6 ("hot") and non-immunogenic TiB75 ("cold") cells were orthotopically implanted into C57BL/6 or BALB/c mice, respectively, to generate focal HCC lesions. After one week, animals were randomly assigned to (A) partial cryoablation (pCryo) (1.2 mm cryoprobe, -40 °C) or (B) no treatment (n = 8 per group and tumor type). Gadolinium 160 (160Gd)-labeled CD8+ antibody was administered intravenously either 1 week after tumor induction (control) or 1-week post (pCryo) (treatment). T1-weighted MRI scans were performed using a 9.4 T MRI scanner. Radiological-pathological correlation included imaging mass cytometry and immunohistochemistry. Results pCryo-treated Hepa1-6 tumors displayed peritumoral ring enhancement on T1-weighted MRI with 160Gd-CD8, correlating with imaging mass cytometry signal patterns. Untreated Hepa1-6 tumors lacked such enhancement. Radiological-pathological correlation confirmed significantly increased tumor-infiltrating CD8+ T lymphocytes in pCryo Hepa1-6 tumors compared with untreated tumors (p <0.001), and a stronger local response compared with systemic lymph nodes (p = 0.0415). Increased T-lymphocyte infiltration was not observed in TiB75 tumors, as indicated by MRI and histopathology. Conclusion pCryo induced increased T-cell infiltration in Hepa1-6 tumors compared to TiB75 tumors. T1-weighted MRI, following 160Gd-CD8 antibody administration, reproducibly detected the ablation-induced changes. These findings encourage further investigation of MRI-based molecular imaging biomarkers to assess immune responses to local tumor therapies. Impact and implications This study successfully established reliable MR-based molecular imaging tools to visualize CD8+ anti-tumor specific T-cell infiltration following partial cryoablation (pCryo) in murine tumor models. The study's significance lies in advancing our understanding of immune responses within induced cirrhosis and distinguishing between "hot" and "cold" tumor phenotypes. The findings not only build upon previous proof-of-principle data but also extend this technology to include different immune cell types in hepatocellular carcinoma. The study reveals that pCryo may exert specific effects on the tumor microenvironment, augmenting the anti-tumor immune response in immunogenic tumors while displaying a weaker local effect in non-immunogenic tumors.
Collapse
Affiliation(s)
- Jessica Gois Santana
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Annabella Shewarega
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
- Department of Radiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität, and Berlin Institute of Health, 10117 Berlin, Germany
| | - David Nam
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - James Duncan
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - David Craig Madoff
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
- Department of Medicine, Section of Medical Oncology, 333 Cedar Street, New Haven, CT 06510 United States
- Department of Surgery, Section of Surgical Oncology, 333 Cedar Street, New Haven, CT 06510, United States
| | - Fahmeed Hyder
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Daniel Coman
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Julius Chapiro
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
2
|
Fazlollahi F, Makary MS. Precision oncology: The role of minimally-invasive ablation therapy in the management of solid organ tumors. World J Radiol 2025; 17:98618. [PMID: 39876886 PMCID: PMC11755905 DOI: 10.4329/wjr.v17.i1.98618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/16/2024] [Accepted: 01/18/2025] [Indexed: 01/21/2025] Open
Abstract
Solid organ tumors present a significant healthcare challenge, both economically and logistically, due to their high incidence and treatment complexity. In 2023, out of the 1.9 million new cancer cases in the United States, over 73% were solid organ tumors. Ablative therapies offer minimally invasive solutions for malignant tissue destruction in situ, often with reduced cost and morbidity compared to surgical resection. This review examines the current Food and Drug Administration-approved locoregional ablative therapies (radiofrequency, microwave, cryogenic, high-intensity focused ultrasound, histotripsy) and their evolving role in cancer care. Data were collected through a comprehensive survey of the PubMed-indexed literature on tumor ablation techniques, their clinical indications, and outcomes. Over time, emerging clinical data will help establish these therapies as the standard of care in solid organ tumor treatment, supported by improved long-term outcomes and progression-free survival.
