1
|
Wang SW, Zheng QY, Hong WF, Tang BF, Hsu SJ, Zhang Y, Zheng XB, Zeng ZC, Gao C, Ke AW, Du SS. Mechanism of immune activation mediated by genomic instability and its implication in radiotherapy combined with immune checkpoint inhibitors. Radiother Oncol 2024; 199:110424. [PMID: 38997092 DOI: 10.1016/j.radonc.2024.110424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Various genetic and epigenetic changes associated with genomic instability (GI), including DNA damage repair defects, chromosomal instability, and mitochondrial GI, contribute to development and progression of cancer. These alterations not only result in DNA leakage into the cytoplasm, either directly or through micronuclei, but also trigger downstream inflammatory signals, such as the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Apart from directly inducing DNA damage to eliminate cancer cells, radiotherapy (RT) exerts its antitumor effects through intracellular DNA damage sensing mechanisms, leading to the activation of downstream inflammatory signaling pathways. This not only enables local tumor control but also reshapes the immune microenvironment, triggering systemic immune responses. The combination of RT and immunotherapy has emerged as a promising approach to increase the probability of abscopal effects, where distant tumors respond to treatment due to the systemic immunomodulatory effects. This review emphasizes the importance of GI in cancer biology and elucidates the mechanisms by which RT induces GI remodeling of the immune microenvironment. By elucidating the mechanisms of GI and RT-induced immune responses, we aim to emphasize the crucial importance of this approach in modern oncology. Understanding the impact of GI on tumor biological behavior and therapeutic response, as well as the possibility of activating systemic anti-tumor immunity through RT, will pave the way for the development of new treatment strategies and improve prognosis for patients.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China
| | - Qiu-Yi Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Wei-Feng Hong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Bu-Fu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Shu-Jung Hsu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Yang Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Xiao-Bin Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chao Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Ai-Wu Ke
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Shi-Suo Du
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China.
| |
Collapse
|
2
|
Yordanov A, Damyanova P, Vasileva-Slaveva M, Hasan I, Kostov S, Shivarov V. Integrated Analysis of Phagocytic and Immunomodulatory Markers in Cervical Cancer Reveals Constellations of Potential Prognostic Relevance. Int J Mol Sci 2024; 25:9117. [PMID: 39201801 PMCID: PMC11354974 DOI: 10.3390/ijms25169117] [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: 07/17/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
Despite improvements in vaccination, screening, and treatment, cervical cancer (CC) remains a major healthcare problem on a global scale. The tumor microenvironment (TME) plays an important and controversial role in cancer development, and the mechanism of the tumor's escape from immunological surveillance is still not clearly defined. We aim to investigate the expression of CD68 and CD47 in patients with different histological variants of CC, tumor characteristics, and burden. This is a retrospective cohort study performed on paraffin-embedded tumor tissues from 191 patients diagnosed with CC between 2014 and 2021 at the Medical University Pleven, Bulgaria. Slides for immunohistochemical (IHC) evaluation were obtained, and the expression of CD68 was scored in intratumoral (IT) and stromal (ST) macrophages (CD68+cells) using a three-point scoring scale. The CD47 expression was reported as an H-score. All statistical analyses were performed using R v. 4.3.1 for Windows. Infiltration by CD68-IT cells in the tumor depended on histological type and the expression of CD47. Higher levels of the CD47 H-score were significantly more frequent among patients in the early stage. Higher levels of infiltration by CD68-ST cells were associated with worse prognosis, and the infiltration of CD68-IT cells was associated with reduced risk of death from neoplastic disease. TME is a complex ecosystem that has a major role in the growth and development of tumors. Macrophages are a major component of innate immunity and, when associated with a tumor process, are defined as TAM. Tumor cells try to escape immunological surveillance in three ways, and one of them is reducing immunogenicity by the overexpression of negative coreceptors by T-lymphocytes and their ligands on the surface of tumor cells. One such mechanism is the expression of CD47 in tumor cells, which sends a "don't eat me" signal to the macrophages and, thus, prevents phagocytosis. To our knowledge, this is the first study that has tried to establish the relationship between the CD47 and CD68 expression levels and some clinicopathologic features in CC. We found that the only clinicopathological feature implicating the level of CD68 infiltration was the histological variant of the tumor, and only for CD68-IT-high levels were these observed in SCC. High levels of CD47 expression were seen more frequently in pT1B than pT2A and pT2B in the FIGO I stage than in the FIGO II and III stages. Infiltration by large numbers of CD68-IT cells was much more common among patients with a high expression of CD47 in tumor cells. A high level of infiltration by CD68-ST cells was associated with a worse prognosis, and a high level of infiltration by CD68-ST cells was associated with a lower risk of death from cancer.
Collapse
Affiliation(s)
- Angel Yordanov
- Department of Gynecologic Oncology, Medical University-Pleven, 5800 Pleven, Bulgaria
| | - Polina Damyanova
- Department of General and Clinical Pathology, Heart and Brain Center of Clinical Excellence, 5800 Pleven, Bulgaria;
| | - Mariela Vasileva-Slaveva
- Research Institute, Medical University Pleven, 5800 Pleven, Bulgaria; (M.V.-S.); (S.K.); (V.S.)
- Department of Breast Surgery, Shterev Hospital, 1000 Sofia, Bulgaria
| | - Ihsan Hasan
- Department of Obstetrics and Gynecology, University Hospital “Sofiamed”, 1750 Sofia, Bulgaria;
| | - Stoyan Kostov
- Research Institute, Medical University Pleven, 5800 Pleven, Bulgaria; (M.V.-S.); (S.K.); (V.S.)
- Department of Gynecology, Hospital “Saint Anna”, Medical University—“Prof. Dr. Paraskev Stoyanov”, 9002 Varna, Bulgaria
| | - Velizar Shivarov
- Research Institute, Medical University Pleven, 5800 Pleven, Bulgaria; (M.V.-S.); (S.K.); (V.S.)
| |
Collapse
|
3
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Sonam S, Jelača S, Laube M, Schädlich J, Pietzsch J, Maksimović-Ivanić D, Mijatović S, Kaluđerović GN, Hey-Hawkins E. Carborane Conjugates with Ibuprofen, Fenoprofen and Flurbiprofen: Synthesis, Characterization, COX Inhibition Potential and In Vitro Activity. ChemMedChem 2024:e202400018. [PMID: 38844420 DOI: 10.1002/cmdc.202400018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 06/03/2024] [Indexed: 11/10/2024]
Abstract
The most effective anticancer drugs currently entail substantial and formidable side effects, and resistance of tumors to chemotherapeutic agents is a further challenge. Thus, the search for new anticancer drugs as well as novel therapeutic methods is still extremely important. Non-steroidal anti-inflammatory drugs (NSAIDs) can inhibit COX (cyclooxygenase), overexpressed in some tumors. Carboranes are emerging as promising pharmacophores. We have therefore combined both moieties in a single molecule to design drugs with a dual mode of action and enhanced effectiveness. The NSAIDs ibuprofen, flurbiprofen, and fenoprofen were connected with 1,2-dicarba-closo-dodecaborane(12) via methylene, ethylene or propylene spacers. Three sets of carborane-NSAID conjugates were synthesized and analyzed through multinuclear (1H, 11B, and 13C) NMR spectroscopy. Conjugates with methylene spacers exhibited the most potent COX inhibition potential, particularly conjugates with flurbiprofen and fenoprofen, displaying higher selectivity towards COX-1. Furthermore, conjugates with methylene and ethylene spacers were more efficient in suppressing the growth of human cancer cell lines than their propylene counterparts. The carborane-flurbiprofen conjugate with an ethylene spacer was the most efficient and selective toward the COX-2-negative cell line HCT116. Its mode of action was basically cytostatic with minor contribution of apoptotic cell death and dominance of cells trapped in the division process.
