1
|
Uinarni H, Oghenemaro EF, Menon SV, Hjazi A, Ibrahim FM, Kaur M, Zafarjonovna AZ, Deorari M, Jabir MS, Zwamel AH. Breaking Barriers: Nucleic Acid Aptamers in Gastrointestinal (GI) Cancers Therapy. Cell Biochem Biophys 2024; 82:1763-1776. [PMID: 38916791 DOI: 10.1007/s12013-024-01367-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 06/26/2024]
Abstract
Conventional cancer therapies can have significant adverse effects as they are not targeted to cancer cells and may damage healthy cells. Single-stranded oligonucleotides assembled in a particular architecture, known as aptamers, enable them to attach selectively to target areas. Usually, they are created by Systematic Evolution of Ligand by Exponential enrichment (SELEX), and they go through a rigorous pharmacological revision process to change their therapeutic half-life, affinity, and specificity. They could thus offer a viable substitute for antibodies in the targeted cancer treatment market. Although aptamers can be a better choice in some situations, antibodies are still appropriate for many other uses. The technique of delivering aptamers is simple and reasonable, and the time needed to manufacture them is relatively brief. Aptamers do not require animals or an immune response to be produced, in contrast to antibodies. When used as a medication, aptamers can directly suppress tumor cells. As an alternative, they can be included in systems for targeted drug delivery that administer medications specifically to tumor cells while reducing toxicity to healthy cells. The most recent and cutting-edge methods for treating gastrointestinal (GI) tract cancer with aptamers will be covered in this review, with a focus on targeted therapy as a means of conquering resistance to traditional medicines.
Collapse
Affiliation(s)
- Herlina Uinarni
- Department of Anatomy, School of Medicine and Health Sciences Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia.
- Radiology department of Pantai Indah Kapuk Hospital Jakarta, Jakarta, Indonesia.
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Fatma Magdi Ibrahim
- Assisstant professor, Community Health Nursing, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
- Lecturer, geriatric nursing, Mansoura University, Mansoura, Egypt
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | | | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Majid S Jabir
- Department of applied sciences, University of technology, Baghdad, Iraq
| | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Ausejo-Mauleon I, Martinez-Velez N, Lacalle A, de la Nava D, Cebollero J, Villanueva H, Casares N, Marco-Sanz J, Laspidea V, Becher O, Patiño-García A, Labiano S, Pastor F, Alonso MM. Combination of locoregional radiotherapy with a TIM-3 aptamer improves survival in diffuse midline glioma models. JCI Insight 2024; 9:e175257. [PMID: 39146023 PMCID: PMC11457855 DOI: 10.1172/jci.insight.175257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
Pediatric diffuse midline gliomas (DMG) with altered H3-K27M are aggressive brain tumors that arise during childhood. Despite advances in genomic knowledge and the significant number of clinical trials testing new targeted therapies, patient outcomes are still poor. Immune checkpoint blockades with small molecules, such as aptamers, are opening new therapeutic options that represent hope for this orphan disease. Here, we demonstrated that a TIM-3 aptamer (TIM-3 Apt) as monotherapy increased the immune infiltration and elicited a strong specific immune response with a tendency to improve the overall survival of treated DMG-bearing mice. Importantly, combining TIM-3 Apt with radiotherapy increased the overall median survival and led to long-term survivor mice in 2 pediatric DMG orthotopic murine models. Interestingly, TIM-3 Apt administration increased the number of myeloid populations and the proinflammatory CD8-to-Tregs ratios in the tumor microenvironment as compared with nontreated groups after radiotherapy. Importantly, the depletion of T cells led to a major loss of the therapeutic effect achieved by the combination. This work uncovers TIM-3 targeting as an immunotherapy approach to improve the radiotherapy outcome in DMGs and offers a strong foundation for propelling a phase I clinical trial using radiotherapy and TIM-3 blockade combination as a treatment for these tumors.
Collapse
Affiliation(s)
- Iker Ausejo-Mauleon
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Naiara Martinez-Velez
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Andrea Lacalle
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Daniel de la Nava
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Cebollero
- Molecular Therapeutics Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
| | - Helena Villanueva
- Molecular Therapeutics Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
| | - Noelia Casares
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Marco-Sanz
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Virginia Laspidea
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Research Department of Hematology and Oncology, University College London, London, UK
| | - Oren Becher
- Jack Martin Fund Division of Pediatric Hematology-Oncology, Mount Sinai, New York, New York, USA
| | - Ana Patiño-García
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Sara Labiano
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| | - Fernando Pastor
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Molecular Therapeutics Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
| | - Marta M. Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain
- Solid Tumor Program, Center for the Applied Medical Research, Pamplona, Navarra, Spain
- Department of Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
3
|
Yıldırım C. Galectin-9, a pro-survival factor inducing immunosuppression, leukemic cell transformation and expansion. Mol Biol Rep 2024; 51:571. [PMID: 38662155 DOI: 10.1007/s11033-024-09563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Leukemia is a malignancy of the bone marrow and blood originating from self-renewing cancerous immature blast cells or transformed leukocytes. Despite improvements in treatments, leukemia remains still a serious disease with poor prognosis because of disease heterogeneity, drug resistance and relapse. There is emerging evidence that differentially expression of co-signaling molecules play a critical role in tumor immune evasion. Galectin-9 (Gal-9) is one of the key proteins that leukemic cells express, secrete, and use to proliferate, self-renew, and survive. It also suppresses host immune responses controlled by T and NK cells, enabling leukemic cells to evade immune surveillance. The present review provides the molecular mechanisms of Gal-9-induced immune evasion in leukemia. Understanding the complex immune evasion machinery driven by Gal-9 expressing leukemic cells will enable the identification of novel therapeutic strategies for efficient immunotherapy in leukemic patients. Combined treatment approaches targeting T-cell immunoglobulin and mucin domain-3 (Tim-3)/Gal-9 and other immune checkpoint pathways can be considered, which may enhance the efficacy of host effector cells to attack leukemic cells.
Collapse
Affiliation(s)
- Cansu Yıldırım
- Atatürk Vocational School of Health Services, Afyonkarahisar Health Sciences University, Afyonkarahisar, Turkey.
| |
Collapse
|
4
|
Kalele K, Nyahatkar S, Mirgh D, Muthuswamy R, Adhikari MD, Anand K. Exosomes: A Cutting-Edge Theranostics Tool for Oral Cancer. ACS APPLIED BIO MATERIALS 2024; 7:1400-1415. [PMID: 38394624 DOI: 10.1021/acsabm.3c01243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Exosomes are a subpopulation of extracellular vesicles (EVs) secreted by cells. In cancer, they are key cellular messengers during cancer development and progression. Tumor-derived exosomes (TEXs) promote cancer progression. In oral cancer, the major complication is oral squamous cell carcinoma (OSCC). Exosomes show strong participation in several OSCC-related activities such as uncontrolled cell growth, immune suppression, angiogenesis, metastasis, and drug and therapeutic resistance. It is also a potential biomarker source for oral cancer. Some therapeutic exosome sources such as stem cells, plants (it is more effective compared to others), and engineered exosomes reduce oral cancer development. This therapeutic approach is effective because of its specificity, biocompatibility, and cell-free therapy (it reduced side effects in cancer treatment). This article highlights exosome-based theranostics signatures in oral cancer, clinical trials, challenges of exosome-based oral cancer research, and future improvements. In the future, exosomes may become an effective and affordable solution for oral cancer.
Collapse
Affiliation(s)
- Ketki Kalele
- Neuron Institute of Applied Research, Rajapeth-Irwin Square Flyover, Amravati, Maharashtra 444601, India
| | - Sidhanti Nyahatkar
- VYWS Dental College & Hospital, WQMV+7X6, Tapovan-Wadali Road, Camp Rd, SRPF Colony, Amravati, Maharashtra 444602, India
| | - Divya Mirgh
- Department of Infectious Diseases, Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Raman Muthuswamy
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - Manab Deb Adhikari
- Department of Biotechnology, University of North Bengal, Darjeeling, West Bengal 734013, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
5
|
Mahmoudian F, Ahmari A, Shabani S, Sadeghi B, Fahimirad S, Fattahi F. Aptamers as an approach to targeted cancer therapy. Cancer Cell Int 2024; 24:108. [PMID: 38493153 PMCID: PMC10943855 DOI: 10.1186/s12935-024-03295-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Conventional cancer treatments can cause serious side effects because they are not specific to cancer cells and can damage healthy cells. Aptamers often are single-stranded oligonucleotides arranged in a unique architecture, allowing them to bind specifically to target sites. This feature makes them an ideal choice for targeted therapeutics. They are typically produced through the systematic evolution of ligands by exponential enrichment (SELEX) and undergo extensive pharmacological revision to modify their affinity, specificity, and therapeutic half-life. Aptamers can act as drugs themselves, directly inhibiting tumor cells. Alternatively, they can be used in targeted drug delivery systems to transport drugs directly to tumor cells, minimizing toxicity to healthy cells. In this review, we will discuss the latest and most advanced approaches to using aptamers for cancer treatment, particularly targeted therapy overcoming resistance to conventional therapies.
Collapse
Affiliation(s)
- Fatemeh Mahmoudian
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Azin Ahmari
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Radiation Oncology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shiva Shabani
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Infectious Diseases, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Bahman Sadeghi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
- Department of Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shohreh Fahimirad
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran.
| | - Fahimeh Fattahi
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran.
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Singh R, Srivastava P, Manna PP. Evaluation of regulatory T-cells in cancer immunotherapy: therapeutic relevance of immune checkpoint inhibition. Med Oncol 2024; 41:59. [PMID: 38238513 DOI: 10.1007/s12032-023-02289-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024]
Abstract
The evolution of the complex immune system is equipped to defend against perilous intruders and concurrently negatively regulate the deleterious effect of immune-mediated inflammation caused by self and nonself antigens. Regulatory T-cells (Tregs) are specialized cells that minimize immune-mediated inflammation, but in malignancies, this feature has been exploited toward cancer progression by keeping the antitumor immune response in check. The modulation of Treg cell infiltration and their induction in the TME (tumor microenvironment) alongside associated inhibitory molecules, both soluble or membranes tethered in the TME, have proven clinically beneficial in boosting the tumoricidal activity of the immune system. Moreover, Treg-associated immune checkpoints pose a greater obstruction in cancer immunotherapy. Inhibiting or blocking active immune checkpoint signaling in combination with other therapies has proven clinically beneficial. This review summarizes the ontogeny of Treg cells and their migration, stability, and function in the TME. We also elucidate the Treg-associated checkpoint moieties that impede effective antitumor activity and harness these molecules for effective and targeted immunotherapy against cancer nuisance.
Collapse
Affiliation(s)
- Ranjeet Singh
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Prateek Srivastava
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Partha Pratim Manna
- Immunobiology Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India.
| |
Collapse
|
7
|
Kejamurthy P, Devi KTR. Immune checkpoint inhibitors and cancer immunotherapy by aptamers: an overview. Med Oncol 2023; 41:40. [PMID: 38158454 DOI: 10.1007/s12032-023-02267-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024]
Abstract
Efforts in cancer immunotherapy aim to counteract evasion mechanisms and stimulate the immune system to recognise and attack cancer cells effectively. Combination therapies that target multiple aspects of immune evasion are being investigated to enhance the overall efficacy of cancer immunotherapy. PD-1 (Programmed Cell Death Protein 1), CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4), LAG-3 (Lymphocyte-Activation Gene 3), and TIM-3 (T Cell Immunoglobulin and Mucin Domain-Containing Protein3) are all immune checkpoint receptors that play crucial roles in regulating the immune response and maintaining self-tolerance often exploited by cancer cells to evade immune surveillance. Antibodies targeted against immune checkpoint inhibitors such as anti-PD-1 antibodies (e.g., pembrolizumab, nivolumab), anti-CTLA-4 antibodies (e.g., Ipilimumab), and experimental drugs targeting LAG-3 and TIM-3, aim to block these interactions and unleash the immune system's ability to recognise and destroy cancer cells. The US FDA has approved different categories of immune checkpoint inhibitors that have been utilised successfully in some patients with metastatic melanoma, renal cell carcinoma, head and neck cancers, and non-small lung cancer. Although several immune checkpoint inhibitor antibodies have been developed, they exhibited immune-related adverse effects, resulting in hypophysitis, diabetes, and neurological issues. These adverse effects of antibodies can be reduced by developing aptamer against the target. Aptamers offer several advantages over traditional antibodies, such as improved specificity, reduced immunogenicity, and flexible design for reduced adverse effects that specifically target and block protein-protein or receptor-ligand interactions involved in immune checkpoint pathways. The current study aims to review the function of particular immune checkpoint inhibitors along with developed aptamer-mediated antitumor cytotoxicity in cancer treatment.
Collapse
Affiliation(s)
- Priyatharcini Kejamurthy
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - K T Ramya Devi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| |
Collapse
|
8
|
Chu J, Liu W, Hu X, Zhang H, Jiang J. P2RY13 is a prognostic biomarker and associated with immune infiltrates in renal clear cell carcinoma: A comprehensive bioinformatic study. Health Sci Rep 2023; 6:e1646. [PMID: 38045624 PMCID: PMC10691167 DOI: 10.1002/hsr2.1646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/03/2023] [Accepted: 10/10/2023] [Indexed: 12/05/2023] Open
Abstract
Background and Aims Clear cell renal cell carcinoma (ccRCC) is a common and aggressive form of cancer with a high incidence globally. This study aimed to investigate the role of P2RY13 in the progression of ccRCC and elucidate its mechanism of action. Methods Gene Expression Omnibus and The Cancer Genome Atlas databases were used to extract gene expression profiles of ccRCC. These profiles were annotated and visualized by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses, as well as Gene Set Enrichment Analysis (GSEA). The STRING database was used to establish a protein-protein interaction network and to analyze the functional similarity. The GEPIA2 database was used to predict survival associated with hub genes. Meanwhile, the TIMER2.0 database was used to assess immune cell infiltration and its link with the hub genes. Immunohistochemistry (IHC) was used to determine the difference between ccRCC and adjacent normal tissue. Results We identified 272 differentially expressed genes (DEGs). GO and KEGG analyses suggested that DEGs were primarily involved in lymphocyte activation, inflammatory response, immunological effector mechanism pathways. By cytohubba, the 20 highest-scoring hub genes were screened to identify critical genes in the protein-protein interaction network linked with ccRCC. Resting dendritic cells, CD8 T cells, and activated mast cells all showed a significant positive correlation with these hub genes. Moreover, a higher immune score was associated with increased prognostic risk scores, which in turn correlated with a poorer prognosis. IHC revealed that P2RY13 was expressed at higher levels in ccRCC compared to para-cancer tissues. Conclusion Identifying the DEGs will aid in the understanding of the causes and molecular mechanisms involved in ccRCC. P2RY13 may play a pivotal role in the progression and prognosis of ccRCC, potentially driving carcinogenesis though immune system mechanisms.
Collapse
Affiliation(s)
- Jie Chu
- Department of OncologyThe First People's Hospital of ZiyangZiyangChina
| | - Wei Liu
- Department of General Family MedicineThe First People's Hospital of NeiJiangNeiJiangChina
| | - Xinyue Hu
- Department of Clinical Laboratory, Kunming First People's HospitalKunming Medical UniversityKunmingChina
| | - Huiling Zhang
- Department of OncologyThe First People's Hospital of ZiyangZiyangChina
| | - Jiudong Jiang
- Department of SurgeryThe First People's Hospital of ZiYangZiyangChina
| |
Collapse
|
9
|
Szymanowski W, Szymanowska A, Bielawska A, Lopez-Berestein G, Rodriguez-Aguayo C, Amero P. Aptamers as Potential Therapeutic Tools for Ovarian Cancer: Advancements and Challenges. Cancers (Basel) 2023; 15:5300. [PMID: 37958473 PMCID: PMC10647731 DOI: 10.3390/cancers15215300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Ovarian cancer (OC) is the most common lethal gynecologic cause of death in women worldwide, with a high mortality rate and increasing incidence. Despite advancements in the treatment, most OC patients still die from their disease due to late-stage diagnosis, the lack of effective diagnostic methods, and relapses. Aptamers, synthetic, short single-stranded oligonucleotides, have emerged as promising anticancer therapeutics. Their ability to selectively bind to target molecules, including cancer-related proteins and receptors, has revolutionized drug discovery and biomarker identification. Aptamers offer unique insights into the molecular pathways involved in cancer development and progression. Moreover, they show immense potential as drug delivery systems, enabling targeted delivery of therapeutic agents to cancer cells while minimizing off-target effects and reducing systemic toxicity. In the context of OC, the integration of aptamers with non-coding RNAs (ncRNAs) presents an opportunity for precise and efficient gene targeting. Additionally, the conjugation of aptamers with nanoparticles allows for accurate and targeted delivery of ncRNAs to specific cells, tissues, or organs. In this review, we will summarize the potential use and challenges associated with the use of aptamers alone or aptamer-ncRNA conjugates, nanoparticles, and multivalent aptamer-based therapeutics for the treatment of OC.
Collapse
Affiliation(s)
- Wojciech Szymanowski
- Department of Biotechnology, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.S.); (A.B.)
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, 15-222 Bialystok, Poland; (W.S.); (A.B.)
| | - Gabriel Lopez-Berestein
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.S.); (G.L.-B.); (C.R.-A.)
| |
Collapse
|
10
|
Bertrand P. Aptamers Targeting the PD-1/PD-L1 Axis: A Perspective. J Med Chem 2023; 66:10878-10888. [PMID: 37561598 DOI: 10.1021/acs.jmedchem.3c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Aptamers have emerged in recent years as alternatives to antibodies or small molecules to interfere with the immune check points by blocking the PD-1/PD-L1 interactions and represent an interesting perspective for immuno-oncology. Aptamers are RNA or DNA nucleotides able to bind to a target with high affinity, with the target ranging from small molecules to proteins and up to cells. Aptamers are identified by the SELEX method that can be modified for specific purposes. The range of applications of aptamers covers therapy as well as new alternative assay technologies similar to ELISA. Aptamers' limited plasma stability can be managed using delivery strategies. The goal of this Perspective is to give an overview of the current development of aptamers targeting the most studied immune checkpoint modulators, PD-1 and PD-L1, and analogous strategies with aptamers for other immuno-related targets.
Collapse
Affiliation(s)
- Philippe Bertrand
- University of Poitiers, IC2MP UMR 7285 CNRS, 4 rue Michel Brunet B27, TSA 51106, 86073 Poitiers cedex 9, France
| |
Collapse
|
11
|
Bailly C, Thuru X, Goossens L, Goossens JF. Soluble TIM-3 as a biomarker of progression and therapeutic response in cancers and other of human diseases. Biochem Pharmacol 2023; 209:115445. [PMID: 36739094 DOI: 10.1016/j.bcp.2023.115445] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023]
Abstract
Immune checkpoints inhibition is a privileged approach to combat cancers and other human diseases. The TIM-3 (T cell immunoglobulin and mucin-domain containing-3) inhibitory checkpoint expressed on different types of immune cells is actively investigated as an anticancer target, with a dozen of monoclonal antibodies in (pre)clinical development. A soluble form sTIM-3 can be found in the plasma of patients with cancer and other diseases. This active circulating protein originates from the proteolytic cleavage by two ADAM metalloproteases of the membrane receptor shared by tumor and non-tumor cells, and extracellular vesicles. In most cancers but not all, overexpression of mTIM-3 at the cell surface leads to high level of sTIM-3. Similarly, elevated levels of sTIM-3 have been reported in chronic autoimmune diseases, inflammatory gastro-intestinal diseases, certain viral and parasitic diseases, but also in cases of organ transplantation and in pregnancy-related pathologies. We have analyzed the origin of sTIM-3, its methods of dosage in blood or plasma, its presence in multiple diseases and its potential role as a biomarker to follow disease progression and/or the treatment response. In contrast to sPD-L1 generated by different classes of proteases and by alternative splicing, sTIM-3 is uniquely produced upon ADAM-dependent shedding, providing a more homogenous molecular entity and a possibly more reliable molecular marker. However, the biological functionality of sTIM-3 remains insufficiently characterized. The review shed light on pathologies associated with an altered expression of sTIM-3 in human plasma and the possibility to use sTIM-3 as a diagnostic or therapeutic marker.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Consulting Scientific Office, Lille (Wasquehal) 59290, France; University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France.
| | - Xavier Thuru
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France
| | - Laurence Goossens
- University of Lille, Faculty of Pharmacy, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), 3 rue du Professeur Laguesse, 59000 Lille, France; University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, 59000 Lille, France
| | - Jean-François Goossens
- University of Lille, CHU Lille, ULR 7365 - GRITA - Groupe de Recherche sur les formes Injectables et les Technologies Associées, 59000 Lille, France
| |
Collapse
|
12
|
Yang J, Tabuchi Y, Katsuki R, Taki M. bioTCIs: Middle-to-Macro Biomolecular Targeted Covalent Inhibitors Possessing Both Semi-Permanent Drug Action and Stringent Target Specificity as Potential Antibody Replacements. Int J Mol Sci 2023; 24:3525. [PMID: 36834935 PMCID: PMC9968108 DOI: 10.3390/ijms24043525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Monoclonal antibody therapies targeting immuno-modulatory targets such as checkpoint proteins, chemokines, and cytokines have made significant impact in several areas, including cancer, inflammatory disease, and infection. However, antibodies are complex biologics with well-known limitations, including high cost for development and production, immunogenicity, a limited shelf-life because of aggregation, denaturation, and fragmentation of the large protein. Drug modalities such as peptides and nucleic acid aptamers showing high-affinity and highly selective interaction with the target protein have been proposed alternatives to therapeutic antibodies. The fundamental limitation of short in vivo half-life has prevented the wide acceptance of these alternatives. Covalent drugs, also known as targeted covalent inhibitors (TCIs), form permanent bonds to target proteins and, in theory, eternally exert the drug action, circumventing the pharmacokinetic limitation of other antibody alternatives. The TCI drug platform, too, has been slow in gaining acceptance because of its potential prolonged side-effect from off-target covalent binding. To avoid the potential risks of irreversible adverse drug effects from off-target conjugation, the TCI modality is broadening from the conventional small molecules to larger biomolecules possessing desirable properties (e.g., hydrolysis resistance, drug-action reversal, unique pharmacokinetics, stringent target specificity, and inhibition of protein-protein interactions). Here, we review the historical development of the TCI made of bio-oligomers/polymers (i.e., peptide-, protein-, or nucleic-acid-type) obtained by rational design and combinatorial screening. The structural optimization of the reactive warheads and incorporation into the targeted biomolecules enabling a highly selective covalent interaction between the TCI and the target protein is discussed. Through this review, we hope to highlight the middle to macro-molecular TCI platform as a realistic replacement for the antibody.
Collapse
Affiliation(s)
- Jay Yang
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
- School of Medicine and Public Health, University of Wisconsin, Madison, WI 53706, USA
- Department of GI Surgery II, