1
|
Kubbara EA, Bolad A, Malibary H. Advances in Liposomal Interleukin and Liposomal Interleukin Gene Therapy for Cancer: A Comprehensive Review of Preclinical Studies. Pharmaceutics 2025; 17:383. [PMID: 40143046 PMCID: PMC11945541 DOI: 10.3390/pharmaceutics17030383] [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: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Preclinical studies on liposomal interleukin (IL) therapy demonstrate considerable promise in cancer treatment. This review explores the achievements, challenges, and future potential of liposomal IL encapsulation, focusing on preclinical studies. METHODS A structured search was conducted using the PubMed and Web of Science databases with the following search terms and Boolean operators: ("liposomal interleukin" OR "liposome-encapsulated interleukin") AND ("gene therapy" OR "gene delivery") AND ("cancer" OR "tumor" OR "oncology") AND ("pre-clinical studies" OR "animal models" OR "in vitro studies". RESULTS Liposomal IL-2 formulations are notable for enhancing delivery and retention at tumor sites. Recombinant human interleukin (rhIL-2) adsorbed onto small liposomes (35-50 nm) substantially reduces metastases in murine models. Hepatic metastasis models demonstrate superior efficacy of liposomal IL-2 over free IL-2 by enhancing immune responses, particularly in the liver. Localized delivery strategies, including nebulized liposomal IL-2 in canine pulmonary metastases and intrathoracic administration in murine sarcoma models, reduce systemic toxicity while promoting immune activation and tumor regression. Liposomal IL gene therapy, delivering cytokine genes directly to tumor sites, represents a notable advancement. Combining IL-2 gene therapy with other cytokines, including IL-6 or double-stranded RNA adjuvants, synergistically enhances macrophage and T-cell activation. Liposomal IL-4, IL-6, and IL-21 therapies show potential across various tumor types. Pairing liposomal IL-2 with chemotherapy or immune agents improves remission and survival. Innovative strategies, including PEGylation and ligand-targeted systems, optimize delivery, release, and therapeutic outcomes. CONCLUSIONS Utilizing immune-stimulatory ILs through advanced liposomal delivery and gene therapy establishes a strong foundation for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Eman A. Kubbara
- Clinical Biochemistry Department, Faculty of Medicine, Rabigh Branch, King Abdulaziz University, Rabigh 21911, Saudi Arabia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Ahmed Bolad
- Department of Microbiology and Unit of Immunology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Husam Malibary
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| |
Collapse
|
2
|
Dong C, Tan D, Sun H, Li Z, Zhang L, Zheng Y, Liu S, Zhang Y, He Q. Interleukin-12 Delivery Strategies and Advances in Tumor Immunotherapy. Curr Issues Mol Biol 2024; 46:11548-11579. [PMID: 39451566 PMCID: PMC11506767 DOI: 10.3390/cimb46100686] [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: 09/25/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Interleukin-12 (IL-12) is considered to be a promising cytokine for enhancing an antitumor immune response; however, recombinant IL-12 has shown significant toxicity and limited efficacy in early clinical trials. Recently, many strategies for delivering IL-12 to tumor tissues have been developed, such as modifying IL-12, utilizing viral vectors, non-viral vectors, and cellular vectors. Previous studies have found that the fusion of IL-12 with extracellular matrix proteins, collagen, and immune factors is a way to enhance its therapeutic potential. In addition, studies have demonstrated that viral vectors are a good platform, and a variety of viruses such as oncolytic viruses, adenoviruses, and poxviruses have been used to deliver IL-12-with testing previously conducted in various cancer models. The local expression of IL-12 in tumors based on viral delivery avoids systemic toxicity while inducing effective antitumor immunity and acting synergistically with other therapies without compromising safety. In addition, lipid nanoparticles are currently considered to be the most mature drug delivery system. Moreover, cells are also considered to be drug carriers because they can effectively deliver therapeutic substances to tumors. In this article, we will systematically discuss the anti-tumor effects of IL-12 on its own or in combination with other therapies based on different delivery strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qing He
- State Key Laboratory of Drug Regulatory Sciences, National Institutes for Food and Drug Control, Beijing 102629, China; (C.D.); (D.T.); (H.S.); (Z.L.); (L.Z.); (Y.Z.); (S.L.); (Y.Z.)
| |
Collapse
|
3
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
4
|
Xu Y, Sun X, Tong Y. Interleukin-12 in multimodal tumor therapies for induction of anti-tumor immunity. Discov Oncol 2024; 15:170. [PMID: 38753073 PMCID: PMC11098992 DOI: 10.1007/s12672-024-01011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
Interleukin-12 (IL-12) can be used as an immunomodulator in cancer immunotherapy. And it has demonstrated enormous potential in inhibiting tumor growth and improving the tumor microenvironment (TME) by several preclinical models. However, some disappointing results have showed in the early clinical trials when IL-12 used as a single agent for systemic cancer therapy. Combination therapy is an effective way to significantly fulfill the great potential of IL-12 as an immunomodulator. Here, we discuss the effects of IL-12 combined with traditional methods (chemotherapy, radiotherapy and surgery), targeted therapy or immunotherapy in the preclinical and clinical studies. Moreover, we summarized the potential mechanism underlying the anti-tumor effect of IL-12 in the combination strategies. And we also discussed the delivery methods and tumor-targeted modification of IL-12 and outlines future prospects for IL-12 as an immunomodulator.
Collapse
Affiliation(s)
- Yulian Xu
- College of Life Sciences, China Jiliang University, 168 Xueyuan Street, Hangzhou, Zhejiang, China
| | - Xueli Sun
- College of Life Sciences, China Jiliang University, 168 Xueyuan Street, Hangzhou, Zhejiang, China
| | - Yunguang Tong
- College of Life Sciences, China Jiliang University, 168 Xueyuan Street, Hangzhou, Zhejiang, China.
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Omigen, Inc, Hangzhou, 310018, Zhejiang, China.
| |
Collapse
|
5
|
Novel strategies exploiting interleukin-12 in cancer immunotherapy. Pharmacol Ther 2022; 239:108189. [DOI: 10.1016/j.pharmthera.2022.108189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 11/24/2022]
|
6
|
Yang Q, Hu J, Jia Z, Zhang S, Wang Q, Wang J, Dao L, Zhang W, Xia X, Gorlick R, Li S. Membrane-Anchored and Tumor-Targeted IL12 (attIL12)-PBMC Therapy for Osteosarcoma. Clin Cancer Res 2022; 28:3862-3873. [PMID: 35727602 PMCID: PMC10142228 DOI: 10.1158/1078-0432.ccr-22-0721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/25/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T-cell therapy has shown great promise for treating hematologic malignancies but requires a long duration of T-cell expansion, is associated with severe toxicity, and has limited efficacy for treating solid tumors. We designed experiments to address those challenges. EXPERIMENTAL DESIGN We generated a cell membrane-anchored and tumor-targeted IL12 (attIL12) to arm peripheral blood mononuclear cells (PBMC) instead of T cells to omit the expansion phase for required CAR T cells. RESULTS This IL12-based attIL12-PBMC therapy showed significant antitumor efficacy in both heterogeneous osteosarcoma patient-derived xenograft tumors and metastatic osteosarcoma tumors with no observable toxic effects. Mechanistically, attIL12-PBMC treatment resulted in tumor-restricted antitumor cytokine release and accumulation of attIL12-PBMCs in tumors. It also induced terminal differentiation of osteosarcoma cells into bone-like cells to impede tumor growth. CONCLUSIONS In summary, attIL12-PBMC therapy is safe and effective against osteosarcoma. Our goal is to move this treatment into a clinical trial. Owing to the convenience of the attIL12-PBMC production process, we believe it will be feasible.
