1
|
Hawly J, Murcar MG, Schcolnik-Cabrera A, Issa ME. Glioblastoma stem cell metabolism and immunity. Cancer Metastasis Rev 2024; 43:1015-1035. [PMID: 38530545 DOI: 10.1007/s10555-024-10183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Despite enormous efforts being invested in the development of novel therapies for brain malignancies, there remains a dire need for effective treatments, particularly for pediatric glioblastomas. Their poor prognosis has been attributed to the fact that conventional therapies target tumoral cells, but not glioblastoma stem cells (GSCs). GSCs are characterized by self-renewal, tumorigenicity, poor differentiation, and resistance to therapy. These characteristics represent the fundamental tools needed to recapitulate the tumor and result in a relapse. The mechanisms by which GSCs alter metabolic cues and escape elimination by immune cells are discussed in this article, along with potential strategies to harness effector immune cells against GSCs. As cellular immunotherapy is making significant advances in a variety of cancers, leveraging this underexplored reservoir may result in significant improvements in the treatment options for brain malignancies.
Collapse
Affiliation(s)
- Joseph Hawly
- Faculty of Medicine and Medical Sciences, University of Balamand, Dekouaneh, Lebanon
| | - Micaela G Murcar
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Mark E Issa
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
2
|
Luan J, Zhang D, Liu B, Yang A, Lv K, Hu P, Yu H, Shmuel A, Zhang C, Ma G. Immune-related lncRNAs signature and radiomics signature predict the prognosis and immune microenvironment of glioblastoma multiforme. J Transl Med 2024; 22:107. [PMID: 38279111 PMCID: PMC10821572 DOI: 10.1186/s12967-023-04823-y] [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: 06/17/2023] [Accepted: 12/22/2023] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common primary malignant brain tumor in adults. This study aimed to construct immune-related long non-coding RNAs (lncRNAs) signature and radiomics signature to probe the prognosis and immune infiltration of GBM patients. METHODS We downloaded GBM RNA-seq data and clinical information from The Cancer Genome Atlas (TCGA) project database, and MRI data were obtained from The Cancer Imaging Archive (TCIA). Then, we conducted a cox regression analysis to establish the immune-related lncRNAs signature and radiomics signature. Afterward, we employed a gene set enrichment analysis (GSEA) to explore the biological processes and pathways. Besides, we used CIBERSORT to estimate the abundance of tumor-infiltrating immune cells (TIICs). Furthermore, we investigated the relationship between the immune-related lncRNAs signature, radiomics signature and immune checkpoint genes. Finally, we constructed a multifactors prognostic model and compared it with the clinical prognostic model. RESULTS We identified four immune-related lncRNAs and two radiomics features, which show the ability to stratify patients into high-risk and low-risk groups with significantly different survival rates. The risk score curves and Kaplan-Meier curves confirmed that the immune-related lncRNAs signature and radiomics signature were a novel independent prognostic factor in GBM patients. The GSEA suggested that the immune-related lncRNAs signature were involved in L1 cell adhesion molecular (L1CAM) interactions and the radiomics signature were involved signaling by Robo receptors. Besides, the two signatures was associated with the infiltration of immune cells. Furthermore, they were linked with the expression of critical immune genes and could predict immunotherapy's clinical response. Finally, the area under the curve (AUC) (0.890,0.887) and C-index (0.737,0.817) of the multifactors prognostic model were greater than those of the clinical prognostic model in both the training and validation sets, indicated significantly improved discrimination. CONCLUSIONS We identified the immune-related lncRNAs signature and tradiomics signature that can predict the outcomes, immune cell infiltration, and immunotherapy response in patients with GBM.
Collapse
Affiliation(s)
- Jixin Luan
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Di Zhang
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Liaocheng, Shandong, China
| | - Bing Liu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Aocai Yang
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kuan Lv
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Pianpian Hu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Hongwei Yu
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Amir Shmuel
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Chuanchen Zhang
- Department of Radiology, Liaocheng People's Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Liaocheng, Shandong, China.
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China.
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Craig AW, Frieboes HB, Videira PA. Advancing cancer immunotherapy: from innovative preclinical models to clinical insights. Sci Rep 2024; 14:1205. [PMID: 38216668 PMCID: PMC10786836 DOI: 10.1038/s41598-024-51704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2024] Open
Affiliation(s)
- Andrew W Craig
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
- Cancer Biology and Genetics Division, Queen's Cancer Research Institute, Kingston, ON, Canada.
