1
|
Nie W, He Y, Mi X, He S, Chen J, Zhang Y, Wang B, Zheng S, Qian Z, Gao X. Immunostimulatory CKb11 gene combined with immune checkpoint PD-1/PD-L1 blockade activates immune response and simultaneously overcomes the immunosuppression of cancer. Bioact Mater 2024; 39:239-254. [PMID: 38832303 PMCID: PMC11145080 DOI: 10.1016/j.bioactmat.2024.05.014] [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: 03/01/2024] [Revised: 05/05/2024] [Accepted: 05/05/2024] [Indexed: 06/05/2024] Open
Abstract
Immunosuppression tumor microenvironment (TME) seriously impedes anti-tumor immune response, resulting in poor immunotherapy effect of cancer. This study develops a folate-modified delivery system to transport the plasmids encoding immune stimulatory chemokine CKb11 and PD-L1 inhibitors to tumor cells, resulting in high CKb11 secretion from tumor cells, successfully activating immune cells and increasing cytokine secretion to reshape the TME, and ultimately delaying tumor progression. The chemokine CKb11 enhances the effectiveness of tumor immunotherapy by increasing the infiltration of immune cells in TME. It can cause high expression of IFN-γ, which is a double-edged sword that inhibits tumor growth while causing an increase in the expression of PD-L1 on tumor cells. Therefore, combining CKb11 with PD-L1 inhibitors can counterbalance the suppressive impact of PD-L1 on anti-cancer defense, leading to a collaborative anti-tumor outcome. Thus, utilizing nanotechnology to achieve targeted delivery of immune stimulatory chemokines and immune checkpoint inhibitors to tumor sites, thereby reshaping immunosuppressive TME for cancer treatment, has great potential as an immunogene therapy in clinical applications.
Collapse
Affiliation(s)
- Wen Nie
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Yihong He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Xue Mi
- Department of Pharmacy, West China Second University Hospital of Sichuan University, 610041, Chengdu, PR China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Jing Chen
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Yunchu Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, 610041, Chengdu, PR China
| | - Songping Zheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, PR China
| |
Collapse
|
2
|
Zong Y, Lin Y, Wei T, Cheng Q. Lipid Nanoparticle (LNP) Enables mRNA Delivery for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2303261. [PMID: 37196221 DOI: 10.1002/adma.202303261] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Messenger RNA (mRNA) has received great attention in the prevention and treatment of various diseases due to the success of coronavirus disease 2019 (COVID-19) mRNA vaccines (Comirnaty and Spikevax). To meet the therapeutic purpose, it is required that mRNA must enter the target cells and express sufficient proteins. Therefore, the development of effective delivery systems is necessary and crucial. Lipid nanoparticle (LNP) represents a remarkable vehicle that has indeed accelerated mRNA applications in humans, as several mRNA-based therapies have already been approved or are in clinical trials. In this review, the focus is on mRNA-LNP-mediated anticancer therapy. It summarizes the main development strategies of mRNA-LNP formulations, discusses representative therapeutic approaches in cancer, and points out current challenges and possible future directions of this research field. It is hoped that these delivered messages can help further improve the application of mRNA-LNP technology in cancer therapy.
Collapse
Affiliation(s)
- Yan Zong
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Yi Lin
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Tuo Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| |
Collapse
|
3
|
Jiang X, Wu L, Zhang M, Zhang T, Chen C, Wu Y, Yin C, Gao J. Biomembrane nanostructures: Multifunctional platform to enhance tumor chemoimmunotherapy via effective drug delivery. J Control Release 2023; 361:510-533. [PMID: 37567505 DOI: 10.1016/j.jconrel.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Chemotherapeutic drugs have been found to activate the immune response against tumors by inducing immunogenic cell death, in addition to their direct cytotoxic effects toward tumors, therefore broadening the application of chemotherapy in tumor immunotherapy. The combination of other therapeutic strategies, such as phototherapy or radiotherapy, could further strengthen the therapeutic effects of immunotherapy. Nanostructures can facilitate multimodal tumor therapy by integrating various active agents and combining multiple types of therapeutics in a single nanostructure. Biomembrane nanostructures (e.g., exosomes and cell membrane-derived nanostructures), characterized by superior biocompatibility, intrinsic targeting ability, intelligent responsiveness and immune-modulating properties, could realize superior chemoimmunotherapy and represent next-generation nanostructures for tumor immunotherapy. This review summarizes recent advances in biomembrane nanostructures in tumor chemoimmunotherapy and highlights different types of engineering approaches and therapeutic mechanisms. A series of engineering strategies for combining different biomembrane nanostructures, including liposomes, exosomes, cell membranes and bacterial membranes, are summarized. The combination strategy can greatly enhance the targeting, intelligence and functionality of biomembrane nanostructures for chemoimmunotherapy, thereby serving as a stronger tumor therapeutic method. The challenges associated with the clinical translation of biomembrane nanostructures for chemoimmunotherapy and their future perspectives are also discussed.