Collapse
Affiliation(s)
- Qing Yang
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Orthopedic Surgery, The First People’s Hospital of Xiangtan City, 411101, Xiangtan, China
| | - Jiemiao Hu
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhiliang Jia
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sheng Zhang
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - LongHoang Dao
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendong Zhang
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xueqing Xia
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Gorlick
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shulin Li
- Department of Pediatrics–Research, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Synthesis, Characterization, and In Vivo Cytokinome Profile of IL-12-Loaded PLGA Nanospheres. J Immunol Res 2022; 2022:6993187. [PMID: 35465347 PMCID: PMC9023212 DOI: 10.1155/2022/6993187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/23/2022] [Indexed: 01/19/2023] Open
Abstract
We report the successful encapsulation and elution of recombinant murine IL-12 (rmIL-12) from poly(lactide-co-glycolic) acid (PLGA) nanospheres (IL-12-NS) synthesized using the double emulsion/solvent evaporation (DESE) technique with microsphere depletion through ultracentrifugation. Images obtained with scanning electron microscopy (SEM) showcased a characteristic spherical shape with a mean particle diameter of 138.1 ± 10.8 nm and zeta potential of −15.1 ± 1.249 mV. These values suggest minimal flocculation when in solution, which was reflected in an in vivo biodistribution study that reported no observed morbidity/mortality. Encapsulation efficiency (EE) was determined to be 0.101 ± 0.009% with average particle concentration obtained per batch of 1.66 × 109 ± 4.45 × 108 particles/mL. Disparate zeta (ζ) potentials obtained from both protein-loaded and protein-unloaded batches suggested surface adsorption of protein, and confocal microscopy of BSA-FITC-loaded nanospheres confirmed the presence of protein within the polymeric shell. Furthermore, elution of rmIL-12 from IL-12-NS at a concentration of 500 million particles/mL was characterized using enzyme-linked immunosorbent assay (ELISA). When IL-12-NS was administered in vivo to female BALB/c mice through retroorbital injection, IL-12-NS produced a favorable systemic cytokine profile for tumoricidal activity within the peripheral blood. Whereas IFN-γ nadir occurred at 72 hours, levels recovered quickly and displayed positive correlations postburst out to 25 days postinjection. IL-12-NS administration induced proinflammatory changes while prompting minimal counterregulatory increases in anti-inflammatory IL-10 and IL-4 cytokine levels. Further, while IL-6 levels increased to 30 folds of the baseline during the burst phase, they normalized by 72 hours and trended negatively throughout the sill phase. Similar trends were observed with IL-1β and CXCL-1, suggesting a decreased likelihood of progression to a systemic inflammatory response syndrome-like state. As IL-12-NS delivers logarithmically lower amounts of IL-12 than previously administered during human clinical trials, our data reflect the importance of IL-12-NS which safely create a systemic immunostimulatory environment.
Collapse
|
8
|
Salem ML, Salman S, Barnawi IO. Brief in vitro IL-12 conditioning of CD8 + T Cells for anticancer adoptive T cell therapy. Cancer Immunol Immunother 2021; 70:2751-2759. [PMID: 33966093 PMCID: PMC10992799 DOI: 10.1007/s00262-021-02887-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy represents a potential treatment approach through non-specific and specific enhancement of the immune responses. Adoptive cell therapy (ACT) is a potential modality of immunotherapy that depends on harvesting T cells from the tumor-bearing host, activating them in vitro and infusing them back to the same host. Several cytokines, in particular IL-2, IL-7 and IL-15, have been used to enhance survival T cells in vitro. Although effective, conditioning of T cells in vitro with these cytokines requires long-term culture which results in the loss of expression of their trafficking receptors mainly CD62L. It also results in exhaustion of the activated T cells and reduction in their functions upon adoptive transfer in vivo. Our recent studies and those of other groups showed that brief (3 days) conditioning of CD8+ T cells by IL-12 in vitro can result in enhancing function of tumor-reactive CD8+ T cells. Adoptive transfer of these IL-12-conditioned CD8+ T cells into tumor-bearing mice, preconditioned with cyclophosphamide, 1 day before ACT, induced tumor eradication that was associated with generation of tumor-specific memory response. In this review, we summarize studies that indicated to the superiority of IL-12 as a potential cytokine for conditioning T cells for ACT. In addition, we discuss some of the cellular and molecular mechanisms that govern how IL-12 programs CD8+ T cells to enhance their functionality especially in vitro and its implication in combination with other ACT modalities, opening a avenue for the clinical application of this cytokine.
Collapse
Affiliation(s)
- Mohamed Labib Salem
- Immunology and Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt.
- Center of Excellence in Cancer Research (CECR), Tanta University, Tanta, Egypt.
| | - Samar Salman
- Department of Dermatology and VenereologyFaculty of MedicineTanta University Hospital, Tanta University, Tanta, Egypt
| | - Ibrahim O Barnawi
- Animal Section, Department of Biological Sciences, Faculty of Science, Taibah University, Medina, Saudi Arabia
| |
Collapse
|
9
|
Pires IS, Hammond PT, Irvine DJ. Engineering Strategies for Immunomodulatory Cytokine Therapies - Challenges and Clinical Progress. ADVANCED THERAPEUTICS 2021; 4:2100035. [PMID: 34734110 PMCID: PMC8562465 DOI: 10.1002/adtp.202100035] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Cytokines are immunoregulatory proteins involved in many pathological states with promising potential as therapeutic agents. A diverse array of cytokines have been studied in preclinical disease models since the 1950s, some of which became successful biopharmaceutical products with the advancement of recombinant protein technology in the 1980s. However, following these early approvals, clinical translation of these natural immune signaling molecules has been limited due to their pleiotropic action in many cell types, and the fact that they have evolved to act primarily locally in tissues. These characteristics, combined with poor pharmacokinetics, have hindered the delivery of cytokines via systemic administration routes due to dose-limiting toxicities. However, given their clinical potential and recent clinical successes in cancer immunotherapy, cytokines continue to be extensively pursued in preclinical and clinical studies, and a range of molecular and formulation engineering strategies are being applied to reduce treatment toxicity while maintaining or enhancing therapeutic efficacy. This review provides a brief background on the characteristics of cytokines and their history as clinical therapeutics, followed by a deeper discussion on the engineering strategies developed for cytokine therapies with a focus on the translational relevance of these approaches.
Collapse
Affiliation(s)
- Ivan S Pires
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
10
|
Li J, Lin W, Chen H, Xu Z, Ye Y, Chen M. Dual-target IL-12-containing nanoparticles enhance T cell functions for cancer immunotherapy. Cell Immunol 2020; 349:104042. [PMID: 32061376 DOI: 10.1016/j.cellimm.2020.104042] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 01/10/2020] [Indexed: 01/26/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) play a major role in cancer immunotherapy. A potent tumor immunotherapy may not only require activation of anti-tumor effector cells but also rely on the use of cytokines to create a controlled environment for the development of anti-tumor T cells. In this study, we fabricated a dual-target immunonanoparticle, e.g. poly(d,l-lactide-co-glycolide) nanoparticle, by loading Interleukin-12 (IL-12) and modifying with CD8 and Glypican-3 antibodies on the surface. Our results demonstrate that the fabricated targeting immunonanoparticles bind specifically to the two target cells of interest, i.e. CD8+ T cells and HepG-2 cells via the antibody-antigen interactions and form T cell-HepG-2 cell clusters, which enhances the cytotoxicity of T cells. IL-12-containing dual-target immunonanoparticles delivered IL-12 specifically to CD8+ T cells, and favored the expansion, activation and cytotoxic activity of CD8+ T lymphocytes. These results suggest that dual-target IL-12-encapsulated nanoparticles are a promising platform for cancer immunotherapy.
Collapse
Affiliation(s)
- Jieyu Li
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Wansong Lin
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Huijing Chen
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Zhiping Xu
- Australian Institute for Bioengineering and Nanotechnology (Building 75), The University of Queensland, Cooper Rd., St Lucia, Brisbane, QLD 4072, Australia
| | - Yunbin Ye
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China.
| | - Mingshui Chen
- Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China.
| |
Collapse
|
11
|
Yi L, Chen L, Guo X, Lu T, Wang H, Ji X, Zhang J, Ren Y, Pan P, Kinghorn AD, Huang X, Wang LS, Fan Z, Caligiuri MA, Yu J. A Synthetic Disaccharide Derivative of Diphyllin, TAARD, Activates Human Natural Killer Cells to Secrete Interferon-Gamma via Toll-Like Receptor-Mediated NF-κB and STAT3 Signaling Pathways. Front Immunol 2018; 9:1509. [PMID: 30072983 PMCID: PMC6058043 DOI: 10.3389/fimmu.2018.01509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 06/18/2018] [Indexed: 11/29/2022] Open
Abstract
Natural products and their derivatives have long been used as pharmacological agents in the fight against cancer. Human natural killer (NK) cells are critical in our immune system in that they are capable of destroying tumor cells directly. However, there are few reports that elucidate the role of natural products in activating NK cells. In this study, we discovered that a synthetic disaccharide derivative of diphyllin, 4-O-{[2′′,3′′,4′′-tri-O-acetyl-α-D-arabinopyranosyl-(1′′→4′)]-2′,3′-di-O-acetyl-α-L-rhamnopyranosyl}diphyllin (TAARD), can alone stimulate interferon (IFN)-γ secretion in primary human NK cells and the NKL cell line. Additionally, it had an additive effect with IL-12 or IL-15 on IFN-γ production, but little adverse effects on NK cells. Mechanistically, TAARD induced the phosphorylation of NF-κB and STAT3, resulting in their binding on the IFNG promoter, which was dependent on TLR1 and TLR3 signaling, respectively. STAT3 and NF-κB knockdown with lentivirus shRNA as well as the NF-κB-specific inhibitor, N-tosyl-l-phenylalaninechloromethyl ketone, significantly suppressed TAARD-induced IFN-γ generation in primary NK cells. Blockade of TLR1 and TLR3 with neutralizing antibodies considerably decreased TAARD-induced activation of NF-κB and STAT3, respectively, as well as IFN-γ generation in NK cells. Collectively, our data suggest that TAARD can induce NK cell IFN-γ production through TLR1-NF-κB and TLR3-STAT3 signaling pathways, rendering its potential use as an agent for cancer prevention or treatment.