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
- U of L Health - Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA.
| | - Paula A Videira
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal.
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal.
| |
Collapse
|
4
|
Pan W, Tao T, Qiu Y, Zhu X, Zhou X. Natural killer cells at the forefront of cancer immunotherapy with immune potency, genetic engineering, and nanotechnology. Crit Rev Oncol Hematol 2024; 193:104231. [PMID: 38070841 DOI: 10.1016/j.critrevonc.2023.104231] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Natural killer (NK) cells are vital components of the human immune system, acting as innate lymphocytes and playing a crucial role in immune surveillance. Their unique ability to independently eliminate target cells without antigen contact or antibodies has sparked interest in immunological research. This review examines recent NK cell developments and applications, encompassing immune functions, interactions with target cells, genetic engineering techniques, pharmaceutical interventions, and implications in cancers. Insights into NK cell regulation emerge, with a focus on promising genetic engineering like CAR-engineered NK cells, enhancing specificity against tumors. Immune checkpoint inhibitors also enhance NK cells' potential in cancer therapy. Nanotechnology's emergence as a tool for targeted drug delivery to improve NK cell therapies is explored. In conclusion, NK cells are pivotal in immunity, holding exciting potential in cancer immunotherapy. Ongoing research promises novel therapeutic strategies, advancing immunotherapy and medical interventions.
Collapse
Affiliation(s)
- Weiyi Pan
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; School of Public Health, Southern Medical University, Guangzhou, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Yishu Qiu
- Department of Biology, College of Arts and Science, New York University, New York, USA
| | - Xiao Zhu
- Computational Systems Biology Lab (CSBL), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
5
|
Quixabeira DCA, Pakola S, Jirovec E, Havunen R, Basnet S, Santos JM, Kudling TV, Clubb JHA, Haybout L, Arias V, Grönberg-Vähä-Koskela S, Cervera-Carrascon V, Kerkelä E, Pasanen A, Anttila M, Tapper J, Kanerva A, Hemminki A. Boosting cytotoxicity of adoptive allogeneic NK cell therapy with an oncolytic adenovirus encoding a human vIL-2 cytokine for the treatment of human ovarian cancer. Cancer Gene Ther 2023; 30:1679-1690. [PMID: 37949944 PMCID: PMC10721546 DOI: 10.1038/s41417-023-00674-3] [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: 04/13/2023] [Revised: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023]
Abstract
Despite good results in the treatment of hematological malignancies, Natural killer (NK) cells have shown limited effectiveness in solid tumors, such as ovarian cancer (OvCa). Here, we assessed the potential of an oncolytic adenovirus expressing a variant interleukin-2 (vIL-2) cytokine, Ad5/3-E2F-d24-vIL2 (vIL-2 virus), also known as TILT-452, to enhance NK cell therapy efficacy in human OvCa ex vivo. Human OvCa surgical specimens were processed into single-cell suspensions and NK cells were expanded from healthy blood donors. OvCa sample digests were co-cultured ex vivo with NK cells and vIL-2 virus and cancer cell killing potential assessed in real time through cell impedance measurement. Proposed therapeutic combination was evaluated in vivo with an OvCa patient-derived xenograft (PDX) in mice. Addition of vIL-2 virus significantly enhanced NK cell therapy killing potential in treated OvCa co-cultures. Similarly, vIL-2 virus in combination with NK cell therapy promoted the best in vivo OvCa tumor control. Mechanistically, vIL-2 virus induced higher percentages of granzyme B in NK cells, and CD8+ T cells, while T regulatory cell proportions remained comparable to NK cell monotherapy in vivo. Ad5/3-E2F-d24-vIL2 virus treatment represents a promising strategy to boost adoptive NK cell therapeutic effect in human OvCa.
Collapse
Affiliation(s)
- D C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - S Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - E Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - S Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - T V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - L Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - V Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - S Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - V Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - E Kerkelä
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - A Pasanen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - M Anttila
- Pathology, Finnish Food Authority, Helsinki, Finland
| | - J Tapper
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - A Kanerva
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - A Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- TILT Biotherapeutics Ltd, Helsinki, Finland.
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| |
Collapse
|
6
|
Maeoka R, Nakazawa T, Matsuda R, Morimoto T, Shida Y, Yamada S, Nishimura F, Nakamura M, Nakagawa I, Park YS, Tsujimura T, Nakase H. Therapeutic Anti-KIR Antibody of 1-7F9 Attenuates the Antitumor Effects of Expanded and Activated Human Primary Natural Killer Cells on In Vitro Glioblastoma-like Cells and Orthotopic Tumors Derived Therefrom. Int J Mol Sci 2023; 24:14183. [PMID: 37762486 PMCID: PMC10531877 DOI: 10.3390/ijms241814183] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastoma (GBM) is the leading malignant intracranial tumor, where prognosis for which has remained extremely poor for two decades. Immunotherapy has recently drawn attention as a cancer treatment, including for GBM. Natural killer (NK) cells are immune cells that attack cancer cells directly and produce antitumor immunity-related cytokines. The adoptive transfer of expanded and activated NK cells is expected to be a promising GBM immunotherapy. We previously established an efficient expansion method that produced highly purified, activated primary human NK cells, which we designated genuine induced NK cells (GiNKs). The GiNKs demonstrated antitumor effects in vitro and in vivo, which were less affected by blockade of the inhibitory checkpoint receptor programmed death 1 (PD-1). In the present study, we assessed the antitumor effects of GiNKs, both alone and combined with an antibody targeting killer Ig-like receptor 2DLs (KIR2DL1 and DL2/3, both inhibitory checkpoint receptors of NK cells) in vitro and in vivo with U87MG GBM-like cells and the T98G GBM cell line. Impedance-based real-time cell growth assays and apoptosis detection assays revealed that the GiNKs exhibited growth inhibitory effects on U87MG and T98G cells by inducing apoptosis. KIR2DL1 blockade attenuated the growth inhibition of the cell lines in vitro. The intracranial administration of GiNKs prolonged the overall survival of the U87MG-derived orthotopic xenograft brain tumor model. The KIR2DL1 blockade did not enhance the antitumor effects; rather, it attenuated it in the same manner as in the in vitro experiment. GiNK immunotherapy directly administered to the brain could be a promising immunotherapeutic alternative for patients with GBM. Furthermore, KIR2DL1 blockade appeared to require caution when used concomitantly with GiNKs.
Collapse
Affiliation(s)
- Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan;
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Yoichi Shida
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Shuichi Yamada
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Fumihiko Nishimura
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Mitsutoshi Nakamura
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Young-Soo Park
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| | - Takahiro Tsujimura
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan;
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Hiroyuki Nakase
- Department of Neurosurgery, Nara Medical University, Nara 634-8521, Japan; (R.M.); (T.M.); (Y.S.); (S.Y.); (F.N.); (M.N.); (I.N.); (Y.-S.P.); (H.N.)
| |
Collapse
|
7
|
Liu J, Wu D, Leng Y, Li Y, Li N. Dietary supplementation with selenium polysaccharide from selenium-enriched Phellinus linteus improves antioxidant capacity, immunity and production performance of laying hens. J Trace Elem Med Biol 2023; 77:127140. [PMID: 36812786 DOI: 10.1016/j.jtemb.2023.127140] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
BACKGROUND Selenium (Se) plays a beneficial role in the physiological function of humans and animals. Selenium polysaccharide, improving enzyme activity and regulating immunity, is the extraction from selenium-rich plants or mushrooms. This study aimed to evaluate the effect of selenium polysaccharide from selenium-enriched Phellinus linteus on the antioxidative ability, immunity, serum biochemistry, and production performance of laying hens. METHODS Three hundred sixty adult laying hens were randomly assigned to 4 groups. The four groups were divided as follows: CK (control group), PS group (4.2 g/kg polysaccharide), Se group (0.5 Se mg/kg), and PSSe group (4.2 g/kg with 0.5 Se mg/kg, Selenium polysaccharide). RESULTS After the 8 weeks, the hens were sampled and the antioxidant ability(total antioxidant (T-AOC), superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), malondialdehyde (MDA), and Nitric Oxide (NO)), immunity(Interleukin-2(IL-2), Immunoglobulin M(IgM), Immunoglobulin A(IgA), Immunoglobulin G(IgG) and interferon-gamma (IFN-γ) and secretory Immunoglobulin A(sIgA)), serum biochemistry(total protein, triglycerides, total cholesterol, glucose, glutamic-pyruvictransaminase (ALT), and aspartate transaminase (AST)) and production performance were assessed. Compared with the control group, T-AOC, SOD, CAT, GSH, IL-2, IgM, IgA, sIgA, IgG, IFN-γ, total protein, average laying rate, average egg weight, and final body were significantly increased in the PS, Se, and PSSe groups, however, the MDA and NO, triglyceride, cholesterol, glucose, AST, ALT, average daily feed consumption, and feed conversion ratio were significantly decreased in the PS, Se, and PSSe groups. The PSSe group in the immune index, antioxidant ability and serum biochemistry was improved the highest. CONCLUSION The result suggested that selenium polysaccharide from selenium-enriched Phellinus linteus can enhance the antioxidant ability and immunity, change serum biochemistry, providing a new method for improving the production performance of laying hens.
Collapse
Affiliation(s)
- Jing Liu
- College of Life Sciences, Changchun Sci-Tech University, Changchun 130600 China
| | - Dan Wu
- Health Monitoring and Inspection Center of Jilin Province, Changchun 130062 China
| | - Yang Leng
- Bureau of Agriculture and Rural Affairs of Wangqing County, Yanbian 133200 China
| | - Yun Li
- College of Life Sciences, Changchun Sci-Tech University, Changchun 130600 China.
| | - Nan Li
- Changchun Academy of Agricultural Science, Changchun 130062 China.
| |
Collapse
|
8
|
Savchenko AA, Kudryavtsev IV, Isakov DV, Sadowski IS, Belenyuk VD, Borisov AG. Recombinant Human Interleukin-2 Corrects NK Cell Phenotype and Functional Activity in Patients with Post-COVID Syndrome. Pharmaceuticals (Basel) 2023; 16:ph16040537. [PMID: 37111294 PMCID: PMC10144656 DOI: 10.3390/ph16040537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/31/2023] [Accepted: 04/01/2023] [Indexed: 04/07/2023] Open
Abstract
Post-COVID syndrome develops in 10–20% of people who have recovered from COVID-19 and it is characterized by impaired function of the nervous, cardiovascular, and immune systems. Previously, it was found that patients who recovered from infection with the SARS-CoV-2 virus had a decrease in the number and functional activity of NK cells. The aim of the study was to assess the effectiveness of recombinant human IL-2 (rhIL-2) administered to correct NK cell phenotype and functional activity in patients with post-COVID syndrome. Patients were examined after 3 months for acute COVID-19 of varying severity. The phenotype of the peripheral blood NK cells was studied by flow cytometry. It was found that disturbances in the cell subset composition in patients with post-COVID syndrome were characterized by low levels of mature (p = 0.001) and cytotoxic NK cells (p = 0.013), with increased release of immature NK cells (p = 0.023). Functional deficiency of NK cells in post-COVID syndrome was characterized by lowered cytotoxic activity due to the decreased count of CD57+ (p = 0.001) and CD8+ (p < 0.001) NK cells. In the treatment of patients with post-COVID syndrome with recombinant IL-2, peripheral blood NK cell count and functional potential were restored. In general, the effectiveness of using rhIL-2 in treatment of post-COVID syndrome has been proven in patients with low levels of NK cells.
Collapse
Affiliation(s)
- Andrei A. Savchenko
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Igor V. Kudryavtsev
- Institute of Experimental Medicine, 197376 St. Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Dmitry V. Isakov
- Institute of Experimental Medicine, Pavlov First St. Petersburg State Medical University of the Russian Federation Ministry of Healthcare, 197022 St. Petersburg, Russia
| | - Ivan S. Sadowski
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Vasily D. Belenyuk
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| | - Alexandr G. Borisov
- Federal Research Center “Krasnoyarsk Science Center” of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660022 Krasnoyarsk, Russia
| |
Collapse
|
9
|
Yalamarty SSK, Filipczak N, Li X, Subhan MA, Parveen F, Ataide JA, Rajmalani BA, Torchilin VP. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers (Basel) 2023; 15:cancers15072116. [PMID: 37046777 PMCID: PMC10093719 DOI: 10.3390/cancers15072116] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer that is difficult to treat due to its resistance to both radiation and chemotherapy. This resistance is largely due to the unique biology of GBM cells, which can evade the effects of conventional treatments through mechanisms such as increased resistance to cell death and rapid regeneration of cancerous cells. Additionally, the blood–brain barrier makes it difficult for chemotherapy drugs to reach GBM cells, leading to reduced effectiveness. Despite these challenges, there are several treatment options available for GBM. The standard of care for newly diagnosed GBM patients involves surgical resection followed by concurrent chemoradiotherapy and adjuvant chemotherapy. Emerging treatments include immunotherapy, such as checkpoint inhibitors, and targeted therapies, such as bevacizumab, that attempt to attack specific vulnerabilities in GBM cells. Another promising approach is the use of tumor-treating fields, a type of electric field therapy that has been shown to slow the growth of GBM cells. Clinical trials are ongoing to evaluate the safety and efficacy of these and other innovative treatments for GBM, intending to improve with outcomes for patients.
Collapse
Affiliation(s)
- Satya Siva Kishan Yalamarty
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Xiang Li
- State Key Laboratory of Innovative Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang 330006, China
| | - Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Farzana Parveen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital, Jhang 35200, Pakistan
| | - Janaína Artem Ataide
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Faculty of Pharmaceutical Sciences, University of Campinas (UNICAMP), Campinas 13083-871, Brazil
| | - Bharat Ashok Rajmalani
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine (CPBN), Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
10
|
Li F, Liu S. Focusing on NK cells and ADCC: A promising immunotherapy approach in targeted therapy for HER2-positive breast cancer. Front Immunol 2022; 13:1083462. [PMID: 36601109 PMCID: PMC9806173 DOI: 10.3389/fimmu.2022.1083462] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast cancer has a high metastatic potential. Monoclonal antibodies (mAbs) that target HER2, such as trastuzumab and pertuzumab, are the cornerstone of adjuvant therapy for HER2-positive breast cancer. A growing body of preclinical and clinical evidence points to the importance of innate immunity mediated by antibody-dependent cellular cytotoxicity (ADCC) in the clinical effect of mAbs on the resulting anti-tumor response. In this review, we provide an overview of the role of natural killer (NK) cells and ADCC in targeted therapy of HER2-positive breast cancer, including the biological functions of NK cells and the role of NK cells and ADCC in anti-HER2 targeted drugs. We then discuss regulatory mechanisms and recent strategies to leverage our knowledge of NK cells and ADCC as an immunotherapy approach for HER2-positive breast cancer.
Collapse
|
11
|
Mendoza-Valderrey A, Alvarez M, De Maria A, Margolin K, Melero I, Ascierto ML. Next Generation Immuno-Oncology Strategies: Unleashing NK Cells Activity. Cells 2022; 11:3147. [PMID: 36231109 PMCID: PMC9562848 DOI: 10.3390/cells11193147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/06/2022] [Accepted: 10/02/2022] [Indexed: 11/19/2022] Open
Abstract
In recent years, immunotherapy has become a powerful therapeutic option against multiple malignancies. The unique capacity of natural killer (NK) cells to attack cancer cells without antigen specificity makes them an optimal immunotherapeutic tool for targeting tumors. Several approaches are currently being pursued to maximize the anti-tumor properties of NK cells in the clinic, including the development of NK cell expansion protocols for adoptive transfer, the establishment of a favorable microenvironment for NK cell activity, the redirection of NK cell activity against tumor cells, and the blockage of inhibitory mechanisms that constrain NK cell function. We here summarize the recent strategies in NK cell-based immunotherapies and discuss the requirement to further optimize these approaches for enhancement of the clinical outcome of NK cell-based immunotherapy targeting tumors.
Collapse
Affiliation(s)
- Alberto Mendoza-Valderrey
- Rosalie and Harold Rae Brown Cancer Immunotherapy Research Program, Borstein Family Melanoma Program, Translational Immunology Department, Saint John’s Cancer Institute, Santa Monica, CA 90404, USA
| | - Maite Alvarez
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Andrea De Maria
- Department of Health Sciences, University of Genoa, 16126 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Kim Margolin
- Borstein Family Melanoma Program, Saint John’s Cancer Institute, Santa Monica, CA 90404, USA
| | - Ignacio Melero
- Program for Immunology and Immunotherapy, CIMA, Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Maria Libera Ascierto
- Rosalie and Harold Rae Brown Cancer Immunotherapy Research Program, Borstein Family Melanoma Program, Translational Immunology Department, Saint John’s Cancer Institute, Santa Monica, CA 90404, USA
| |
Collapse
|