Collapse
Affiliation(s)
- Xianghe Jiang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Cuimin Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China.
| | - Chuan Yin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
4
|
Park SH, Eun R, Heo J, Lim YT. Nanoengineered drug delivery in cancer immunotherapy for overcoming immunosuppressive tumor microenvironment. Drug Deliv Transl Res 2023; 13:2015-2031. [PMID: 36581707 DOI: 10.1007/s13346-022-01282-8] [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] [Accepted: 12/08/2022] [Indexed: 12/31/2022]
Abstract
Almost like a living being in and of itself, tumors actively interact with and modify their environment to escape immune responses. Owing to the pre-formation of cancer-favorable microenvironment prior to anti-cancer treatment, the numerous attempts that followed propose limited efficacy in oncology. Immunogenicity by activation of immune cells within the tumor microenvironment or recruitment of immune cells from nearby lymph nodes is quickly offset as the immunosuppressive environment, rapidly converting immunogenic cells into immune suppressive cells, overriding the immune system. Tumor cells, as well as regulatory cells, namely M2 macrophages, Treg cells, and MDSCs, derived by the immunosuppressive environment, also cloak from potential anti-tumoral factors by directly or indirectly secreting cytokines, such as IL-10 and TGF-β, related to immune regulation. Enzymes and other metabolic or angiogenetic constituents - VEGF, IDO1, and iNOS - are also employed directed for anti-cancer immune cell malfunctioning. Therefore, the conversion of "cold" immunosuppressive environment into "hot" immune responsive environment is of paramount importance, bestowing the advances in the field of cancer immunotherapy the opportunity to wholly fulfill its intended purpose. This paper reviews the mechanisms by which tumors wield to exercise immune suppression and the nanoengineered delivery strategies being developed to overcome this suppression.
Collapse
Affiliation(s)
- Sei Hyun Park
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Ryounho Eun
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Janghun Heo
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Science and Technology, Department of Nano Engineering, School of Chemical Engineering, and Biomedical Institute for Convergence at SKKU, Sungkyunkwan University (SKKU), Suwon, Gyeonggi-Do, 16419, Republic of Korea.
| |
Collapse
|
5
|
Kim IY, Kim HY, Song HW, Park JO, Choi YH, Choi E. Functional enhancement of exosomes derived from NK cells by IL-15 and IL-21 synergy against hepatocellular carcinoma cells: The cytotoxicity and apoptosis in vitro study. Heliyon 2023; 9:e16962. [PMID: 37484408 PMCID: PMC10361042 DOI: 10.1016/j.heliyon.2023.e16962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 07/25/2023] Open
Abstract
Exosomes are released by various cells, including natural killer (NK) cells and transport signaling molecules for the intercellular communication. Hepatocellular carcinoma (HCC), also known as primary liver cancer, is often inoperable and difficult to accurate diagnosis. Notably, the prognosis and underlying mechanisms of HCC are not fully understood. Exosomes-derived NK cells (NK-exos) express unique cytotoxic proteins with a killing ability in tumors and can easily penetrate tumor tissues to improve their targeting ability. NK cell functions, inducing cellular cytotoxicity are modulated by cytokines such as interleukin (IL)-15 and IL-21. However, the mechanisms and effects of cytokines-stimulated NK-exos for the treatment of liver cancer, including HCC, are not well known. In this study, we aimed to investigate the synergistic anti-tumor effects of NK-exos stimulated with IL-15 and IL-21 (NK-exosIL-15/21) in Hep3B cells. Our findings revealed that NK-exosIL-15/21 expressed cytotoxic proteins (perforin and granzyme B) and contained typical exosome markers (CD9 and CD63) within the size range of 100-150 nm. Moreover, we demonstrated that NK-exosIL-15/21 induced the enhancement of cytotoxicity and apoptotic activity in Hep3B cells by activating the specific pro-apoptotic proteins (Bax, cleaved caspase 3, cleaved PARP, perforin, and granzyme B) and inhibiting the anti-apoptotic protein (Bcl-2). In summary, our results suggest that NK-exosIL-15/21 regulate strong anti-tumor effects of HCC cells, by increasing the cytotoxicity and apoptosis through the activation of specific cytotoxic molecules.
