1
|
Amosu MM, Jankowski AM, McCright JC, Yang BE, Grano de Oro Fernandez J, Moore KA, Gadde HS, Donthi M, Kaluzienski ML, Maisel K. Plasmacytoid Dendritic Cells Mediate CpG-ODN-induced Increase in Survival in a Mouse Model of Lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2024; 71:519-533. [PMID: 38990702 PMCID: PMC11568470 DOI: 10.1165/rcmb.2023-0410oc] [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: 11/21/2023] [Accepted: 07/11/2024] [Indexed: 07/13/2024] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a devastating disease primarily found in women of reproductive age that leads to cystic destruction of the lungs. Recent work has shown that LAM causes immunosuppression and that checkpoint inhibitors can be used as LAM treatment. Toll-like receptor (TLR) agonists can also reactivate immunity, and the TLR9 agonist CpG oligodeoxynucleotide (CpG-ODN) has been effective in treating lung cancer in animal models. In this study, we investigated the use of TLR9 agonist CpG-ODN as LAM immunotherapy in combination with checkpoint inhibitor anti-PD1 and standard of care rapamycin, and determined the immune mechanisms underlying therapeutic efficacy. We used survival studies, flow cytometry, ELISA, and histology to assess immune response and survival after intranasal treatment with CpG-ODN in combination with rapamycin or anti-PD1 therapy in a mouse model of metastatic LAM. We found that local administration of CpG-ODN enhances survival in a mouse model of LAM. We found that a lower dose led to longer survival, likely because of fewer local side effects, but increased LAM nodule count and size compared with the higher dose. CpG-ODN treatment also reduced regulatory T cells and increased the number of T-helper type 17 cells as well as cytotoxic T cells. These effects appear to be mediated in part by plasmacytoid dendritic cells because depletion of plasmacytoid dendritic cells reduces survival and abrogates T-helper type 17 cell response. Finally, we found that CpG-ODN treatment is effective in early-stage and progressive disease and is additive with anti-PD1 therapy and rapamycin. In summary, we have found that TLR9 agonist CpG-ODN can be used as LAM immunotherapy and effectively synergizes with rapamycin and anti-PD1 therapy in LAM.
Collapse
Affiliation(s)
- Mayowa M Amosu
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Ashleigh M Jankowski
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Jacob C McCright
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Bennett E Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | | | - Kaitlyn A Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Havish S Gadde
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Mehul Donthi
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Michele L Kaluzienski
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| | - Katharina Maisel
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland
| |
Collapse
|
2
|
Zang X, Li G, Zhu J, Dong X, Zhai Y. Evaluation of the adjuvant effect of imiquimod and CpG ODN 1826 in chimeric DNA vaccine against Japanese encephalitis. Int Immunopharmacol 2024; 140:112816. [PMID: 39083930 DOI: 10.1016/j.intimp.2024.112816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/15/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Vaccines represent a significant milestone in the history of human medical science and serve as the primary means for controlling infectious diseases. In recent years, the geographical distribution of Japanese encephalitis viruses (JEV) of various genotypes has become increasingly complex, which provides a rationale for the development of safer and more effective vaccines. The advent of subunit and nucleic acid vaccines, especially propelled by advancements in genetic engineering since the 1980s, has accelerated the application of novel adjuvants. These novel vaccine adjuvants have diversified into toll-like receptor (TLR) agonists, complex adjuvants, nanoparticles and so on. However, the efficacy of adjuvant combinations can vary depending on the host system, disease model, or vaccine formulation, sometimes resulting in competitive or counteractive effects. In our previous study, we constructed a pJME-LC3 chimeric DNA vaccine aimed at inducing an immune response through autophagy induction. Building on this, we investigated the impact of the TLR7/8 agonist imiquimod (IMQ) and the TLR9 agonist CpG ODN 1826 as adjuvants on the immunogenicity of the Japanese encephalitis chimeric DNA vaccine. Our findings indicate that the combination of the pJME-LC3 vaccine with IMQ and CpG ODN 1826 adjuvants enhanced the innate immune response, promoting the maturation and activation of antigen-presenting cells in the early immune response. Furthermore, it played a regulatory and optimizing role in subsequent antigen-specific immune responses, resulting in effective cellular and humoral immunity and providing prolonged immune protection. The synergistic effect of IMQ and CpG ODN 1826 as adjuvants offers a novel approach for the development of Japanese encephalitis nucleic acid vaccines.