Collapse
Affiliation(s)
- Farbod Fazlollahi
- College of Medicine, The Ohio State University, Columbus, OH 43210, United States
| | - Mina S Makary
- Department of Radiology, The Ohio State University Medical Center, Columbus, OH 43210, United States
| |
Collapse
|
3
|
Bodard S, Geevarghese R, Razakamanantsoa L, Frandon J, Petre EN, Marcelin C, Cornelis FH. Percutaneous cryoablation in soft tissue tumor management: an educational review. Insights Imaging 2024; 15:278. [PMID: 39556172 PMCID: PMC11573955 DOI: 10.1186/s13244-024-01822-5] [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: 04/16/2024] [Accepted: 09/06/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND Percutaneous cryoablation (PCA), having shown effectiveness in treating liver, lung, prostate, breast, and kidney tumors, is now gaining attention for the treatment of soft tissue tumors. PCA functions by freezing tissue, which induces ice crystal formation and cell death without damaging collagen structures. Technical considerations include the selection and handling of cryoprobes and cryogenic agents, procedural duration, and choice of image guidance for precision. This review aims to synthesize the mechanisms, applications, and technical aspects of PCA in the treatment of soft tissue tumors. METHODS Adhering to PRISMA 2020 guidelines, a review was conducted of studies published prior to March 2024 that investigated PCA of soft tissue tumors. The review focused on technical and procedural aspects of cryoablation, cryobiological principles, cellular and tissue responses to extreme cold, intra- and post-procedure physiological mechanisms during and post-procedure, and main clinical applications. RESULTS PCA is efficient in treating soft tissue tumors, including desmoid tumors, vascular malformations, and abdominal wall endometriosis. Several cryobiological mechanisms are involved, notably ice crystal formation, cellular dehydration, osmotic effects, and the inflammatory response, all of which contribute to its efficacy. Key technical aspects include the choice of cryoprobes, cryogenic agents (argon gas or liquid nitrogen), and the duration and control of freezing/thawing cycles. PCA also frequently outperformed traditional treatments like surgery and radiotherapy in terms of pain reduction, tumor size reduction, and patient outcomes. Moreover, its nerve sideration properties make it effective under local anesthesia. CONCLUSION Demonstrating substantial pain reduction, tumor size decrease, and high technical success rates, PCA offers a promising and minimally invasive alternative for soft tissue tumor treatment. CRITICAL RELEVANCE STATEMENT Percutaneous cryoablation provides a minimally invasive, precise alternative for soft tissue tumor management, advancing clinical radiology by offering effective treatment with reduced patient risk and enhanced outcomes through image-guided procedures. KEY POINTS Percutaneous cryoablation (PCA) offers a promising, minimally invasive alternative for managing soft tissue tumors. PCA employs image-guided techniques to accurately target and treat tumors, ensuring high precision and control. PCA preserves structures like collagen, reduces pain, decreases tumor size, and generally enhances patient outcomes.
Collapse
Affiliation(s)
- Sylvain Bodard
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
- Department of Radiology, Necker Hospital, University of Paris Cité, 149 rue de Sèvre, 75015, Paris, France.
- Laboratoire d'Imagerie Biomédicale, Sorbonne University, CNRS UMR 7371, INSERM U 1146, 75006, Paris, France.
| | - Ruben Geevarghese
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Leo Razakamanantsoa
- Department of Interventional Radiology and Oncology, Sorbonne University, Tenon Hospital, 4 rue de la Chine, 75020, Paris, France
| | - Julien Frandon
- Radiology Department, Nimes University Hospital, Nimes, France
| | - Elena N Petre
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Clement Marcelin
- Department of Radiology, Centre Hospitalo-Universitaire de Bordeaux, 33076, Bordeaux, France
| | - François H Cornelis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
- Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.
- Department of Interventional Radiology and Oncology, Sorbonne University, Tenon Hospital, 4 rue de la Chine, 75020, Paris, France.
| |
Collapse
|
4
|
Li ZH, Ma YY, Niu LZ, Xu KC. Cryoablation for intrapulmonary bronchial cyst: A case report. World J Radiol 2024; 16:616-620. [PMID: 39494135 PMCID: PMC11525826 DOI: 10.4329/wjr.v16.i10.616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Bronchial cysts are congenital malformations usually located in the mediastinum, and intrapulmonary localization is very rare. Cryoablation is a novel therapeutic approach that promotes tumor necrosis and stimulates anti-tumor immune responses. CASE SUMMARY This article reports a case of a 68-year-old male patient who was diagnosed with an intrapulmonary bronchogenic cyst by computed tomography examination and pathology, and the patient subsequently underwent cryoablation therapy and achieved complete response with after 3 months of follow-up. CONCLUSION Intrapulmonary bronchogenic cysts are very rare, cryoablation therapy is feasible, safe, and effective for intrapulmonary bronchial cysts.
