1
|
Wang XY, Yan Y, Guo XR, Lu A, Jiang LX, Zhu YJ, Shi YJ, Liu XY, Wang JC. Enhanced Tumor Immunotherapy by Triple Amplification Effects of Nanomedicine on the STING Signaling Pathway in Dendritic Cells. Adv Healthc Mater 2024:e2403143. [PMID: 39440648 DOI: 10.1002/adhm.202403143] [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: 08/21/2024] [Revised: 10/02/2024] [Indexed: 10/25/2024]
Abstract
Insufficient activation of stimulator of interferon genes (STING) signaling pathway in tumor-associated dendritic cells limits the efficiency of tumor immunotherapy. Herein, the "three-in-one" IAHA-LaP/siPTPN6 NPs containing lanthanum ions (La3+), cGAMP, and PTPN6 siRNA are developed for triple amplification of the STING pathway. In vitro results demonstrate that La3+ significantly promotes cGAMP-mediated activation of the STING pathway by enhancing the phosphorylation of STING, TBK1, IRF3, and NF-κB p65. Moreover, the IAHA-LaP/siPTPN6 NPs further significantly enhance the phosphorylation of STING and NF-κB p65 and augment K63-linked ubiquitination of STING protein via siPTPN6-mediated downregulation of SHP-1 protein. Furthermore, NPs improve the secretion of IFNβ (2.4-fold), IL-6 (1.5-fold), and TNF-α (1.4-fold), thereby promoting DCs maturation compared to the mixture of La3+ and cGAMP. In vivo results show that the IAHA-LaP/siPTPN6 NPs remarkably inhibit primary tumor growth by increasing the percentage of mature DCs in tumor-draining lymph nodes, polarizing M2/M1 phenotype in TME, and promoting the infiltration of CD8+T cells into tumors. Moreover, these NPs dramatically prevent the growth of distal tumor by inducing systemic anti-tumor immunity and generating a long-term anti-tumor memory for protection against tumor recurrence in mice bearing bilateral B16F10. These IAHA-LaP/siPTPN6 NPs may offer a promising platform for robust anti-tumor immune responses.
Collapse
Affiliation(s)
- Xiang-Yu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiao-Ru Guo
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lin-Xia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yuan-Jun Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yu-Jie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiao-Yan Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Peking University Ningbo Institute of Marine Medicine, Ningbo, 315832, China
| |
Collapse
|
2
|
Takagi K, Sukhbaatar A, Inaba Y, Mori S, Kodama T. A combination of lymphatic drug delivery of anti-CTLA-4 antibody and local radiotherapy for solid-tumor treatment. Cancer Sci 2024. [PMID: 39380185 DOI: 10.1111/cas.16369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024] Open
Abstract
The combination of radiotherapy and immunotherapy is a promising approach that has been shown in clinical trials to improve significantly survival and response rates compared with monotherapy against solid tumor. Since anti-CTLA-4 antibodies block immunosuppressive signals mainly in the lymph nodes (LNs), efficient drug delivery to the lymphatic system is desirable. However, the immune checkpoint inhibitors, especially anti-CTLA-4 are currently administered intravenously (i.v.), resulting in limited efficacy in controlling solid tumor and inhibiting metastases, and the method of administration has not been optimized. Here, we show that a combination of local radiotherapy and administration of anti-CTLA-4 antibodies using a lymphatic drug delivery system (LDDS) suppresses solid tumor and metastases. We compared the efficacy of LDDS-based immunotherapy or radioimmunotherapy with i.v. administration in a solid-tumor model created by subcutaneous inoculation into LN-swollen mice with osteosarcoma cells. Tumor-bearing mice were divided into various groups (no treatment, immunotherapy [i.v. or LDDS], radiotherapy, and radioimmunotherapy [i.v. or LDDS]) and were observed for 28 days. Immunotherapy was administered with a cumulative dose of 10 mg/kg of anti-CTLA-4 monoclonal antibody, and radiotherapy was administered with a cumulative 8 Gy of fractionated X-ray irradiation. For immunotherapy alone, LDDS provided slight tumor growth inhibition but did not inhibit distant metastasis. For radioimmunotherapy, however, tumor growth was delayed and distant metastasis was suppressed compared with radiotherapy alone. In particular, the LDDS group achieved a high tumor-suppressive effect with T cell-mediated immune activity, indicating the efficacy of LDDS in radioimmunotherapy.