Collapse
Affiliation(s)
- In-Young Kim
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Ho Yong Kim
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Hyeong-woo Song
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - You Hee Choi
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Eunpyo Choi
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
- School of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| |
Collapse
|
6
|
Xiao M, Tang Q, Zeng S, Yang Q, Yang X, Tong X, Zhu G, Lei L, Li S. Emerging biomaterials for tumor immunotherapy. Biomater Res 2023; 27:47. [PMID: 37194085 DOI: 10.1186/s40824-023-00369-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND The immune system interacts with cancer cells in various intricate ways that can protect the individual from overproliferation of cancer cells; however, these interactions can also lead to malignancy. There has been a dramatic increase in the application of cancer immunotherapy in the last decade. However, low immunogenicity, poor specificity, weak presentation efficiency, and off-target side effects still limit its widespread application. Fortunately, advanced biomaterials effectively contribute immunotherapy and play an important role in cancer treatment, making it a research hotspot in the biomedical field. MAIN BODY This review discusses immunotherapies and the development of related biomaterials for application in the field. The review first summarizes the various types of tumor immunotherapy applicable in clinical practice as well as their underlying mechanisms. Further, it focuses on the types of biomaterials applied in immunotherapy and related research on metal nanomaterials, silicon nanoparticles, carbon nanotubes, polymer nanoparticles, and cell membrane nanocarriers. Moreover, we introduce the preparation and processing technologies of these biomaterials (liposomes, microspheres, microneedles, and hydrogels) and summarize their mechanisms when applied to tumor immunotherapy. Finally, we discuss future advancements and shortcomings related to the application of biomaterials in tumor immunotherapy. CONCLUSION Research on biomaterial-based tumor immunotherapy is booming; however, several challenges remain to be overcome to transition from experimental research to clinical application. Biomaterials have been optimized continuously and nanotechnology has achieved continuous progression, ensuring the development of more efficient biomaterials, thereby providing a platform and opportunity for breakthroughs in tumor immunotherapy.
Collapse
Affiliation(s)
- Minna Xiao
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shiying Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinming Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Xinying Tong
- Department of Hemodialysis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
7
|
Jin X, Bi J. Prospects for NK-based immunotherapy of chronic HBV infection. Front Immunol 2022; 13:1084109. [PMID: 36591230 PMCID: PMC9797727 DOI: 10.3389/fimmu.2022.1084109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/01/2022] [Indexed: 12/16/2022] Open
Abstract
Effective and long-term treatment is required for controlling chronic Hepatitis B Virus (HBV) infection. Natural killer (NK) cells are antiviral innate lymphocytes and represent an essential arm of current immunotherapy. In chronic HBV (CHB), NK cells display altered changes in phenotypes and functions, but preserve antiviral activity, especially for cytolytic activity. On the other hand, NK cells might also cause liver injury in the disease. NK -based immunotherapy, including adoptive NK cell therapy and NK -based checkpoint inhibition, could potentially exploit the antiviral aspect of NK cells for controlling CHB infection while preventing liver tissue damage. Here, we review recent progress in NK cell biology under the context of CHB infection, and discuss potential NK -based immunotherapy strategies for the disease.