Collapse
Affiliation(s)
- Sonam Sonam
- Institute of Bioanalytical Chemistry, Centre for Biotechnology and Biomedicine (BBZ), Faculty of Chemistry and Mineralogy, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217, Merseburg, Germany
| | - Sanja Jelača
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, 11108, Belgrade, Serbia
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Jonas Schädlich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Mommsenstrasse 4, 01062, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, Mommsenstrasse 4, 01062, Dresden, Germany
| | - Danijela Maksimović-Ivanić
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, 11108, Belgrade, Serbia
| | - Sanja Mijatović
- Institute for Biological Research "Siniša Stanković", National Institute of the Republic of Serbia, University of Belgrade, 11108, Belgrade, Serbia
| | - Goran N Kaluđerović
- Department of Engineering and Natural Sciences, University of Applied Sciences Merseburg, Eberhard-Leibnitz-Str. 2, 06217, Merseburg, Germany
| | - Evamarie Hey-Hawkins
- Institute of Bioanalytical Chemistry, Centre for Biotechnology and Biomedicine (BBZ), Faculty of Chemistry and Mineralogy, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
| |
Collapse
|
5
|
Yordanov A, Shivarov V, Kostov S, Ivanova Y, Dimitrova P, Popovska S, Tsoneva E, Vasileva-Slaveva M. Prognostic Utility of CD47 in Cancer of the Uterine Cervix and the Sensitivity of Immunohistochemical Scores. Diagnostics (Basel) 2022; 13:diagnostics13010052. [PMID: 36611344 PMCID: PMC9818840 DOI: 10.3390/diagnostics13010052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Cancer of the uterine cervix (CUC) is still one of the most frequent oncological diagnoses in women. The specific interactions between the tumor cells of CUC and the cells and tissues in the tumor microenvironment can affect cancer cells' invasive and metastatic potential and can modulate tumor's progression and death. CD47 is a trans-membranous immunoglobulin, expressed in many cells. It protects the cells from being destroyed by the circulating macrophages. AIM We aimed to evaluate the prognostic role of CD47 expressed in the tumor tissues of patients with CUC for tumor progression and to find the most sensitive immunohistochemical score for defining the cut-off significantly associated with tumor biology and progression. MATERIALS AND METHODS Paraffin-embedded tumor tissues from 86 patients with CUC were included in the study. Clinico-morphological data for patients, such as age and stage at diagnosis according to FIGO and TNM classification, were obtained from the hospital electronic medical records. Immunohistochemical staining was performed with rabbit recombinant monoclonal CD47 antibody (Clone SP279). The final result was interpreted based on three reporting models in immunohistochemistry: H-score, Allred score and combined score. RESULTS The expression of CD47 was higher in tumors limited in the cervix compared with those invading other structures, and it did not depend on the nodal status. The results of immunohistochemical staining were similar regardless of which immunohistochemical method was used. The most significant correlation with TNM stage was observed with the H-score (p = 0.00018). The association with the Allred and combined score was less significant, with p values of 0.0013 and 0.0002, respectively. CONCLUSION The expression of CD47 in the cancer cells is prognostic for tumor invasion in the surrounding structures, independent of lymph node engagement. The H-score is the most sensitive immunohistochemical score to describe tumor stage. To the best of our knowledge, this is the first study evaluating the significance of CD47 expression in CUC.
Collapse
Affiliation(s)
- Angel Yordanov
- Department of Gynaecological Oncology, Medical University Pleven, 5800 Pleven, Bulgaria
- Correspondence:
| | - Velizar Shivarov
- Research Institute, Medical University Pleven, 5800 Pleven, Bulgaria
| | - Stoyan Kostov
- Department of Gynecology, St. Anna University Hospital, Medical University—Varna “Prof. Dr. Paraskev Stoyanov”, 9000 Varna, Bulgaria
| | - Yonka Ivanova
- Department of Gynecology, St. Anna University Hospital, Medical University—Varna “Prof. Dr. Paraskev Stoyanov”, 9000 Varna, Bulgaria
| | - Polina Dimitrova
- Department of Pathology, Medical University–Pleven, 5800 Pleven, Bulgaria
| | - Savelina Popovska
- Department of Pathology, Medical University–Pleven, 5800 Pleven, Bulgaria
| | - Eva Tsoneva
- Department of Obstetrics and Gynecology, Shterev Hospital, 1000 Sofia, Bulgaria
| | - Mariela Vasileva-Slaveva
- Research Institute, Medical University Pleven, 5800 Pleven, Bulgaria
- Department of Breast Surgery, Shterev Hospital, 1000 Sofia, Bulgaria
| |
Collapse
|
6
|
Géczi Z, Róth I, Kőhidai Z, Kőhidai L, Mukaddam K, Hermann P, Végh D, Zelles T. The use of Trojan-horse drug delivery system in managing periodontitis. Int Dent J 2022; 73:346-353. [PMID: 36175203 DOI: 10.1016/j.identj.2022.08.003] [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: 06/19/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022] Open
Abstract
The aim of this review is to evaluate the possibility of delivering a silver-acid complex via a Trojan-horse mechanism for managing periodontits. We theroised that the complex could be an effective treatment option for bacterial inflammatory processes in the oral cavity. Searches were conducted using MEDLINE, Embase, Web of Science Core Collection, and Google Scholar search engines. We also reviewed several reference lists of the included studies or relevant reviews identified by the search. By using Medical Subject Headings (MeSH) terminology, a comprehensive search was performed for the following keywords: silver, folic acid, periodontitis, macrophages, Trojan-horse mechanism, toxicity, and targeting. Using the keywords mentioned earlier, we selected 110 articles and after appropriate elimination the review was written based on 37 papers. Accordingly the we noted that silver isons were an effective approach to kill oral pathogens. Secondly the Trojan-horse mechanism. could be used by macrophages (as the Trojan horse) to deliver silver ions in large quantities to the inflammatory focus to kill the periodontopathogens. The Trojan-horse mechanism has never been described in the field of dentistry before. The proposed novel approach using the principle of Trojan Horse delivery of drugs/chemicals could be used to manage oral inflammatory conditions. This method can be used to supplement regular treatments.
Collapse
Affiliation(s)
- Zoltán Géczi
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary.
| | - Ivett Róth
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary
| | - Zsófia Kőhidai
- Department of Oral Diagnostics, Semmelweis University, Budapest, Hungary
| | - László Kőhidai
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Khaled Mukaddam
- Department of Oral Surgery, University Center for Dental Medicine Basel, University of Basel, Basel, Switzerland
| | - Péter Hermann
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary
| | - Dániel Végh
- Department of Prosthodontics, Semmelweis University,Budapest, Hungary
| | - Tivadar Zelles
- Department of Oral Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
7
|
Robinson BW, Redwood AJ, Creaney J. How Our Continuing Studies of the Pre-clinical Inbred Mouse Models of Mesothelioma Have Influenced the Development of New Therapies. Front Pharmacol 2022; 13:858557. [PMID: 35431929 PMCID: PMC9008447 DOI: 10.3389/fphar.2022.858557] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/24/2022] [Indexed: 11/17/2022] Open
Abstract
Asbestos-induced preclinical mouse models of mesothelioma produce tumors that are very similar to those that develop in humans and thus represent an ideal platform to study this rare, universally fatal tumor type. Our team and a number of other research groups have established such models as a stepping stone to new treatments, including chemotherapy, immunotherapy and other approaches that have been/are being translated into clinical trials. In some cases this work has led to changes in mesothelioma treatment practice and over the last 30 years these models and studies have led to trials which have improved the response rate in mesothelioma from less than 10% to over 50%. Mouse models have had a vital role in that improvement and will continue to play a key role in the future success of mesothelioma immunotherapy. In this review we focus only on these original inbred mouse models, the large number of preclinical studies conducted using them and their contribution to current and future clinical therapy for mesothelioma.
Collapse
Affiliation(s)
- Bruce W.S. Robinson
- Medicine, University of Western Australia, Perth, WA, Australia
- Institute for Respiratory Health, University of Western Australia, Perth, WA, Australia
- *Correspondence: Bruce W.S. Robinson,
| | - Alec J. Redwood
- Institute for Respiratory Health, University of Western Australia, Perth, WA, Australia
- Biomedical Science, University of Western Australia, Perth, WA, Australia
| | - Jenette Creaney
- Institute for Respiratory Health, University of Western Australia, Perth, WA, Australia
- Biomedical Science, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
8
|
Thakral D, Gupta R, Khan A. Leukemic stem cell signatures in Acute myeloid leukemia- targeting the Guardians with novel approaches. Stem Cell Rev Rep 2022; 18:1756-1773. [DOI: 10.1007/s12015-022-10349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2022] [Indexed: 11/09/2022]
|
9
|
Wang Y. Tumor Immune Escape and Treatment. BIO WEB OF CONFERENCES 2022. [DOI: 10.1051/bioconf/20225501010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Tumor immune escape is one of the ten characteristics of tumor occurrence and development. Immunotherapy targeting immune escape has achieved remarkable success in recent years. Immunotherapy involves many factors and links, which are related to the changes of tumor cells themselves and tumor microenvironment, and the mechanism is complex. At present, it still faces great challenges in clinical practice. This article introduces the mechanism of tumor immune escape from several aspects, including the changes of tumor itself, the changes of tumor induced microenvironment, and the tumor microenvironment promoting tumor development. At the same time, in view of these mechanisms, the current treatment strategies were sorted out, including the predicament and progress of immune checkpoint inhibitors, CAR-T therapy and immune cell therapy, aiming to clarify the ideas for the next development of tumor immunotherapy.
Collapse
|
10
|
Maulhardt H, Marin A, Hesseltine H, diZerega G. Submicron particle docetaxel intratumoral injection in combination with anti-mCTLA-4 into 4T1-Luc orthotopic implants reduces primary tumor and metastatic pulmonary lesions. Med Oncol 2021; 38:106. [PMID: 34331595 PMCID: PMC8325653 DOI: 10.1007/s12032-021-01555-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/24/2021] [Indexed: 01/19/2023]
Abstract
We describe here characterization of the response of local and metastatic disease and immunomodulation following intratumoral (IT) injection of submicron particle docetaxel (SPD) administered alone or in combination with systemic antibody anti-mCTLA-4 (anti-mCTLA-4) in the metastatic 4T1-Luc2-1A4 (4T1) murine breast cancer model. In-life assessments of treatment tolerance, tumor volume (TV), and metastasis were performed (n = 10 animals/group). At study end, immune cell populations in tumor-site tissues and peripheral blood were analyzed using flow cytometry. Signs of distress typical of this aggressive tumor model occurred across all animals except for the combination treated which were asymptomatic and gained weight. TV at study end was significantly reduced in the combination group versus untreated [43% reduced (p < 0.05)] and vehicle controls [54% reduced (p < 0.0001)]. No evidence of thoracic metastasis was found in 40% of combination group animals and thoracic bioluminescence imaging (BLI) was reduced vs. untreated controls (p < 0.01). Significant elevations (p < 0.05) in CD4 + T, CD4 + helper T, Treg, and NKT cells were found in tumor and blood in SPD or combination treatment compared to controls or anti-mCTLA-4. Combination treatment increased tumor-associated CD8 + T cells (p < 0.01), peripheral B cells (p < 0.01), and tumor associated and circulating dendritic cells (DC) (p < 0.05). Tumor-associated NK cells were significantly increased in SPD ± anti-mCTLA-4 treatments (p < 0.01). Myeloid-derived suppressor cells (MDSC) were reduced in bloods in SPD ± anti-mCTLA-4 groups (p < 0.05). These data demonstrate that both SPD and anti-mCTLA-4 produce local anti-tumor effects as well as reductions in metastasis which are significantly enhanced when administered in combination.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Combined Modality Therapy
- Docetaxel/administration & dosage
- Docetaxel/chemistry
- Docetaxel/pharmacology
- Female
- Immune Checkpoint Inhibitors/pharmacology
- Injections, Intralesional
- Killer Cells, Natural/immunology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lymphocytes, Tumor-Infiltrating/immunology
- Mammary Neoplasms, Animal/drug therapy
- Mammary Neoplasms, Animal/immunology
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice
- Mice, Inbred BALB C
- Myeloid-Derived Suppressor Cells/immunology
- Particle Size
- T-Lymphocytes, Regulatory/immunology
- Tumor Burden
Collapse
Affiliation(s)
- Holly Maulhardt
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, USA
| | - Alyson Marin
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, USA
| | - Holly Hesseltine
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, USA
| | - Gere diZerega
- US Biotest, Inc., 231 Bonetti Drive, Suite 240, San Luis Obispo, CA, USA.
- NanOlogy, LLC., 3909 Hulen Street, Fort Worth, TX, USA.
| |
Collapse
|
11
|
Jafarzadeh L, Khakpoor-Koosheh M, Mirzaei H, Mirzaei HR. Biomarkers for predicting the outcome of various cancer immunotherapies. Crit Rev Oncol Hematol 2021; 157:103161. [DOI: 10.1016/j.critrevonc.2020.103161] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/21/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022] Open
|
12
|
Błauż A, Rychlik B, Plazuk D, Peccati F, Jiménez-Osés G, Steinke U, Sierant M, Trzeciak K, Skorupska E, Miksa B. Biotin-phenosafranin as a new photosensitive conjugate for targeted therapy and imaging. NEW J CHEM 2021. [DOI: 10.1039/d0nj06170k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A biotinylated phenazine compound as a phenosafranin conjugate (Biot-PSF) was synthesized and reported for the first time.
Collapse
Affiliation(s)
- Andrzej Błauż
- Cytometry Laboratory
- Department of Molecular Biophysics
- Faculty of Biology & Environmental Protection
- University of Lodz
- 90-236 Lodz
| | - Błażej Rychlik
- Cytometry Laboratory
- Department of Molecular Biophysics
- Faculty of Biology & Environmental Protection
- University of Lodz
- 90-236 Lodz
| | - Damian Plazuk
- Department of Organic Chemistry
- Faculty of Chemistry
- University of Lodz
- 91-403 Lodz
- Poland
| | - Francesca Peccati
- CIC bioGUNE
- Center for Cooperative Research in Bioscience
- Bizkaia Science and Technology Park
- Computational Chemistry Lab
- 48160 Derio-Bizkaia
| | - Gonzalo Jiménez-Osés
- CIC bioGUNE
- Center for Cooperative Research in Bioscience
- Bizkaia Science and Technology Park
- Computational Chemistry Lab
- 48160 Derio-Bizkaia
| | - Urszula Steinke
- Centre of Molecular and Macromolecular Studies Polish Academy of Science
- 90-363 Lodz
- Poland
| | - Malgorzata Sierant
- Centre of Molecular and Macromolecular Studies Polish Academy of Science
- 90-363 Lodz
- Poland
| | - Katarzyna Trzeciak
- Centre of Molecular and Macromolecular Studies Polish Academy of Science
- 90-363 Lodz
- Poland
| | - Ewa Skorupska
- Centre of Molecular and Macromolecular Studies Polish Academy of Science
- 90-363 Lodz
- Poland
| | - Beata Miksa
- Centre of Molecular and Macromolecular Studies Polish Academy of Science
- 90-363 Lodz
- Poland
| |
Collapse
|
13
|
Acebes-Fernández V, Landeira-Viñuela A, Juanes-Velasco P, Hernández AP, Otazo-Perez A, Manzano-Román R, Gongora R, Fuentes M. Nanomedicine and Onco-Immunotherapy: From the Bench to Bedside to Biomarkers. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1274. [PMID: 32610601 PMCID: PMC7407304 DOI: 10.3390/nano10071274] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022]
Abstract
The broad relationship between the immune system and cancer is opening a new hallmark to explore for nanomedicine. Here, all the common and synergy points between both areas are reviewed and described, and the recent approaches which show the progress from the bench to the beside to biomarkers developed in nanomedicine and onco-immunotherapy.
Collapse
Affiliation(s)
- Vanessa Acebes-Fernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Angela-Patricia Hernández
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Andrea Otazo-Perez
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Raúl Manzano-Román
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| | - Rafael Gongora
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
| | - Manuel Fuentes
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (V.A.-F.); (A.L.-V.); (P.J.-V.); (A.-P.H.); (A.O.-P.); (R.G.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain;
| |
Collapse
|
14
|
Therapeutic Mesenchymal Stromal Cells for Immunotherapy and for Gene and Drug Delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 16:204-224. [PMID: 32071924 PMCID: PMC7012781 DOI: 10.1016/j.omtm.2020.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mesenchymal stromal cells (MSCs) possess several fairly unique properties that, when combined, make them ideally suited for cellular-based immunotherapy and as vehicles for gene and drug delivery for a wide range of diseases and disorders. Key among these are: (1) their relative ease of isolation from a variety of tissues; (2) the ability to be expanded in culture without a loss of functionality, a property that varies to some degree with tissue source; (3) they are relatively immune-inert, perhaps obviating the need for precise donor/recipient matching; (4) they possess potent immunomodulatory functions that can be tailored by so-called licensing in vitro and in vivo; (5) the efficiency with which they can be modified with viral-based vectors; and (6) their almost uncanny ability to selectively home to damaged tissues, tumors, and metastases following systemic administration. In this review, we summarize the latest research in the immunological properties of MSCs, their use as immunomodulatory/anti-inflammatory agents, methods for licensing MSCs to customize their immunological profile, and their use as vehicles for transferring both therapeutic genes in genetic disease and drugs and genes designed to destroy tumor cells.
Collapse
|
15
|
Panizza BJ, de Souza P, Cooper A, Roohullah A, Karapetis CS, Lickliter JD. Phase I dose-escalation study to determine the safety, tolerability, preliminary efficacy and pharmacokinetics of an intratumoral injection of tigilanol tiglate (EBC-46). EBioMedicine 2019; 50:433-441. [PMID: 31810818 PMCID: PMC6921293 DOI: 10.1016/j.ebiom.2019.11.037] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/12/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tigilanol tiglate, a short-chain diterpene ester, is being developed as intratumoral treatment of a broad range of cancers. We conducted the first-in-human study of intratumoral tigilanol tiglate in patients with solid tumors. METHODS Tigilanol tiglate was administered in a multicentre, non randomized, single-arm study, with escalating doses beginning with 0·06 mg/m2 in tumors estimated to be at least twice the volume of injection (dose-escalation cohorts). Patients with smaller tumors were assigned to the local effects cohort and received the appropriate dose for tumor size. FINDINGS Twenty-two patients were enrolled. The maximum dose was 3·6 mg/m2 and the maximum tolerated dose was not reached. There was one report of dose-limiting toxicity (upper airway obstruction), two serious adverse events (upper airway obstruction and septicemia), 160 treatment-emergent adverse events, and no deaths. Injection site reactions in all tumors and tumor types occurred even at the lowest dose. Six of the 22 patients experienced a treatment response, with four of the six patients achieving complete response. INTERPRETATION Intratumoral tigilanol tiglate was generally well tolerated, the maximum tolerated dose was not reached, and clinical activity was observed in 9 tumor types including complete response in four patients. These results support the continued development of tigilanol tiglate for intratumoral administration. FUNDING QBiotics Group Limited Brisbane, Queensland, Australia was the sponsor of the study.
Collapse
Affiliation(s)
- Benedict J Panizza
- Department of Otolaryngology-Head and Neck Surgery, Princess Alexandra Hospital and Faculty of Medicine, University of Queensland, Brisbane, Australia.
| | - Paul de Souza
- Medical Oncology, University of Western Sydney, Sydney, Australia
| | - Adam Cooper
- Medical Oncology, University of Western Sydney, Sydney, Australia
| | - Aflah Roohullah
- Medical Oncology, University of Western Sydney, Sydney, Australia
| | - Christos S Karapetis
- Department of Medical Oncology, Flinders Medical Center and Flinders Center for Innovation in Cancer, Flinders University, Adelaide, Australia
| | | |
Collapse
|
16
|
Uher O, Caisova V, Hansen P, Kopecky J, Chmelar J, Zhuang Z, Zenka J, Pacak K. Coley's immunotherapy revived: Innate immunity as a link in priming cancer cells for an attack by adaptive immunity. Semin Oncol 2019; 46:385-392. [PMID: 31739997 DOI: 10.1053/j.seminoncol.2019.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 12/11/2022]
Abstract
There is no doubt that immunotherapy lies in the spotlight of current cancer research and clinical trials. However, there are still limitations in the treatment response in certain types of tumors largely due to the presence of the complex network of immunomodulatory and immunosuppressive pathways. These limitations are not likely to be overcome by current immunotherapeutic options, which often target isolated steps in immune pathways preferentially involved in adaptive immunity. Recently, we have developed an innovative anti-cancer immunotherapeutic strategy that initially elicits a strong innate immune response with subsequent activation of adaptive immunity in mouse models. Robust primary innate immune response against tumor cells is induced by toll-like receptor ligands and anti-CD40 agonistic antibodies combined with the phagocytosis-stimulating ligand mannan, anchored to a tumor cell membrane by biocompatible anchor for membrane. This immunotherapeutic approach results in a dramatic therapeutic response in large established murine subcutaneous tumors including melanoma, sarcoma, pancreatic adenocarcinoma, and pheochromocytoma. Additionally, eradication of metastases and/or long-lasting resistance to subsequent re-challenge with tumor cells was also accomplished. Current and future advantages of this immunotherapeutic approach and its possible combinations with other available therapies are discussed in this review.
Collapse
Affiliation(s)
- Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA; Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Veronika Caisova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA
| | - Per Hansen
- Immunoaction LLC, Charlotte, Vermont, VT 05445, USA
| | - Jan Kopecky
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Jindrich Chmelar
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Zhengping Zhuang
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, MD 20814, USA
| | - Jan Zenka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, MD 20814, USA.
| |
Collapse
|
17
|
Local Injection of Submicron Particle Docetaxel is Associated with Tumor Eradication, Reduced Systemic Toxicity and an Immunologic Response in Uro-Oncologic Xenografts. Cancers (Basel) 2019; 11:cancers11040577. [PMID: 31022918 PMCID: PMC6520999 DOI: 10.3390/cancers11040577] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/20/2022] Open
Abstract
Intratumoral (IT) administration of submicron particle docetaxel (NanoDoce®, NanOlogy LLC, Fort Worth, TX, USA) and its efficacy against genitourinary-oncologic xenografts in rats and mice, xenograft-site docetaxel concentrations and immune-cell infiltration were studied. IT-NanoDoce®, IV-docetaxel and IT-vehicle were administered to clear cell renal carcinoma (786-O: rats), transitional cell bladder carcinoma (UM-UC-3: mice) and prostate carcinoma (PC-3: mice). Treatments were given every 7 days with 1, 2, or 3 doses administered. Animals were followed for tumor growth and clinical signs. At necropsy, 786-O and UM-UC-3 tumor-site tissues were evaluated by H&E and IHC and analyzed by LC-MS/MS for docetaxel concentration. Two and 3 cycles of IT-NanoDoce® significantly reduced UM-UC-3 tumor volume (p < 0.01) and eliminated most UM-UC-3 and 786-O tumors. In both models, NanoDoce® treatment was associated with (peri)tumor-infiltrating immune cells. Lymphoid structures were observed in IT-NanoDoce®-treated UM-UC-3 animals adjacent to tumor sites. IT-vehicle and IV-docetaxel exhibited limited immune-cell infiltration. In both studies, high levels of docetaxel were detected in NanoDoce®-treated animals up to 50 days post-treatment. In the PC-3 study, IT-NanoDoce® and IV-docetaxel resulted in similar tumor reduction. NanoDoce® significantly reduced tumor volume compared to IT-vehicle in all xenografts (p < 0.0001). We hypothesize that local, persistent, therapeutic levels of docetaxel from IT-NanoDoce® reduces tumor burden while increasing immune-cell infiltration. IT NanoDoce® treatment of prostate, renal and bladder cancer may result in enhanced tumoricidal effects.
Collapse
|
18
|
Nanofluidic drug-eluting seed for sustained intratumoral immunotherapy in triple negative breast cancer. J Control Release 2018; 285:23-34. [DOI: 10.1016/j.jconrel.2018.06.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/15/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
|
19
|
Caisová V, Uher O, Nedbalová P, Jochmanová I, Kvardová K, Masáková K, Krejčová G, Paďouková L, Chmelař J, Kopecký J, Ženka J. Effective cancer immunotherapy based on combination of TLR agonists with stimulation of phagocytosis. Int Immunopharmacol 2018; 59:86-96. [PMID: 29635103 DOI: 10.1016/j.intimp.2018.03.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 03/22/2018] [Accepted: 03/30/2018] [Indexed: 01/03/2023]
Abstract
Immunotherapy emerges as a fundamental approach in cancer treatment. Up to date, the efficacy of numerous different immunotherapies has been evaluated. The use of microorganisms or their parts for immune cell activation, referred to as Pathogen-Associated Molecular Patterns (PAMPs), represents highly promising concept. The therapeutic effect of PAMPs can be further amplified by suitable combination of different types of PAMPs such as Toll like receptor (TLR) agonists and phagocytosis activating ligands. Previously, we used the combination of phagocytosis activating ligand (mannan) and mixture of TLR agonists (resiquimod (R-848), poly(I:C), inactivated Listeria monocytogenes) for successful treatment of melanoma in murine B16-F10 model. In the present study, we optimized the composition and timing of previously used mixture. Therapeutic mixture based on well-defined chemical compounds consisted of mannan anchoring to tumor cell surface by biocompatible anchor for membranes (BAM) and TLR agonists resiquimod, poly(I:C), and lipoteichoic acid (LTA). The optimization resulted in (1) eradication of advanced stage progressive melanoma in 83% of mice, (2) acquisition of resistance to tumor re-transplantation, and (3) potential anti-metastatic effect. After further investigation of mechanisms, underlying anti-tumor responses, we concluded that both innate and adaptive immunity are activated and involved in these processes. We tested the efficacy of our treatment in Panc02 murine model of aggressive pancreatic tumor as well. Simultaneous application of agonistic anti-CD40 antibody was necessary to achieve effective therapeutic response (80% recovery) in this model. Our results suggest that herein presented immunotherapeutic approach is a promising cancer treatment strategy with the ability to eradicate not only primary tumors but also metastases.