Collapse
Affiliation(s)
- Xin Zang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Guozhen Li
- Department of Gastroenterology, Wuhan Red Cross Hospital, Wuhan 430015, China
| | - Junyao Zhu
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaoying Dong
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yongzhen Zhai
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
3
|
Grindel AL, Fretellier N, Soares M, Bouzakher N, Millot Maysounabe V, Santus R, Bawa O, Wintrebert M, Couquelet C, Robert P, Emile JF, Capron C. Antitumoral effect of local injection of TLR-9 agonist emulsified in Lipiodol with systemic anti-PD-1 in a murine model of colorectal carcinoma. Front Immunol 2024; 14:1272246. [PMID: 38292484 PMCID: PMC10825566 DOI: 10.3389/fimmu.2023.1272246] [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: 08/03/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Local treatments of cancer, including transarterial chemoembolization, could enhance responses to systemic immune checkpoint inhibitors such as anti-PD-1 antibodies. Lipiodol, a radiopaque oil, is widely used for transarterial chemoembolization as a tumor-targeting drug carrier and could be used in emulsion with immunomodulators. This study aimed at evaluating the antitumoral effect of intra-tumoral injection of Lipiodol-immunomodulator emulsions combined with systemic anti-PD-1 therapy in a murine model of colorectal carcinoma. Method Mice (male BALB/c) with anti-PD-1-resistant subcutaneous CT26 tumors were injected with immunomodulators, emulsified or not with Lipiodol (N=10-12/group). Results The TLR-9 agonist CpG displayed antitumor effects, while Poly I:C and QS21 did not. The Lipiodol-CpG emulsion appeared to be stable and maintained CpG within tumors for a longer time. Repeated intra-tumoral injections, combined with anti-PD-1, induced responses towards the tumor as well as to a distant metastatic-like nodule. This treatment was associated with an increase in proliferative CD8+ T cells and of IFN-γ expression, a decrease in proliferative regulatory T cells but also, surprisingly, an increase in myeloid derived suppressor cells. Conclusions Local administration of CpG emulsified with Lipiodol led to an effective antitumoral effect when combined to systemic anti-PD-1 therapy. Lipiodol, apart from its radiopaque properties, is an efficient drug-delivery system. The formulated oil-in-water emulsion allows efficient loading and control release of CpG, which induces favorable immune modifications in this murine tumor model.