Collapse
|
8
|
Nash AM, Aghlara-Fotovat S, Castillio B, Hernandez A, Pugazenthi A, Lee HS, Jang HJ, Nguyen A, Lu A, Burt BM, Ghanta RK, Veiseh O. Activation of Adaptive and Innate Immune Cells via Localized IL2 Cytokine Factories Eradicates Mesothelioma Tumors. Clin Cancer Res 2022; 28:5121-5135. [PMID: 35993913 PMCID: PMC9713361 DOI: 10.1158/1078-0432.ccr-22-1493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/21/2022] [Accepted: 08/12/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE IL2 immunotherapy has the potential to elicit immune-mediated tumor lysis via activation of effector immune cells, but clinical utility is limited due to pharmacokinetic challenges as well as vascular leak syndrome and other life-threatening toxicities experienced by patients. We developed a safe and clinically translatable localized IL2 delivery system to boost the potency of therapy while minimizing systemic cytokine exposure. EXPERIMENTAL DESIGN We evaluated the therapeutic efficacy of IL2 cytokine factories in a mouse model of malignant mesothelioma. Changes in immune populations were analyzed using time-of-flight mass cytometry (CyTOF), and the safety and translatability of the platform were evaluated using complete blood counts and serum chemistry analysis. RESULTS IL2 cytokine factories enabled 150× higher IL2 concentrations in the local compartment with limited leakage into the systemic circulation. AB1 tumor burden was reduced by 80% after 1 week of monotherapy treatment, and 7 of 7 of animals exhibited tumor eradication without recurrence when IL2 cytokine factories were combined with anti-programmed cell death protein 1 (aPD1). Furthermore, CyTOF analysis showed an increase in CD69+CD44+ and CD69-CD44+CD62L- T cells, reduction of CD86-PD-L1- M2-like macrophages, and a corresponding increase in CD86+PD-L1+ M1-like macrophages and MHC-II+ dendritic cells after treatment. Finally, blood chemistry ranges in rodents demonstrated the safety of cytokine factory treatment and reinforced its potential for clinical use. CONCLUSIONS IL2 cytokine factories led to the eradication of aggressive mouse malignant mesothelioma tumors and protection from tumor recurrence, and increased the therapeutic efficacy of aPD1 checkpoint therapy. This study provides support for the clinical evaluation of this IL2-based delivery system. See related commentary by Palanki et al., p. 5010.
Collapse
Affiliation(s)
- Amanda M. Nash
- Department of Bioengineering, Rice University, Houston, Texas
| | | | | | | | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Hyun-Sung Lee
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Hee-Jin Jang
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Annie Nguyen
- Department of Bioengineering, Rice University, Houston, Texas
| | - Alexander Lu
- Department of Bioengineering, Rice University, Houston, Texas
| | - Bryan M. Burt
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Ravi K. Ghanta
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, Texas
| |
Collapse
|
9
|
Jeong S, Kim YG, Kim S, Kim K. Enhanced anticancer efficacy of primed natural killer cells via coacervate-mediated exogenous interleukin-15 delivery. Biomater Sci 2022; 10:5968-5979. [PMID: 36048163 DOI: 10.1039/d2bm00876a] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Effective exogenous delivery of interleukin (IL)-15 to natural killer (NK) cells with subsequent anticancer efficacy could be a promising immune cell-based cancer immunotherapy. For the protection of encapsulated cargo IL-15 while maintaining its bioactivity under physiological conditions, we utilized a coacervate (Coa) consisting of a cationic methoxy polyethylene glycol-poly(ethylene arginyl aspartate diglyceride) (mPEG-PEAD) polymer, anionic counterpart heparin, and cargo IL-15. mPEGylation into the backbone cation effectively preserved the colloidal stability of Coa in harsh environments and enhanced the protection of cargo IL-15 than normal Coa without mPEGylation. Proliferation and anticancer efficacy of primed NK cells through co-culture with multiple cancer cell lines were enhanced in the mPEG-Coa group due to the maintained bioactivity of cargo IL-15 during the ex vivo expansion of NK cells. These facilitated functions of NK cells were also supported by the increased expression of mRNAs related to anticancer effects of NK cells, including cytotoxic granules, death ligands, anti-apoptotic proteins, and activation receptors. In summary, our Coa-mediated exogenous IL-15 delivery could be an effective ex vivo priming technique for NK cells with sustained immune activation that can effectively facilitate its usage for cancer immunotherapy.