Collapse
Affiliation(s)
- Long Yi
- Research Center for Nutrition and Food Safety and Third Affiliated Hospital, Third Military Medical University, Chongqing, China.,The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Luxi Chen
- Biomedical Sciences Graduate Program, Medical Scientist Training Program, The Ohio State University, Columbus, OH, United States
| | - Xiaofeng Guo
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.,State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Ting Lu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
| | - Haixia Wang
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Xiaotian Ji
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Jianying Zhang
- Center for Biostatistics, Department of Bioinformatics, The Ohio State University, Columbus, OH, United States
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Pan Pan
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Xiaohua Huang
- Department of Chemistry, The University of Memphis, Memphis, TN, United States
| | - Li-Shu Wang
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Zhijin Fan
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin, China
| | - Michael A Caligiuri
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States.,The James Cancer Hospital, Columbus, OH, United States
| | - Jianhua Yu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States.,Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, United States.,The James Cancer Hospital, Columbus, OH, United States
| |
Collapse
|
12
|
Song X, He J, Xu H, Hu XP, Wu XL, Wu HQ, Liu LZ, Liao CH, Zeng Y, Li Y, Hao Y, Xu CS, Fan L, Zhang J, Zhang HJ, He ZD. The antiviral effects of acteoside and the underlying IFN-γ-inducing action. Food Funct 2018; 7:3017-30. [PMID: 27326537 DOI: 10.1039/c6fo00335d] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
There are many herbal teas that are found in nature that may be effective at treating the symptoms and also shortening the duration of viral infections. When combating viral infections, T lymphocytes are an indispensable part of human acquired immunity. However, studies on the use of natural products in stimulating lymphocyte-mediated interferon-gamma (IFN-γ) production are very limited. In this study, we found that acteoside, a natural phenylpropanoid glycoside from Kuding Tea, enhanced IFN-γ production in mouse lymphocytes in a dose-dependent manner, particularly in the CD4+ and CD8+ subsets of T lymphocytes. To this end, we suggest that the antiviral activity of acteoside was highly correlated to its inducing ability of IFN-γ production. Mechanistically, the activation of T-bet enhanced the promoter of IFN-γ and subsequently resulted in an increased IFN-γ production in T cells. Collectively, we have found a natural product with the capacity to selectively enhance mouse T cell IFN-γ production. Given the role of IFN-γ in the immune system, further studies to clarify the role of acteoside in inducing IFN-γ and prevention of viral infection are needed.
Collapse
Affiliation(s)
- Xun Song
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China. and School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, P. R. China.
| | - Jiang He
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Hong Xu
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Xiao-Peng Hu
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Xu-Li Wu
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Hai-Qiang Wu
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Li-Zhong Liu
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Cheng-Hui Liao
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Yong Zeng
- The First Affiliated Hospital of Kunming Medical University, Kunming 650032, P. R. China
| | - Yan Li
- The First Affiliated Hospital of Kunming Medical University, Kunming 650032, P. R. China
| | - Yue Hao
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Chen-Shu Xu
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Long Fan
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Jian Zhang
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| | - Hong-Jie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, P. R. China.
| | - Zhen-Dan He
- Department of Pharmacy, Shenzhen Key Laboratory of Novel Natural Health Care Products, Innovation Platform for Natural Small Molecule Drugs, Engineering Laboratory of Shenzhen Natural Small Molecule Innovative Drugs, School of Medicine, College of Life Science, Shenzhen University, Shenzhen 518060, P. R. China.
| |
Collapse
|
13
|
Pedrotti LP, Sena AA, Rodriguez Galán MC, Cejas H, Correa SG. Intestinal mononuclear cells primed by systemic interleukin-12 display long-term ability to aggravate colitis in mice. Immunology 2017; 150:290-300. [PMID: 27891587 PMCID: PMC5290244 DOI: 10.1111/imm.12685] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/21/2016] [Accepted: 10/23/2016] [Indexed: 12/20/2022] Open
Abstract
To address whether the burst of systemic interleukin-12 (IL-12) influences intestinal inflammation elicited by luminal stimuli, we induced IL-12 release by cDNA injection in C57BL/6 mice and simultaneously started dextran sulphate sodium administration. The sequence of the inflammatory response triggered by IL-12 release was characterized by assessing myeloperoxidase activity and histological damage in colon samples on days 1, 3, 5 and 7 after colitis induction. To evaluate the persistence of IL-12 priming, colitis was induced in mice 7 or 60 days after cDNA injection. Under IL-12 influence, the development of acute colitis presented a faster and selective infiltration of inflammatory mononuclear cells in the lamina propria. Recruitment was driven by systemic cytokines rather than luminal antigens. Interestingly, when colitis was triggered 7 or 60 days after the cytokine storm, cells maintained the ability to worsen clinical signs of intestinal inflammation. Together, a systemic IL-12 burst effectively primed intestinal cells that became more prone to develop inflammatory responses. Activation was long-lasting because intestinal cells maintained their inflammatory potential and their ability to aggravate colitis.
Collapse
Affiliation(s)
- Luciano P. Pedrotti
- ImmunologyDepartment of Clinical Biochemistry‐CIBICI (CONICET)Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| | - Angela A. Sena
- ImmunologyDepartment of Clinical Biochemistry‐CIBICI (CONICET)Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| | - María Cecilia Rodriguez Galán
- ImmunologyDepartment of Clinical Biochemistry‐CIBICI (CONICET)Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| | - Hugo Cejas
- ImmunologyDepartment of Clinical Biochemistry‐CIBICI (CONICET)Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| | - Silvia G. Correa
- ImmunologyDepartment of Clinical Biochemistry‐CIBICI (CONICET)Facultad de Ciencias Químicas, Universidad Nacional de CórdobaCórdobaArgentina
| |
Collapse
|
14
|
Chertok B, Langer R, Anderson DG. Spatial Control of Gene Expression by Nanocarriers Using Heparin Masking and Ultrasound-Targeted Microbubble Destruction. ACS NANO 2016; 10:7267-7278. [PMID: 27472268 PMCID: PMC5240524 DOI: 10.1021/acsnano.6b01199] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
We developed a method to spatially control gene expression following nonviral delivery of DNA. This method includes surface-modifying DNA nanocarriers with heparin to inhibit passive gene transfer in both the target and the off-target tissues and using ultrasound-targeted microbubble destruction (UTMD) to selectively activate heparin-inhibited gene transfer at the target site. We observed that the engraftment of heparin onto the surface of cationic liposomes reduced off-target gene expression in the liver, a major site of nanoplex accumulation, by more than 700-fold compared to the nonheparinized PEGylated liposomes. We further observed that tumor-directed UTMD increased gene transfer with heparin-modified nanoplexes by more than 10-fold. This method augmented tumor-to-liver selectivity of gene expression by 4000-fold compared to controls. We conclude that heparinization of DNA nanocarriers in conjunction with localized activation of gene transfer by UTMD may enable greater spatial control over genetic therapy.
Collapse
Affiliation(s)
- Beata Chertok
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan , Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering, College of Engineering, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Robert Langer
- Department of Chemical Engineering, MIT , Cambridge, Massachusetts 02139, United States
- David H. Koch Institute for Integrative Cancer Research, MIT , Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering & Science, MIT , Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts 02139, United States
| | - Daniel G Anderson
- Department of Chemical Engineering, MIT , Cambridge, Massachusetts 02139, United States
- David H. Koch Institute for Integrative Cancer Research, MIT , Cambridge, Massachusetts 02139, United States
- Institute for Medical Engineering & Science, MIT , Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
15
|
Mann K, Kullberg M. Trastuzumab-targeted gene delivery to Her2-overexpressing breast cancer cells. Cancer Gene Ther 2016; 23:221-8. [PMID: 27199219 PMCID: PMC4946976 DOI: 10.1038/cgt.2016.21] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 04/06/2016] [Accepted: 04/07/2016] [Indexed: 12/23/2022]
Abstract
We describe a novel gene delivery system that specifically targets human epidermal growth factor receptor 2 (Her2)-overexpressing breast cancer cells. The targeting complexes consist of a PEGylated polylysine core that is bound to DNA molecules coding for either green fluorescent protein or shrimp luciferase. The complex is disulfide linked to the monoclonal antibody trastuzumab and to a pore-forming protein, Listeriolysin O (LLO). Trastuzumab is responsible for specific targeting of Her2 receptors and uptake of the gene delivery complex into endosomes of recipient cells, whereas LLO ensures that the DNA molecules are capable of transit from the endosomes into the cytoplasm. Omission of either trastuzumab or LLO from the nanocomplexes results in minimal gene product in targeted cells. Treatment of isogeneic MCF7 and MCF7/Her18 cell lines, differing only in number of Her2 receptors, with the complete gene delivery system results in a 30-fold greater expression of luciferase activity in the Her2-overexpressing MCF7/Her18 cells. Our nanocomplexes are small (150–250 nm), stable to storage, nontoxic and generic in make-up such that any plasmid DNA or antibody specific for cell-surface receptors can be coupled to the PEGylated polylysine core.
