151
|
Nasr SA, Saad AAEM. Evaluation of the cytotoxic anticancer effect of polysaccharide of Nepeta septemcrenata. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021. [DOI: 10.1186/s43088-021-00135-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Promoting cancer cells apoptosis is one of the effective methods to treat cancer. Human hepatocellular carcinoma (HepG2) and colorectal cancer (HCT-116) cell lines were used in the present study to evaluate the cytotoxic and anticancer properties of Nepeta septemcrenata Polysaccharide (NSP).
Result
Treatment of the two examined cells with NSP displayed a significant cytotoxicity towards HepG2 in a dose-dependent manner; meanwhile, its effect on HCT-116 was obtained under the influence of low doses. The quantitative real- time PCR (QRT-PCR) investigation revealed that NSP significantly up-regulated the expression levels of p53, p16, Fas, Fas-L, Bax, caspases-3, caspase-9, and TNF-α in association with down-regulation of cyclin D1, TERT, and BCL2. These findings declare the apoptotic characteristic of NSP.NSP, can also inhibit the development of cancer cells through the down-regulation of TGF-β and VEGF.
Conclusions
Our results suggested that the polysaccharides isolated from N. septemcrenata possess anticancer properties that could be explored for the development of novel anticancer agents.
Collapse
|
152
|
Aghlara-Fotovat S, Nash A, Kim B, Krencik R, Veiseh O. Targeting the extracellular matrix for immunomodulation: applications in drug delivery and cell therapies. Drug Deliv Transl Res 2021; 11:2394-2413. [PMID: 34176099 DOI: 10.1007/s13346-021-01018-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2021] [Indexed: 12/12/2022]
Abstract
Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.
Collapse
Affiliation(s)
| | - Amanda Nash
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Robert Krencik
- Center for Neuroregeneration, Department of Neurosurgery, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
153
|
Abadi B, Yazdanpanah N, Nokhodchi A, Rezaei N. Smart biomaterials to enhance the efficiency of immunotherapy in glioblastoma: State of the art and future perspectives. Adv Drug Deliv Rev 2021; 179:114035. [PMID: 34740765 DOI: 10.1016/j.addr.2021.114035] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022]
Abstract
Glioblastoma multiform (GBM) is considered as the most lethal tumor among CNS malignancies. Although immunotherapy has achieved remarkable advances in cancer treatment, it has not shown satisfactory results in GBM patients. Biomaterial science, along with nanobiotechnology, is able to optimize the efficiency of immunotherapy in these patients. They can be employed to provide the specific activation of immune cells in tumor tissue and combinational therapy as well as preventing systemic adverse effects resulting from hyperactivation of immune responses and off-targeting effect. Advance biomaterials in this field are classified into targeting nanocarriers and localized delivery systems. This review will offer an overview of immunotherapy strategies for glioblastoma and advance delivery systems for immunotherapeutics that may have a high potential in glioblastoma treatment.
Collapse
|
154
|
Guven DC, Acar R, Yekeduz E, Bilgetekin I, Baytemur NK, Erol C, Ceylan F, Sendur MA, Demirci U, Urun Y, Karadurmus N, Erman M, Kilickap S. The association between antibiotic use and survival in renal cell carcinoma patients treated with immunotherapy: a multi-center study. Curr Probl Cancer 2021; 45:100760. [PMID: 34130864 DOI: 10.1016/j.currproblcancer.2021.100760] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/07/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Immunotherapy improves overall survival (OS) in the second and later lines of renal cell carcinoma (RCC) treatment. Recent studies have suggested that antibiotic (ATB) use either shortly before or after the start of immunotherapy could lead to decreased OS. Herein, we evaluate the impact of ATB use on OS in RCC patients treated with nivolumab in a multi-center cohort from Turkey. METHODS The data of 93 metastatic RCC patients treated with nivolumab in the second line or later were retrospectively collected from 6 oncology centers. Previous treatments, sites of metastases, International Metastatic RCC Database Consortium risk classification, and ATB use in the three months before (-3) or three months after (+3) the start of immunotherapy were recorded together with survival data. The association of clinical factors with OS and progression-free survival (PFS) was analyzed with univariate and multivariable analyses. RESULTS The median age was 61 (interquartile range 54-67), and 76.3% of the patients were male. The median OS of the cohort was 23.75 ± 4.41, and the PFS was 8.44 ± 1.61 months. Thirty-one (33.3%) patients used ATBs in the 3 months before (-3) or 3 months after (+3) nivolumab initiation. In the multivariable analyses, ATB exposure (HR: 2.306, 95% confidence interval [CI]: 1.155-4.601, P = 0.018) and the presence of brain metastases at the baseline (HR: 2.608, 95% CI: 1.200-5.666, P = 0.015) had a statistically significant association with OS, while ATB exposure was the only statistically significant parameter associated with PFS (HR: 2.238, 95% CI: 1.284-3.900, P = 0.004). CONCLUSION In our study, patients with ATB exposure in the 3 months before or 3 months after the start of immunotherapy had shorter OS. Our findings further support meticulous risk-benefit assessments of prescribing ATBs for patients who are either receiving or are expected to receive immunotherapy.
Collapse
Affiliation(s)
- Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey.
| | - Ramazan Acar
- Department of Medical Oncology, Health Sciences University, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Emre Yekeduz
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Irem Bilgetekin
- Department of Medical Oncology, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Ankara, Turkey
| | | | - Cihan Erol
- Department of Medical Oncology, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Furkan Ceylan
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey
| | - Mehmet Ali Sendur
- Department of Medical Oncology, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Umut Demirci
- Department of Medical Oncology, Memorial Ankara Hospital, Ankara, Turkey
| | - Yuksel Urun
- Department of Medical Oncology, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Nuri Karadurmus
- Department of Medical Oncology, Health Sciences University, Gulhane Training and Research Hospital, Ankara, Turkey
| | - Mustafa Erman
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey
| | - Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Oncology Hospital, Ankara, Turkey; Department of Medical Oncology, Istinye University Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
155
|
Kumar AR, Devan AR, Nair B, Vinod BS, Nath LR. Harnessing the immune system against cancer: current immunotherapy approaches and therapeutic targets. Mol Biol Rep 2021; 48:8075-8095. [PMID: 34671902 PMCID: PMC8605995 DOI: 10.1007/s11033-021-06752-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a rapidly evolving concept that has been given the tag "fifth pillar" of cancer therapy while radiation therapy, chemotherapy, surgery and targeted therapy remain the other four pillars. This involves the stimulation of the immune system to control tumor growth and it specifically targets the neoplastic cells rather than the normal cells. Conventional chemotherapy has many limitations which include drug resistance, recurrence of cancer and severe adverse effects. Immunology has made major treatment breakthroughs for several cancers such as colorectal cancer, prostate cancer, breast cancer, lung cancer, liver cancer, kidney cancer, stomach cancer, acute lymphoblastic leukaemia etc. Currently, therapeutic strategies harnessing the immune system involve Checkpoint inhibitors, Chimeric antigen receptor T cells (CAR T cells), Monoclonal antibodies, Cancer vaccines, Cytokines, Radio-immunotherapy and Oncolytic virus therapy. The molecular characterization of several tumor antigens (TA) indicates that these TA can be utilized as promising candidates in cancer immunotherapy strategies. Here in this review, we highlight and summarize the different categories of emerging cancer immunotherapies along with the immunologically recognized tumor antigens involved in the tumor microenvironment.
Collapse
Affiliation(s)
- Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Balachandran S Vinod
- Department of Biochemistry, Sree Narayana College, Kollam, Kerala, 691001, India.
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India.
| |
Collapse
|
156
|
Ali AL, Nailwal NP, Doshi GM. Emerging Role of Interleukins for the Assessment and Treatment of Liver Diseases. Endocr Metab Immune Disord Drug Targets 2021; 22:371-382. [PMID: 34819013 DOI: 10.2174/1871530321666211124102837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/15/2021] [Accepted: 10/25/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The most common liver diseases are fibrosis, alcoholic liver disease, non-alcoholic fatty disease, viral hepatitis, and hepatocellular carcinoma. These liver diseases account for approximately 2 million deaths per year worldwide, with cirrhosis accounting for 2.1% of the worldwide burden. The most widely used liver function tests for diagnosis are alanine transaminase, aspartate transaminase, serum proteins, serum albumin, and serum globulins, whereas antivirals and corticosteroids have been proven to be useful for the treatment of liver diseases. A major disadvantage of these diagnostic measures is the lack of specificity to a particular tissue or cell type, as these enzymes are common to one or more tissues. The major adverse effect of current treatment methods is drug resistance. To overcome these issues, interleukins have been investigated. The balance of these interleukins determines the outcome of an immune response. Interleukins are considered interesting therapeutic targets for the treatment of liver diseases. In this review, we summarize the current state of knowledge regarding interleukins in the diagnosis, treatment, and pathogenesis of different acute and chronic liver diseases. OBJECTIVE To understand the role of interleukins in the assessment and treatment of different types of liver diseases. METHODS A literature search was conducted using PubMed, Science Direct, and NCBI with the following keywords: Interleukins, Acute Liver Failure, Alcoholic Liver Disease, Non-Alcoholic Fatty Liver Disease, Liver Fibrosis, Hepatocellular Carcinoma, Inflammation, Liver injury, Hepatoprotective effect. Clinical trial data on these interleukins have been searched on Clinicaltrials.gov. RESULTS Existing literature and preclinical and clinical trial data demonstrate that interleukins play a crucial role in the pathogenesis of liver diseases. CONCLUSION Our findings indicate that IL-1, IL-6, IL-10, IL-17, IL-22, IL-35, and IL-37 are involved in the progression and control of various liver conditions via the regulation of cell signaling pathways. However, further investigation on the involvement of these interleukins is necessary for their use as a targeted therapy in liver diseases.
Collapse
Affiliation(s)
- Aaliya L Ali
- Department of Pharmacology, SVKM'S Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, Vile Parle (W), Mumbai-400056. India
| | - Namrata P Nailwal
- Department of Pharmacology, SVKM'S Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, Vile Parle (W), Mumbai-400056. India
| | - Gaurav M Doshi
- Department of Pharmacology, SVKM'S Dr. Bhanuben Nanavati College of Pharmacy, Mithibai Campus, Vile Parle (W), Mumbai-400056. India
| |
Collapse
|
157
|
Yang Y, Liu Q, Shi X, Zheng Q, Chen L, Sun Y. Advances in plant-derived natural products for antitumor immunotherapy. Arch Pharm Res 2021; 44:987-1011. [PMID: 34751930 DOI: 10.1007/s12272-021-01355-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 10/29/2021] [Indexed: 12/28/2022]
Abstract
In recent years, immunotherapy has emerged as a novel antitumor strategy in addition to traditional surgery, radiotherapy and chemotherapy. It uniquely focuses on immune cells and immunomodulators in the tumor microenvironment and helps eliminate tumors at the root by rebuilding the immune system. Despite remarkable breakthroughs, cancer immunotherapy still faces many challenges: lack of predictable and prognostic biomarkers, adverse side effects, acquired treatment resistance, high costs, etc. Therefore, more efficacious and efficient, safer and cheaper antitumor immunomodulatory drugs have become an urgent requirement. For decades, plant-derived natural products obtained from land and sea have provided the most important source for the development of antitumor drugs. Currently, more attention is being paid to the discovery of potential cancer immunotherapy modulators from plant-derived natural products, such as polysaccharides, phenols, terpenoids, quinones and alkaloids. Some of these agents have outstanding advantages of multitargeting and low side effects and low cost compared to conventional immunotherapeutic agents. We intend to summarize the progress of comprehensive research on these plant-derived natural products and their derivatives and discuss their possible mechanisms in regulating the immune system and their efficacy as monotherapies or in combination with regular chemotherapeutic agents.