Collapse
Affiliation(s)
- Anne-Laure Grindel
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
| | | | - Miguel Soares
- Guerbet, Research and Innovation Division, Aulnay-sous-Bois, France
| | - Nabiha Bouzakher
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
| | | | - Robin Santus
- Guerbet, Research and Innovation Division, Aulnay-sous-Bois, France
| | - Olivia Bawa
- INSERM US23 Analyse Moléculaire, Modélisation et Imagerie de la Maladie Cancéreuse (AMMICA), Villejuif, France
| | | | | | - Philippe Robert
- Guerbet, Research and Innovation Division, Aulnay-sous-Bois, France
| | - Jean-Francois Emile
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
- Département d’anatomie Pathologique et de Cytologie, Hôpital Ambroise-Paré, Boulogne-Billancourt, France
| | - Claude Capron
- EA4340 Biomarqueurs en oncologie et onco-hématologie (BECCOH), Université Paris Saclay, Versailles, France
- Immunology and Hematology Department, Hôpital Ambroise Paré, Boulogne-Billancourt, France
| |
Collapse
|
4
|
Sun M, Liu Z, Wu L, Yang J, Ren J, Qu X. Bioorthogonal-Activated In Situ Vaccine Mediated by a COF-Based Catalytic Platform for Potent Cancer Immunotherapy. J Am Chem Soc 2023; 145:5330-5341. [PMID: 36815731 DOI: 10.1021/jacs.2c13010] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Personalized tumor vaccines have become a promising modality for cancer immunotherapy. However, in situ personalized tumor vaccines generated from immunogenic cancer cell death (ICD) and adjuvants are mired by toxic side effects and unsatisfactory efficiency. Herein, by functionalizing the reticular structure to optimize the catalytic activity of the materials, a series of biocompatible covalent organic framework (COF)-based catalysts have been designed and screened for establishing a bioorthogonal-activated in situ cancer vaccine in an efficient and safe way. Especially, pro-doxorubicin (pro-DOX) could be bioorthogonally activated in situ by the COF-based Fe(II) catalysts, which elicited ICD and released tumor-associated antigens (TAAs). This in situ prodrug activation strategy could minimize drug side effects and maximize treatment effects. More importantly, the system could also catalytically activate pro-imiquimod (pro-IMQ, a TLR7/8 immune agonist), which served as an adjuvant to amplify the antitumor immunity. Notably, this bioorthogonal-activated in situ cancer vaccine not only facilitated a strong antitumor immune response but also prevented the dose-dependent side effects of chemotherapeutic drugs, including systemic inflammation caused by the random distribution of adjuvants. To the best of our knowledge, it is the first time to devise an efficient catalytic platform for generating an in situ bioorthogonal-activated cancer vaccine, which would provide a paradigm for achieving secure and robust immunotherapy.
Collapse
Affiliation(s)
- Mengyu Sun
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Zhengwei Liu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China
| | - Li Wu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong 226019, Jiangsu, P. R. China
| | - Jie Yang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, Jilin, P. R. China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| |
Collapse
|
5
|
Shan Y, Zhang B, Chen L, Zhang H, Jiang C, You Q, Li Y, Han H, Zhu J. Herpesvirus entry mediator regulates the transduction of Tregs via STAT5/Foxp3 signaling pathway in ovarian cancer cells. Anticancer Drugs 2023; 34:73-80. [PMID: 35946515 DOI: 10.1097/cad.0000000000001336] [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: 11/25/2022]
Abstract
The ratio of regulatory T cells (Treg) in peripheral blood of cancer patients has a closely correlation to the occurrence and development of ovarian cancer. In this study, our aim to explore the expression of herpesvirus entry mediator (HVEM) in ovarian cancer and its correlation with Tregs. The expression of HVEM in peripheral blood of ovarian cancer patients was detected by ELISA, and the ratio of CD4+ CD25 + Foxp3 positive Tregs cells was detected by flow cytometry. Ovarian cancer cell lines with high- and low-HVEM expression were constructed. CD4+ cells were co-cultured with ovarian cancer (OC) cells, and the expressions of IL-2 and TGF-β1 in the supernatant of cells were detected by ELISA, and western blot was used to detect the expressions of STAT5, p-STAT5, and Foxp3. The results indicated that the number of Treg cells in the peripheral blood of OC patients increased, and the expression of HVEM increased, the two have a certain correlation. At the same time, the overexpression of HVEM promoted the expression of cytokines IL-2 and TGF- β1, promoted the activation of STAT5 and the expression of Foxp3, leading to an increase in the positive rate of Treg, while the HVEM gene silence group was just the opposite. Our results showed that the expression of HVEM in OC cells has a positive regulation effect on Tregs through the STAT5/Foxp3 signaling pathway. To provide experimental basis and related mechanism for the clinical treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ying Shan
- Department of Obstetrics and Gynecology
| | | | - Li Chen
- Department of Obstetrics and Gynecology
| | - Hu Zhang
- Department of Obstetrics and Gynecology
| | - Cui Jiang
- Department of Obstetrics and Gynecology
| | - Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yanyi Li
- Department of Obstetrics and Gynecology
- Department of Health Science, Graduate School of Medical, Osaka University, Osaka, Japan
| | | | | |
Collapse
|
6
|
Helicobacter pylori promotes gastric cancer progression through the tumor microenvironment. Appl Microbiol Biotechnol 2022; 106:4375-4385. [PMID: 35723694 DOI: 10.1007/s00253-022-12011-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 02/05/2023]
Abstract
Gastric cancer (GC) is a leading type of cancer. Although immunotherapy has yielded important recent progress in the treatment of GC, the prognosis remains poor due to drug resistance and frequent recurrence and metastasis. There are multiple known risk factors for GC, and infection with Helicobacter pylori is one of the most significant. The mechanisms underlying the associations of H. pylori and GC remain unclear, but it is well known that infection can alter the tumor microenvironment (TME). The TME and the tumor itself constitute a complete ecosystem, and the TME plays critical roles in tumor progression, metastasis, and drug resistance. H. pylori infection can act synergistically with the TME to cause DNA damage and abnormal expression of multiple genes and activation of signaling pathways. It also modulates the host immune system in ways that enhance the proliferation and metastasis of tumor cells, promote epithelial-mesenchymal transition, inhibit apoptosis, and provide energy support for tumor growth. This review elaborates myriad ways that H. pylori infections promote the occurrence and progression of GC by influencing the TME, providing new directions for immunotherapy treatments for this important disease. KEY POINTS: • H. pylori infections cause DNA damage and affect the repair of the TME to DNA damage. • H. pylori infections regulate oncogenes or activate the oncogenic signaling pathways. • H. pylori infections modulate the immune system within the TME.
Collapse
|
7
|
Bellanti JA. Seasonal-specific Variations of the Upper Airway Microbiome and Childhood Asthma: Friend or Foe? J Allergy Clin Immunol 2022; 150:62-63. [PMID: 35623413 DOI: 10.1016/j.jaci.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Joseph A Bellanti
- Professor of Pediatrics and Microbiology-Immunology (Emeritus), Director, International Center for Interdisciplinary Studies of Immunology (ICISI), Georgetown University Medical Center, 3900 Reservoir Road, NW, Room 308 NW, Washington, DC 20057
| |
Collapse
|
8
|
Marques HS, de Brito BB, da Silva FAF, Santos MLC, de Souza JCB, Correia TML, Lopes LW, Neres NSDM, Dórea RSDM, Dantas ACS, Morbeck LLB, Lima IS, de Almeida AA, Dias MRDJ, de Melo FF. Relationship between Th17 immune response and cancer. World J Clin Oncol 2021; 12:845-867. [PMID: 34733609 PMCID: PMC8546660 DOI: 10.5306/wjco.v12.i10.845] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/21/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is the second leading cause of death worldwide and epidemiological projections predict growing cancer mortality rates in the next decades. Cancer has a close relationship with the immune system and, although Th17 cells are known to play roles in the immune response against microorganisms and in autoimmunity, studies have emphasized their roles in cancer pathogenesis. The Th17 immune response profile is involved in several types of cancer including urogenital, respiratory, gastrointestinal, and skin cancers. This type of immune response exerts pro and antitumor functions through several mechanisms, depending on the context of each tumor, including the protumor angiogenesis and exhaustion of T cells and the antitumor recruitment of T cells and neutrophils to the tumor microenvironment. Among other factors, the paradoxical behavior of Th17 cells in this setting has been attributed to its plasticity potential, which makes possible their conversion into other types of T cells such as Th17/Treg and Th17/Th1 cells. Interleukin (IL)-17 stands out among Th17-related cytokines since it modulates pathways and interacts with other cell profiles in the tumor microenvironment, which allow Th17 cells to prevail in tumors. Moreover, the IL-17 is able to mediate pro and antitumor processes that influence the development and progression of various cancers, being associated with variable clinical outcomes. The understanding of the relationship between the Th17 immune response and cancer as well as the singularities of carcinogenic processes in each type of tumor is crucial for the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Hanna Santos Marques