Collapse
Affiliation(s)
- K Mann
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK, USA.,WWAMI Medical Education Program, University of Alaska Anchorage, Anchorage, AK, USA
| | - M Kullberg
- WWAMI Medical Education Program, University of Alaska Anchorage, Anchorage, AK, USA
| |
Collapse
|
16
|
Vo JL, Yang L, Kurtz SL, Smith SG, Koppolu BP, Ravindranathan S, Zaharoff DA. Neoadjuvant immunotherapy with chitosan and interleukin-12 to control breast cancer metastasis. Oncoimmunology 2015; 3:e968001. [PMID: 25964864 PMCID: PMC4352958 DOI: 10.4161/21624011.2014.968001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/17/2014] [Indexed: 12/15/2022] Open
Abstract
Metastasis accounts for approximately 90% of breast cancer-related deaths. Therefore, novel approaches which prevent or control breast cancer metastases are of significant clinical interest. Interleukin-12 (IL-12)-based immunotherapies have shown promise in controlling metastatic disease, yet modest responses and severe toxicities due to systemic administration of IL-12 in early trials have hindered clinical application. We hypothesized that localized delivery of IL-12 co-formulated with chitosan (chitosan/IL-12) could elicit tumor-specific immunity and provide systemic protection against metastatic breast cancer while minimizing systemic toxicity. Chitosan is a biocompatible polysaccharide derived primarily from the exoskeletons of crustaceans. In a clinically relevant resection model, mice bearing spontaneously metastatic 4T1 mammary adenocarcinomas received intratumoral injections of chitosan/IL-12, or appropriate controls, prior to tumor resection. Neoadjuvant chitosan/IL-12 immunotherapy resulted in long-term tumor-free survival in 67% of mice compared to only 24% or 0% of mice treated with IL-12 alone or chitosan alone, respectively. Antitumor responses following chitosan/IL-12 treatment were durable and provided complete protection against rechallenge with 4T1, but not RENCA renal adenocarcinoma, cells. Lymphocytes from chitosan/IL-12-treated mice demonstrated robust tumor-specific lytic activity and interferon-γ production. Cell-mediated immune memory was confirmed in vivo via clinically relevant delayed-type hypersensitivity (DTH) assays. Comprehensive hematology and toxicology analyses revealed that chitosan/IL-12 induced transient, reversible leukopenia with no changes in critical organ function. Results of this study suggest that neoadjuvant chitosan/IL-12 immunotherapy prior to breast tumor resection is a promising translatable strategy capable of safely inducing to tumor-specific immunity and, in the long term, reducing breast cancer mortality due to progressive recurrences.
Collapse
Key Words
- ALT, alanine aminotransferase
- CBC, complete blood count
- CTL, cytotoxic T lymphocyte
- DTH, delayed-type hypersensitivity
- ELISPOT, enzyme-linked immunosorbent spot
- Gy, gray
- IFNγ, interferon-γ;. IL-12, interleukin-12
- RENCA, renal cell adenocarcinoma
- TNBC, triple-negative breast cancer
- chitosan
- cytokine delivery
- i.p., intraperitoneal
- i.t., intratumoral
- interleukin-12
- metastatic breast cancer
- neoadjuvant immunotherapy
- s.c., subcutaneous
Collapse
Affiliation(s)
- Jimmy Ln Vo
- Department of Biomedical Engineering; University of Arkansas ; Fayetteville, AR USA
| | - Lirong Yang
- Department of Biomedical Engineering; University of Arkansas ; Fayetteville, AR USA
| | - Samantha L Kurtz
- Department of Biomedical Engineering; University of Arkansas ; Fayetteville, AR USA
| | - Sean G Smith
- Department of Biomedical Engineering; University of Arkansas ; Fayetteville, AR USA
| | | | | | - David A Zaharoff
- Department of Biomedical Engineering; University of Arkansas ; Fayetteville, AR USA
| |
Collapse
|
17
|
Deng Y, Chu J, Ren Y, Fan Z, Ji X, Mundy-Bosse B, Yuan S, Hughes T, Zhang J, Cheema B, Camardo AT, Xia Y, Wu LC, Wang LS, He X, Kinghorn AD, Li X, Caligiuri MA, Yu J. The natural product phyllanthusmin C enhances IFN-γ production by human NK cells through upregulation of TLR-mediated NF-κB signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:2994-3002. [PMID: 25122922 DOI: 10.4049/jimmunol.1302600] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Natural products are a major source for cancer drug development. NK cells are a critical component of innate immunity with the capacity to destroy cancer cells, cancer-initiating cells, and clear viral infections. However, few reports describe a natural product that stimulates NK cell IFN-γ production and unravel a mechanism of action. In this study, through screening, we found that a natural product, phyllanthusmin C (PL-C), alone enhanced IFN-γ production by human NK cells. PL-C also synergized with IL-12, even at the low cytokine concentration of 0.1 ng/ml, and stimulated IFN-γ production in both human CD56(bright) and CD56(dim) NK cell subsets. Mechanistically, TLR1 and/or TLR6 mediated PL-C's activation of the NF-κB p65 subunit that in turn bound to the proximal promoter of IFNG and subsequently resulted in increased IFN-γ production in NK cells. However, IL-12 and IL-15Rs and their related STAT signaling pathways were not responsible for the enhanced IFN-γ secretion by PL-C. PL-C induced little or no T cell IFN-γ production or NK cell cytotoxicity. Collectively, we identify a natural product with the capacity to selectively enhance human NK cell IFN-γ production. Given the role of IFN-γ in immune surveillance, additional studies to understand the role of this natural product in prevention of cancer or infection in select populations are warranted.
Collapse
Affiliation(s)
- Youcai Deng
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210; Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Jianhong Chu
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210
| | - Zhijin Fan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | - Xiaotian Ji
- State Key Laboratory of Elemento-Organic Chemistry, Nankai University, Tianjin 300071, China
| | | | - Shunzong Yuan
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210; Department of Lymphoma, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Tiffany Hughes
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Jianying Zhang
- Center for Biostatistics, The Ohio State University, Columbus, OH 43210
| | - Baljash Cheema
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210
| | - Andrew T Camardo
- The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210
| | - Yong Xia
- Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Lai-Chu Wu
- Department of Molecular and Cellular Biochemistry, The Ohio State University College of Medicine, Columbus, OH 43210
| | - Li-Shu Wang
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing 400038, China;
| | - Michael A Caligiuri
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210; The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210;
| |
Collapse
|
18
|
Salem ML, Shoukry NM, Zidan AAA, Vournakis J. Immunomodulatory effects of IL-12 released from poly-N-acetyl glucosamine gel matrix during schistosomiasis infection. Cytotechnology 2014; 66:667-675. [PMID: 23884721 PMCID: PMC4082772 DOI: 10.1007/s10616-013-9620-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 07/15/2013] [Indexed: 02/07/2023] Open
Abstract
We have reported recently that Interleukin-12 (IL-12) released from poly-N-acetyl glucosamine gel matrix (F2 gel/IL-12) is more effective than free IL-12 to enhance vaccination of mice with Schistosoma soluble worm antigen preparation. The aim of this study is to evaluate the effect of F2 gel/IL-12 on the inflammatory responses in mice undergoing schistosomiasis infection in absence of vaccination. To achieve this, mice undergoing Schistosoma mansoni infection or cured from this infection, after treatment with praziquantil (PZQ), were treated with subcutaneous injection of IL-12 for 3 consecutive days or once with F2 gel loaded with IL-12 (F2 gel/IL-12). The treatment was started on day 35 days after infection. For infection, mice were infected with 100 cercariae of S. mansoni using tail immersion method. We found that treatment with F2 gel/IL-12 induced significant decreases in the egg burden with a moderate reduction in the size of granuloma and decrease in the cellular granulomatous reaction in the lung as compared to infected mice treated with IL-12. These effects of F2 gel/IL-12 were more pronounced in infected mice previously treated with the anti-schistosomal drug PZQ. The total numbers of white blood cells in all treated mice showed similar profile. Treatment with IL-12 or F2 gel/IL-12, however, showed significant reduction in the number of mononuclear cells when compared with non-treated infected mice. In conclusion, this study showed the ability of IL-12 released from F2 gel to lower the inflammatory response to Schistosoma infection even in absence of vaccination.