Collapse
Affiliation(s)
- Yi Yang
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Xianai Shi
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China
| | - Li Chen
- Fujian Provincial Key Laboratory of Medical Instrument and Pharmaceutical Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, 350108, China.
| | - Yang Sun
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
- Department of Gyn-Surgical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, 350014, China.
| |
Collapse
|
158
|
Identification of a Prognosis-Related Risk Signature for Bladder Cancer to Predict Survival and Immune Landscapes. J Immunol Res 2021; 2021:3236384. [PMID: 34708131 PMCID: PMC8545590 DOI: 10.1155/2021/3236384] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/22/2022] Open
Abstract
Background Bladder cancer is the tenth most common cancer worldwide. Valuable biomarkers in the field of diagnostic bladder cancer are urgently required. Method Here, the gene expression matrix and clinical data were obtained from The Cancer Genome Atlas (TCGA), GSE13507, GSE32894, and Mariathasan et al. Five prognostic genes were identified by the univariate, robust, and multivariate Cox's regression and were used to develop a prognosis-related model. The Kaplan-Meier survival curves and receiver operating characteristics were used to evaluate the model's effectiveness. The potential biological functions of the selected genes were analyzed using CIBERSORT and ESTIMATE algorithms. Cancer Therapeutics Response Portal (CTRP) and PRISM datasets were used to identify drugs with high sensitivity. Subsequently, using the bladder cancer (BLCA) cell lines, the role of TNFRSF14 was determined by Western blotting, cell proliferation assay, and 5-ethynyl-20-deoxyuridine assay. Results GSDMB, CLEC2D, APOL2, TNFRSF14, and GBP2 were selected as prognostic genes in bladder cancer patients. The model's irreplaceable reliability was validated by the training and validation cohorts. CD8+ T cells were highly infiltrated in the high-TNFRSF14-expression group, and M2 macrophages were the opposite. Higher expression of TNFRSF14 was associated with higher expression levels of LCK, interferon, MHC-I, and MHC-II, while risk score was the opposite. Many compounds with higher sensitivity for treating bladder cancer patients in the low-TNFRSF14-expression group were identified, with obatoclax being a potential drug most likely to treat patients in the low-TNFRSF14-expression group. Finally, the proliferation of BLCA cell lines was increased in the TNFRSF14-reduced group, and the differential expression was identified. TNFRSF14 plays a role in bladder cancer progression through the Wnt/β-catenin-dependent pathway. TNFRSF14 is a potential protective biomarker involved in cell proliferation in BLCA. Conclusion We conducted a study to establish a 5-gene score model, providing reliable prediction for the outcome of bladder cancer patients and therapeutic drugs to individualize therapy. Our findings provide a signature that might help determine the optimal treatment for individual patients with bladder cancer.
Collapse
|
159
|
Sahu M, Suryawanshi H. Immunotherapy: The future of cancer treatment. J Oral Maxillofac Pathol 2021; 25:371. [PMID: 34703141 PMCID: PMC8491352 DOI: 10.4103/0973-029x.325257] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/02/2020] [Accepted: 03/31/2021] [Indexed: 12/01/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are one of the most common cancers worldwide. A large number of patients are diagnosed with locally advanced disease and require multimodal treatment approaches. Standard treatment modalities ranging from surgery to chemotherapy and radiation are yielding mixed results. To overcome this hurdle, newer innovative approaches are required to reduce the morbidity and mortality of the patients. In the last few decades, immunotherapy has become an important part of treating some types of cancer. The immune system plays a key role in the development, establishment and progression of HNSCC. A greater understanding of the dysregulation and evasion of the immune system in the evolution and progression of HNSCC provides the basis for improved therapies and outcomes for patients. Newer types of immune treatments are now being studied, and they will impact how we treat cancer in the future. This article provides a brief overview of the current immunotherapeutic strategies for cancer with emphasis on HNSCC.
Collapse
Affiliation(s)
- Manisha Sahu
- Department of Oral Pathology and Microbiology, Chhattisgarh Dental College and Research Institute, Rajnandgaon, Chhattisgarh, India
| | - Hemakumari Suryawanshi
- Department of Oral Pathology and Microbiology, Chhattisgarh Dental College and Research Institute, Rajnandgaon, Chhattisgarh, India
| |
Collapse
|
160
|
Kłos P, Dabravolski SA. The Role of Mitochondria Dysfunction in Inflammatory Bowel Diseases and Colorectal Cancer. Int J Mol Sci 2021; 22:11673. [PMID: 34769108 PMCID: PMC8584106 DOI: 10.3390/ijms222111673] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/30/2022] Open
Abstract
Inflammatory bowel disease (IBD) is one of the leading gut chronic inflammation disorders, especially prevalent in Western countries. Recent research suggests that mitochondria play a crucial role in IBD development and progression to the more severe disease-colorectal cancer (CRC). In this review, we focus on the role of mitochondrial mutations and dysfunctions in IBD and CRC. In addition, main mitochondria-related molecular pathways involved in IBD to CRC transition are discussed. Additionally, recent publications dedicated to mitochondria-targeted therapeutic approaches to cure IBD and prevent CRC progression are discussed.
Collapse
Affiliation(s)
- Patrycja Kłos
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 72 Al. Powstańców Wlkp., 70-111 Szczecin, Poland;
| | - Siarhei A. Dabravolski
- Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
| |
Collapse
|
161
|
Sex-Based Differences in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:499-533. [PMID: 34664253 DOI: 10.1007/978-3-030-73119-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Cancers are heterogeneous multifactorial diseases consisting of a major public health issue worldwide. Sex disparities are evidenced in cancer incidence, mortality, expression of prognosis factor, response to treatment, and survival. For both sexes, an interplay of intrinsic and environmental factors influences cancer cells and tumor microenvironment (TME) components. The TME cumulates both supportive and communicative functions, contributing to cancer development, progression, and metastasis dissemination. The frontline topics of this chapter are focused on the contribution of sex, via steroid hormones, such as estrogens and androgens, on the following components of the TME: cancer-associated fibroblasts (CAFs), extracellular matrix (ECM), blood and lymphatic endothelial cells, and immunity/inflammatory system.
Collapse
|
162
|
Hong S, You JY, Paek K, Park J, Kang SJ, Han EH, Choi N, Chung S, Rhee WJ, Kim JA. Inhibition of tumor progression and M2 microglial polarization by extracellular vesicle-mediated microRNA-124 in a 3D microfluidic glioblastoma microenvironment. Am J Cancer Res 2021; 11:9687-9704. [PMID: 34646393 PMCID: PMC8490520 DOI: 10.7150/thno.60851] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/14/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Glioblastoma (GBM) is one of the most aggressive types of brain cancer. GBM progression is closely associated with microglia activation; therefore, understanding the regulation of the crosstalk between human GBM and microglia may help develop effective therapeutic strategies. Elucidation of efficient delivery of microRNA (miRNA) via extracellular vesicles (EVs) and their intracellular communications is required for therapeutic applications in GBM treatment. Methods: We used human GBM cells (U373MG) and human microglia. MiRNA-124 was loaded into HEK293T-derived EVs (miR-124 EVs). Various anti-tumor effects (proliferation, metastasis, chemosensitivity, M1/M2 microglial polarization, and cytokine profile) were investigated in U373MG and microglia. Anti-tumor effect of miR-124 EVs was also investigated in five different patient-derived GBM cell lines (SNU-201, SNU-466, SNU-489, SNU-626, and SNU-1105). A three-dimensional (3D) microfluidic device was used to investigate the interactive microenvironment of the tumor and microglia. Results: MiR-124 EVs showed highly efficient anti-tumor effects both in GBM cells and microglia. The mRNA expression levels of tumor progression and M2 microglial polarization markers were decreased in response to miR-124 EVs. The events were closely related to signal transducer and activator of transcription (STAT) 3 signaling in both GBM and microglia. In 3D microfluidic experiments, both U373MG and microglia migrated to a lesser extent and showed less-elongated morphology in the presence of miR-124 EVs compared to the control. Analyses of changes in cytokine levels in the microfluidic GBM-microglia environment showed that the treatment with miR-124 EVs led to tumor suppression and anti-cancer immunity, thereby recruiting natural killer (NK) cells into the tumor. Conclusions: In this study, we demonstrated that EV-mediated miR-124 delivery exerted synergistic anti-tumor effects by suppressing the growth of human GBM cells and inhibiting M2 microglial polarization. These findings provide new insights toward a better understanding of the GBM microenvironment and provide substantial evidence for the development of potential therapeutic strategies using miRNA-loaded EVs.
Collapse
|
163
|
Moro-Pérez L, Lozada-Chang SL, Rivas-García G, Álvarez C, Rojas-Pérez L, Boggiano-Ayo T, González-González Y. Purification and Conformational Characterization of a Novel Interleukin-2 Mutein. Protein J 2021; 40:917-928. [PMID: 34643845 DOI: 10.1007/s10930-021-10025-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 11/29/2022]
Abstract
Toxicity of high-dose IL-2-based therapies have motivated the development of the IL-2 mutein, which has low expansion properties for regulatory T lymphocytes. The development of two variants (A and B) for the IL-2 mutein purification as well as a conformational comparative study by Circular dichroism (CD) and fluorescence spectroscopy of these products were evaluated. For the first time, in our center, were used of DTT and 2% SDS in the solubilization step to decrease the aggregates on intermediate product, which favors that disulfide bridges are correctly formed during re-folding. A molecular weight of 18 kDa to the monomeric form and of 25-37 kDa to the oligomeric species were estimated by SDS-PAGE. IL-2 mutein showed similar far-UV CD spectral characteristic typical of cytokines with 41% of α-helix content. Batches obtained by Process B showed similar conformational features according near-UV CD and FS studies. However, those obtained by Process A differed in their folding. IL-2 mutein showed that conformational features by near-UV CD were affected by 2% SDS, no variations on secondary structure were observed. Melting temperature values by far-UV CD were higher than 95 °C, indicating a high thermal stability. Finally, the drug product obtained by Process B showed similar conformational characteristics by near-UV CD and FS, and higher biological activity values (7.0 × 103 ng/mL) in the cell proliferation assay with respect to Process A. Also, the recovery was 15% higher than in the Process A and exhibited a 78.48% of purity. Indeed, Process B was selected for the purification.
Collapse
Affiliation(s)
- Leina Moro-Pérez
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba.
| | - Sum Lai Lozada-Chang
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| | - Gabriela Rivas-García
- Center of Molecular Immunology, Quality Control Direction, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| | - Carlos Álvarez
- Biology Faculty, Center for Protein Studies, University of Havana, 25 St. No. 455, Vedado, 10400, Havana, Cuba
| | - Laritza Rojas-Pérez
- Biology Faculty, Center for Protein Studies, University of Havana, 25 St. No. 455, Vedado, 10400, Havana, Cuba
| | - Tammy Boggiano-Ayo
- Bioprocess R&D Department, Center of Molecular Immunology, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| | - Yamile González-González
- Center of Molecular Immunology, EPOVAC Facility, 216 Street and 15 Avenue, Atabey, Playa, P.O. Box 16040, 11600, Havana, Cuba
| |
Collapse
|
164
|
Bhattacharjee O, Ayyangar U, Kurbet AS, Lakshmanan V, Palakodeti D, Ginhoux F, Raghavan S. Epithelial-Macrophage Crosstalk Initiates Sterile Inflammation in Embryonic Skin. Front Immunol 2021; 12:718005. [PMID: 34721382 PMCID: PMC8553113 DOI: 10.3389/fimmu.2021.718005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/13/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages are highly responsive to the environmental cues and are the primary responders to tissue stress and damage. While much is known about the role of macrophages during inflammatory disease progression; the initial series of events that set up the inflammation remains less understood. In this study, we use next generation sequencing (NGS) of embryonic skin macrophages and the niche cells - skin epithelia and stroma in the epidermis specific knockout of integrin beta 1 (Itgβ1) model to uncover specific roles of each cell type and identify how these cell types communicate to initiate the sterile inflammatory response. We demonstrate that while the embryonic skin fibroblasts in the Itgβ1 knockout skin are relatively inactive, the keratinocytes and macrophages are the critical responders to the sterile inflammatory cues. The epidermis expresses damage associated molecular patterns (DAMPs), stress response genes, pro-inflammatory cytokines, and chemokines that aid in eliciting the inflammatory response. The macrophages, in-turn, respond by acquiring enhanced M2-like characteristics expressing ECM remodeling and matrisome signatures that exacerbate the basement membrane disruption. Depletion of macrophages by blocking the CSF1 receptor (CSF1R) results in improved basement membrane integrity and reduced ECM remodeling activity in the KO skin. Further, blocking the skin inflammation with celecoxib reveals that the acquired fate of macrophages in the KO skin is dependent on its interaction with the epidermal compartment through COX2 dependent cytokine production. Taken together, our study highlights a critical crosstalk between the epithelia and the dermal macrophages that shapes macrophage fate and initiates sterile inflammation in the skin. The insights gained from our study can be extrapolated to other inflammatory disorders to understand the early events that set up the disease.