Collapse
Affiliation(s)
- Sehwan Jeong
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| | - Young Guk Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Rallis KS, Corrigan AE, Dadah H, Stanislovas J, Zamani P, Makker S, Szabados B, Sideris M. IL-10 in cancer: an essential thermostatic regulator between homeostatic immunity and inflammation - a comprehensive review. Future Oncol 2022; 18:3349-3365. [PMID: 36172856 DOI: 10.2217/fon-2022-0063] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Cytokines are soluble proteins that mediate intercellular signaling regulating immune and inflammatory responses. Cytokine modulation represents a promising cancer immunotherapy approach for immune-mediated tumor regression. However, redundancy in cytokine signaling and cytokines' pleiotropy, narrow therapeutic window, systemic toxicity, short half-life and limited efficacy represent outstanding challenges for cytokine-based cancer immunotherapies. Recently, there has been interest in the paradoxical role of IL-10 in cancer, its controversial prognostic utility and novel strategies to enhance its therapeutic profile. Here, the authors review the literature surrounding the role of IL-10 within the tumor microenvironment, its prognostic correlates to cancer patient outcomes and its pro- and antitumor effects, and they assess the legitimacy of potential therapeutic strategies harnessing IL-10 by outlining the notable preclinical and clinical evidence to date.
Collapse
Affiliation(s)
- Kathrine S Rallis
- Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK.,Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Amber E Corrigan
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Hashim Dadah
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Justas Stanislovas
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Parisa Zamani
- GKT School of Medicine, King's College London, London, SE1 9RT, UK
| | - Shania Makker
- Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, E1 2AD, UK
| | - Bernadett Szabados
- Barts Cancer Institute, Queen Mary University of London, London, EC1M 5PZ, UK
| | - Michail Sideris
- Women's Health Research Unit, Queen Mary University of London, London, E1 2AB, UK
| |
Collapse
|
11
|
Chu H, Cao T, Dai G, Liu B, Duan H, Kong C, Tian N, Hou D, Sun Z. Recent advances in functionalized upconversion nanoparticles for light-activated tumor therapy. RSC Adv 2021; 11:35472-35488. [PMID: 35493151 PMCID: PMC9043211 DOI: 10.1039/d1ra05638g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/28/2021] [Indexed: 01/16/2023] Open
Abstract
Upconversion nanoparticles (UCNPs) are a class of optical nanocrystals doped with lanthanide ions that offer great promise for applications in controllable tumor therapy. In recent years, UCNPs have become an important tool for studying the treatment of various malignant and nonmalignant cutaneous diseases. UCNPs convert near-infrared (NIR) radiation into shorter-wavelength visible and ultraviolet (UV) radiation, which is much better than conventional UV activated tumor therapy as strong UV-light can be damaging to healthy surrounding tissue. Moreover, UV light generally does not penetrate deeply into the skin, an issue that UCNPs can now address. However, the current studies are still in the early stage of research, with a long way to go before clinical implementation. In this paper, we systematically analysed recent advances in light-activated tumor therapy using functionalized UCNPs. We summarized the purpose and mechanism of UCNP-based photodynamic therapy (PDT), gene therapy, immunotherapy, chemo-therapy and integrated therapy. We believe the creation of functional materials based on UCNPs will offer superior performance and enable innovative applications, increasing the scope and opportunities for cancer therapy in the future.