Collapse
Affiliation(s)
- Mohamed L Salem
- Immunology and Biotechnology Division, Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt,
| | | | | | | |
Collapse
|
19
|
Kullberg M, McCarthy R, Anchordoquy TJ. Gene delivery to Her-2+ breast cancer cells using a two-component delivery system to achieve specificity. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 10:1253-62. [PMID: 24632244 DOI: 10.1016/j.nano.2014.02.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 01/31/2014] [Accepted: 02/28/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED Current liposomal gene delivery systems predominately utilize cationic lipids, which efficiently bind and deliver DNA plasmid, but also result in nonspecific gene expression in lung and liver tissue. To improve specificity, a two-component delivery strategy employing neutral liposomes was used to target breast cancers positive for the human epidermal growth factor receptor 2 (Her-2). The first component consisted of plasmid DNA condensed with cationic polyethylene glycol (PEG) modified polylysine (PL/DNA). The second component was a neutral Her-2 targeting liposome conjugated to the pore-forming protein, Listeriolysin O (LLO). Independently, PL/DNA delivery resulted in low expression of plasmid DNA. However, when PL/DNA and LLO/liposomes co-localized within an endosome, LLO disrupted endosome integrity, leading to cytoplasmic delivery and expression of the plasmid. When used to deliver a plasmid encoding the luciferase gene, this two-component system resulted in gene expression that was 268-fold greater in Her-2 positive cells than in Her-2 negative cells. FROM THE CLINICAL EDITOR In this paper a novel two-component gene delivery method is presented using PL/DNA and LLO liposomes, demonstrating strongly significant results in a model system.
Collapse
Affiliation(s)
- Max Kullberg
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO.
| | - Ryan McCarthy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO
| | - Thomas J Anchordoquy
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO
| |
Collapse
|
20
|
Fraser CK, Diener KR, Brown MP, Hayball JD. Improving vaccines by incorporating immunological coadjuvants. Expert Rev Vaccines 2014; 6:559-78. [PMID: 17669010 DOI: 10.1586/14760584.6.4.559] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
While vaccination continues to be the most successful interventionist health policy to date, infectious disease remains a significant cause of death worldwide. A primary reason that vaccination is not able to generate effective immunity is a lack of appropriate adjuvants capable of initiating the desired immune response. Adjuvant combinations can potentially overcome this problem; however, the possible permutations to consider, which include the route and kinetics of vaccination, as well as combinations of adjuvants, are practically limitless. This review aims to summarize the current understanding of adjuvants and related immunological processes and how this knowledge can and has been applied to the strategic selection of adjuvant combinations as components of vaccines against human infectious disease.
Collapse
Affiliation(s)
- Cara K Fraser
- Experimental Therapeutics Laboratory, Hanson Institute, and School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Australia.
| | | | | | | |
Collapse
|
21
|
Kullberg M, McCarthy R, Anchordoquy TJ. Systemic tumor-specific gene delivery. J Control Release 2013; 172:730-6. [PMID: 24035974 DOI: 10.1016/j.jconrel.2013.08.300] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 08/20/2013] [Accepted: 08/22/2013] [Indexed: 12/19/2022]
Abstract
The objective of a systemically administered cancer gene therapy is to achieve gene expression that is isolated to the tumor tissue. Unfortunately, viral systems have strong affinity for the liver, and delivery from non-viral cationic systems often results in high expression in the lungs. Non-specific delivery to these organs must be overcome if tumors are to be aggressively treated with genes such as IL-12 which activates a tumor immune response, and TNF-alpha which can induce tumor cell apoptosis. Techniques which have led to specific expression in tumor tissue include receptor targeting through ligand conjugation, utilization of tumor specific promoters and viral mutation in order to take advantage of proteins overexpressed in tumor cells. This review analyzes these techniques applied to liposomal, PEI, dendrimer, stem cell and viral gene delivery systems in order to determine the techniques that are most effective in achieving tumor specific gene expression after systemic administration.
Collapse
Affiliation(s)
- Max Kullberg
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, 12850 Montview Boulevard, Aurora, Colorado 80045, USA.
| | | | | |
Collapse
|
22
|
Abstract
ISG15 is a well-known intracellular ubiquitin-like molecule involved in ISGylation. However, a recent study has revived the notion first put forward two decades ago that ISG15 is also a secreted molecule. Human neutrophils, monocytes and lymphocytes can release ISG15, even though this protein has no detectable signal peptide sequence. ISG15 has also been found in the secretory granules of granulocytes. The mechanism underlying ISG15 secretion is unknown. Secreted ISG15 acts on at least T and natural killer (NK) lymphocytes, in which it induces interferon (IFN)-γ production. However, the mechanism by which ISG15 stimulates these cells also remains unclear. ISG15 and IFN-γ seem to define an innate circuit that operates preferentially, but not exclusively, between granulocytes and NK cells. Inherited ISG15 deficiency is associated with severe mycobacterial disease in both mice and humans. This infectious phenotype probably results from the lack of secreted ISG15, because patients and mice with other inborn errors of IFN-γ immunity also display mycobacterial diseases. In addition to raising mechanistic issues, the studies described here pave the way for clinical studies of various aspects, ranging from the use of recombinant ISG15 in patients with infectious diseases to the use of ISG15-blocking agents in patients with inflammatory diseases.
Collapse
|
23
|
Christian DA, Hunter CA. Particle-mediated delivery of cytokines for immunotherapy. Immunotherapy 2012; 4:425-41. [PMID: 22512636 DOI: 10.2217/imt.12.26] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The ability of cytokines to direct the immune response to vaccination, infection and tumors has motivated their use in therapy to augment or shape immunity. To avoid toxic side effects associated with systemic cytokine administration, several approaches have been developed using particle-encapsulated cytokines to deliver this cargo to specific cell types and tissues. Initial work used cytokine-loaded particles to deliver proinflammatory cytokines to phagocytes to enhance antimicrobial and antitumor responses. These particles have also been used to create a cytokine depot at a local site to supplement prophylactic or antitumor vaccines or injected directly into solid tumors to activate immune cells to eliminate established tumors. Finally, recent advances have revealed that paracrine delivery of cytokines directly to T cells has the potential to enhance T-cell mediated therapies. The studies reviewed here highlight the progress in the last 30 years that has established the potential of particle-mediated cytokine immunotherapy.
Collapse
Affiliation(s)
- David A Christian
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
24
|
Xu Q, Guo L, Gu X, Zhang B, Hu X, Zhang J, Chen J, Wang Y, Chen C, Gao B, Kuang Y, Wang S. Prevention of colorectal cancer liver metastasis by exploiting liver immunity via chitosan-TPP/nanoparticles formulated with IL-12. Biomaterials 2012; 33:3909-18. [PMID: 22374455 DOI: 10.1016/j.biomaterials.2012.02.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 02/06/2012] [Indexed: 11/30/2022]
Abstract
The development of effective therapies for the prevention of colorectal cancer (CRC) liver metastasis is of great importance. Recently, chitosan (CS) nanoparticles have been utilized as carriers of interluekin-12 (IL-12) administered locally to deliver therapeutic proteins and genes. In this study, we encapsulated IL-12 by incorporation using tripolyphosphate (TPP) as the coacervated crosslinking agent to form CS-TPP/IL-12 nanoparticles. We further characterized the association efficiency, rate of release, liver-targeting, and toxicity, which were predominantly dependent on the factors of particle size, zeta potential, pH of solution, and whether or not modified with TPP. Systemic delivery of CS-TPP/IL-12 nanoparticles significantly reduced the number and volume of CRC liver metastasis foci compared to the CS-TPP treated mouse group. Although delivery of IL-12 alone also inhibited the number of CRC liver metastasis observed, further study of the change in hepatic metastasis volume demonstrated no significant differences between the groups treated with CS-TPP or IL-12 alone. Mechanistically, CS-TPP nanoparticles blocked the toxicity of IL-12 and induced infiltration of NK cells and some T cells, which are most likely the effector cells that mediate tumor metastasis inhibition during CS-TPP/IL-12 immunotherapy. The results obtained from this study demonstrate the potential benefit of using chitosan modification technology as a cytokine delivery system for the successful prevention of CRC liver metastasis by exploiting liver immunity.
Collapse
Affiliation(s)
- Qiongming Xu
- Department of Pharmaceutical Chemistry, Soochow University College of Pharmaceutical Science, Suzhou 215123, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Alenzi FQ, Alenazi FA, Al-Kaabi Y, Salem ML. The use of growth factors to modulate the activities of antigen-specific CD8+ T cells in vitro. J Med Life 2011; 4:399-406. [PMID: 22514573 PMCID: PMC3227152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 09/29/2011] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Adoptive T cell therapy depends on the harvesting of the cells from the host, their activation in vitro, and their infusion back to the same host. The way of activating the T cells in vitro is a critical factor for their homing, survival and function in vivo. Sustaining T cell homing molecules, particularly CD62L, is benefic for the trafficking of the adoptive transferred cells. OBJECTIVE The aim of the present study is to test whether insulin-like growth factor-1 (IGF-1), thymosin- α1 (T-α1) as well as all-trans retinoid acid (ATRA) alone or in combination with IL-2, IL-12, IL-15 can enhance the activation and survival phenotypes of antigen-activated T cells in vitro. METHODS & RESULTS To this end, OT-1 transgenic T cells were used as a model. These CD8+ T cells recognize OVA peptide presented by MHC class-I. The results showed that antigen stimulation of OT1 cells resulted in their activation as evidenced by the decrease in surface expression of CD62L, analyzed for 3 days after antigen stimulation and was more pronounced on day 5. The addition of IL-12 or IGF-1 alone but not of IL-2, IL-15 augmented OT-1 cell activation measured on day 5. Interestingly, the combination of IL-12 with IGF-1 sustained the expression of CD62L on OT1 cells. Although the addition of ATRA alone or in combination with IL-12 resulted in decreases in CD62L expression on day 3, they showed a dose-dependent effect on the restoration of CD62L expression on day 5. The analysis of the activation-induced cell death (apoptosis) of OT1 cells showed an increased rate of death on day 5 than on day 3-post antigen stimulation. The addition of only IL-12 or IGF-1 alone, but not of IL-2, IL-15 or T- α1, decreased OT1 cell apoptosis on day 3. These anti-apoptotic effects of IL-12 and IGF- 1, however, were recovered on day 5-post stimulation. DISCUSSION In conclusion, these results indicate that the activation phenotype and the survival of antigen-specific T cells can be differently modulated by immunomodulatory factors, where, interleukin-12 and IGF-1 induced the favorable effect. These results have a significant implication for T cell adoptive immunotherapy in different settings.