Collapse
Affiliation(s)
- Oindrila Bhattacharjee
- Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
| | - Uttkarsh Ayyangar
- Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
| | - Ambika S. Kurbet
- Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
| | - Vairavan Lakshmanan
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Integrative Chemical Biology, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Dasaradhi Palakodeti
- Integrative Chemical Biology, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Srikala Raghavan
- Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
- Agency for Science, Technology and Research (A*STAR) Skin Research Lab (A*SRL), Singapore, Singapore
| |
Collapse
|
165
|
Cao X, Liang Y, Hu Z, Li H, Yang J, Hsu EJ, Zhu J, Zhou J, Fu YX. Next generation of tumor-activating type I IFN enhances anti-tumor immune responses to overcome therapy resistance. Nat Commun 2021; 12:5866. [PMID: 34620867 PMCID: PMC8497482 DOI: 10.1038/s41467-021-26112-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/14/2021] [Indexed: 01/01/2023] Open
Abstract
Type I interferon is promising in treating different kinds of tumors, but has been limited by its toxicity, lack of tumor targeting, and very short half-life. To target tumors, reduce systemic toxicity, and increase half-life, here we engineer a masked type I IFN-Fc (ProIFN) with its natural receptor connected by a cleavable linker that can be targeted by tumor-associated proteases. ProIFN has a prolonged serum half-life and shows an improved tumor-targeting effect. Interestingly, ProIFN-treated mice show enhanced DC cross-priming and significant increased CD8+ infiltration and effector function in the tumor microenvironment. ProIFN is able to improve checkpoint blockade efficacy in established tumors, as well as radiation efficacy for both primary and metastatic tumors. ProIFN exhibits superior long-term pharmacokinetics with minimal toxicity in monkeys. Therefore, this study demonstrates an effective tumor-activating IFN that can increase targeted immunity against primary tumor or metastasis and reduce periphery toxicity to the host.
Collapse
Affiliation(s)
- Xuezhi Cao
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yong Liang
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhenxiang Hu
- LivzonBio, Inc., Zhuhai, Guangdong, 519045, China
| | - Huiyu Li
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jiaming Yang
- LivzonBio, Inc., Zhuhai, Guangdong, 519045, China
| | - Eric J Hsu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jiankun Zhu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jin Zhou
- LivzonBio, Inc., Zhuhai, Guangdong, 519045, China
| | - Yang-Xin Fu
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
166
|
Seitz I, Shaukat A, Nurmi K, Ijäs H, Hirvonen J, Santos HA, Kostiainen MA, Linko V. Prospective Cancer Therapies Using Stimuli-Responsive DNA Nanostructures. Macromol Biosci 2021; 21:e2100272. [PMID: 34614301 DOI: 10.1002/mabi.202100272] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/28/2021] [Indexed: 11/08/2022]
Abstract
Nanostructures based on DNA self-assembly present an innovative way to address the increasing need for target-specific delivery of therapeutic molecules. Currently, most of the chemotherapeutics being used in clinical practice have undesired and exceedingly high off-target toxicity. This is a challenge in particular for small molecules, and hence, developing robust and effective methods to lower these side effects and enhance the antitumor activity is of paramount importance. Prospectively, these issues could be tackled with the help of DNA nanotechnology, which provides a route for the fabrication of custom, biocompatible, and multimodal structures, which can, to some extent, resist nuclease degradation and survive in the cellular environment. Similar to widely employed liposomal products, the DNA nanostructures (DNs) are loaded with selected drugs, and then by employing a specific stimulus, the payload can be released at its target region. This review explores several strategies and triggers to achieve targeted delivery of DNs. Notably, different modalities are explained through which DNs can interact with their respective targets as well as how structural changes triggered by external stimuli can be used to achieve the display or release of the cargo. Furthermore, the prospects and challenges of this technology are highlighted.
Collapse
Affiliation(s)
- Iris Seitz
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland
| | - Ahmed Shaukat
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland
| | - Kurt Nurmi
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Heini Ijäs
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland.,Nanoscience Center, Department of Biological and Environmental Science, University of Jyväskylä, P.O. Box 35, Jyväskylä, 40014, Finland
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland.,Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, University Medical Center Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Mauri A Kostiainen
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland.,HYBER Centre, Department of Applied Physics, Aalto University, P.O. Box 15100, Aalto, 00076, Finland
| | - Veikko Linko
- Biohybrid Materials, Department of Bioproducts and Biosystems, Aalto University, P.O. Box 16100, Aalto, 00076, Finland.,HYBER Centre, Department of Applied Physics, Aalto University, P.O. Box 15100, Aalto, 00076, Finland
| |
Collapse
|
167
|
Short Review on Advances in Hydrogel-Based Drug Delivery Strategies for Cancer Immunotherapy. Tissue Eng Regen Med 2021; 19:263-280. [PMID: 34596839 DOI: 10.1007/s13770-021-00369-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy has become the new paradigm of cancer treatment. The introduction and discovery of various therapeutic agents have also accelerated the application of immunotherapy in clinical trials. However, despite the significant potency and demonstrated advantages of cancer immunotherapy, its clinical application to patients faces several safety and efficacy issues, including autoimmune reactions, cytokine release syndrome, and vascular leak syndrome-related issues. In addressing these problems, biomaterials traditionally used for tissue engineering and drug delivery are attracting attention. Among them, hydrogels can be easily injected into tumors with drugs, and they can minimize side effects by retaining immune therapeutics at the tumor site for a long time. This article reviews the status of functional hydrogels for effective cancer immunotherapy. First, we describe the basic mechanisms of cancer immunotherapy and the advantages of using hydrogels to apply these mechanisms. Next, we summarize recent advances in the development of functional hydrogels designed to locally release various immunotherapeutic agents, including cytokines, cancer immune vaccines, immune checkpoint inhibitors, and chimeric antigen receptor-T cells. Finally, we briefly discuss the current problems and possible prospects of hydrogels for effective cancer immunotherapy.
Collapse
|
168
|
Shafique QUA, Rehman HM, Zaheer T, Tahir RA, Bhinder MA, Gul R, Saleem M. A Computational Approach to Modeling an Antagonistic Angiogenic VEGFR1-IL2 Fusion Protein for Cancer Therapy. Bioinform Biol Insights 2021; 15:11779322211043297. [PMID: 34566410 PMCID: PMC8458685 DOI: 10.1177/11779322211043297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022] Open
Abstract
In cancer treatment, immunotherapy has great potential for improving the prognosis of patients with hematologic and solid malignancies. In this study, various bioinformatics tools and servers were used to design an antiangiogenic fusion protein. After comprehensive evaluation, an antiangiogenic fusion protein was designed using a soluble extracellular domain of human vascular endothelial growth factor receptor 1 (sVEGFR-1) and human interleukin-2 (IL-2) joined by a flexible linker. The final construct was composed of 875 amino acids. The secondary structure of the fusion protein, obtained by CFSSP, PSIPRED, and SOPMA tools, consisted of 14.17% helices, 29.71% extended strands, 4.69% beta turns and 51.43% random coils. Tertiary structure prediction by Raptor X showed that the fusion protein comprises 3 domains with 875 modeled amino acids, out of which 26 positions (2%) were considered disordered. The Ramachandran plot revealed 89.3%, 7.1%, and 3.6% amino acid residues in favored, allowed, and outlier regions, respectively. Physical features of the Molecular Dynamic (MD) simulated system such as root mean square deviation, root mean square fluctuation, solvent-on hand surface region, and radius of gyration identified the fusion construct as a stable and compact protein with few fluctuations in its overall structure. Docking of the fusion protein showed that interaction between sVEGFR-1/VEGFA and IL-2/IL-2R still exists. In silico analysis revealed that the fusion protein comprising IL-2 and sVEGFR-1 has stable structure and the selected linker can efficiently separate the two domains. These observations may be helpful in determining protein stability prior to protein expression.
Collapse
Affiliation(s)
| | - Hafiz Muzzammel Rehman
- Institute of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan.,Department of Human Genetics and Molecular Biology, University of Health Sciences, Lahore, Pakistan
| | - Tahreem Zaheer
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rana Adnan Tahir
- Department of Biosciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Munir Ahmad Bhinder
- Department of Human Genetics and Molecular Biology, University of Health Sciences, Lahore, Pakistan
| | - Roquyya Gul
- Faculty of Life Sciences, Gulab Devi Educational Complex, Lahore, Pakistan
| | - Mahjabeen Saleem
- Institute of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
169
|
Di Trani CA, Fernandez-Sendin M, Cirella A, Segués A, Olivera I, Bolaños E, Melero I, Berraondo P. Advances in mRNA-based drug discovery in cancer immunotherapy. Expert Opin Drug Discov 2021; 17:41-53. [PMID: 34496689 DOI: 10.1080/17460441.2021.1978972] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors and adoptive T-cell therapy based on chimeric antigen receptors are the spearhead strategies to exploit the immune system to fight cancer. To take advantage of the full potential of the immune system, cancer immunotherapy must incorporate new biotechnologies such as mRNA technology that may synergize with already approved immunotherapies and act more effectively on immune targets. AREAS COVERED This review describes the basics of mRNA biotechnology and provides insight into the recent advances in the use of mRNA for the local and systemic delivery of immunostimulatory antibodies, proinflammatory cytokines or for optimizing adoptive T-cell therapy. EXPERT OPINION mRNA-based nanomedicines have great potential to expand the arsenal of immunotherapy tools due to their ability to simplify and accelerate drug development and their suitability for transient and local expression of immunostimulatory molecules, whose systemic and sustained expression would be toxic. The success of mRNA-based COVID-19 vaccines has highlighted the feasibility of this approach. Continuous advances in the delivery and construction of RNA-based vectors hold promise for improvements in clinical efficacy.
Collapse
Affiliation(s)
- Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain
| | - Myriam Fernandez-Sendin
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain
| | - Aina Segués
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.,Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh UK
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain.,Centro De Investigación Biomédica En Red De Cáncer (Ciberonc), Spain.,Departments of Oncology and Immunology, Clínica Universidad De Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad De Navarra, Pamplona, Spain.,Navarra Institute for Health Research (Idisna), Pamplona, Spain.,Centro De Investigación Biomédica En Red De Cáncer (Ciberonc), Spain
| |
Collapse
|
170
|
Dakhel S, Lizak C, Matasci M, Mock J, Villa A, Neri D, Cazzamalli S. An Attenuated Targeted-TNF Localizes to Tumors In Vivo and Regains Activity at the Site of Disease. Int J Mol Sci 2021; 22:10020. [PMID: 34576184 PMCID: PMC8469155 DOI: 10.3390/ijms221810020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 11/16/2022] Open
Abstract
Antibody-cytokine fusion proteins (immunocytokines) are gaining importance for cancer therapy, but those products are often limited by systemic toxicity related to the activity of the cytokine payload in circulation and in secondary lymphoid organs. Tumor necrosis factor (TNF) is used as a pro-inflammatory payload to trigger haemorrhagic necrosis and boost anti-cancer immunity at the tumor site. Here we describe a depotentiated version of TNF (carrying the single point mutation I97A), which displayed reduced binding affinity to its cognate receptor tumor necrosis factor receptor 1 (TNFR-1) and lower biocidal activity. The fusion of the TNF(I97A) mutant to the L19 antibody promoted restoration of anti-tumor activity upon accumulation on the cognate antigen, the alternatively spliced EDB domain of fibronectin. In vivo administration of high doses (375 μg/Kg) of the fusion protein showed a potent anti-tumor effect without apparent toxicity compared with the wild type protein. L19-TNFI97A holds promise for the targeted delivery of TNF activity to neoplastic lesions, helping spare normal tissues.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/metabolism
- Cricetulus
- Cytokines/genetics
- Cytokines/metabolism
- Female
- Fibronectins/genetics
- Fibronectins/metabolism
- Fluorescent Antibody Technique
- Immunotherapy
- Mice, Inbred BALB C
- Mutation
- Protein Structure, Secondary
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Mice
Collapse
Affiliation(s)
- Sheila Dakhel
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Christian Lizak
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Mattia Matasci
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Jacqueline Mock
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Alessandra Villa
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| | - Dario Neri
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
- Philogen S.p.A., Piazza La Lizza, 7, 53100 Siena, Italy
| | - Samuele Cazzamalli
- Philochem AG, CH-8112 Otelfingen, Switzerland; (S.D.); (C.L.); (M.M.); (J.M.); (A.V.); (D.N.)
| |
Collapse
|
171
|
Hotz C, Wagenaar TR, Gieseke F, Bangari DS, Callahan M, Cao H, Diekmann J, Diken M, Grunwitz C, Hebert A, Hsu K, Bernardo M, Karikó K, Kreiter S, Kuhn AN, Levit M, Malkova N, Masciari S, Pollard J, Qu H, Ryan S, Selmi A, Schlereth J, Singh K, Sun F, Tillmann B, Tolstykh T, Weber W, Wicke L, Witzel S, Yu Q, Zhang YA, Zheng G, Lager J, Nabel GJ, Sahin U, Wiederschain D. Local delivery of mRNA-encoded cytokines promotes antitumor immunity and tumor eradication across multiple preclinical tumor models. Sci Transl Med 2021; 13:eabc7804. [PMID: 34516826 DOI: 10.1126/scitranslmed.abc7804] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
| | | | | | | | | | - Hui Cao
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | | | - Mustafa Diken
- BioNTech, 55131 Mainz, Germany.,Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH (TRON), 55131 Mainz, Germany
| | | | - Andy Hebert
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Karl Hsu
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Marie Bernardo
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | | | - Sebastian Kreiter
- BioNTech, 55131 Mainz, Germany.,Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH (TRON), 55131 Mainz, Germany
| | | | - Mikhail Levit
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | | | | | - Jack Pollard
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Hui Qu
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Sue Ryan
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Abderaouf Selmi
- Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH (TRON), 55131 Mainz, Germany
| | | | - Kuldeep Singh
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Fangxian Sun
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Bodo Tillmann
- Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH (TRON), 55131 Mainz, Germany
| | | | - William Weber
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | | | - Sonja Witzel
- Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH (TRON), 55131 Mainz, Germany
| | - Qunyan Yu
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Yu-An Zhang
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Gang Zheng
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Joanne Lager
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Gary J Nabel
- Research and Development, Sanofi, Cambridge, MA 02139, USA
| | - Ugur Sahin
- BioNTech, 55131 Mainz, Germany.,Translational Oncology at the University Medical Center of Johannes Gutenberg University GmbH (TRON), 55131 Mainz, Germany
| | | |
Collapse
|
172
|
The evolution of commercial drug delivery technologies. Nat Biomed Eng 2021; 5:951-967. [PMID: 33795852 DOI: 10.1038/s41551-021-00698-w] [Citation(s) in RCA: 658] [Impact Index Per Article: 164.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Drug delivery technologies have enabled the development of many pharmaceutical products that improve patient health by enhancing the delivery of a therapeutic to its target site, minimizing off-target accumulation and facilitating patient compliance. As therapeutic modalities expanded beyond small molecules to include nucleic acids, peptides, proteins and antibodies, drug delivery technologies were adapted to address the challenges that emerged. In this Review Article, we discuss seminal approaches that led to the development of successful therapeutic products involving small molecules and macromolecules, identify three drug delivery paradigms that form the basis of contemporary drug delivery and discuss how they have aided the initial clinical successes of each class of therapeutic. We also outline how the paradigms will contribute to the delivery of live-cell therapies.
Collapse
|
173
|
Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The Role of Curcumin in Cancer Treatment. Biomedicines 2021; 9:1086. [PMID: 34572272 PMCID: PMC8464730 DOI: 10.3390/biomedicines9091086] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Curcumin is a polyphenol extracted from the rhizomes of the turmeric plant, Curcuma longa which has anti-inflammatory, and anticancer properties. Chronic inflammation is associated with the development of cancer. Curcumin acts on the regulation of various immune modulators, including cytokines, cyclooxygenase-2 (COX-2), and reactive oxygen species (ROS), which partly explains its anticancer effects. It also takes part in the downregulation of growth factors, protein kinases, oncogenic molecules and various signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 3 (STAT3) signaling. Clinical trials of curcumin have been completed or are ongoing for various types of cancer. This review presents the molecular mechanisms of curcumin in different types of cancer and the evidence from the most recent clinical trials.
Collapse
Affiliation(s)
- Vasiliki Zoi
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Vasiliki Galani
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Georgios D. Lianos
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Spyridon Voulgaris
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
| | - Athanasios P. Kyritsis
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
| | - George A. Alexiou
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
| |
Collapse
|
174
|
Kim HY, Han D, Kim J, Yoo MS, Lee JS, Kim H, Kim HM, Jeong HJ. An anti-cancer effect of Sambou bamboo salt TM in melanoma skin cancer both in vivo and in vitro models. J Food Biochem 2021; 45:e13903. [PMID: 34387368 DOI: 10.1111/jfbc.13903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 01/10/2023]
Abstract
Bamboo salt has anti-allergic, anti-inflammatory, anti-oxidant, diabetics, anti-aging, and immune-enhancing effects, which are closely related to anti-cancer effect. The aim of this study was to investigate the anti-cancer effects of Sambou bamboo saltTM (SBS) in melanoma skin cancer in vivo and in vitro models. SBS-administered mice effectively reduced tumor growth and increased survival rate compared with B16F10 cell-inoculated mice without tissue damage, hepatotoxicity, and nephrotoxicity. SBS enhanced levels of immune-enhancing mediators, such as interferon-γ, interleukin (IL)-2, IL-6, IL-12, tumor necrosis factor-α, and IgE in serum and melanoma tissues. Furthermore, SBS enhanced activities of caspases and levels of Bax and p53, whereas decreased levels of Bcl-2. This reduction was a consequence of apoptosis signaling pathway. In conclusion, these results suggest that SBS is a potential substance for cancer therapy. SBS has the potential to be developed either as Korean traditional medicine or as a health functional food for cancer therapy. PRACTICAL APPLICATIONS: In these days cancer is one of the world's largest health problems. Bamboo salt is used as a Korean traditional food or medicine and has beneficial effect on inflammation. We have identified Sambou bamboo saltTM (SBS) is a potential substance for cancer therapy. These insights suggest that SBS can potentially be utilized for health functional foods for cancer treatment as well as improve various cancer diseases such as melanoma skin cancer.
Collapse
Affiliation(s)
- Hee-Yun Kim
- Biochip Research Center, Hoseo University, Asan, Republic of Korea
| | - Dongkyun Han
- Department of Food Science & Technology, Hoseo University, Asan, Republic of Korea
| | - Jungeun Kim
- Department of Food Science & Technology, Hoseo University, Asan, Republic of Korea
| | - Min-Sun Yoo
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jin-Soo Lee
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyunyong Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hyung-Min Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.,Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun-Ja Jeong
- Biochip Research Center, Hoseo University, Asan, Republic of Korea.,Department of Food Science & Technology, Hoseo University, Asan, Republic of Korea
| |
Collapse
|
175
|
Xu Y, Campos Carrascosa L, Yeung YA, Chu MLH, Yang W, Djuretic I, Pappas DC, Zeytounian J, Ge Z, de Ruiter V, Starbeck-Miller GR, Patterson J, Rigas D, Chen SH, Kraynov E, Boor PP, Noordam L, Doukas M, Tsao D, Ijzermans JN, Guo J, Grünhagen DJ, Erdmann J, Verheij J, van Royen ME, Doornebosch PG, Feldman R, Park T, Mahmoudi S, Dorywalska M, Ni I, Chin SM, Mistry T, Mosyak L, Lin L, Ching KA, Lindquist KC, Ji C, Londono LM, Kuang B, Rickert R, Kwekkeboom J, Sprengers D, Huang TH, Chaparro-Riggers J. An Engineered IL15 Cytokine Mutein Fused to an Anti-PD-1 Improves Intratumoral T-Cell Function and Antitumor Immunity. Cancer Immunol Res 2021; 9:1141-1157. [PMID: 34376502 DOI: 10.1158/2326-6066.cir-21-0058] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/04/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022]
Abstract
The use of cytokines for immunotherapy shows clinical efficacy but is frequently accompanied by severe adverse events caused by excessive and systemic immune activation. Here, we set out to address these challenges by engineering a fusion protein of a single, potency-reduced, IL15 mutein and a PD-1-specific antibody (anti-PD1-IL15m). This immunocytokine was designed to deliver PD-1-mediated, avidity-driven IL2/15 receptor stimulation to PD-1+ tumor-infiltrating lymphocytes (TILs) while minimally affecting circulating peripheral natural killer (NK) cells and T cells. Treatment of tumor-bearing mice with a mouse cross-reactive fusion, anti-mPD1-IL15m demonstrated potent antitumor efficacy without exacerbating body weight loss in B16 and MC38 syngeneic tumor models. Moreover, anti-mPD1-IL15m was more efficacious than an IL15 superagonist, an anti-mPD-1, or the combination thereof in the B16 melanoma model. Mechanistically, anti-PD1-IL15m preferentially targeted CD8+ TILs and scRNA-seq analyses revealed that anti-mPD1-IL15m treatment induced the expansion of an exhausted CD8+ TILs cluster with high proliferative capacity and effector-like signatures. Antitumor efficacy of anti-mPD1-IL15m was dependent on CD8+ T cells, as depletion of CD8+ cells resulted in the loss of antitumor activity, whereas depletion of NK cells had little impact on efficacy. The impact of anti-hPD1-IL15m on primary human TILs from cancer patients was also evaluated. Anti-hPD1-IL15m robustly enhanced the proliferation, activation, and cytotoxicity of CD8+ and CD4+ TILs from human primary cancers in vitro, whereas tumor-derived regulatory T cells were largely unaffected. Taken together, we showed that anti-PD1-IL15m exhibits a high translational promise with improved efficacy and safety of IL15 for cancer immunotherapy via targeting PD-1+ TILs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Irene Ni
- Oncology Research Unit, Pfizer (United States)
| | | | | | | | | | - Keith A Ching
- Computational Biology/Oncology Research Unit, Pfizer Global R & D
| | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Yu Y, Wu S, Zhang J, Li J, Yao C, Wu W, Wang Y, Ji H, Wei W, Gao M, Li Y, Yao S, Huang Y, Bi Q, Qu H, Guo DA. Structurally diverse diterpenoid alkaloids from the lateral roots of Aconitum carmichaelii Debx. and their anti-tumor activities based on in vitro systematic evaluation and network pharmacology analysis. RSC Adv 2021; 11:26594-26606. [PMID: 35480028 PMCID: PMC9037614 DOI: 10.1039/d1ra04223h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022] Open
Abstract
Thirty-seven diterpenoid alkaloids (DAs) with diverse structures were isolated and identified from the lateral roots of Aconitum carmichaelii Debx., comprising eight C20-DAs and twenty-nine C19-DAs. Besides the 31 known DAs identified by comparing the 1H NMR and 13C NMR data with those reported in the literature, the structures of four new compounds (1, 14, 17, and 25), and two other compounds (26 and 37) which were reported to be synthesized previously, were also elucidated based on the comprehensive analysis of their HR-ESI-MS, 1D and 2D NMR spectra, including 1H-1H COSY, HSQC and HMBC and NOESY/ROESY. Among them, compound 1 represents the first example of a C20-DA glucoside. Besides, the anti-tumor activities of all the isolated compounds against human non-small-cell lung cancer A549 and H460 cells were systematically evaluated by MTT methods. The results revealed that all of the C19-DAs possessed moderate activities against both of the two cell lines with IC50 values ranging from 7.97 to 28.42 μM, and their structure-activity relationships indicated the active sites of C-8, C-10, and C-14 positions and the nitrogen atom in the C19-DA skeleton. In addition, all of the isolated DAs, with chemical structures confirmed, were further applied for network pharmacology analysis, in order to give an insight into the possible mechanisms of their anti-tumor activities. As a result, 173 potential targets and three most important pathways related to non-small-cell lung carcinoma were finally unearthed.