Collapse
Affiliation(s)
- Zhong-Hai Li
- Department of Radiology, Guangzhou Fuda Cancer Hospital, Guangzhou 510665, Guangdong Province, China
| | - Yang-Yang Ma
- Central Laboratory, Guangzhou Fuda Cancer Hospital, Guangzhou 510665, Guangdong Province, China
| | - Li-Zhi Niu
- Department of Oncology, Guangzhou Fuda Cancer Hospital, Guangzhou 510665, Guangdong Province, China
| | - Ke-Cheng Xu
- Department of Oncology, Guangzhou Fuda Cancer Hospital, Guangzhou 510665, Guangdong Province, China
| |
Collapse
|
5
|
Zecca F. Editorial Comment: Percutaneous Cryoablation Can Safely Control Foci of Treatment-Refractory Soft-Tissue Sarcoma. AJR Am J Roentgenol 2024; 223:e2431841. [PMID: 39109681 DOI: 10.2214/ajr.24.31841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Affiliation(s)
- Fabio Zecca
- University Hospital of Cagliari, Cagliari, Italy
| |
Collapse
|
6
|
Liu Q, Chen X, Qi M, Li Y, Chen W, Zhang C, Wang J, Han Z, Zhang C. Combined cryoablation and PD-1 inhibitor synergistically enhance antitumor immune responses in Lewis lung adenocarcinoma mice via the PI3K/AKT/mTOR pathway. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167262. [PMID: 38815768 DOI: 10.1016/j.bbadis.2024.167262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024]
Abstract
Cryoablation is a therapeutic modality for lung adenocarcinoma that destroys target tumors using lethal levels of cold, resulting in the release of large amounts of specific antigens that activate immune responses. However, tumor immune checkpoint escape mechanisms prevent these released self-antigens from inducing effective anti-tumor immune responses. To overcome this challenge, we propose the use of immune checkpoint inhibitors to relieve T cell inhibition by immune checkpoints and enhance the anti-tumor immune response mediated by cryoablation. We used bilateral tumor-bearing mouse models and a specific cryoablation instrument to study the efficacy of cryoablation combined with PD-1 inhibitors in Lewis lung adenocarcinoma model mice. We found that cryoablation combined with PD-1 inhibitors significantly inhibited the growth of mouse lung adenocarcinoma, prolonged mouse survival, and enhanced the anti-tumor immune response. Moreover, this combined regimen could synergistically promote the activation and proliferation of T cells via the PI3K/AKT/mTOR pathway. The present study provides a strong theoretical basis for the clinical combination of cryoablation and PD-1 inhibitors.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100091, China
| | - Man Qi
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100091, China; Beijing Key Laboratory of OTIR, Beijing 100091, China
| | - Yongqun Li
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100091, China
| | - Wei Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100091, China
| | - Caiyun Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Jiaxin Wang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100091, China; Beijing Key Laboratory of OTIR, Beijing 100091, China; Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei 230032, Anhui Province, China.
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese PLA General Hospital, Beijing 100048, China; College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing 100091, China.
| |
Collapse
|
7
|
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
|
8
|
Pan H, Tian Y, Pei S, Yang W, Zhang Y, Gu Z, Zhu H, Zou N, Zhang J, Jiang L, Hu Y, Shen S, Wang K, Jin H, Li Z, Zhang Y, Xiao Y, Luo Q, Wang H, Huang J. Combination of percutaneous thermal ablation and adoptive Th9 cell transfer therapy against non-small cell lung cancer. Exp Hematol Oncol 2024; 13:52. [PMID: 38760861 PMCID: PMC11100251 DOI: 10.1186/s40164-024-00520-8] [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: 10/29/2023] [Accepted: 05/07/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is one of the predominant malignancies globally. Percutaneous thermal ablation (PTA) has gained widespread use among NSCLC patients, with the potential to elicit immune responses but limited therapeutic efficacies for advanced-stage disease. T-helper type 9 (Th9) cells are a subset of CD4+ effector T cells with robust and persistent anti-tumor effects. This study proposes to develop PTA-Th9 cell integrated therapy as a potential strategy for NSCLC treatment. METHODS The therapeutic efficacies were measured in mice models with subcutaneously transplanted, recurrence, or lung metastatic tumors. The tumor microenvironments (TMEs) were evaluated by flow cytometry. The cytokine levels were assessed by ELISA. The signaling molecules were determined by quantitative PCR and Western blotting. The translational potential was tested in the humanized NSCLC patient-derived xenograft (PDX) model. RESULTS We find that PTA combined with adoptive Th9 cell transfer therapy substantially suppresses tumor growth, recurrence, and lung metastasis, ultimately extending the survival of mice with NSCLC grafts, outperforming both PTA and Th9 cell transfer monotherapy. Analysis of TMEs indicates that combinatorial therapy significantly augments tumor-infiltrating Th9 cells, boosts anti-tumor effects of CD8+ T cells, and remodels tumor immunosuppressive microenvironments. Moreover, combinatorial therapy significantly strengthens the regional and circulation immune response of CD8+ T cells in mice with tumor lung metastasis and induces peripheral CD8+ T effector memory cells in mice with tumor recurrence. Mechanically, PTA reinforces the anti-tumor ability of Th9 cells primarily through upregulating interleukin (IL)-1β and subsequently activating the downstream STAT1/IRF1 pathway, which could be effectively blocked by intercepting IL-1β signaling. Finally, the enhanced therapeutic effect of combinatorial therapy is validated in humanized NSCLC PDX models. CONCLUSIONS Collectively, this study demonstrates that combinatorial therapy displays robust and durable anti-tumor efficacy and excellent translational potential, offering excellent prospects for translation and emerging as a promising approach for NSCLC treatment.