Collapse
Affiliation(s)
- Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| | - Tingming Cao
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| | - Guangming Dai
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| | - Bei Liu
- School of Science, Minzu University of China Beijing 100081 PR China
| | - Huijuan Duan
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| | - Chengcheng Kong
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| | - Na Tian
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| | - Dailun Hou
- Department of Radiology, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University Beijing 101149 PR China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute Beijing 101149 PR China
| |
Collapse
|
12
|
Lejeune P, Cruciani V, Berg-Larsen A, Schlicker A, Mobergslien A, Bartnitzky L, Berndt S, Zitzmann-Kolbe S, Kamfenkel C, Stargard S, Hammer S, Jørgensen JS, Jackerott M, Nielsen CH, Schatz CA, Hennekes H, Karlsson J, Cuthbertson AS, Mumberg D, Hagemann UB. Immunostimulatory effects of targeted thorium-227 conjugates as single agent and in combination with anti-PD-L1 therapy. J Immunother Cancer 2021; 9:jitc-2021-002387. [PMID: 34615703 PMCID: PMC8496392 DOI: 10.1136/jitc-2021-002387] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2021] [Indexed: 12/25/2022] Open
Abstract
Background Targeted thorium-227 conjugates (TTCs) are an emerging class of targeted alpha therapies (TATs). Their unique mode of action (MoA) is the induction of difficult-to-repair clustered DNA double-strand breaks. However, thus far, their effects on the immune system are largely unknown. Here, we investigated the immunostimulatory effects of the mesothelin-targeted thorium-227 conjugate (MSLN-TTC) in vitro and in vivo in monotherapy and in combination with an inhibitor of the immune checkpoint programmed death receptor ligand 1 (PD-L1) in immunocompetent mice. Methods The murine cell line MC38 was transfected with the human gene encoding for MSLN (hMSLN) to enable binding of the non-cross-reactive MSLN-TTC. The immunostimulatory effects of MSLN-TTC were studied in vitro on human cancer cell lines and MC38-hMSLN cells. The efficacy and MoA of MSLN-TTC were studied in vivo as monotherapy or in combination with anti-PD-L1 in MC38-hMSLN tumor-bearing immunocompetent C57BL/6 mice. Experiments were supported by RNA sequencing, flow cytometry, immunohistochemistry, mesoscale, and TaqMan PCR analyses to study the underlying immunostimulatory effects. In vivo depletion of CD8+ T cells and studies with Rag2/Il2Rg double knockout C57BL/6 mice were conducted to investigate the importance of immune cells to the efficacy of MSLN-TTC. Results MSLN-TTC treatment induced upregulation of DNA sensing pathway transcripts (IL-6, CCL20, CXCL10, and stimulator of interferon genes (STING)-related genes) in vitro as determined by RNASeq analysis. The results, including phospho-STING activation, were confirmed on the protein level. Danger-associated molecular pattern molecules were upregulated in parallel, leading to dendritic cell (DC) activation in vitro. MSLN-TTC showed strong antitumor activity (T:C 0.38, p<0.05) as a single agent in human MSLN-expressing MC38 tumor-bearing immunocompetent mice. Combining MSLN-TTC with anti-PD-L1 further enhanced the efficacy (T:C 0.08, p<0.001) as evidenced by the increased number of tumor-free surviving animals. MSLN-TTC monotherapy caused migration of CD103+ cDC1 DCs and infiltration of CD8+ T cells into tumors, which was enhanced on combination with anti-PD-L1. Intriguingly, CD8+ T-cell depletion decreased antitumor efficacy. Conclusions These in vitro and in vivo data on MSLN-TTC demonstrate that the MoA of TTCs involves activation of the immune system. The findings are of relevance for other targeted radiotherapies and may guide clinical combination strategies.
Collapse
|
13
|
Murakami J, Wu L, Kohno M, Chan ML, Zhao Y, Yun Z, Cho BCJ, de Perrot M. Triple-modality therapy maximizes antitumor immune responses in a mouse model of mesothelioma. Sci Transl Med 2021; 13:13/589/eabd9882. [PMID: 33853932 DOI: 10.1126/scitranslmed.abd9882] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 03/15/2021] [Indexed: 12/20/2022]
Abstract
Malignant pleural mesothelioma (MPM) is an intractable disease with an extremely poor prognosis. Our clinical protocol for MPM of subablative radiotherapy (RT) followed by radical surgery achieved better survival compared to other multimodal treatments, but local relapse and metastasis remain a problem. This subablative RT elicits an antitumoral immune response that is limited by the immunosuppressive microenvironment generated by regulatory T (Treg) cells. The antitumor effect of immunotherapy to simultaneously modulate the immune activation and the immune suppression after subablative RT has not been investigated in MPM. Herein, we demonstrated a rationale to combine interleukin-15 (IL-15) superagonist (IL-15SA) and glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) agonist (DTA-1) with subablative RT in mesothelioma. IL-15SA boosted the systemic expansion of specific antitumoral memory CD8+ T cells that were induced by RT in mice. Their effect, however, was limited by the up-regulation and activation of Treg cells in the radiated tumor microenvironment. Hence, selective depletion of intratumoral Treg cells through DTA-1 enhanced the benefit of subablative RT in combination with IL-15SA. The addition of surgical resection of the radiated tumor in combination with IL-15SA and DTA-1 maximized the benefit of RT and was accompanied by a reproducible abscopal response in a concomitant tumor model. These data support the development of clinical trials in MPM to test such treatment options for patients with locally advanced or metastatic tumors.