Collapse
Affiliation(s)
- Koki Takagi
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Ariunbuyan Sukhbaatar
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
- Division of Oral and Maxillofacial Oncology and Surgical Sciences, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Yohei Inaba
- Department of Radiological Technology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Department of Radiation Disaster Medicine, International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| | - Shiro Mori
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
- Division of Oral and Maxillofacial Oncology and Surgical Sciences, Graduate School of Dentistry, Tohoku University, Sendai, Miyagi, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| | - Tetsuya Kodama
- Laboratory of Biomedical Engineering for Cancer, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
- Biomedical Engineering Cancer Research Center, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
3
|
Liang J, Ma M, Feng W, Xu Q, Chen D, Lai J, Chen J. Anti-PD-L1 blockade facilitates antitumor effects of radiofrequency ablation by improving tumor immune microenvironment in hepatocellular carcinoma. Apoptosis 2024:10.1007/s10495-024-02019-3. [PMID: 39327353 DOI: 10.1007/s10495-024-02019-3] [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] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Hepatocellular carcinoma (HCC) is a complex disease with advanced presentation that significantly affects survival rates. Therefore, novel therapeutic strategies are needed. In this study, we investigate the tumor microenvironment (TME) in HCC by analyzing 13 HCC samples at single cell level. We identified key cell populations, including CD8 + T cells, Tregs, M1/M2 macrophages, and CD4 + memory T cells, and explored their roles and interactions. Our research revealed an early enrichment of CD8 + T cells, which could potentially lead to their exhaustion and facilitate tumor progression. We also investigated the impact of percutaneous radiofrequency ablation (RFA) on the immune microenvironment. Using a dual tumor mouse model, we demonstrated that RFA induces necrosis, enhancing antigen presentation and altering immune responses. Our results indicate that RFA increases PD-L1 expression in residual liver tissue, suggesting potential immune escape mechanisms. Furthermore, the combination of RFA and anti-PD-L1 therapy in the mouse model resulted in significant improvements in immune modulation. This included increased CD8 + T cell efficacy and decreased Treg infiltration. This combination shows promise as an approach to counteract HCC progression by altering the immune landscape. This study highlights the critical interaction within the TME of HCC and suggests the possibility of improving patient outcomes by targeting immune evasion mechanisms through combined therapeutic strategies.
Collapse
Affiliation(s)
- Jiahua Liang
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Medicine II, Medical Faculty Mannheim, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Mingjian Ma
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Feng
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiongcong Xu
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jiaming Lai
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Jiancong Chen
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
4
|
Li T, Sun S, Li Y, Zhang Y, Wei L. Immunotherapy revolutionizing brain metastatic cancer treatment: personalized strategies for transformative outcomes. Front Immunol 2024; 15:1418580. [PMID: 39136027 PMCID: PMC11317269 DOI: 10.3389/fimmu.2024.1418580] [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: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Brain metastatic cancer poses a significant clinical challenge, with limited treatment options and poor prognosis for patients. In recent years, immunotherapy has emerged as a promising strategy for addressing brain metastases, offering distinct advantages over conventional treatments. This review explores the evolving landscape of tumor immunotherapy in the context of brain metastatic cancer, focusing on the intricate interplay between the tumor microenvironment (TME) and immunotherapeutic approaches. By elucidating the complex interactions within the TME, including the role of immune cells, cytokines, and extracellular matrix components, this review highlights the potential of immunotherapy to reshape the treatment paradigm for brain metastases. Leveraging immune checkpoint inhibitors, cellular immunotherapies, and personalized treatment strategies, immunotherapy holds promise in overcoming the challenges posed by the blood-brain barrier and immunosuppressive microenvironment of brain metastases. Through a comprehensive analysis of current research findings and future directions, this review underscores the transformative impact of immunotherapy on the management of brain metastatic cancer, offering new insights and opportunities for personalized and precise therapeutic interventions.
Collapse
Affiliation(s)
- Ting Li
- Medical Oncology Department of Thoracic Cancer 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Shichen Sun
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yubing Li
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yanyu Zhang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Linlin Wei
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Rahnea-Nita RA, Rebegea LF, Toma RV, Mocanu H, Soare I, Mihailov R, Nechifor A, Guliciuc M, Constantin GB, Rahnea-Nita G. Immunotherapy Combined with Radiation in Malignant Melanoma without BRAF Mutations Brain Metastases-Favorable Response after Immunotherapy Continued beyond Progression. J Pers Med 2024; 14:86. [PMID: 38248787 PMCID: PMC10817469 DOI: 10.3390/jpm14010086] [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: 11/27/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
We present the case of a patient who was diagnosed in 2018 with nodular Malignant Melanoma (MM) without BRAF V 600 mutations stage 3 C (pT4b pN1a M0), and who underwent adjuvant citokines treatment with Interferon alpha 2b-48 weeks. Immunotherapy was initiated in January 2021 for lung and lymph node metastases. In June 2021, there was a partial response of the lung and lymph node metastases, but there was also progression to brain metastases. Immunotherapy was continued and Whole Brain Radiotherapy (WBRT) was performed. In September 2023, the imaging investigations revealed a favorable response, with no lesions suggestive of secondary determinations. The combination of Radiotherapy (RT) and Immunotherapy (IT) with Immune Checkpoint Inhibitors (ICI) has an abscopal effect. There is a coordinated action in the combination of RT and IT in order to obtain a common result, with the antitumor effect being greater than if RT or IT acted separately.