Collapse
Affiliation(s)
- Hanbo Pan
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yu Tian
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Siyu Pei
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200030, China
| | - Wanlin Yang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200030, China
| | - Yanyang Zhang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zenan Gu
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Hongda Zhu
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ningyuan Zou
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Jiaqi Zhang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Long Jiang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yingjie Hu
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Shengping Shen
- Department of Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Kai Wang
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Haizhen Jin
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ziming Li
- Department of Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yanyun Zhang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200030, China
| | - Yichuan Xiao
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200030, China.
| | - Qingquan Luo
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Hui Wang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Jia Huang
- Department of Thoracic Surgical Oncology, Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
9
|
Liu H, Wang C, Wang R, Cao H, Cao Y, Huang T, Lu Z, Xiao H, Hu M, Wang H, Zhao J. New insights into mechanisms and interventions of locoregional therapies for hepatocellular carcinoma. Chin J Cancer Res 2024; 36:167-194. [PMID: 38751435 PMCID: PMC11090796 DOI: 10.21147/j.issn.1000-9604.2024.02.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/07/2024] [Indexed: 05/18/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is responsible for a significant number of cancer-related deaths worldwide and its incidence is increasing. Locoregional treatments, which are precision procedures guided by imaging to specifically target liver tumors, play a critical role in the management of a substantial portion of HCC cases. These therapies have become an essential element of the HCC treatment landscape, with transarterial chemoembolization (TACE) being the treatment of choice for patients with intermediate to advanced stages of the disease. Other locoregional therapies, like radiofrequency ablation, are highly effective for small, early-stage HCC. Nevertheless, the advent of targeted immunotherapy has challenged these established treatments. Tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) have shown remarkable efficacy in clinical settings. However, their specific uses and the development of resistance in subsequent treatments have led clinicians to reevaluate the future direction of HCC therapy. This review concentrates on the distinct features of both systemic and novel locoregional therapies. We investigate their effects on the tumor microenvironment at the molecular level and discuss how targeted immunotherapy can be effectively integrated with locoregional therapies. We also examine research findings from retrospective studies and randomized controlled trials on various combined treatment regimens, assessing their validity to determine the future evolution of locoregional therapies within the framework of personalized, comprehensive treatment.