Collapse
Affiliation(s)
- Junichi Murakami
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Department of Surgery and Clinical Science, Division of Chest Surgery, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Mikihiro Kohno
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada.,Division of Thoracic Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C4, Canada
| | - Mei-Lin Chan
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Yidan Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Zhihong Yun
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - B C John Cho
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C1, Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 1L7, Canada. .,Division of Thoracic Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 2C4, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
14
|
Jiang L, Zhang G, Li Y, Shi G, Li M. Potential Application of Plant-Based Functional Foods in the Development of Immune Boosters. Front Pharmacol 2021; 12:637782. [PMID: 33959009 PMCID: PMC8096308 DOI: 10.3389/fphar.2021.637782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/15/2021] [Indexed: 11/13/2022] Open
Abstract
Immune dysfunction, which is responsible for the development of human diseases including cancer, is caused by a variety of factors. Therefore, regulation of the factors influencing the immune response is a potentially effective strategy to counter diseases. Presently, several immune adjuvants are used in clinical practice to enhance the immune response and host defense ability; however, synthetic drugs can exert negative side effects. Thus, the search for natural products of plant origin as new leads for the development of potent and safe immune boosters is gaining considerable research interest. Plant-based functional foods have been shown to exert several immunomodulatory effects in humans; therefore, the application of new agents to enhance immunological and specific host defenses is a promising approach. In this comprehensive review, we have provided an up-to-date report on the use as well as the known and potential mechanisms of bioactive compounds obtained from plant-based functional foods as natural immune boosters. Plant-based bioactive compounds promote immunity through multiple mechanisms, including influencing the immune organs, cellular immunity, humoral immunity, nonspecific immunity, and immune-related signal transduction pathways. Enhancement of the immune response in a natural manner represents an excellent prospect for disease prevention and treatment and is worthy of further research and development using approaches of modern science and technology.
Collapse
Affiliation(s)
- Linlin Jiang
- Department of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Guoqing Zhang
- Inner Mongolia Hospital of Traditional Chinese Medicine, Hohhot, China.,Department of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Ye Li
- Inner Mongolia Hospital of Traditional Chinese Medicine, Hohhot, China
| | | | - Minhui Li
- Department of Pharmacy, Inner Mongolia Medical University, Hohhot, China.,Pharmaceutical Laboratory, Inner Mongolia Institute of Traditional Chinese Medicine, Hohhot, China.,Inner Mongolia Key Laboratory of Characteristic Geoherbs Resources Protection and Utilization, Baotou Medical College, Baotou, China
| |
Collapse
|
15
|
Sun S, Ding Z, Yang X, Zhao X, Zhao M, Gao L, Chen Q, Xie S, Liu A, Yin S, Xu Z, Lu X. Nanobody: A Small Antibody with Big Implications for Tumor Therapeutic Strategy. Int J Nanomedicine 2021; 16:2337-2356. [PMID: 33790553 PMCID: PMC7997558 DOI: 10.2147/ijn.s297631] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
The development of monoclonal antibody treatments for successful tumor-targeted therapies took several decades. However, the efficacy of antibody-based therapy is still confined and desperately needs further improvement. Nanobodies are the recombinant variable domains of heavy-chain-only antibodies, with many unique properties such as small size (~15kDa), excellent solubility, superior stability, ease of manufacture, quick clearance from blood, and deep tissue penetration, which gain increasing acceptance as therapeutical tools and are considered also as building blocks for chimeric antigen receptors as well as for targeted drug delivery. Thus, one of the promising novel developments that may address the deficiency of monoclonal antibody-based therapies is the utilization of nanobodies. This article provides readers the significant factors that the structural and biochemical properties of nanobodies and the research progress on nanobodies in the fields of tumor treatment, as well as their application prospect.