Collapse
Affiliation(s)
- Veronika Caisová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Ondřej Uher
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Pavla Nedbalová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Ivana Jochmanová
- 1st Department of Internal Medicine, Medical Faculty of P. J. Šafárik University in Košice, Košice, Slovakia
| | - Karolína Kvardová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Kamila Masáková
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Gabriela Krejčová
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lucie Paďouková
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Jindřich Chmelař
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Jan Kopecký
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic; Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, v.v.i., České Budějovice, Czech Republic
| | - Jan Ženka
- Department of Medical Biology, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic.
| |
Collapse
|
20
|
Ni G, Wang T, Yang L, Wang Y, Liu X, Wei MQ. Combining anaerobic bacterial oncolysis with vaccination that blocks interleukin-10 signaling may achieve better outcomes for late stage cancer management. Hum Vaccin Immunother 2017; 12:599-606. [PMID: 26367244 DOI: 10.1080/21645515.2015.1089008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Late stage solid tumors cause significant cancer mortality rates worldwide and effective therapy remains a big challenge. Cancer therapeutic vaccines elicit tumor specific T cells that kill tumor cells yet often fail to result in tumor destruction because of the limited T cell response and the local immune-suppressive environment. Blocking interleukin 10 (IL-10) signaling at the time of therapeutic vaccination elicits much stronger T cell responses than vaccination without IL-10 blocking. Anaerobic oncolytic bacteria target hypoxic regions of the late stage tumor tissues which not only stops tumor growth but also provides a pro-inflammatory environment that may increase the effectiveness of a therapeutic vaccine by recruiting more effector T cells to tumor site. In this review, we argue that combining both bacterial and vaccine therapies may improve the efficiency of late stage cancer management.
Collapse
Affiliation(s)
- Guoying Ni
- a School of Medical Science and Griffith Health Institute, Griffith University , Gold Coast , QLD , Australia.,d Tangshan Supervision Institute of Health , Tangshan , China
| | - Tianfang Wang
- c Genecology Research Center, University of the Sunshine Coast , Maroochydore DC , QLD , Australia
| | - Lin Yang
- f Department of Surgical Oncology , Tangshan Gongren Hospital , Tangshan , Hebei , China
| | - Yuejian Wang
- e Cancer Research Institute, Foshan First People's Hospital , Foshan, Guangdong , China
| | - Xiaosong Liu
- b Inflammation and Healing Research Cluster, University of the Sunshine Coast , Maroochydore DC , QLD , Australia.,e Cancer Research Institute, Foshan First People's Hospital , Foshan, Guangdong , China
| | - Ming Q Wei
- a School of Medical Science and Griffith Health Institute, Griffith University , Gold Coast , QLD , Australia
| |
Collapse
|
21
|
Schachtschneider KM, Schwind RM, Newson J, Kinachtchouk N, Rizko M, Mendoza-Elias N, Grippo P, Principe DR, Park A, Overgaard NH, Jungersen G, Garcia KD, Maker AV, Rund LA, Ozer H, Gaba RC, Schook LB. The Oncopig Cancer Model: An Innovative Large Animal Translational Oncology Platform. Front Oncol 2017; 7:190. [PMID: 28879168 PMCID: PMC5572387 DOI: 10.3389/fonc.2017.00190] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/10/2017] [Indexed: 12/20/2022] Open
Abstract
Despite an improved understanding of cancer molecular biology, immune landscapes, and advancements in cytotoxic, biologic, and immunologic anti-cancer therapeutics, cancer remains a leading cause of death worldwide. More than 8.2 million deaths were attributed to cancer in 2012, and it is anticipated that cancer incidence will continue to rise, with 19.3 million cases expected by 2025. The development and investigation of new diagnostic modalities and innovative therapeutic tools is critical for reducing the global cancer burden. Toward this end, transitional animal models serve a crucial role in bridging the gap between fundamental diagnostic and therapeutic discoveries and human clinical trials. Such animal models offer insights into all aspects of the basic science-clinical translational cancer research continuum (screening, detection, oncogenesis, tumor biology, immunogenicity, therapeutics, and outcomes). To date, however, cancer research progress has been markedly hampered by lack of a genotypically, anatomically, and physiologically relevant large animal model. Without progressive cancer models, discoveries are hindered and cures are improbable. Herein, we describe a transgenic porcine model—the Oncopig Cancer Model (OCM)—as a next-generation large animal platform for the study of hematologic and solid tumor oncology. With mutations in key tumor suppressor and oncogenes, TP53R167H and KRASG12D, the OCM recapitulates transcriptional hallmarks of human disease while also exhibiting clinically relevant histologic and genotypic tumor phenotypes. Moreover, as obesity rates increase across the global population, cancer patients commonly present clinically with multiple comorbid conditions. Due to the effects of these comorbidities on patient management, therapeutic strategies, and clinical outcomes, an ideal animal model should develop cancer on the background of representative comorbid conditions (tumor macro- and microenvironments). As observed in clinical practice, liver cirrhosis frequently precedes development of primary liver cancer or hepatocellular carcinoma. The OCM has the capacity to develop tumors in combination with such relevant comorbidities. Furthermore, studies on the tumor microenvironment demonstrate similarities between OCM and human cancer genomic landscapes. This review highlights the potential of this and other large animal platforms as transitional models to bridge the gap between basic research and clinical practice.
Collapse
Affiliation(s)
| | - Regina M Schwind
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | | | | | - Mark Rizko
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nasya Mendoza-Elias
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Paul Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Daniel R Principe
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Alex Park
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nana H Overgaard
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gregers Jungersen
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kelly D Garcia
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL, United States
| | - Ajay V Maker
- Department of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, United States
| | - Laurie A Rund
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Howard Ozer
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Ron C Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States.,Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| |
Collapse
|
22
|
Rossi S, Toschi L, Castello A, Grizzi F, Mansi L, Lopci E. Clinical characteristics of patient selection and imaging predictors of outcome in solid tumors treated with checkpoint-inhibitors. Eur J Nucl Med Mol Imaging 2017; 44:2310-2325. [PMID: 28815334 DOI: 10.1007/s00259-017-3802-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/04/2017] [Indexed: 12/13/2022]
Abstract
The rapidly evolving knowledge on tumor immunology and the continuous implementation of immunotherapy in cancer have recently led to the FDA and EMA approval of several checkpoint inhibitors as immunotherapic agents in clinical practice. Anti-CTLA-4, anti-PD-1, and anti-PDL-1 antibodies are becoming standard of care in advanced melanoma, as well as in relapsed or metastatic lung and kidney cancer, demonstrating higher and longer response compared to standard chemotherapy. These encouraging results have fostered the evaluation of these antibodies either alone or in combination with other therapies in several dozen clinical trials for the treatment of multiple tumor types. However, not all patients respond to immune checkpoint inhibitors, hence, specific biomarkers are necessary to guide and monitor therapy. The utility of PD-L1 expression as a biomarker has varied in different clinical trials, but, to date, no consensus has been reached on whether PD-L1 expression is an ideal marker for response and patient selection; approximately 20-25% of patients will respond to immunotherapy with checkpoint inhibitors despite a negative PD-L1 staining. On the other hand, major issues concern the evaluation of objective response in patients treated with immunotherapy. Pure morphological criteria as commonly used in solid tumors (i.e. RECIST) are not sufficient because change in size is not an early and reliable marker of tumor response to biological therapies. Thus, the scientific community has required a continuous adaptation of immune-related response criteria (irRC) to overcome the problem. In this context, metabolic information and antibody-based imaging with positron emission tomography (PET) have been investigated, providing a powerful approach for an optimal stratification of patients at staging and during the evaluation of the response to therapy. In the present review we provide an overview on the clinical characteristics of patient selection when using imaging predictors of outcome in solid tumors treated with checkpoint-inhibitors.
Collapse
Affiliation(s)
- Sabrina Rossi
- Medical Oncology, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Luca Toschi
- Medical Oncology, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Angelo Castello
- Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Fabio Grizzi
- Immunology and Inflammation, Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Luigi Mansi
- Nuclear Medicine, Seconda Università di Napoli, Naples, Italy
| | - Egesta Lopci
- Nuclear Medicine, Humanitas Clinical and Research Hospital, Rozzano, Italy.