Collapse
Affiliation(s)
- Hanyuan Liu
- Department of General surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210019, China
| | - Chunmei Wang
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Ruiqiang Wang
- School of Public Health, China Medical University, Shenyang 110122, China
| | - Hengsong Cao
- Department of General surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210019, China
| | - Yongfang Cao
- Department of General surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210019, China
| | - Tian Huang
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing 210024, China
| | - Zhengqing Lu
- Hepatobiliary/Liver Transplantation Center, the First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Transplantation, Chinese Academy of Medical Sciences, Nanjing 210024, China
| | - Hua Xiao
- Department of General surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210019, China
| | - Mengcheng Hu
- Department of Gastroenterology, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing 211103, China
| | - Hanjin Wang
- Department of General surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210019, China
| | - Jun Zhao
- Department of Nuclear Medicine, the Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou 213001, China
| |
Collapse
|
10
|
Cao Q, Zhu J, Wu X, Li J, Chen Y, You Y, Li X, Huang X, Zhang Y, Li R, Han D. Efficacy and Safety Assessment of Intrathoracic Perfusion Chemotherapy Combined with immunological factor Interleukin-2 in the Treatment of Advanced Non-Small Cell Lung Cancer: A Retrospective Cohort Study. J Cancer 2024; 15:2024-2032. [PMID: 38434976 PMCID: PMC10905414 DOI: 10.7150/jca.92624] [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: 11/26/2023] [Accepted: 01/12/2024] [Indexed: 03/05/2024] Open
Abstract
Objective: This study evaluated the efficacy and safety of the gemcitabine and oxaliplatin intrathoracic perfusion chemotherapy (IPCGOR) regimen combined with interleukin-2 (IL-2) for advanced non-small cell lung cancer (NSCLC). Methods: We conducted a retrospective analysis of 460 advanced NSCLC patients from the Yunnan Province Early Cancer Diagnosis and Treatment Project (June 2020-October 2022), assessing the IPCGOR and IL-2 combination. Outcomes were measured based on RECIST 1.1 criteria, focusing on objective response rate (ORR), disease control rate (DCR), median progression-free survival (mPFS), median overall survival (MOS), and treatment safety. Results: The treatment demonstrated an ORR of 67.4%, a DCR of 97.4%, an mPFS of 8.5 months, and an MOS of 12.5 months. 14 patients underwent successful surgery post-treatment. Common adverse reactions were manageable, with no treatment-related deaths reported. Conclusion: The IPCGOR combined with IL-2 regimen shows promising efficacy and a tolerable safety profile for advanced NSCLC. These findings suggest its potential as a reference for treating advanced NSCLC. However, the study's retrospective nature and single-center design pose limitations. Future research should focus on prospective studies, randomized controlled trials, and long-term outcome assessments, particularly in diverse patient subgroups, to further validate and refine the clinical application of this regimen.
Collapse
Affiliation(s)
- Qiang Cao
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- School of Medicine, Macau University of Science and Technology, 999078, Macau, Macao
- Department of Earth Sciences, Kunming University of Science and Technology, 650093, Kunming, China
| | - Jinyi Zhu
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xinyan Wu
- Department of Earth Sciences, Kunming University of Science and Technology, 650093, Kunming, China
- College of Veterinary Medicine, Sichuan Agricultural University, 610000, Chengdu, China
| | - Jiapeng Li
- Undergraduate Department, University of Toronto, M2J4A6, Toronto, Canada
| | - Yuquan Chen
- Institute of Medical Information/Library, Chinese Academy of Medical Sciences, 100020 Beijing, China
| | - Yanwei You
- Division of Sports Science & Physical Education, Tsinghua University, Beijing 100084, China
| | - Xiaochen Li
- Department of Earth Sciences, Kunming University of Science and Technology, 650093, Kunming, China
- The Third Affiliated Hospital of Shandong First Medical University, Jinan, 250000, Shandong, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Yujie Zhang
- College of Agriculture, Henan University of Science and Technology, 471023, Luoyang, China
| | - Rizhu Li
- Department of Cardiothoracic Surgery, the Affiliated Hospital of Youjiang Medical University for Nationalities, 18 zhongshan 2nd Road, Baise, Guangxi Province, China
| | - Dan Han
- Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
11
|
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
|
12
|
Azuma R, Sato K, Sunadoi H, Ishii Y, Tomaru U, Motohashi M. Primary cardiac myxofibrosarcoma of the left atrium and pericardium: a case report. J Cardiothorac Surg 2023; 18:334. [PMID: 37974190 PMCID: PMC10655424 DOI: 10.1186/s13019-023-02441-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 11/04/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Primary cardiac myxofibrosarcoma is rare and commonly occurs in the left atrium. Myxofibrosarcoma is aggressive and has a high mortality rate due to its high rate of recurrence. Complete surgical resection is considered important; however, effective treatment options have not been established. CASE PRESENTATION We report the case of a 75-year-old woman who developed a myxofibrosarcoma spreading to the left atrium and pericardium. We performed surgical resection of the tumor to prevent sudden death due to mitral valve obstruction or cerebral infarction due to embolism of the scattered mass. However, we were unable to complete the resection of the tumors. The patient developed brain metastasis 2 months after surgery and eventually died due to brain hemorrhage 3 months after surgery. CONCLUSIONS In this report, we described a rare case of primary cardiac myxofibrosarcoma located not only in the left atrium but also in the pericardium. Considering preoperative laboratory findings, surgical and adjuvant therapy, and the patient's wishes are important for the best therapeutic course for an individual.
Collapse
Affiliation(s)
- Ryota Azuma
- Department of Cardiovascular Surgery, Hakodate Central General Hospital, 33-2 Honcho, Hakodate, Hokkaido, 040-8585, Japan.
| | - Kazuyoshi Sato
- Department of Cardiovascular Surgery, Hakodate Central General Hospital, 33-2 Honcho, Hakodate, Hokkaido, 040-8585, Japan
| | - Hiroki Sunadoi
- Department of Cardiovascular Surgery, Hakodate Central General Hospital, 33-2 Honcho, Hakodate, Hokkaido, 040-8585, Japan
| | - Yasushi Ishii
- Department of Pathology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Utano Tomaru
- Department of Pathology, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Masatoshi Motohashi
- Department of Cardiovascular Surgery, Hakodate Central General Hospital, 33-2 Honcho, Hakodate, Hokkaido, 040-8585, Japan
| |
Collapse
|
13
|
Xiong H, Han X, Cai L, Zheng H. Natural polysaccharides exert anti-tumor effects as dendritic cell immune enhancers. Front Oncol 2023; 13:1274048. [PMID: 37876967 PMCID: PMC10593453 DOI: 10.3389/fonc.2023.1274048] [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: 08/07/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
With the development of immunotherapy, the process of tumor treatment is also moving forward. Polysaccharides are biological response modifiers widely found in plants, animals, fungi, and algae and are mainly composed of monosaccharides covalently linked by glycosidic bonds. For a long time, polysaccharides have been widely used clinically to enhance the body's immunity. However, their mechanisms of action in tumor immunotherapy have not been thoroughly explored. Dendritic cells (DCs) are a heterogeneous population of antigen presenting cells (APCs) that play a crucial role in the regulation and maintenance of the immune response. There is growing evidence that polysaccharides can enhance the essential functions of DCs to intervene the immune response. This paper describes the research progress on the anti-tumor immune effects of natural polysaccharides on DCs. These studies show that polysaccharides can act on pattern recognition receptors (PRRs) on the surface of DCs and activate phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), mitogen-activated protein kinase (MAPK), nuclear factor-κB (NF-κB), Dectin-1/Syk, and other signalling pathways, thereby promoting the main functions of DCs such as maturation, metabolism, antigen uptake and presentation, and activation of T cells, and then play an anti-tumor role. In addition, the application of polysaccharides as adjuvants for DC vaccines, in combination with adoptive immunotherapy and immune checkpoint inhibitors (ICIs), as well as their co-assembly with nanoparticles (NPs) into nano drug delivery systems is also introduced. These results reveal the biological effects of polysaccharides, provide a new perspective for the anti-tumor immunopharmacological research of natural polysaccharides, and provide helpful information for guiding polysaccharides as complementary medicines in cancer immunotherapy.
Collapse
Affiliation(s)
- Hongtai Xiong
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinpu Han
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liu Cai
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Honggang Zheng
- Department of Oncology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
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
|
Grazioso TP, Djouder N. The forgotten art of cold therapeutic properties in cancer: A comprehensive historical guide. iScience 2023; 26:107010. [PMID: 37332670 PMCID: PMC10275721 DOI: 10.1016/j.isci.2023.107010] [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] [Indexed: 06/20/2023] Open
Abstract
Cold therapy has been used for centuries, from Julius Caesar to Mohandas Gandhi, as a potent therapeutic approach. However, it has been largely forgotten in modern medicine. This review explores the history of cold therapy and its potential application as a therapeutic strategy against various diseases, including cancer. We examine the different techniques of cold exposure and the use of other therapeutical approaches, such as cryoablation, cryotherapy, cryoimmunotherapy, cryothalectomy, and delivery of cryogen agents. While clinical trials using cold therapy for cancer treatment are still limited, recent research shows promising results in experimental animal cancer models. This area of research is becoming increasingly significant and warrants further investigation.
Collapse
Affiliation(s)
- Tatiana P. Grazioso
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, ES-28029 Madrid, Spain
- Gynecological, Genitourinary and Skin Cancer Unit HM, Clara Campal Comprehensive Cancer Center, CIOCC, Department of Basic Medical Sciences, Hospital Universitario HM Sanchinarro, ES-28050 Madrid, Spain
- Institute of Applied Molecular Medicine, IMMA, Facultad de Medicina, Universidad San Pablo CEU, ES-28668 Madrid, Spain
| | - Nabil Djouder
- Molecular Oncology Programme, Growth Factors, Nutrients and Cancer Group, Centro Nacional de Investigaciones Oncológicas, CNIO, ES-28029 Madrid, Spain
| |
Collapse
|