Collapse
Affiliation(s)
- Shuyang Sun
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Ziqiang Ding
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiaomei Yang
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xinyue Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Minlong Zhao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Li Gao
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Qu Chen
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shenxia Xie
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Aiqun Liu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Shihua Yin
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhiping Xu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Xiaoling Lu
- International Nanobody Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
- School of Stomatology, Guangxi Medical University, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
16
|
Chen Z, Yang Y, Neo SY, Shi H, Chen Y, Wagner AK, Larsson K, Tong L, Jakobsson PJ, Alici E, Wu J, Cao Y, Wang K, Liu LL, Mao Y, Sarhan D, Lundqvist A. Phosphodiesterase 4A confers resistance to PGE2-mediated suppression in CD25 + /CD54 + NK cells. EMBO Rep 2021; 22:e51329. [PMID: 33480074 PMCID: PMC7926252 DOI: 10.15252/embr.202051329] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/22/2022] Open
Abstract
Inadequate persistence of tumor‐infiltrating natural killer (NK) cells is associated with poor prognosis in cancer patients. The solid tumor microenvironment is characterized by the presence of immunosuppressive factors, including prostaglandin E2 (PGE2), that limit NK cell persistence. Here, we investigate if the modulation of the cytokine environment in lung cancer with IL‐2 or IL‐15 renders NK cells resistant to suppression by PGE2. Analyzing Cancer Genome Atlas (TCGA) data, we found that high NK cell gene signatures correlate with significantly improved overall survival in patients with high levels of the prostaglandin E synthase (PTGES). In vitro, IL‐15, in contrast to IL‐2, enriches for CD25+/CD54+ NK cells with superior mTOR activity and increased expression of the cAMP hydrolyzing enzyme phosphodiesterase 4A (PDE4A). Consequently, this distinct population of NK cells maintains their function in the presence of PGE2 and shows an increased ability to infiltrate lung adenocarcinoma tumors in vitro and in vivo. Thus, strategies to enrich CD25+/CD54+ NK cells for adoptive cell therapy should be considered.
Collapse
Affiliation(s)
- Ziqing Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.,Department of Respiratory Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.,School of Medicine, Zhejiang University, Hangzhou, China
| | - Shi Y Neo
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hao Shi
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yi Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Arnika K Wagner
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Johan Jakobsson
- Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Evren Alici
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jing Wu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kai Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Lisa L Liu
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yumeng Mao
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Di Sotto A, Mancinelli R, Gullì M, Eufemi M, Mammola CL, Mazzanti G, Di Giacomo S. Chemopreventive Potential of Caryophyllane Sesquiterpenes: An Overview of Preliminary Evidence. Cancers (Basel) 2020; 12:E3034. [PMID: 33081075 PMCID: PMC7603190 DOI: 10.3390/cancers12103034] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Chemoprevention is referred to as a strategy to inhibit, suppress, or reverse tumor development and progression in healthy people along with high-risk subjects and oncologic patients through using pharmacological or natural substances. Numerous phytochemicals have been widely described in the literature to possess chemopreventive properties, although their clinical usefulness remains to be defined. Among them, caryophyllane sesquiterpenes are natural compounds widely occurring in nature kingdoms, especially in plants, fungi, and marine environments. Several structures, characterized by a common caryophyllane skeleton with further rearrangements, have been identified, but those isolated from plant essential oils, including β-caryophyllene, β-caryophyllene oxide, α-humulene, and isocaryophyllene, have attracted the greatest pharmacological attention. Emerging evidence has outlined a complex polypharmacological profile of caryophyllane sesquiterpenes characterized by blocking, suppressing, chemosensitizing, and cytoprotective properties, which suggests a possible usefulness of these natural substances in cancer chemoprevention for both preventive and adjuvant purposes. In the present review, the scientific knowledge about the chemopreventive properties of caryophyllane sesquiterpenes and the mechanisms involved have been collected and discussed; moreover, possible structure-activity relationships have been highlighted. Although further high-quality studies are required, the promising preclinical findings and the safe pharmacological profile encourage further studies to define a clinical usefulness of caryophyllane sesquiterpenes in primary, secondary, or tertiary chemoprevention.
Collapse
Affiliation(s)
- Antonella Di Sotto
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| | - Romina Mancinelli
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (R.M.); (C.L.M.)
| | - Marco Gullì
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| | - Margherita Eufemi
- Department of Biochemical Science “A. Rossi Fanelli”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy;
| | - Caterina Loredana Mammola
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (R.M.); (C.L.M.)
| | - Gabriela Mazzanti
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| | - Silvia Di Giacomo
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy; (M.G.); (S.D.G.)
| |
Collapse
|