Collapse
Affiliation(s)
- Yang Yu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Shifei Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Jianqing Zhang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Jiayuan Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Changliang Yao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Wenyong Wu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Yingying Wang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Hongjian Ji
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Wenlong Wei
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Min Gao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Yun Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University Nanjing 210009 China
| | - Shuai Yao
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Yong Huang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Qirui Bi
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Hua Qu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine Nanjing 210023 China
| | - De-An Guo
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
177
|
Liang JL, Luo GF, Chen WH, Zhang XZ. Recent Advances in Engineered Materials for Immunotherapy-Involved Combination Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007630. [PMID: 34050564 DOI: 10.1002/adma.202007630] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/18/2020] [Indexed: 06/12/2023]
Abstract
Immunotherapy that can activate immunity or enhance the immunogenicity of tumors has emerged as one of the most effective methods for cancer therapy. Nevertheless, single-mode immunotherapy is still confronted with several critical challenges, such as the low immune response, the low tumor infiltration, and the complex immunosuppression tumor microenvironment. Recently, the combination of immunotherapy with other therapeutic modalities has emerged as a powerful strategy to augment the therapeutic outcome in fighting against cancer. In this review, recent research advances of the combination of immunotherapy with chemotherapy, phototherapy, radiotherapy, sonodynamic therapy, metabolic therapy, and microwave thermotherapy are summarized. Critical challenges and future research direction of immunotherapy-based cancer therapeutic strategy are also discussed.
Collapse
Affiliation(s)
- Jun-Long Liang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Guo-Feng Luo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
178
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
179
|
Yang Z, Xu Y, Bi Y, Zhang N, Wang H, Xing T, Bai S, Shen Z, Naz F, Zhang Z, Yin L, Shi M, Wang L, Wang L, Wang S, Xu L, Su X, Wu S, Yu C. Immune escape mechanisms and immunotherapy of urothelial bladder cancer. J Clin Transl Res 2021; 7:485-500. [PMID: 34541363 PMCID: PMC8445627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 05/12/2021] [Accepted: 06/25/2021] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND AND AIM Urothelial bladder cancer (UBC) is a common malignant tumor of the urogenital system with a high rate of recurrence. Due to the sophisticated and largely unexplored mechanisms of tumorigenesis of UBC, the classical therapeutic approaches including transurethral resection and radical cystectomy combined with chemotherapy have remained unchanged for decades. However, with increasingly in-depth understanding of the microenvironment and the composition of tumor-infiltrating lymphocytes of UBC, novel immunotherapeutic strategies have been developed. Bacillus Calmette-Guerin (BCG) therapy, immune checkpoint blockades, adoptive T cell immunotherapy, dendritic cell (DC) vaccines, etc., have all been intensively investigated as immunotherapies for UBC. This review will discuss the recent progress in immune escape mechanisms and immunotherapy of UBC. METHODS Based on a comprehensive search of the PubMed and ClinicalTrials.gov database, this review included the literature reporting the immune escape mechanisms of UBC and clinical trials assessing the effect of immunotherapeutic strategies on tumor or immune cells in UBC patients published in English between 1999 and 2020. RESULTS Immune surveillance, immune balance, and immune escape are the three major processes that occur during UBC tumorigenesis. First, the role of immunosuppressive cells, immunosuppressive molecules, immunosuppressive signaling molecules, and DCs in tumor microenvironment is introduced elaborately in the immune escape mechanisms of UBC section. In addition, recent progress of immunotherapies including BCG, checkpoint inhibitors, cytokines, adoptive T cell immunotherapy, DCs, and macrophages on UBC patients are summarized in detail. Finally, the need to explore the mechanisms, molecular characteristics and immune landscape during UBC tumorigenesis and development of novel and robust immunotherapies for UBC are also proposed and discussed. CONCLUSION At present, BCG and immune checkpoint blockades have been approved by the US Food and Drug Administration for the treatment of UBC patients and have achieved encouraging therapeutic results, expanding the traditional chemotherapy and surgery-based treatment for UBC. RELEVANCE FOR PATIENTS Immunotherapy has achieved desirable results in the treatment of UBC, which not only improve the overall survival but also reduce the recurrence rate and the occurrence of treatment-related adverse events of UBC patients. In addition, the indicators to predict the effectiveness and novel therapy strategies, such as combination regimen of checkpoint inhibitor with checkpoint inhibitor or chemotherapy, should be further studied.
Collapse
Affiliation(s)
- Zhao Yang
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,2Department of Bioscience, College of Life Science, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar 843300, Xinjiang, China,
Corresponding authors: Zhao Yang College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.College of Life Science, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar 843300, Xinjiang, China. E-mail:
| | - Yinyan Xu
- 3Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China
| | - Ying Bi
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Nan Zhang
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Haifeng Wang
- 4Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming 650101, China
| | - Tianying Xing
- 5Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Suhang Bai
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zongyi Shen
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Faiza Naz
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zichen Zhang
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Liqi Yin
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mengran Shi
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Luyao Wang
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lei Wang
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shihui Wang
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lida Xu
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xin Su
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Song Wu
- 3Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China,
Song Wu Department of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, Shenzhen 518000, China.
| | - Changyuan Yu
- 1Department of Biomedical Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,
Changyuan Yu College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
180
|
Tang L, Zhang R, Zhang X, Yang L. Personalized Neoantigen-Pulsed DC Vaccines: Advances in Clinical Applications. Front Oncol 2021; 11:701777. [PMID: 34381724 PMCID: PMC8350509 DOI: 10.3389/fonc.2021.701777] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
In the past few decades, great progress has been made in the clinical application of dendritic cell (DC) vaccines loaded with personalized neoantigens. Personalized neoantigens are antigens arising from somatic mutations in cancers, with specificity to each patient. DC vaccines work based on the fundamental characteristics of DCs, which are professional antigen-presenting cells (APCs), responsible for the uptake, processing, and presentation of antigens to T cells to activate immune responses. Neoantigens can exert their antitumor effects only after they are taken up by APCs and presented to T cells. In recent years, neoantigen-based personalized tumor therapeutic vaccines have proven to be safe, immunogenic and feasible treatment strategies in patients with melanoma and glioblastoma that provide new hope in the treatment of cancer patients and a new approach to cure cancer. In addition, according to ClinicalTrials.gov, hundreds of registered DC vaccine trials are either completed or ongoing worldwide, of which 9 are in early phase I, 191 in phase I, 166 in phase II and 8 in phase III. Hundreds of clinical studies on therapeutic tumor vaccines globally have proven that DC vaccines are stable, reliable and very safe. However, in this process, many other factors still limit the effectiveness of the vaccine. This review will focus on the current research progress on personalized neoantigen-pulsed DC vaccines, their limitations and future research directions of DC vaccines loaded with neoantigens. This review aims to provide a better understanding of DCs biology and manipulation of activated DCs for DCs researchers to produce the next generation of highly efficient cancer vaccines for patients.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Rui Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Xiaoyu Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
181
|
Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther 2021; 6:263. [PMID: 34248142 PMCID: PMC8273155 DOI: 10.1038/s41392-021-00658-5] [Citation(s) in RCA: 1291] [Impact Index Per Article: 322.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 05/11/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer development and its response to therapy are regulated by inflammation, which either promotes or suppresses tumor progression, potentially displaying opposing effects on therapeutic outcomes. Chronic inflammation facilitates tumor progression and treatment resistance, whereas induction of acute inflammatory reactions often stimulates the maturation of dendritic cells (DCs) and antigen presentation, leading to anti-tumor immune responses. In addition, multiple signaling pathways, such as nuclear factor kappa B (NF-kB), Janus kinase/signal transducers and activators of transcription (JAK-STAT), toll-like receptor (TLR) pathways, cGAS/STING, and mitogen-activated protein kinase (MAPK); inflammatory factors, including cytokines (e.g., interleukin (IL), interferon (IFN), and tumor necrosis factor (TNF)-α), chemokines (e.g., C-C motif chemokine ligands (CCLs) and C-X-C motif chemokine ligands (CXCLs)), growth factors (e.g., vascular endothelial growth factor (VEGF), transforming growth factor (TGF)-β), and inflammasome; as well as inflammatory metabolites including prostaglandins, leukotrienes, thromboxane, and specialized proresolving mediators (SPM), have been identified as pivotal regulators of the initiation and resolution of inflammation. Nowadays, local irradiation, recombinant cytokines, neutralizing antibodies, small-molecule inhibitors, DC vaccines, oncolytic viruses, TLR agonists, and SPM have been developed to specifically modulate inflammation in cancer therapy, with some of these factors already undergoing clinical trials. Herein, we discuss the initiation and resolution of inflammation, the crosstalk between tumor development and inflammatory processes. We also highlight potential targets for harnessing inflammation in the treatment of cancer.
Collapse
|
182
|
Botticelli A, Zizzari IG, Scagnoli S, Pomati G, Strigari L, Cirillo A, Cerbelli B, Di Filippo A, Napoletano C, Scirocchi F, Rughetti A, Nuti M, Mezi S, Marchetti P. The Role of Soluble LAG3 and Soluble Immune Checkpoints Profile in Advanced Head and Neck Cancer: A Pilot Study. J Pers Med 2021; 11:651. [PMID: 34357118 PMCID: PMC8304359 DOI: 10.3390/jpm11070651] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Unresectable recurrent and/or metastatic head and neck squamous cell carcinoma (R/M HNSCC) has a very poor prognosis. Soluble immune checkpoints (sICs) are circulating proteins that result from the alternative splicing of membrane proteins and can modulate the immune response to cancer cells. The aim of our pilot study was to determine the possible role of a comprehensive evaluation of sICs in the classification of prognosis and response to treatment in patients with advanced disease. We evaluated several sICs (CD137, CTLA-4, PD-1, PD-L1, PD-L2, TIM3, LAG3, GITR, HVEM, BTLA, IDO, CD80, CD27, and CD28) from peripheral blood at baseline and investigated the association with clinical characteristics and outcomes. A high baseline soluble LAG3 (sLAG3 > 377 pg/mL) resulted in an association with poor PFS and OS (p = 0.047 and p = 0.003, respectively). Moreover, sLAG3 emerged as an independent prognostic factor using an MVA (p = 0.005). The evaluation of sICs, in particular sLAG3, may be relevant for identifying patients with worse prognoses, or resistance to treatments, and may lead to the development of novel targeted strategies.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Oncology, University of Rome “Sapienza”, 00185 Rome, Italy; (A.B.); (P.M.)
| | - Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (C.N.); (F.S.); (A.R.); (M.N.)
| | - Simone Scagnoli
- Department of Medical and Surgical Sciences and Translational Medicine, University of Rome “Sapienza”, 00185 Rome, Italy
| | - Giulia Pomati
- Department of Molecular Medicine, University of Rome “Sapienza”, 00161 Rome, Italy;
| | - Lidia Strigari
- Medical Physics Unit, “S. Orsola-Malpighi” Hospital, 40138 Bologna, Italy;
| | - Alessio Cirillo
- Department of Radiological, Oncological and Anatomo-Pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (S.M.)
| | - Bruna Cerbelli
- Department of Radiological, Oncological and Anatomo-Pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (S.M.)
| | - Alessandra Di Filippo
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (C.N.); (F.S.); (A.R.); (M.N.)
| | - Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (C.N.); (F.S.); (A.R.); (M.N.)
| | - Fabio Scirocchi
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (C.N.); (F.S.); (A.R.); (M.N.)
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (C.N.); (F.S.); (A.R.); (M.N.)