- Nuclear Medicine, Humanitas Cancer Center, Humanitas Clinical and Research Hospital, Via Manzoni 56, 20089, Rozzano, MI, Italy.
| |
Collapse
|
23
|
Li N, Zheng Z, Li J, Fan J, Wang T, Zhang J, Wang H, Chen J, Lv Y, Yi J, Huang M, Ling R. Immediate breast reconstruction with omental flap for luminal breast cancer patients: Ten clinical case reports. Medicine (Baltimore) 2017; 96:e7797. [PMID: 28816969 PMCID: PMC5571706 DOI: 10.1097/md.0000000000007797] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
RATIONALE Luminal subtype breast cancer, accounting for 70 to 80% of all breast cancers, has been reported to be associated with good prognosis. However, for the patients with large mass or worse mass position, omental flap transplantation may provide a new option for breast reconstruction. PATIENT CONCERNS Ten patients (6 luminal B1, 2 luminal B2, 2 luminal A), were enrolled into the study, between January 23, 2015 and August 22, 2016. The mean age was 34.6 ± 6.96 (24-44) years old. Immunohistochemistry demonstrated that the tumor cells were positive for estrogen receptor and progestrone receptor. DIAGNOSES According to the clinicopathological features, diagnosis of breast cancer patients were made. INTERVENTIONS Breast-conserving surgery, laparoscopic greater omentum harvest and vascular anas-tomosis were carried out orderly. Postoperative operative results, cosmetic outcomes, complications, as well as blood supply were investigated for surgery evaluation. Reasonable chemotherapy and irradia-tion were adopted to patients according to the pathological condition. OUTCOMES We successfully accomplished breast reconstruction by omental flap transplantation, ex-cept one failed case because of the necrosis of omentum and changed to fat transplantation. The volumes and symmetry of breasts were all satisfied. The blood supply was detected to be fluent. Only one case of slight hematoma and another case of one distant metastasis were observed during fol-low-up period. No arm mordities or arm movement restriction occurred after surgery. Moreover, radia-tion therapy and chemotherapy had no clear effects on the reconstructed breast. LESSONS Immediate breast reconstruction surgery by transplanting omental flap for luminal breast cancer patients can be considered successful based on the excellent clinic outcome.
Collapse
Affiliation(s)
- Nanlin Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Zhao Zheng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University
| | - Jipeng Li
- Department of Digestive Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jing Fan
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Ting Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Juliang Zhang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Hui Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Jianghao Chen
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Yonggang Lv
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Jun Yi
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Meiling Huang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| | - Rui Ling
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University
| |
Collapse
|
24
|
Maccalli C, Giannarelli D, Chiarucci C, Cutaia O, Giacobini G, Hendrickx W, Amato G, Annesi D, Bedognetti D, Altomonte M, Danielli R, Calabrò L, Di Giacomo AM, Marincola FM, Parmiani G, Maio M. Soluble NKG2D ligands are biomarkers associated with the clinical outcome to immune checkpoint blockade therapy of metastatic melanoma patients. Oncoimmunology 2017; 6:e1323618. [PMID: 28811958 DOI: 10.1080/2162402x.2017.1323618] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/20/2017] [Accepted: 04/21/2017] [Indexed: 12/23/2022] Open
Abstract
The introduction of immune checkpoint blockade into the clinical practice resulted in improvement of survival of a significant portion of melanoma patients. Consequently, predictive biomarkers of response are needed to optimize patient's stratification and the development of combination therapies. The aim of this study was to determine whether levels of soluble NKG2D ligands (MICA, MICB, ULBP1, 2 and 3; sNKG2DLs) in the serum of melanoma patients can serve as useful predictors of response to the treatment with immune checkpoint blockade. sNKG2DLs were measured by ELISA in baseline and post-treatment serum and these results were correlated with the clinical outcome of melanoma patients (N = 194). The same determinations were performed also in a cohort of patients (N = 65) treated with either chemotherapy, radiotherapy, or mutated BRAF inhibitors (BRAFi). Absence of soluble MICB and ULBP-1 in baseline serum correlated with improved survival (OS = 21.6 and 25.3 mo and p = 0.02 and 0.01, respectively) of patients treated with immunological therapies while detectable levels of these molecules were found in poor survivors (OS = 8.8 and 12.1 mo, respectively). Multivariate analysis showed that LDH (p <0.0001), sULBP-1 (p = 0.02), and sULBP-2 (p = 0.02) were independent predictors of clinical outcome for the cohort of melanoma patients treated with immune checkpoint blockade. Only LDH but not sNKG2DLs was significantly associated with the clinical outcome of patients treated with standard or BRAFi regimens. These findings highlight the relevance of sNKG2DLs in the serum of melanoma patients as biomarkers for patients' stratification and optimization of immune checkpoint inhibition regimens.
Collapse
Affiliation(s)
- Cristina Maccalli
- Research Branch, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Diana Giannarelli
- Unit of Statistics, Regina Elena National Cancer Institute, Rome, Italy
| | - Carla Chiarucci
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.,University of Siena, Siena, Italy
| | - Ornella Cutaia
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.,University of Siena, Siena, Italy
| | - Gianluca Giacobini
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.,University of Siena, Siena, Italy
| | - Wouter Hendrickx
- Research Branch, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Giovanni Amato
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Diego Annesi
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Davide Bedognetti
- Research Branch, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Maresa Altomonte
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Riccardo Danielli
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Luana Calabrò
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Anna Maria Di Giacomo
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Francesco M Marincola
- Office of the Chief Research Officer (CRO), Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Giorgio Parmiani
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy.,Italian Network for Bio-therapy of Tumors-(NIBIT)-Laboratory, Siena, Italy
| | - Michele Maio
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| |
Collapse
|
25
|
Shi Y, Du L, Lin L, Wang Y. Tumour-associated mesenchymal stem/stromal cells: emerging therapeutic targets. Nat Rev Drug Discov 2016; 16:35-52. [PMID: 27811929 DOI: 10.1038/nrd.2016.193] [Citation(s) in RCA: 322] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells, also known as mesenchymal stromal cells (MSCs), exist in many tissues and are known to actively migrate to sites of tissue injury, where they participate in wound repair. Tumours can be considered "wounds that never heal" and, in response to cues from a tumour, MSCs are continuously recruited to and become integral components of the tumour microenvironment. Recently, it has become apparent that such tumour-associated MSCs (TA-MSCs) have an active role in tumour initiation, promotion, progression and metastasis. In this Review, we discuss recent advances in our understanding of the pathogenic role of TA-MSCs in regulating the survival, proliferation, migration and drug resistance of tumour cells, as well as the influence of MSCs on the immune status of the tumour microenvironment. Moreover, we discuss therapeutic approaches that target TA-MSC upstream or downstream modulators or use MSCs as vehicles for the delivery of tumoricidal agents. It is anticipated that new insights into the functions of TA-MSCs will lead to the development of novel therapeutic strategies against tumours.
Collapse
Affiliation(s)
- Yufang Shi
- The First Affiliated Hospital of Soochow University and Jiangsu Engineering Research Center for Tumor Immunotherapy, Institutes for Translational Medicine and Suzhou Key Laboratory of Tumor Microenvironment and Pathology, Soochow University, 199 Renai Road, Suzhou, Jiangsu 215123, China.,Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901, USA.,Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Liming Du
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Liangyu Lin
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China.,Shanghai Jiao Tong University School of Medicine, 280 Chongqing Road, Shanghai 200025, China
| | - Ying Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| |
Collapse
|
26
|
Yang L, Liu H, Zhang L, Hu J, Chen H, Wang L, Yin X, Li Q, Qi Y. Effect of IL-17 in the development of colon cancer in mice. Oncol Lett 2016; 12:4929-4936. [PMID: 28101230 PMCID: PMC5228117 DOI: 10.3892/ol.2016.5329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 06/16/2016] [Indexed: 11/20/2022] Open
Abstract
Cytokine therapy is commonly used for tumor immunotherapy. Although early studies focused directly on the tumor, current investigations are more attentive of the tumor microenvironment. Various immune cells and related cytokines in the tumor microenvironment play an important role in the occurrence and development of tumor. Interleukin (IL)-17 is the characteristic cytokine produced by Th17 cells. IL-17 has been associated with various immune responses. The results of previous studies showed that IL-17 can significantly reduce the size of transplanted tumors in tumor-bearing mice, albeit it has no effect on the survival time of mice. By investigating the effect of IL-17 in the number and distribution of lymphocyte infiltration in tumor tissues, the expression of cytokines and transcription factors associated with the subsets of CD4+T cells in tumor tissues, the distribution of subsets of spleen lymphocyte in tumor-bearing mice, a preliminary investigation of the possible antitumor mechanism of IL-17 was performed. In conclusion, the antitumor effect of IL-17 gene transfection in the colon cancer of mice may be associated with the mechanisms whereby IL-17 gene transfection can change the distribution of different subsets of spleen lymphocytes in mice. IL-17 gene transfection can increase the number of lymphocyte infiltration in tumor tissues. IL-17 gene transfection can promote the high expression of interferon-γ in tumor tissue, while reducing the expression of IL-10 and IL-13 factors, thus exerting an antitumor effect.