- Campus Vitória da Conquista, Universidade Estadual do Sudoeste da Bahia, Vitória da Conquista 45083-900, Bahia, Brazil
| | - Breno Bittencourt de Brito
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Maria Luísa Cordeiro Santos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Júlio César Braga de Souza
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Thiago Macêdo Lopes Correia
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Luana Weber Lopes
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Nayara Silva de Macêdo Neres
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Anna Carolina Saúde Dantas
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Lorena Lôbo Brito Morbeck
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Iasmin Souza Lima
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Amanda Alves de Almeida
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Maiara Raulina de Jesus Dias
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
9
|
Liu S, Liu H, Song X, Jiang A, Deng Y, Yang C, Sun D, Jiang K, Yang F, Zheng Y. Adoptive CD8 +T-cell grafted with liposomal immunotherapy drugs to counteract the immune suppressive tumor microenvironment and enhance therapy for melanoma. NANOSCALE 2021; 13:15789-15803. [PMID: 34528979 DOI: 10.1039/d1nr04036g] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The immunosuppressive tumor microenvironment has become a formidable obstacle to the treatment of tumors using adoptive T cell therapy, in particular solid tumors. For the purposes of addressing this issue, effector OT-1 CD8+T cells conjugated with liposomal immune regulators (CD8-T-LP-CpG/CD8-T-LP-BMS-202) were developed. An anionic liposome formulation was employed to avoid T cell aggregation and prevent unfavorable side-effects. The inclusion of EGCG in the LP-CpG formulation facilitated the formation of compact complexes with poly lysine (PLL) and is thus expected to increase the stability. CD8-T-LP-CpG administered with a median dose of CpG (20 μg per mouse) markedly reduced the frequency of tumor infiltrating polymorphonuclear leukocyte myeloid-derived suppressor cells (PMN-MDSCs) (20-folds), M2-like macrophages (8-folds), regulatory T-cells (Treg) (2.7-folds), and consequently increased the frequency of cytotoxic CD8+T cells in tumor-infiltrating lymphocytes (TILs) (2-folds) and splenic effector memory CD8+T cells (3-folds) relative to the phosphate buffered saline (PBS) control group. Furthermore, the absolute number of tumor infiltrating lymphocyte subtypes altered followed a consistent trend. The difference remained significant compared to the OT-1 CD8+T cells and the drug-loaded liposome combination group. According to in vivo imaging of CD8-T-LP-DiD, we assumed that the improvement in regulation of the tumor microenvironment of LP-CpG/LP-BMS-202 was attributed to the enhanced drug transportation to the tumor site aided by tumor-specific OT-1 CD8+T cells. In addition, CD8-T-LP-BMS-202 administered with a low dose of BMS-202 (1.5 mg per kg body weight) exerted a dramatically improved therapeutic effect by reducing the tumor infiltrating PMN-MDSCs and M2-like macrophages and the corresponding promoted cytotoxic CD8+T cell recruitment in the TILs and effector memory CD8+T cells mediated anti-tumor immunity. In summary, immune therapy drugs backpacked onto adoptive T cell therapy provides a feasible strategy to improve the therapeutic effect and could result in future clinical translation.
Collapse
Affiliation(s)
- Simeng Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Huimin Liu
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Xiaoshuang Song
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Ailing Jiang
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Yuchuan Deng
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Chengli Yang
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Dan Sun
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| | - Kun Jiang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Fan Yang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Zheng
- State Key Laboratory of Biotherapy/Collaborative Innovation Centre of Biotherapy, West China Hospital, Sichuan University, 17#, Section 3, Ren Min Nan Road, Chengdu, Sichuan, 610041, PR China.
| |
Collapse
|