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (C.N.); (F.S.); (A.R.); (M.N.)
| | - Silvia Mezi
- Department of Radiological, Oncological and Anatomo-Pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (S.M.)
| | - Paolo Marchetti
- Department of Clinical and Molecular Oncology, University of Rome “Sapienza”, 00185 Rome, Italy; (A.B.); (P.M.)
| |
Collapse
|
183
|
Mansurov A, Lauterbach A, Budina E, Alpar AT, Hubbell JA, Ishihara J. Immunoengineering approaches for cytokine therapy. Am J Physiol Cell Physiol 2021; 321:C369-C383. [PMID: 34232748 DOI: 10.1152/ajpcell.00515.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Since the discovery of cytokines, much effort has been put forth to achieve therapeutic translation for treatment of various diseases, including cancer and autoimmune diseases. Despite these efforts, very few cytokines have cleared regulatory approval, and those that were approved are not commonly used due to their challenging toxicity profile and/or limited therapeutic efficacy. The main limitation in translation has been that wild-type cytokines have unfavorable pharmacokinetic and pharmacodynamic profiles, either eliciting unwanted systemic side effects or insufficient residence in secondary lymphoid organs. In this review, we address protein-engineering approaches that have been applied to both proinflammatory and anti-inflammatory cytokines to enhance their therapeutic indices, and we highlight diseases in which administration of engineered cytokines is especially relevant.
Collapse
Affiliation(s)
- Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Abigail Lauterbach
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Erica Budina
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Aaron T Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Jun Ishihara
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois.,Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
184
|
Combinatorial therapy in tumor microenvironment: Where do we stand? Biochim Biophys Acta Rev Cancer 2021; 1876:188585. [PMID: 34224836 DOI: 10.1016/j.bbcan.2021.188585] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/28/2021] [Accepted: 06/23/2021] [Indexed: 01/09/2023]
Abstract
The tumor microenvironment plays a pivotal role in tumor initiation and progression by creating a dynamic interaction with cancer cells. The tumor microenvironment consists of various cellular components, including endothelial cells, fibroblasts, pericytes, adipocytes, immune cells, cancer stem cells and vasculature, which provide a sustained environment for cancer cell proliferation. Currently, targeting tumor microenvironment is increasingly being explored as a novel approach to improve cancer therapeutics, as it influences the growth and expansion of malignant cells in various ways. Despite continuous advancements in targeted therapies for cancer treatment, drug resistance, toxicity and immune escape mechanisms are the basis of treatment failure and cancer escape. Targeting tumor microenvironment efficiently with approved drugs and combination therapy is the solution to this enduring challenge that involves combining more than one treatment modality such as chemotherapy, surgery, radiotherapy, immunotherapy and nanotherapy that can effectively and synergistically target the critical pathways associated with disease pathogenesis. This review shed light on the composition of the tumor microenvironment, interaction of different components within tumor microenvironment with tumor cells and associated hallmarks, the current status of combinatorial therapies being developed, and various growing advancements. Furthermore, computational tools can also be used to monitor the significance and outcome of therapies being developed. We addressed the perceived barriers and regulatory hurdles in developing a combinatorial regimen and evaluated the present status of these therapies in the clinic. The accumulating depth of knowledge about the tumor microenvironment in cancer may facilitate further development of effective treatment modalities. This review presents the tumor microenvironment as a sweeping landscape for developing novel cancer therapies.
Collapse
|
185
|
Hu W, Xiao T, Li D, Fan Y, Xing L, Wang X, Li Y, Shi X, Shen M. Intelligent Molybdenum Disulfide Complexes as a Platform for Cooperative Imaging-Guided Tri-Mode Chemo-Photothermo-Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100165. [PMID: 34145978 PMCID: PMC8292874 DOI: 10.1002/advs.202100165] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/27/2021] [Indexed: 05/27/2023]
Abstract
Design of new nanoplatforms that integrates multiple imaging and therapeutic components for precision cancer nanomedicine remains to be challenging. Here, a facile strategy is reported to prepare polydopamine (PDA)-coated molybdenum disulfide (MoS2 ) nanoflakes as a nanocarrier to load dual drug cisplatin (Pt) and 1-methyl-tryptophan (1-MT) for precision tumor theranostics. Preformed MoS2 nanoflakes are coated with PDA, modified with methoxy-polyethylene glycol (PEG)-amine, and loaded with 1-MT and Pt. The formed functional 1-MT-Pt-PPDA@MoS2 (the second P stands for PEG) complexes exhibit good colloidal stability and photothermal conversion efficiency (47.9%), dual pH-, and photothermal-sensitive drug release profile, and multimodal thermal, computed tomography and photoacoustic imaging capability. Due to the respective components of Pt, MoS2 , and 1-MT that can block the immune checkpoint associated to tumoral indoleamine 2,3-dioxygenase-induced tryptophan metabolism, tri-mode chemo-photothermo-immunotherapy of tumors can be realized. In particular, under the near infrared laser irradiation, fast release of both drugs can be facilitated to achieve cooperative tumor therapy effect, and the combined immunogenic cell death induced by the dual-mode chemo-photothermo treatment and the 1-MT-induced immune checkpoint blockade can boost enhanced antitumor immune response to generate significant cytotoxic T cells for tumor killing. The developed 1-MT-Pt-PPDA@MoS2 complexes may be used as an intelligent nanoplatform for cooperative precision imaging-guided combinational tumor therapy.
Collapse
Affiliation(s)
- Wei Hu
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
- Department of Gynecology and ObstetricsXinHua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Tingting Xiao
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Du Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Yu Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| | - Lingxi Xing
- Department of Gynecology and ObstetricsXinHua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Xipeng Wang
- Department of Gynecology and ObstetricsXinHua Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghai200092P. R. China
| | - Yulin Li
- The Key Laboratory for Ultrafine Materials of Ministry of EducationState Key Laboratory of Bioreactor EngineeringEngineering Research Center for Biomedical Materials of Ministry of EducationSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237P. R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
- CQM‐Centro de Quimica da MadeiraUniversidade da MadeiraFunchal9020‐105Portugal
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Chemistry, Chemical Engineering and BiotechnologyDonghua UniversityShanghai201620P. R. China
| |
Collapse
|
186
|
Ghods A, Mehdipour F, Shariat M, Talei AR, Ghaderi A. Regulatory T cells express Tumor Necrosis Factor Receptor 2 with the highest intensity among CD4 + T cells in the draining lymph nodes of breast cancer. Mol Immunol 2021; 137:52-56. [PMID: 34214829 DOI: 10.1016/j.molimm.2021.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/27/2021] [Accepted: 06/20/2021] [Indexed: 12/16/2022]
Abstract
Tumor Necrosis Factor Receptor 2 (TNFR2) is one of the receptors of TNF-α, which is expressed on various cell types. TNFR2 signaling has a balancing role between regulatory and effector functions of T cells. Herein, we investigated the expression of TNFR2 on regulatory T cells (Tregs) and non-Tregs in breast tumor-draining lymph nodes. Mononuclear cells were isolated from 16 axillary lymph nodes, and the expressions of TNFR2, Foxp3 and CD25 were assessed in CD4+ T cells by flow cytometry. Our results showed that the majority of TNFR2+CD4+ T cells were Foxp3-CD25-. However, the percentage of TNFR2+ cells was significantly higher in Foxp3+CD25+CD4+ Tregs compared to Foxp3-CD25-CD4+, Foxp3+CD25-CD4+, and Foxp3-CD25+CD4+ T cell subsets. Among these subsets, Foxp3+CD25+TNFR2+CD4+ T cells were found to have the highest intensity of TNFR2 expression. The intensity of Foxp3 expression in Foxp3+CD25+TNFR2+CD4+ Treg cells was significantly higher than in their TNFR2- counterpart. Collectively, we showed that most of TNFR2+CD4+ T lymphocytes were Foxp3-CD25-, while the majority of Foxp3+CD25+CD4+ Tregs were TNFR2+, and they expressed TNFR2 with the highest intensity. This report highlights the importance of TNFR2 expression on Tregs and paves the way for further investigation of the effects of TNF-α on the suppressive activity of Tregs in the tumor microenvironment.
Collapse
Affiliation(s)
- Atri Ghods
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Mehdipour
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahmoud Shariat
- Department of Pathology, Shiraz Central Hospital, Shiraz, Iran
| | - Abdol-Rasoul Talei
- Breast Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
187
|
Yoon SJ, Lee CB, Chae SU, Jo SJ, Bae SK. The Comprehensive "Omics" Approach from Metabolomics to Advanced Omics for Development of Immune Checkpoint Inhibitors: Potential Strategies for Next Generation of Cancer Immunotherapy. Int J Mol Sci 2021; 22:6932. [PMID: 34203237 PMCID: PMC8268114 DOI: 10.3390/ijms22136932] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/11/2022] Open
Abstract
In the past decade, immunotherapies have been emerging as an effective way to treat cancer. Among several categories of immunotherapies, immune checkpoint inhibitors (ICIs) are the most well-known and widely used options for cancer treatment. Although several studies continue, this treatment option has yet to be developed into a precise application in the clinical setting. Recently, omics as a high-throughput technique for understanding the genome, transcriptome, proteome, and metabolome has revolutionized medical research and led to integrative interpretation to advance our understanding of biological systems. Advanced omics techniques, such as multi-omics, single-cell omics, and typical omics approaches, have been adopted to investigate various cancer immunotherapies. In this review, we highlight metabolomic studies regarding the development of ICIs involved in the discovery of targets or mechanisms of action and assessment of clinical outcomes, including drug response and resistance and propose biomarkers. Furthermore, we also discuss the genomics, proteomics, and advanced omics studies providing insights and comprehensive or novel approaches for ICI development. The overview of ICI studies suggests potential strategies for the development of other cancer immunotherapies using omics techniques in future studies.
Collapse
Affiliation(s)
| | | | | | | | - Soo Kyung Bae
- College of Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon 14662, Korea; (S.J.Y.); (C.B.L.); (S.U.C.); (S.J.J.)
| |
Collapse
|
188
|
Abdelbaky SB, Ibrahim MT, Samy H, Mohamed M, Mohamed H, Mustafa M, Abdelaziz MM, Forrest ML, Khalil IA. Cancer immunotherapy from biology to nanomedicine. J Control Release 2021; 336:410-432. [PMID: 34171445 DOI: 10.1016/j.jconrel.2021.06.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/16/2021] [Accepted: 06/19/2021] [Indexed: 02/06/2023]
Abstract
With the significant drawbacks of conventional cancer chemotherapeutics, cancer immunotherapy has demonstrated the ability to eradicate cancer cells and circumvent multidrug resistance (MDR) with fewer side effects than traditional cytotoxic therapies. Various immunotherapeutic agents have been investigated for that purpose including checkpoint inhibitors, cytokines, monoclonal antibodies and cancer vaccines. All these agents aid immune cells to recognize and engage tumor cells by acting on tumor-specific pathways, antigens or cellular targets. However, immunotherapeutics are still associated with some concerns such as off-target side effects and poor pharmacokinetics. Nanomedicine may resolve some limitations of current immunotherapeutics such as localizing delivery, controlling release and enhancing the pharmacokinetic profile. Herein, we discuss recent advances of immunotherapeutic agents with respect to their development and biological mechanisms of action, along with the advantages that nanomedicine strategies lend to immunotherapeutics by possibly improving therapeutic outcomes and minimizing side effects.