Collapse
Affiliation(s)
- Lijuan Yang
- Department of Immunology, College of Basic Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; The Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei 050017, P.R. China
| | - Hao Liu
- Department of the Nuclear Medicine, The First Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Lili Zhang
- The Second Hospital of Shijiazhuang, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jie Hu
- School of Nursing, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Haixia Chen
- Department of Electroencephalogram, Xingtai People's Hospital, Xingtai, Hebei 054031, P.R. China
| | - Lei Wang
- Department of Pathogenic Biology and Immunology, Cangzhou Medical College, Cangzhou, Hebei 061001, P.R. China
| | - Xiaolin Yin
- Department of Immunology, College of Basic Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; The Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, Hebei 050017, P.R. China
| | - Quanhai Li
- Department of Immunology, College of Basic Medical Sciences, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China; Cell Therapy Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050031, P.R. China
| | - Yixin Qi
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
27
|
Saetang J, Puseenam A, Roongsawang N, Voravuthikunchai SP, Sangkhathat S, Tipmanee V. Immunologic Function and Molecular Insight of Recombinant Interleukin-18. PLoS One 2016; 11:e0160321. [PMID: 27483370 PMCID: PMC4970725 DOI: 10.1371/journal.pone.0160321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/18/2016] [Indexed: 11/18/2022] Open
Abstract
In recent years, cytokine-mediated therapy has emerged as further advance alternative in cancer therapy. Interleukin-18 (IL-18) has exhibited interesting anti-cancer properties especially when combined with IL-12. We engineered IL-18 in order to improve its activity using single point mutagenesis. IL-18 mutants were constructed according to binding residues and polarity which we tried to increase polarity in M33Q and M60Q, enhanced cationicity in E6K, and flexibility in T63A. All IL-18 proteins were expressed in Pichia pastoris, purified, and then measured the activity by treating with the NK-92MI cell line to evaluate interferon-γ (IFN-γ) stimulation. The E6K and T63A mutant forms showed higher activity with respect to native proteins at the concentration of 200 ng mL-1 by inducing the expression of IFN-γ, about factors of 9 and 4, respectively. Meanwhile, M33Q and M60Q had no significant activity to induce IFN-γ. Interestingly, the combination of E6K and T63A mutations could synergize the induction activity of IL-18 to be 16 times at 200 ng mL-1. Furthermore, molecular dynamics studies have elucidated the effect due to mutation on conformation of the binding site of IL-18. The results turn out that E6K provides structural perseverance against mutation, while M33Q and M60Q promote vivid overall change in protein conformation, especially at the binding site. For T63A, mutation yields small difference in structure but clearly increases structural flexibility. However, a small structural change was observed when T63A was combined with E6K. Our research resulted in a novel version of IL-18 which could be a new key candidate for cytokine-mediated therapy.
Collapse
MESH Headings
- Amino Acid Sequence
- Binding Sites
- Cell Line, Tumor
- Cloning, Molecular
- Gene Expression
- Humans
- Interferon-gamma/biosynthesis
- Interferon-gamma/metabolism
- Interleukin-18/chemistry
- Interleukin-18/genetics
- Interleukin-18/immunology
- Interleukin-18/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Kinetics
- Lymphocyte Activation/drug effects
- Models, Molecular
- Molecular Weight
- Pichia/genetics
- Pichia/metabolism
- Protein Binding
- Protein Conformation, alpha-Helical
- Protein Conformation, beta-Strand
- Protein Engineering
- Protein Interaction Domains and Motifs
- Receptors, Interleukin-18/chemistry
- Receptors, Interleukin-18/genetics
- Receptors, Interleukin-18/immunology
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Sequence Alignment
- Structure-Activity Relationship
- Substrate Specificity
Collapse
Affiliation(s)
- Jirakrit Saetang
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
- Graduate School, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Aekkachai Puseenam
- Microbial Cell Factory Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Niran Roongsawang
- Microbial Cell Factory Laboratory, Bioresources Technology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 113 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, Pathum Thani, 12120, Thailand
| | - Supayang Piyawan Voravuthikunchai
- Department of Microbiology and Natural Product Research Center of Excellence, Faculty of Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Surasak Sangkhathat
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
- Department of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Varomyalin Tipmanee
- Department of Biomedical Sciences, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
- * E-mail:
| |
Collapse
|
28
|
Long-Term Clinical Responses of Neoadjuvant Dendritic Cell Infusions and Radiation in Soft Tissue Sarcoma. Sarcoma 2015; 2015:614736. [PMID: 26880867 PMCID: PMC4735941 DOI: 10.1155/2015/614736] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 11/14/2015] [Accepted: 11/29/2015] [Indexed: 11/17/2022] Open
Abstract
Purpose. Patients with large >5 cm, high-grade resectable soft tissue sarcomas (STS) have the highest risk of distant metastases. Previously we have shown that dendritic cell (DC) based vaccines show consistent immune responses. Methods. This was a Phase I single institution study of neoadjuvant radiation with DC injections on 18 newly diagnosed high-risk STS patients. Neoadjuvant treatment consisted of 50 Gy of external beam radiation (EBRT), given in 25 fractions delivered five days/week, combined with four intratumoral injections of DCs followed by complete resection. The primary endpoint was to establish the immunological response to neoadjuvant therapy and obtain data on its clinical safety and outcomes. Results. There were no unexpected toxicities or serious adverse events. Twelve out of 18 (67%) patients were alive, of which an encouraging 11/18 (61%) were alive with no systemic recurrence over a period of 2-8 years. Favorable immunological responses correlated with clinical responses in some cases. Conclusions. This study provides clinical support to using dendritic cell injections along with radiation in sarcomas, which when used optimally in combination can help clinical outcomes in soft tissue sarcoma. Study registration number is NCT00365872.
Collapse
|
29
|
Dahlberg CIM, Sarhan D, Chrobok M, Duru AD, Alici E. Natural Killer Cell-Based Therapies Targeting Cancer: Possible Strategies to Gain and Sustain Anti-Tumor Activity. Front Immunol 2015; 6:605. [PMID: 26648934 PMCID: PMC4663254 DOI: 10.3389/fimmu.2015.00605] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022] Open
Abstract
Natural killer (NK) cells were discovered 40 years ago, by their ability to recognize and kill tumor cells without the requirement of prior antigen exposure. Since then, NK cells have been seen as promising agents for cell-based cancer therapies. However, NK cells represent only a minor fraction of the human lymphocyte population. Their skewed phenotype and impaired functionality during cancer progression necessitates the development of clinical protocols to activate and expand to high numbers ex vivo to be able to infuse sufficient numbers of functional NK cells to the cancer patients. Initial NK cell-based clinical trials suggested that NK cell-infusion is safe and feasible with almost no NK cell-related toxicity, including graft-versus-host disease. Complete remission and increased disease-free survival is shown in a small number of patients with hematological malignances. Furthermore, successful adoptive NK cell-based therapies from haploidentical donors have been demonstrated. Disappointingly, only limited anti-tumor effects have been demonstrated following NK cell infusion in patients with solid tumors. While NK cells have great potential in targeting tumor cells, the efficiency of NK cell functions in the tumor microenvironment is yet unclear. The failure of immune surveillance may in part be due to sustained immunological pressure on tumor cells resulting in the development of tumor escape variants that are invisible to the immune system. Alternatively, this could be due to the complex network of immune-suppressive compartments in the tumor microenvironment, including myeloid-derived suppressor cells, tumor-associated macrophages, and regulatory T cells. Although the negative effect of the tumor microenvironment on NK cells can be transiently reverted by ex vivo expansion and long-term activation, the aforementioned NK cell/tumor microenvironment interactions upon reinfusion are not fully elucidated. Within this context, genetic modification of NK cells may provide new possibilities for developing effective cancer immunotherapies by improving NK cell responses and making them less susceptible to the tumor microenvironment. Within this review, we will discuss clinical trials using NK cells with a specific reflection on novel potential strategies, such as genetic modification of NK cells and complementary therapies aimed at improving the clinical outcome of NK cell-based immune therapies.