Collapse
Affiliation(s)
- Roxana-Andreea Rahnea-Nita
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania; (R.-A.R.-N.); (R.-V.T.)
- The Oncology-Palliative Care Department, “Sf. Luca” Chronic Disease Hospital, 041915 Bucharest, Romania;
| | - Laura-Florentina Rebegea
- The Clinical Department, The Faculty of Medicine and Pharmacy, “Dunarea de Jos” University in Galati, 800008 Galati, Romania; (L.-F.R.); (R.M.); (A.N.); (M.G.)
- The Radiotherapy Department, “Sf. Ap. Andrei” County Emergency Clinical Hospital, 800579 Galati, Romania
- The Research Center in the Field of Medical and Pharmaceutical Sciences, ReFORM-UDJ, 800010 Galati, Romania
| | - Radu-Valeriu Toma
- The Clinical Department, The Faculty of Medicine, The University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania; (R.-A.R.-N.); (R.-V.T.)
- The Radiotherapy Department, The Oncological Institute, 022328 Bucharest, Romania
| | - Horia Mocanu
- The Clinical Department, The Faculty of Medicine, “Titu Maiorescu” University, 040051 Bucharest, Romania; (H.M.); (I.S.)
- The E.N.T Department, Gaesti City Hospital, 135200 Gaesti, Romania
| | - Ioana Soare
- The Clinical Department, The Faculty of Medicine, “Titu Maiorescu” University, 040051 Bucharest, Romania; (H.M.); (I.S.)
| | - Raul Mihailov
- The Clinical Department, The Faculty of Medicine and Pharmacy, “Dunarea de Jos” University in Galati, 800008 Galati, Romania; (L.-F.R.); (R.M.); (A.N.); (M.G.)
| | - Alexandru Nechifor
- The Clinical Department, The Faculty of Medicine and Pharmacy, “Dunarea de Jos” University in Galati, 800008 Galati, Romania; (L.-F.R.); (R.M.); (A.N.); (M.G.)
| | - Mădălin Guliciuc
- The Clinical Department, The Faculty of Medicine and Pharmacy, “Dunarea de Jos” University in Galati, 800008 Galati, Romania; (L.-F.R.); (R.M.); (A.N.); (M.G.)
- The Urology Department, “Sf. Ap. Andrei” County Emergency Clinical Hospital, 800579 Galati, Romania
| | - Georgiana Bianca Constantin
- The Morphological and Functional Sciences Department, The Faculty of Medicine and Pharmacy, “Dunarea de Jos” University in Galati, 800008 Galati, Romania
| | - Gabriela Rahnea-Nita
- The Oncology-Palliative Care Department, “Sf. Luca” Chronic Disease Hospital, 041915 Bucharest, Romania;
- The Clinical Department, The Faculty of Midwifery and Nursing, The University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania
| |
Collapse
|
6
|
Botticella A, Dhermain F. Combination of radiosurgery and immunotherapy in brain metastases: balance between efficacy and toxicities. Curr Opin Neurol 2023; 36:587-591. [PMID: 37865858 DOI: 10.1097/wco.0000000000001217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
PURPOSE OF REVIEW The incidence of brain metastasis is high and still increasing. Among local therapies, stereotactic radiosurgery (SRS) is an effective treatment option, optimally sparing normal brain, even for multiple brain metastases. Immune checkpoint inhibitors (ICIs) become the new standard of care in an increasing number of cancers, and the combination SRS and ICI is often proposed to patients, but few data have been published on the efficacy and the toxicity of this association. RECENT FINDINGS Explaining this lack of consensus: retrospective studies with different primary cancers, various treatment lines and unknown levels of steroid exposure. Concerning the toxicity, the independent association of radionecrosis with brain-PTV volume was confirmed, and a decreased dose of SRS is now tested in a randomized study. Finally, a 'concurrent' delivery of SRS and ICI (within a 4 weeks' interval) seems the optimal schedule; fractionated radiosurgery for large brain metastasis should be favored. Radio-sensitizing nanoparticles and devices aiming to increase the permeability of the blood brain barrier should be considered in future combinations. SUMMARY The efficacy/toxicity balance of SRS-ICI combination should be regularly re-evaluated, anticipating continued progress in ICI and SRS delivery, with more long-survivors potentially exposed to long-term toxicities. Patients should be included in clinical trials and clearly informed to participate more closely in the final choice.
Collapse
Affiliation(s)
- Angela Botticella
- Department of Radiation Oncology, International Center for Thoracic Cancers (CICT), Gustave Roussy Cancer Campus, Villejuif, France
| | | |
Collapse
|