Collapse
Affiliation(s)
- F Q Alenzi
- College of Applied Medical Sciences, Prince Salman University, Saudi Arabia.
| | | | | | | |
Collapse
|
26
|
Giuliani N, Airoldi I. Novel insights into the role of interleukin-27 and interleukin-23 in human malignant and normal plasma cells. Clin Cancer Res 2011; 17:6963-70. [PMID: 21880791 DOI: 10.1158/1078-0432.ccr-11-1724] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multiple myeloma is a monoclonal postgerminal center tumor that has phenotypic features of plasmablasts and/or plasma cells and usually localizes at multiple sites in the bone marrow. The pathogenesis of multiple myeloma is complex and dependent on the interactions between tumor cells and their microenvironment. Different cytokines, chemokines, and proangiogenic factors released in the tumor microenvironment are known to promote multiple myeloma cell growth. Here, we report recent advances on the role of 2 strictly related immunomodulatory cytokines, interleukin-27 (IL-27) and IL-23, in human normal and neoplastic plasma cells, highlighting their ability to (i) act directly against multiple myeloma cells, (ii) influence the multiple myeloma microenvironment by targeting osteoclast and osteoblast cells, and (iii) modulate normal plasma cell function. Finally, the therapeutic implication of these studies is discussed.
Collapse
Affiliation(s)
- Nicola Giuliani
- Hematology and Blood and Marrow Transplantation (BMT) Center, University of Parma, Parma, Italy
| | | |
Collapse
|
27
|
Abstract
IL-12 is a potent antitumor cytokine that exhibits significant clinical toxicities after systemic administration. We hypothesized that intratumoral (i.t.) administration of IL-12 coformulated with the biodegradable polysaccharide chitosan could enhance the antitumor activity of IL-12 while limiting its systemic toxicity. Noninvasive imaging studies monitored local retention of IL-12, with and without chitosan coformulation, after i.t. injection. Antitumor efficacy of IL-12 alone and IL-12 coformulated with chitosan (chitosan/IL-12) was assessed in mice bearing established colorectal (MC32a) and pancreatic (Panc02) tumors. Additional studies involving depletion of immune cell subsets, tumor rechallenge, and CTL activity were designed to elucidate mechanisms of regression and tumor-specific immunity. Coformulation with chitosan increased local IL-12 retention from 1 to 2 days to 5 to 6 days. Weekly i.t. injections of IL-12 alone eradicated ≤10% of established MC32a and Panc02 tumors, while i.t. chitosan/IL-12 immunotherapy caused complete tumor regression in 80% to 100% of mice. Depletion of CD4(+) or Gr-1(+) cells had no impact on chitosan/IL-12-mediated tumor regression. However, CD8(+) or NK cell depletion completely abrogated antitumor activity. I.t. chitosan/IL-12 immunotherapy generated systemic tumor-specific immunity, as >80% of mice cured with i.t. chitosan/IL-12 immunotherapy were at least partially protected from tumor rechallenge. Furthermore, CTLs from spleens of cured mice lysed MC32a and gp70 peptide-loaded targets. Chitosan/IL-12 immunotherapy increased local retention of IL-12 in the tumor microenvironment, eradicated established, aggressive murine tumors, and generated systemic tumor-specific protective immunity. Chitosan/IL-12 is a well-tolerated, effective immunotherapy with considerable potential for clinical translation.
Collapse
|
28
|
Whitworth JM, Alvarez RD. Evaluating the role of IL-12 based therapies in ovarian cancer: a review of the literature. Expert Opin Biol Ther 2011; 11:751-62. [DOI: 10.1517/14712598.2011.566854] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
29
|
Salem ML, Demcheva M, Gillanders WE, Cole DJ, Vournakis JN. Poly-N-acetyl glucosamine gel matrix as a non-viral delivery vector for DNA-based vaccination. Anticancer Res 2010; 30:3889-3894. [PMID: 21036699 PMCID: PMC3398836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intramuscular administration of plasmid DNA vaccines is one of the main delivery approaches that can generate antigen specific T cell responses. However, major limitations of the intramuscular delivery strategy are the low level of myocyte transfection, resulting in a minimal level of protein expression; the inability to directly target antigen presenting cells, in particular dendritic cells, which are critical for establishment of efficacious antigen-specific immune responses. Although several viral vectors have been designed to improve plasmid DNA delivery, they have limitations, including the generation of neutralizing antibodies in addition to lacking the simplicity and versatility required for universal clinical application. We have developed an inexpensive non-viral delivery vector based on the polysaccharide polymer poly-N-acetyl glucosamine with the capability to target dendritic cells. This vector is fully biocompatible, biodegradable, and nontoxic. The advantage of the application of this delivery system relative to other approaches is discussed.
Collapse
Affiliation(s)
- Mohamed L Salem
- Zoology Department, Faculty of Science, Tanta University, Egypt.
| | | | | | | | | |
Collapse
|
30
|
Ferretti E, Di Carlo E, Cocco C, Ribatti D, Sorrentino C, Ognio E, Montagna D, Pistoia V, Airoldi I. Direct inhibition of human acute myeloid leukemia cell growth by IL-12. Immunol Lett 2010; 133:99-105. [PMID: 20705102 DOI: 10.1016/j.imlet.2010.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 07/22/2010] [Accepted: 08/02/2010] [Indexed: 11/19/2022]
Abstract
Acute myeloid leukemia is a haematopoietic malignancy originating from the transformation of myeloid progenitors that proliferate and accumulate in the bone marrow. In AML patients the survival rate at 5 years is 40-50% highlighting the need for novel therapies. In this study we have asked whether IL-12, an immuno-modulatory cytokine with anti-tumor activity, may inhibit directly AML cell growth. We show that the human AML cell lines U937, K562 and THP-1 expressed both chains of the IL-12 receptor (R), i.e. IL-12Rβ1 and IL-12Rβ2. IL-12 inhibited the angiogenic potential of AML cells in vitro, but did not affect their survival or proliferation. In vivo experiments were performed using SCID-NOD mice injected intraperitoneally (i.p.) with the human U937 AML cell line and subsequently treated with human recombinant IL-12 or PBS i.p. Histological, immunohistochemical and flow cytometric analyses on explanted tumors revealed that IL-12 reduced new vessel formation, induced apoptosis and inhibited tumor cell proliferation. Studies on a panel of angiogenesis related genes in explanted tumors using PCR arrays showed significantly down-regulated expression of numerous pro-angiogenic genes including VEGF-C, IL-6, IL-8, CXCL1, CXCL6 and alanyl aminopeptidase in IL-12 vs PBS treated mice. This study shows for the first time that IL-12 targets directly AML cell growth and paves the way to further investigation of IL-12 as potential drug for AML treatment.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/pharmacology
- Humans
- Interleukin-12/administration & dosage
- Interleukin-12/pharmacology
- K562 Cells
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Mice
- Mice, SCID
- Neoplasm Transplantation
- Neovascularization, Pathologic/genetics
- Receptors, Interleukin-12/genetics
- Receptors, Interleukin-12/immunology
- Receptors, Interleukin-12/metabolism
- U937 Cells
Collapse
Affiliation(s)
- Elisa Ferretti
- Laboratory of Oncology, G. Gaslini Institute, Largo Gaslini 5, 16148 Genova, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fewell JG, Matar MM, Rice JS, Brunhoeber E, Slobodkin G, Pence C, Worker M, Lewis DH, Anwer K. Treatment of disseminated ovarian cancer using nonviral interleukin-12 gene therapy delivered intraperitoneally. J Gene Med 2009; 11:718-28. [PMID: 19507172 DOI: 10.1002/jgm.1356] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The poor prognosis associated with ovarian cancer is primarily the result of delayed diagnosis and the lack of an effective treatment for advanced disease. Use of novel immunotherapy strategies are being evaluated that work to enhance local and systemic immune responses against cancer cells and can possibly work together with traditional cytotoxic chemotherapy regimens to produce more effective treatment options. METHODS In the present study, we describe a gene-based therapy whereby the anticancer cytokine interleukin-12 gene (pmIL-12) is formulated with a synthetic polymeric delivery vehicle (PPC) and administered intraperitoneally into a mouse model of disseminated ovarian cancer. RESULTS The administration of pmIL-12/PPC in tumor-bearing mice was associated with a shift towards a Th1 immune state, including significant increases in murine IL-12 (mIL-12) and interferon (IFN)-gamma (mIFN-gamma) in ascites fluid, with little change in systemic levels of these proteins. The mIL-12 protein was detectable for several days and could be reintroduced with subsequent injections. We show that treatment delayed the onset of ascites formation and improved survival in a dose-dependent manner. A significant decrease in vascular endothelial growth factor was associated with pmIL-12/PPC delivery and believed to play a predominant role in inhibiting ascites accumulation. Administration of pmIL-12/PPC was associated with minimal toxicity and, when combined with standard chemotherapies, resulted in additive improvement in survival. CONCLUSIONS Taken together, these results suggest that pmIL-12/PPC may be an effective strategy for inhibiting progression of disseminated ovarian cancer and may offer a new option for treatment of advanced disease that can be used to complement standard therapies.
Collapse
|
32
|
Pistoia V, Cocco C, Airoldi I. Interleukin-12 receptor beta2: from cytokine receptor to gatekeeper gene in human B-cell malignancies. J Clin Oncol 2009; 27:4809-16. [PMID: 19720917 DOI: 10.1200/jco.2008.21.3579] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin (IL) -12 is a cytokine that has been extensively characterized for its immunoregulatory activities. IL-12 binds to a heterodimeric receptor composed of the beta1 and beta2 chains. In this review article, we discuss recent findings on the expression and function of IL-12 receptor (IL-12R) in malignant B cells frozen at various stages of differentiation and in their normal counterparts. These studies, together with others performed in Il12rb2 knockout mice, have established the concept that the IL-12Rbeta2 gene is a gatekeeper from cancer. We will delineate three paradigms reflecting the differential expression of IL-12Rbeta2 in different groups of malignant B cells and discuss the therapeutic perspectives stemming from these studies.
Collapse
Affiliation(s)
- Vito Pistoia
- Laboratory of Oncology, Department of Experimental and Laboratory Medicine, G. Gaslini Institute, 16147 Genova, Italy.
| | | | | |
Collapse
|
33
|
Zaharoff DA, Hoffman BS, Hooper HB, Benjamin CJ, Khurana KK, Hance KW, Rogers CJ, Pinto PA, Schlom J, Greiner JW. Intravesical immunotherapy of superficial bladder cancer with chitosan/interleukin-12. Cancer Res 2009; 69:6192-9. [PMID: 19638573 PMCID: PMC2788203 DOI: 10.1158/0008-5472.can-09-1114] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intravesical BCG has been used successfully to treat superficial bladder cancer for three decades. However, 20% to 30% of patients will fail initial BCG therapy and 30% to 50% of patients will develop recurrent tumors within 5 years. Alternative or complementary strategies for the management of superficial bladder cancer are needed. Interleukin-12 (IL-12) is a potent T(H)1 cytokine with robust antitumor activity and the ability to potentiate immunologic memory. Unfortunately, intravesical IL-12 did not show antitumor efficacy in a recent clinical study of patients with recurrent superficial bladder cancer. We hypothesized that coformulation of IL-12 with chitosan, a biocompatible, mucoadhesive polysaccharide, could improve intravesical IL-12 delivery and provide an effective and durable alternative for the treatment of superficial bladder cancer. In antitumor studies, 88% to 100% of mice bearing orthotopic bladder tumors were cured after four intravesical treatments with chitosan/IL-12. In contrast, only 38% to 60% of mice treated with IL-12 alone and 0% treated with BCG were cured. Antitumor responses following chitosan/IL-12 treatments were durable and provided complete protection from intravesical tumor rechallenge. Urinary cytokine analysis showed that chitosan/IL-12 induced multiple T(H)1 cytokines at levels significantly higher than either IL-12 alone or BCG. Immunohistochemistry revealed moderate to intense tumor infiltration by T cells and macrophages following chitosan/IL-12 treatments. Bladder submucosa from cured mice contained residual populations of immune cells that returned to baseline levels after several months. Intravesical chitosan/IL-12 is a well-tolerated, effective immunotherapy that deserves further consideration for testing in humans for the management of superficial bladder cancer.
Collapse
Affiliation(s)
- David A. Zaharoff
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Benjamin S. Hoffman
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - H. Brooks Hooper
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Compton J. Benjamin
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kiranpreet K. Khurana
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kenneth W. Hance
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Connie J. Rogers
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter A. Pinto
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - John W. Greiner
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
34
|
IL-12 delivered intratumorally by multilamellar liposomes reactivates memory T cells in human tumor microenvironments. Clin Immunol 2009; 132:71-82. [PMID: 19395317 DOI: 10.1016/j.clim.2009.03.516] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 02/16/2009] [Accepted: 03/17/2009] [Indexed: 01/07/2023]
Abstract
Using a novel loading technique, IL-12 is reported here to be efficiently encapsulated within large multilamellar liposomes. The preclinical efficacy of the cytokine loaded liposomes to deliver IL-12 into human tumors and to reactive tumor-associated T cells in situ is tested using a human tumor xenograft model. IL-12 is released in vivo from these liposomes in a biologically active form when injected into tumor xenografts that are established by the subcutaneous implantation of non-disrupted pieces of human lung, breast or ovarian tumors into immunodeficient mice. The histological architecture of the original tumor tissue, including tumor-associated leukocytes, tumor cells and stromal cells is preserved anatomically and the cells remain functionally responsive to cytokines in these xenografts. The local and sustained release of IL-12 into the tumor microenvironment reactivates tumor-associated quiescent effector memory T cells to proliferate, produce and release IFN-gamma resulting in the killing of tumor cells in situ. Very little IL-12 is detected in the serum of mice for up to 5 days after an intratumoral injection of the IL-12 liposomes. We conclude that IL-12 loaded large multilamellar liposomes provide a safe method for the local and sustained delivery of IL-12 to tumors and a therapeutically effective way of reactivating existing tumor-associated T cells in human solid tumor microenvironments. The potential of this local in situ T cell re-stimulation to induce a systemic anti-tumor immunity is discussed.
Collapse
|
35
|
Liu C, Towler MJ, Medrano G, Cramer CL, Weathers PJ. Production of mouse interleukin-12 is greater in tobacco hairy roots grown in a mist reactor than in an airlift reactor. Biotechnol Bioeng 2009; 102:1074-86. [PMID: 18988263 DOI: 10.1002/bit.22154] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
We compared the growth and productivity of a tobacco line of hairy roots that produces murine interleukin 12 (mIL-12) grown in three different culture systems: shake flasks, an airlift reactor, and a scalable mist reactor. Of the total mIL-12 produced by cultures grown in shake flasks ( approximately 434.8 microg L(-1)), almost 21% was recovered from the medium. In contrast to roots harvested from shake flasks and the mist reactor, roots were not uniformly distributed in the airlift reactor. Roots formed a dense ring around the wall of the reactor and surrounding the central rising column of fine aeration bubbles. Root quality was also better in both the shake flasks and mist reactor than in the airlift reactor. There were more pockets of dark roots in the airlift reactor suggesting some of the roots were nutrient starved. Although the best root growth (7 g DW L(-1)) was in the shake flasks, both reactors produced about the same, but less dry mass, nearly 5 g DW L(-1). Total mIL-12 concentration was highest in the mist reactor at 5.3 microg g(-1) FW, but productivity, 31 microg g(-1) FW day(-1) was highest in shake flasks. Roots grown in the mist reactor produced about 49.5% more mIL-12 than roots grown in the airlift reactor. Protease activity in the media increased steadily during culture of the roots in all three systems. The comparisons of protease activity, protein and mIL-12 levels done in the shake flask system suggest that the increase in proteases associated with progression into stationary phase is most detrimental to mIL-12 concentration. This is the first description of the design and operation of a scalable version of a mist bioreactor that uses a plastic bag. This also the first report of reasonable production levels of functional mIL-12, or any protein, produced by hairy roots grown in a mist reactor. Results will prove useful for further optimization and scale-up studies of plant-produced therapeutic proteins.
Collapse
Affiliation(s)
- Chunzhao Liu
- Arkansas Biosciences Institute, Arkansas State University, State University, Arkansas 72467-0639, USA.
| | | | | | | | | |
Collapse
|
36
|
Liu J, Dolan MC, Reidy M, Cramer CL. Expression of bioactive single-chain murine IL-12 in transgenic plants. J Interferon Cytokine Res 2008; 28:381-92. [PMID: 18593333 DOI: 10.1089/jir.2007.0129] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Interleukin-12 (IL-12), an important immunomodulator for cell-mediated immunity, shows significant potential as a vaccine adjuvant and anticancer therapeutic. However, its clinical application is limited in part by lack of an effective bioproduction system for this complex heterodimeric glycoprotein. Transgenic plants show promise as scalable bioproduction platforms for challenging biopharmaceutical proteins. To test the potential of plants to effectively produce bioactive IL-12, we developed transgenic tobacco plant lines and derived root cultures yielding high levels of mouse IL-12 (MuIL-12). Functional IL-12 is a heterodimer consisting of two disulfide-linked subunits, p35 and p40. To ensure the stoichiometric expression and assembly of p35 and p40, we expressed a single-chain version of MuIL-12. Plant-derived single-chain MuIL-12 was characterized and purified for in vitro bioactivity assays. Our results demonstrated precise cleavage of the endogenous mouse p40 signal peptide in plants as well as addition of N-linked glycans. Plant-derived MuIL-12 triggered induction of interferon-gamma (IFN-gamma) secretion from mouse splenocytes and stimulated splenocyte proliferation with comparable activities to those observed for commercially available animal cell-derived MuIL-12. These studies indicate that plants produce fully functional MuIL-12 at levels compatible with commercial production and may serve as an effective bioproduction platform for bioactive IL-12s from other species for human or veterinary vaccine and therapeutic applications.
Collapse
Affiliation(s)
- Jianyun Liu
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR 72467, USA
| | | | | | | |
Collapse
|
37
|
Díaz-Montero CM, El Naggar S, Al Khami A, El Naggar R, Montero AJ, Cole DJ, Salem ML. Priming of naive CD8+ T cells in the presence of IL-12 selectively enhances the survival of CD8+CD62Lhi cells and results in superior anti-tumor activity in a tolerogenic murine model. Cancer Immunol Immunother 2008; 57:563-572. [PMID: 17726606 PMCID: PMC3406410 DOI: 10.1007/s00262-007-0394-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2007] [Accepted: 08/15/2007] [Indexed: 02/07/2023]
Abstract
During the antigen-dependant activation process several subsets CD8+ T cells appear with different phenotypic and functional characteristics. Recent studies indicate that the state of T cell differentiation radically affects their ability to effectively respond to tumor challenge, with early effector CD8+ T (CD62Lhigh) cells having better anti-tumor activity. Thus strategies aimed at optimizing the generation of such subpopulations could significantly enhance the effectiveness of adoptive cell therapy (ACT) for cancer. In this study, we show that priming of naïve CD8+ T cells in the presence of IL-12 selectively rescued early CD8+ CD62L(hi) from activation induced cell death and resulted in the increased accumulation of this subset of CD8+ T cells. Furthermore, we demonstrated that IL-12 directly modulated the expression of CD62L on activated CD8+ T cells. When used for ACT, naïve CD8+ T cells primed in vitro in the presence of IL-12 showed superior anti-tumor activity toward B16 melanoma. Importantly, using the Pmel-1 model, priming pmel-1 cells in vitro with IL-12 reduced the state of functional tolerance associated with the non-mutated "self" tumor antigen gp100, as demonstrated by significant tumor responses in the absence of vaccination. Together, our results suggest that in vitro conditioning of naïve CD8+ T cells with IL-12 prior to ACT could significantly enhance their anti-tumor activity.
Collapse
Affiliation(s)
- C Marcela Díaz-Montero
- Department of Surgery, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Simpson-Abelson M, Bankert RB. Targeting the TCR signaling checkpoint: a therapeutic strategy to reactivate memory T cells in the tumor microenvironment. Expert Opin Ther Targets 2008; 12:477-90. [DOI: 10.1517/14728222.12.4.477] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
39
|
Induction of cytolytic activity and interferon-γ production in murine natural killer cells by polymyxins B and E. Int Immunopharmacol 2008; 8:508-13. [DOI: 10.1016/j.intimp.2007.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 11/01/2007] [Accepted: 11/01/2007] [Indexed: 11/20/2022]
|
40
|
Weiss JM, Subleski JJ, Wigginton JM, Wiltrout RH. Immunotherapy of cancer by IL-12-based cytokine combinations. Expert Opin Biol Ther 2007; 7:1705-21. [PMID: 17961093 DOI: 10.1517/14712598.7.11.1705] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Cancer is a multi-faceted disease comprising complex interactions between neoplastic and normal cells. Over the past decade, there has been considerable progress in defining the molecular, cellular and environmental contributions to the pathophysiology of tumor development. Despite these advances, the conventional treatment of patients still generally involves surgery, radiotherapy and/or chemotherapy, and the clinical outcome for many of these efforts remains unsatisfactory. Recent studies have highlighted the feasibility of using immunotherapeutic approaches that seek to enhance host immune responses to developing tumors. These strategies include immunomodulatory cytokines, with TNF-alpha, type I or type II IFNs, IL-2, IL-12, IL-15 and IL-18 being among the most potent inducers of anti-tumor activity in a variety of preclinical studies. More recently, some exciting new cytokines have been characterized, such as IL-21, IL-23, IL-27 and their immunomodulatory and antitumor effects in vitro and in vivo suggest that they may have considerable promise for future immunotherapy protocols. The promise of cytokine therapy does indeed derive from the identification of these novel cytokines but even more fundamentally, the field is greatly benefiting from the ever-expanding amount of preclinical data that convincingly demonstrate synergistic and/or novel biologic effects, which may be achieved through the use of several combinations of cytokines with complementary immune-stimulating capabilities. One cytokine in particular, IL-12, holds considerable promise by virtue of the fact that it plays a central role in regulating both innate and adaptive immune responses, can by itself induce potent anticancer effects, and synergizes with several other cytokines for increased immunoregulatory and antitumor activities. This review discusses the antitumor activity of IL-12, with a special emphasis on its ability to synergize with other cytokines for enhancement of immune effector cell populations and regulation of host-tumor cell interactions and the overall tumor microenvironment.
Collapse
Affiliation(s)
- Jonathan M Weiss
- National Cancer Institute, Cancer and Inflammation Program, Laboratory of Experimental Immunology, Center for Cancer Research, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
41
|
Abstract
With respect to CD8 effector T cells, interleukin-12 (IL-12) and transforming growth factor beta (TGFbeta) are 2 cytokines that exert opposing effects. IL-12 promotes antitumor immune responses by augmenting activated CD8 T-cell proliferation and interferon-gamma secretion. Conversely, TGFbeta generates a permissive environment for cancer growth, in part by antagonizing the effects of immunomodulatory cytokines, including IL-12. We demonstrate that TGFbeta-resistant T cells are capable of sustaining IL-12-induced mitogenesis and interferon-gamma secretion in a TGFbeta-rich milieu. Furthermore, in 2 murine tumor models associated with high TGFbeta1 levels in the local microenvironment, treatment with IL-12 and adoptively transferred TGFbeta-resistant T cells provided improved survival times. These results suggest that combining IL-12 with TGFbeta neutralization strategies may be effective in enhancing antitumor immune responses.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Line
- Cell Proliferation
- Female
- Immunotherapy, Adoptive
- Interferon-gamma/biosynthesis
- Interleukin-12/immunology
- Interleukin-12/pharmacology
- Male
- Melanoma, Experimental/mortality
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/mortality
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Papilloma, Choroid Plexus/mortality
- Papilloma, Choroid Plexus/pathology
- Papilloma, Choroid Plexus/therapy
- Receptors, Transforming Growth Factor beta/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Transforming Growth Factor beta1/immunology
Collapse
Affiliation(s)
- Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, 1008 West Hazelwood Drive, Urbana, IL 61801, USA.
| | | | | |
Collapse
|
42
|
Yoshida T, Tuder RM. Pathobiology of cigarette smoke-induced chronic obstructive pulmonary disease. Physiol Rev 2007; 87:1047-82. [PMID: 17615396 DOI: 10.1152/physrev.00048.2006] [Citation(s) in RCA: 375] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary diseases (COPD), comprised of pulmonary emphysema, chronic bronchitis, and structural and inflammatory changes of small airways, is a leading cause of morbidity and mortality in the world. A better understanding of the pathobiology of COPD is critical for the developing of novel therapies, as the majority of patients with the disease have little therapeutic options at the present time. The pathobiology of COPD encompasses multiple injurious processes including inflammation (excessive or inappropriate innate and adaptive immunity), cellular apoptosis, altered cellular and molecular alveolar maintenance program, abnormal cell repair, extracellular matrix destruction (protease and anti-protease imbalance), and oxidative stress (oxidant and antioxidant imbalance). These processes are triggered by urban and rural air pollutants and active and/or passive cigarette smoke and modified by cellular senescence and infection. A series of receptor-mediated signal transduction pathways are activated by reactive oxygen species and tobacco components, resulting in impairment of a variety of cell signaling and cytokine networks, subsequently leading to chronic airway responses with mucus production, airway remodeling, and alveolar destruction. The authors provide an updated insight into the molecular and cellular pathobiology of COPD based on human and/or animal data.
Collapse
Affiliation(s)
- Toshinori Yoshida
- Division of Cardiopulmonary Pathology, Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | |
Collapse
|