Collapse
Affiliation(s)
- Carin I M Dahlberg
- Cell Therapies Institute, Nova Southeastern University , Fort Lauderdale, FL , USA ; Cell and Gene Therapy Group, Center for Hematology and Regenerative Medicine (HERM), Karolinska University Hospital Huddinge, NOVUM , Stockholm , Sweden
| | - Dhifaf Sarhan
- Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet , Stockholm , Sweden ; Division of Hematology, Oncology and Transplantation, Masonic Cancer Research Center, University of Minnesota , Minnesota, MN , USA
| | - Michael Chrobok
- Cell Therapies Institute, Nova Southeastern University , Fort Lauderdale, FL , USA ; Cell and Gene Therapy Group, Center for Hematology and Regenerative Medicine (HERM), Karolinska University Hospital Huddinge, NOVUM , Stockholm , Sweden
| | - Adil D Duru
- Cell Therapies Institute, Nova Southeastern University , Fort Lauderdale, FL , USA ; Cell and Gene Therapy Group, Center for Hematology and Regenerative Medicine (HERM), Karolinska University Hospital Huddinge, NOVUM , Stockholm , Sweden
| | - Evren Alici
- Cell Therapies Institute, Nova Southeastern University , Fort Lauderdale, FL , USA ; Cell and Gene Therapy Group, Center for Hematology and Regenerative Medicine (HERM), Karolinska University Hospital Huddinge, NOVUM , Stockholm , Sweden ; Hematology Center, Karolinska University Hospital Huddinge , Stockholm , Sweden
| |
Collapse
|
30
|
Diao Y, Wang X, Wan Y, Zhong J, Gao D, Liu Y, Gao N, Li W, Liu B, Huang X, Jin Z, Peng B, Wang Z, Fu L, Chen S, Jin G. Antitumor activity of a novel small molecule TLR7 agonist via immune response induction and tumor microenvironment modulation. Oncol Rep 2015; 35:793-800. [PMID: 26718332 DOI: 10.3892/or.2015.4436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 10/11/2015] [Indexed: 11/06/2022] Open
Abstract
Immunotherapy is emerging as a powerful and active tumor-specific approach against cancer via triggering the immune system. Toll-like receptors (TLRs) are fundamental elements of the immune system, which facilitate our understanding of the innate and adaptive immune pathways. TLR agonists used as single agents can effectively eradicate tumors due to their potent stimulation of innate and adaptive immunity. We examined the effects of a novel adenine type of TLR7 agonists on both innate and adaptive immune activation in vitro and in vivo. We established the local and distant tumor‑bearing mice derived from murine mammary carcinoma cell line (4T1) to model metastatic disease. Our data demonstrated that SZU101 was able to stimulate innate immune cells to release cytokines at the very high level compared with LPS at the same or lower concentration. Locally intratumoral SZU101 injection can elicit a systemic antitumor effect on murine breast tumor model. SZU101 affected the frequency of intratumoral immune cell infiltration, including the percentage of CD4+ and CD8+ increase, and the ratio of Tregs decrease. Our data reveal that the antitumor effect of SZU101 is associated with multiple mechanisms, inducing tumor‑specific immune response, activation of innate immune cells and modulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Yuwen Diao
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Xiaodong Wang
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Yanyan Wan
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Jingjing Zhong
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Dong Gao
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Yu Liu
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Ningning Gao
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Wang Li
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Bing Liu
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Xinping Huang
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Zhenchao Jin
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Boya Peng
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Zhulin Wang
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Li Fu
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Siping Chen
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Guangyi Jin
- Cancer Research Center, National‑Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Medicine, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| |
Collapse
|
31
|
Chen S, Ni G, Wu X, Zhu B, Liao Z, Wang Y, Liu X. Blocking IL-10 signalling at the time of immunization renders the tumour more accessible to T cell infiltration in mice. Cell Immunol 2015; 300:9-17. [PMID: 26607604 DOI: 10.1016/j.cellimm.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 11/05/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022]
Abstract
We recently reported that blockade of IL-10 signalling at the time of a human papillomavirus (HPV) long E7 peptide/LPS immunization leads to the regression of established HPV-16 immortalized tumours in mice similar to that induced by long E7 peptide/incomplete Freund's adjuvant (IFA)-based vaccination. In this paper, we demonstrated that blockade of IL-10 signalling at the time of long E7 peptide/LPS could elicit stronger T cells responses and render the tumour more accessible for immune cell infiltration than vaccination with long E7 peptide/IFA. Furthermore, priming with long E7 peptide/LPS and IL10 signalling blockade then boosting with long E7 peptide/IFA elicits stronger CD8+ T cell responses than long E7 peptide/IFA immunization. The results suggest that priming with long E7 peptide/LPS and IL10 signalling inhibitor, then boosting with long E7 peptide/IFA elicits may lead to better HPV infection related tumour regression in clinic.
Collapse
Affiliation(s)
- Shu Chen
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Guoying Ni
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia
| | - Xiaolian Wu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Bin Zhu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Zaowen Liao
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Yuejian Wang
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China.
| | - Xiaosong Liu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China; Inflammation and Healing Research Cluster, School of Health and Sport Sciences, University of Sunshine Coast, Maroochydore DC, QLD 4558, Australia.
| |
Collapse
|
32
|
The immunohistochemical analysis of membrane-bound CD55, CD59 and fluid-phase FH and FH-like complement inhibitors in cancers of ovary and corpus uteri origin. Cent Eur J Immunol 2015; 40:349-53. [PMID: 26648780 PMCID: PMC4655386 DOI: 10.5114/ceji.2015.54598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/15/2015] [Indexed: 12/19/2022] Open
Abstract
One of the potential therapeutic methods of cancer treatment is the immunotherapy with monoclonal antibodies. This kind of therapy, although devoid of serious side effects, has often insufficient efficacy. The presence of complement inhibitors on the cancer cells, which are able to inactivate complement-mediated immune response represents one of the main reasons for the inefficiency of such therapy. In our studies we investigated the expression of main membrane–bound and fluid-phase complement regulators: CD55, CD59 and factor H/factor H-like in tumour samples of ovarian and corpus uteri cancer. Tissue samples were collected from 50 patients and stained immunohistochemically, with the use of peroxidase-based immunodetection system. Immunohistochemical analysis revealed that complement inhibitors are present in examined tumors although their presence is heterogenous. The most prevalent is the presence of factor H/H-like, localized mostly in tumor stroma and within vascular structures. Membrane bound complement inhibitors are less prominently expressed by cancer cells. CD55 was detected in low percentage of cells, predominantly within cancer tubules. CD59 immunoreactivity was more prevalent in cancer cells, and was localized particularly at the margin of cancer cell tubules. Our results demonstrate that the most prominent complement inhibitor in cancer of ovary and corpus uteri origin is factor H/factor H-like. Blocking or downregulation of this inhibitor should be taken into consideration with regards to improving the efficiency of immunotherapy with monoclonal antibodies.
Collapse
|
33
|
Ni G, Wang T, Walton S, Zhu B, Chen S, Wu X, Wang Y, Wei MQ, Liu X. Manipulating IL-10 signalling blockade for better immunotherapy. Cell Immunol 2015; 293:126-9. [PMID: 25596475 DOI: 10.1016/j.cellimm.2014.12.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/29/2014] [Indexed: 01/21/2023]
Abstract
Interleukin 10 is a cytokine with the ability to reduce or terminate inflammation. Chronic viral infection, such as infection of chronic hepatitis B, hepatitis C and HIV, has increased levels of interleukin 10 in peripheral blood. Serum IL-10 levels are also high in certain cancers. Blocking IL-10 signalling at the time of immunisation clears chronic viral infection and prevents tumour growth in animal models. We review recent advances in this area, with the emphasis on potential use of this novel strategy to treat chronic viral infection and cancer in human.
Collapse
Affiliation(s)
- Guoying Ni
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia
| | - Tianfang Wang
- Genecology Research Centre, University of Sunshine Coast, Sippy Downs 4556, QLD, Australia
| | - Shelley Walton
- Inflammation and Healing Research Cluster, University of Sunshine Coast, Sippy Downs 4556, Australia
| | - Bin Zhu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Shu Chen
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Xiaolian Wu
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China
| | - Yuejian Wang
- Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China.
| | - Ming Q Wei
- School of Medical Science, Griffith Health Institute, Griffith University, Gold Coast, QLD 4222, Australia.
| | - Xiaosong Liu
- Inflammation and Healing Research Cluster, University of Sunshine Coast, Sippy Downs 4556, Australia; Cancer Research Institute, Foshan First People's Hospital, Foshan, Guangdong 528000, China.
| |
Collapse
|
34
|
Drakes ML, Stiff PJ. Harnessing immunosurveillance: current developments and future directions in cancer immunotherapy. Immunotargets Ther 2014; 3:151-65. [PMID: 27471706 PMCID: PMC4918242 DOI: 10.2147/itt.s37790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite improved methods of cancer detection and disease management over the last few decades, cancer remains a major public health problem in many societies. Conventional therapies, such as chemotherapy, radiation, and surgery, are not usually sufficient to prevent disease recurrence. Therefore, efforts have been focused on developing novel therapies to manage metastatic disease and to prolong disease-free and overall survival, by modulating the immune system to alleviate immunosuppression, and to enhance antitumor immunity. This review discusses protumor mechanisms in patients that circumvent host immunosurveillance, and addresses current immunotherapy modalities designed to target these mechanisms. Given the complexity of cancer immunosuppressive mechanisms, we propose that identification of novel disease biomarkers will drive the development of more targeted immunotherapy. Finally, administration of different classes of immunotherapy in combination regimens, will be the ultimate route to impact low survival rates in advanced cancer patients.
Collapse
Affiliation(s)
- Maureen L Drakes
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Patrick J Stiff
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|