Collapse
Affiliation(s)
- Salma B Abdelbaky
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Molecular, Cellular, and Developmental Biology, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, United States of America
| | - Mayar Tarek Ibrahim
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry, Center for Scientific Computation, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75275, United States of America
| | - Hebatallah Samy
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Menatalla Mohamed
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Hebatallah Mohamed
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario L8S 4M1, Canada
| | - Mahmoud Mustafa
- University of Science and Technology, Zewail City, 6th of October City, Giza 12578, Egypt
| | - Moustafa M Abdelaziz
- Department of Bioengineering, School of Engineering, The University of Kansas, Lawrence, KS 66045, USA
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, Lawrence, KS 66047, USA.
| | - Islam A Khalil
- Department of Pharmaceutics, College of Pharmacy and Drug Manufacturing, Misr University of Science and Technology (MUST), 6th of October, Giza 12582, Egypt.
| |
Collapse
|
189
|
Radulescu R, Totan AR, Imre MM, Miricescu D, Didilescu A, Greabu M. Mediators of extracellular matrix degradation and inflammation: A new team of possible biomarkers for oral squamous cell carcinoma stage. Exp Ther Med 2021; 22:877. [PMID: 34194555 PMCID: PMC8237384 DOI: 10.3892/etm.2021.10309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Oral cancer represents one of the most common types of cancer worldwide, with oral squamous cell carcinoma (OSCC) being the most frequently diagnosed. Cytokines play a crucial role in inflammation, apoptosis and metastasis. Interleukin (IL)-8 promotes the direct migration of inflammatory cells. IL-6 induces tumor cell proliferation, increases expression of invasiveness and angiogenetic factors or matrix metalloproteinases (MMPs), promoting metastasis. Tissue inhibitor of metalloproteinases (TIMPs) blocks the action of MMPs controlling extracellular matrix degradation and inhibiting metastasis. The aim of our study was to analyze the existence of correlations between inflammation markers (IL-6 and IL-8) and extracellular degradation protection markers such as TIMP-1 in OSCC tumors. Our study included 20 patients (12 females and 8 males) diagnosed with OSCC, recruited from January to April, 2020. IL-8, IL-6 and TIMP-1 levels were measured in the tumor cell lysates by ELISA technique, using relevant assay kits. Our results showed a positive and significant correlation between IL-6 and IL-8 (P=0.005, R=0.517) indicating that high IL-8 levels can be associated with high IL-6 levels. We also found a significant and high negative correlation (P<0.001, R=-0.673) between IL-6 and TIMP-1 and a significant and high negative correlation (P<0.001, R=-0.684) between IL-8 and TIMP-1 indicating that high levels of IL-8 and IL-6 are significantly associated with lower levels of TIMP-1. In conclusion, our study confirms the available literature data on IL-6 and IL-8 as potential markers for oral cancers such as OSCC and affect the tumor microenvironment by decreasing TIMPs. All three biomarkers included in this study have the potential to be used as detection or prognostic factors for oral cancer.
Collapse
Affiliation(s)
- Radu Radulescu
- Department of Biochemistry, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alexandra Ripszky Totan
- Department of Biochemistry, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Marina Melescanu Imre
- Department of Complete Denture, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 032799 Bucharest, Romania
| | - Daniela Miricescu
- Department of Biochemistry, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Andreea Didilescu
- Department of Embryology, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Maria Greabu
- Department of Biochemistry, Faculty of Dental Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
190
|
Paterson K, Zanivan S, Glasspool R, Coffelt SB, Zagnoni M. Microfluidic technologies for immunotherapy studies on solid tumours. LAB ON A CHIP 2021; 21:2306-2329. [PMID: 34085677 PMCID: PMC8204114 DOI: 10.1039/d0lc01305f] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/09/2021] [Indexed: 05/10/2023]
Abstract
Immunotherapy is a powerful and targeted cancer treatment that exploits the body's immune system to attack and eliminate cancerous cells. This form of therapy presents the possibility of long-term control and prevention of recurrence due to the memory capabilities of the immune system. Various immunotherapies are successful in treating haematological malignancies and have dramatically improved outcomes in melanoma. However, tackling other solid tumours is more challenging, mostly because of the immunosuppressive tumour microenvironment (TME). Current in vitro models based on traditional 2D cell monolayers and animal models, such as patient-derived xenografts, have limitations in their ability to mimic the complexity of the human TME. As a result, they have inadequate translational value and can be poorly predictive of clinical outcome. Thus, there is a need for robust in vitro preclinical tools that more faithfully recapitulate human solid tumours to test novel immunotherapies. Microfluidics and lab-on-a-chip technologies offer opportunities, especially when performing mechanistic studies, to understand the role of the TME in immunotherapy, and to expand the experimental throughput when using patient-derived tissue through its miniaturization capabilities. This review first introduces the basic concepts of immunotherapy, presents the current preclinical approaches used in immuno-oncology for solid tumours and then discusses the underlying challenges. We provide a rationale for using microfluidic-based approaches, highlighting the most recent microfluidic technologies and methodologies that have been used for studying cancer-immune cell interactions and testing the efficacy of immunotherapies in solid tumours. Ultimately, we discuss achievements and limitations of the technology, commenting on potential directions for incorporating microfluidic technologies in future immunotherapy studies.
Collapse
Affiliation(s)
- K Paterson
- Centre for Microsystems and Photonics, EEE Department, University of Strathclyde, Glasgow, UK.
| | - S Zanivan
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK and Cancer Research UK Beatson Institute, Glasgow, UK
| | - R Glasspool
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK and Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - S B Coffelt
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK and Cancer Research UK Beatson Institute, Glasgow, UK
| | - M Zagnoni
- Centre for Microsystems and Photonics, EEE Department, University of Strathclyde, Glasgow, UK.
| |
Collapse
|
191
|
Frazzini S, Riva F, Amadori M. Therapeutic and Prophylactic Use of Oral, Low-Dose IFNs in Species of Veterinary Interest: Back to the Future. Vet Sci 2021; 8:109. [PMID: 34208413 PMCID: PMC8231284 DOI: 10.3390/vetsci8060109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 12/17/2022] Open
Abstract
Cytokines are important molecules that orchestrate the immune response. Given their role, cytokines have been explored as drugs in immunotherapy in the fight against different pathological conditions such as bacterial and viral infections, autoimmune diseases, transplantation and cancer. One of the problems related to their administration consists in the definition of the correct dose to avoid severe side effects. In the 70s and 80s different studies demonstrated the efficacy of cytokines in veterinary medicine, but soon the investigations were abandoned in favor of more profitable drugs such as antibiotics. Recently, the World Health Organization has deeply discouraged the use of antibiotics in order to reduce the spread of multi-drug resistant microorganisms. In this respect, the use of cytokines to prevent or ameliorate infectious diseases has been highlighted, and several studies show the potential of their use in therapy and prophylaxis also in the veterinary field. In this review we aim to review the principles of cytokine treatments, mainly IFNs, and to update the experiences encountered in animals.
Collapse
Affiliation(s)
- Sara Frazzini
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Federica Riva
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, 26900 Lodi, Italy
| | - Massimo Amadori
- Rete Nazionale di Immunologia Veterinaria, 25125 Brescia, Italy;
| |
Collapse
|
192
|
Lee SH, Lim YJ, Kim CJ, Yu D, Lee JJ, Won Hong J, Baek YJ, Jung JY, Shin DJ, Kim SK. Safety and immunological effects of recombinant canine IL-15 in dogs. Cytokine 2021; 148:155599. [PMID: 34103211 DOI: 10.1016/j.cyto.2021.155599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/03/2021] [Accepted: 05/26/2021] [Indexed: 11/19/2022]
Abstract
Interleukin-15 (IL-15) is a pleiotropic cytokine that plays pivotal roles in innate and adaptive immunity. It is also a promising cytokine for treating cancer. Despite growing interest in its use as an immunotherapeutic, its safety and immunological effects in dogs have not been reported. In this study, healthy dogs were given recombinant canine IL-15 (rcIL-15) intravenously at a daily dose of 20 μg/kg for 8 days and monitored for 32 days to determine the safety and immunological effects of rcIL-15. The repeated administration of rcIL-15 was well tolerated, did not cause any serious side effects, and promoted the selective proliferation and activation of canine anti-cancer effector cells, including CD3+CD8+ cytotoxic T lymphocytes, CD3+CD5dimCD21-, and non-B/non-T NK cell populations, without stimulating Treg lymphocytes. The rcIL-15 injections also stimulated the expression of molecules and transcription factors associated with the activation and effector functions of NK cells, including CD16, NKG2D, NKp30, NKp44, NKp46, perforin, granzyme B, Ly49, T-bet, and Eomes. These results suggest that rcIL-15 might be a valuable therapeutic adjuvant to improve immunity against cancer in dogs.
Collapse
Affiliation(s)
- Soo-Hyeon Lee
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Yu-Jin Lim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Cheol-Jung Kim
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Dohyeon Yu
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Je-Jung Lee
- Department of Hemotology-Oncology, Chonnam National Univresity Hwasun Hospital, Hwasun, Jeollanamdo, Republic of Korea
| | - Jeong Won Hong
- Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Yeon-Ju Baek
- Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Ji-Youn Jung
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea
| | - Dong-Jun Shin
- Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; SD Medic Co, Gwangju, Republic of Korea.
| | - Sang-Ki Kim
- Department of Integrated Life Science and Technology, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Department of Companion and Laboratory Animal Science, College of Industrial Science, Kongju National University, Yesan-gun, Chungnam, Republic of Korea; Research Institute for Natural Products, Kongju National University, Yesan-gun, Chungnam, Republic of Korea.
| |
Collapse
|
193
|
Chauhan DS, Dhasmana A, Laskar P, Prasad R, Jain NK, Srivastava R, Jaggi M, Chauhan SC, Yallapu MM. Nanotechnology synergized immunoengineering for cancer. Eur J Pharm Biopharm 2021; 163:72-101. [PMID: 33774162 PMCID: PMC8170847 DOI: 10.1016/j.ejpb.2021.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 12/26/2022]
Abstract
Novel strategies modulating the immune system yielded enhanced anticancer responses and improved cancer survival. Nevertheless, the success rate of immunotherapy in cancer treatment has been below expectation(s) due to unpredictable efficacy and off-target effects from systemic dosing of immunotherapeutic(s). As a result, there is an unmet clinical need for improving conventional immunotherapy. Nanotechnology offers several new strategies, multimodality, and multiplex biological targeting advantage to overcome many of these challenges. These efforts enable programming the pharmacodynamics, pharmacokinetics, and delivery of immunomodulatory agents/co-delivery of compounds to prime at the tumor sites for improved therapeutic benefits. This review provides an overview of the design and clinical principles of biomaterials driven nanotechnology and their potential use in personalized nanomedicines, vaccines, localized tumor modulation, and delivery strategies for cancer immunotherapy. In this review, we also summarize the latest highlights and recent advances in combinatorial therapies availed in the treatment of cold and complicated tumors. It also presents key steps and parameters implemented for clinical success. Finally, we analyse, discuss, and provide clinical perspectives on the integrated opportunities of nanotechnology and immunology to achieve synergistic and durable responses in cancer treatment.
Collapse
Affiliation(s)
- Deepak S Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nishant K Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA.
| |
Collapse
|
194
|
Peng X, Wang J, Zhou F, Liu Q, Zhang Z. Nanoparticle-based approaches to target the lymphatic system for antitumor treatment. Cell Mol Life Sci 2021; 78:5139-5161. [PMID: 33963442 PMCID: PMC11072902 DOI: 10.1007/s00018-021-03842-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/14/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Immunotherapies have been established as safe and efficient modalities for numerous tumor treatments. The lymphatic system, which is an important system, can modulate the immune system via a complex network, which includes lymph nodes, vessels, and lymphocytes. With the deepening understanding of tumor immunology, a plethora of immunotherapies, which include vaccines, photothermal therapy, and photodynamic therapy, have been established for antitumor treatments. However, the deleterious off-target effects and nonspecific targeting of therapeutic agents result in low efficacy of immunotherapy. Fortunately, nanoparticle-based approaches for targeting the lymphatic system afford a unique opportunity to manufacture drugs that can simultaneously tackle both aspects, thereby improving tumor treatments. Over the past decades, great strides have been made in the development of DC vaccines and nanomedicine as antitumor treatments in the field of lymphatic therapeutics and diagnosis. In this review, we summarize the current strategies through which nanoparticle technology has been designed to target the lymphatic system and describe applications of lymphatic imaging for the diagnosis and image-guided surgery of tumor metastasis. Moreover, improvements in the tumor specificity of nanovaccines and medicines, which have been realized through targeting or stimulating the lymphatic system, can provide amplified antitumor immune responses and reduce side effects, thereby promoting the paradigm of antitumor treatment into the clinic to benefit patients.
Collapse
Affiliation(s)
- Xingzhou Peng
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China
| | - Junjie Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Feifan Zhou
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China
| | - Qian Liu
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China.
| | - Zhihong Zhang
- School of Biomedical Engineering, Hainan University, Haikou, 570228, Hainan, China.
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, School of Engineering Sciences, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China.
| |
Collapse
|
195
|
Persano S, Das P, Pellegrino T. Magnetic Nanostructures as Emerging Therapeutic Tools to Boost Anti-Tumour Immunity. Cancers (Basel) 2021; 13:2735. [PMID: 34073106 PMCID: PMC8198238 DOI: 10.3390/cancers13112735] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy has shown remarkable results in various cancer types through a range of immunotherapeutic approaches, including chimeric antigen receptor-T cell (CAR-T) therapy, immune checkpoint blockade (ICB), and therapeutic vaccines. Despite the enormous potential of cancer immunotherapy, its application in various clinical settings has been limited by immune evasion and immune suppressive mechanisms occurring locally or systemically, low durable response rates, and severe side effects. In the last decades, the rapid advancement of nanotechnology has been aiming at the development of novel synthetic nanocarriers enabling precise and enhanced delivery of immunotherapeutics, while improving drug stability and effectiveness. Magnetic nanostructured formulations are particularly intriguing because of their easy surface functionalization, low cost, and robust manufacturing procedures, together with their suitability for the implementation of magnetically-guided and heat-based therapeutic strategies. Here, we summarize and discuss the unique features of magnetic-based nanostructures, which can be opportunely designed to potentiate classic immunotherapies, such as therapeutic vaccines, ICB, adoptive cell therapy (ACT), and in situ vaccination. Finally, we focus on how multifunctional magnetic delivery systems can facilitate the anti-tumour therapies relying on multiple immunotherapies and/or other therapeutic modalities. Combinatorial magnetic-based therapies are indeed offering the possibility to overcome current challenges in cancer immunotherapy.
Collapse
Affiliation(s)
- Stefano Persano
- Nanomaterials for Biomedical Applications, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy;
| | | | - Teresa Pellegrino
- Nanomaterials for Biomedical Applications, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy;
| |
Collapse
|
196
|
Autenshlyus A, Arkhipov S, Mikhaylova E, Marinkin I, Arkhipova V, Varaksin N, Vavilin V, Lyahovich V. Analyzing the relationship between the cytokine profile of invasive breast carcinoma, its histopathological characteristics and metastasis to regional lymph nodes. Sci Rep 2021; 11:11359. [PMID: 34059727 PMCID: PMC8166970 DOI: 10.1038/s41598-021-90930-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
This study was aimed at analyzing the relations of metastasis to regional lymph nodes (RLNs) with histopathological indicators of invasive breast carcinoma of no special type (IC-NST) and its cytokine profile. Enzyme-linked immunosorbent assays were performed to determine concentrations of IL-2, IL-6, IL-8, IL-10, IL-17, IL-18, IL-1β, IL-1Ra, TNF-α, IFN-γ, G-CSF, GM-CSF, VEGF-A, and MCP-1 in the culture supernatant of IC-NST samples from 48 female patients. Histopathological indicators (degree of tumor cell differentiation, mitoses, and others) and ER, PR, Her2/neu, Ki-67, and CD34 expression levels were determined. By means of three types of neural network models, it was shown that for different parameters of the output layer, different groups of parameters are involved that have predictive value regarding metastasis to RLNs. As a result of multi-dimensional cluster analysis, three clusters were formed with different cytokines profiles of IC-NST. Different correlations between indicators of cytokine production by IC-NST and its histopathological parameters were revealed in groups with different cytokine profiles. It was shown that at simultaneous evaluation of the production of even two cytokines, the importance of which relationship with metastasis was revealed by neural network modeling, can increase the probability of determining the presence of metastasis in the RLNs.
Collapse
Affiliation(s)
- Alexander Autenshlyus
- Department of Scientific Work, Central Research Laboratory, Laboratory of Immunohistochemistry Biochemistry and Pharmacology, Novosibirsk State Medical University, Krasny Prospect 52, Novosibirsk, 630091, Russian Federation.,Institute of Molecular Biology and Biophysics - subdivision of FRC FTM, Novosibirsk, Russia
| | - Sergey Arkhipov
- Department of Scientific Work, Central Research Laboratory, Laboratory of Immunohistochemistry Biochemistry and Pharmacology, Novosibirsk State Medical University, Krasny Prospect 52, Novosibirsk, 630091, Russian Federation. .,Institute of Molecular Biology and Biophysics - subdivision of FRC FTM, Novosibirsk, Russia.
| | - Elena Mikhaylova
- Department of Scientific Work, Central Research Laboratory, Laboratory of Immunohistochemistry Biochemistry and Pharmacology, Novosibirsk State Medical University, Krasny Prospect 52, Novosibirsk, 630091, Russian Federation.,Institute of Molecular Biology and Biophysics - subdivision of FRC FTM, Novosibirsk, Russia
| | - Igor Marinkin
- Department of Scientific Work, Central Research Laboratory, Laboratory of Immunohistochemistry Biochemistry and Pharmacology, Novosibirsk State Medical University, Krasny Prospect 52, Novosibirsk, 630091, Russian Federation
| | - Valentina Arkhipova
- Department of Scientific Work, Central Research Laboratory, Laboratory of Immunohistochemistry Biochemistry and Pharmacology, Novosibirsk State Medical University, Krasny Prospect 52, Novosibirsk, 630091, Russian Federation
| | | | - Valentin Vavilin
- Institute of Molecular Biology and Biophysics - subdivision of FRC FTM, Novosibirsk, Russia
| | - Vyacheslav Lyahovich
- Institute of Molecular Biology and Biophysics - subdivision of FRC FTM, Novosibirsk, Russia
| |
Collapse
|
197
|
Rückert M, Flohr AS, Hecht M, Gaipl US. Radiotherapy and the immune system: More than just immune suppression. STEM CELLS (DAYTON, OHIO) 2021; 39:1155-1165. [PMID: 33961721 DOI: 10.1002/stem.3391] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/20/2021] [Indexed: 11/07/2022]
Abstract
Radiotherapy (RT) is still one of the standard cancer therapies, with up to two third of all cancer patients with solid tumors being irradiated in the course of their disease. The aim of using ionizing radiation in fractionated treatment schedules was always to achieve local tumor control by inducing DNA damage which can be repaired by surrounding normal tissue but leads to cell death in tumor cells. Meanwhile, it is known that RT also has immunological effects reshaping the tumor microenvironment. Nevertheless, RT alone often fails to elicit potent antitumor immune responses as these effects can be immunostimulatory as well as immunosuppressive. Here, we discuss how immunotherapies can be exploited in combined therapies to boost RT-induced antitumor immune responses or to counteract preexisting and RT-mediated immunosuppression to improve local and systemic tumor control. Furthermore, we highlight some parameters of radioimmunotherapies (RITs) which are under investigation for potential optimizations and how RIT approaches are tested in first phases II and III trials. Finally, we discuss how RT might affect normal and cancer stem cells.
Collapse
Affiliation(s)
- Michael Rückert
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Ann-Sophie Flohr
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| | - Udo S Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.,Deutsches Zentrum Immuntherapie, Erlangen, Germany
| |
Collapse
|
198
|
Abstract
PURPOSE OF REVIEW The use of cytokines in harnessing the immune system to eradicate cancer has been an important treatment modality. However, the dose-limiting toxicities of these cytokines limited their usage in clinic. Here, we review the basic biology of cytokines involved in the treatment of melanoma and discuss their therapeutic applications. Moreover, we describe several innovative technological approaches that have been developed to improve the pharmacokinetics, safety, and efficacy of these cytokines. RECENT FINDINGS The safety and the anti-tumor activity of newly engineered cytokines including PEGylated IL-2 (NKTR-214), PEGylated IL-10 (AM0010), and IL-15 super agonist (ALT-803) have been evaluated in clinical trials with encouraging clinical activity and acceptable safety profile, both as single agents and in combination with immuno-oncology agents. A greater understanding of the mechanisms of action and effective dosing of these newly engineered cytokine together with determination of optimum combination therapy regimens may yield greater clinical benefits in the future.
Collapse
|
199
|
Faraji N, Esrafili A, Esfandiari B, Abednezhad A, Naghizadeh M, Arasteh J. Synthesis of pH-sensitive hyaluronic acid nanogels loaded with paclitaxel and interferon gamma: Characterization and effect on the A549 lung carcinoma cell line. Colloids Surf B Biointerfaces 2021; 205:111845. [PMID: 34015733 DOI: 10.1016/j.colsurfb.2021.111845] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 04/17/2021] [Accepted: 05/11/2021] [Indexed: 11/15/2022]
Abstract
Lung cancer is one of the deadliest cancers in various populations. Apart from the effects that anticancer drugs such as paclitaxel (PTX) have on cancer cells, they also have many side effects on healthy cells. Interferon gamma (IFN-γ) is also one of the cytokines used in the treatment of cancer. Current research is focused on providing new drug carriers to find new therapeutic goals. After synthesis of nanogels and loading of PTX and IFN-γ, the cytotoxicity of these nanogels on A549 and HEK293 healthy cell line was measured by MTT assay and flow cytometry analysis. Finally, the expression of STAT1 gene was investigated using Real time-PCR. The results of MTT assay showed that the survival rate of healthy cells treated with PTX and IFN-γ-loaded nanogels was 2.15 and 2.39 times higher than cancer cells, respectively. The results also showed that the gene expression STAT1 in A549 cells exposed to these nanogels was higher than healthy cells (p < 0.05). Based on flow cytometry results, the death rate of healthy cells treated with the mentioned nanogels was lower than cancer cells (p < 0.05). Therefore, Studies showed that synthesized nanogels have positive effects on cancer cells and also have fewer side effects on healthy cells.
Collapse
Affiliation(s)
- Nima Faraji
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ali Esrafili
- Department of Environmental Health Engineering, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Esfandiari
- Department of Biology, Islamshahr Branch, Islamic Azad University, Tehran, Iran
| | - Arash Abednezhad
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Naghizadeh
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Javad Arasteh
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
200
|
Isvoranu G, Surcel M, Munteanu AN, Bratu OG, Ionita-Radu F, Neagu MT, Chiritoiu-Butnaru M. Therapeutic potential of interleukin-15 in cancer (Review). Exp Ther Med 2021; 22:675. [PMID: 33986840 PMCID: PMC8112152 DOI: 10.3892/etm.2021.10107] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
The immune system is dysfunctional in cancer, and therapeutic approaches designated to restore immunity and increase long-term overall survival are desirable. The role of immunotherapy is to trigger the immune system to recognize and destroy tumor cells. Interleukin-15 (IL-15) is a member of the common gamma-chain (γc) cytokines that promote the differentiation and expansion of T cells, B cells and natural killer (NK) cells, leading to enhanced antitumor responses. This suggests that IL-15 is a promising candidate for anticancer therapy. Renewed interest in cancer immunotherapy has led to an increased number of preclinical studies and clinical trials that have investigated the reliability and potency of IL-15-based agents, not only as single therapy, but also in combination with others. This review provides a description of these studies which show the advantages and disadvantages of IL-15 as an immunotherapeutic agent. We present here the role of IL-15 and pharmacologically improved IL-15 superagonists as a single treatment or in combination with other therapeutic agents.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Department of Animal Husbandry, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Surcel
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania
| | - Adriana Narcisa Munteanu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Ovidiu Gabriel Bratu
- Department of Clinical Department III, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Clinic of Urology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania.,Academy of Romanian Scientists, 050094 Bucharest, Romania
| | - Florentina Ionita-Radu
- Department of Gastroenterology, 'Dr. Carol Davila' University Emergency Central Military Hospital, 010825 Bucharest, Romania
| | - Monica Teodora Neagu
- Department of Immunology, 'Victor Babes' National Institute of Pathology, 050096 Bucharest, Romania.,Department of Doctoral School of Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Marioara Chiritoiu-Butnaru
- Department of Molecular Cell Biology, Institute of Biochemistry of the Romanian Academy (IBAR), 060031 Bucharest, Romania
| |
Collapse
|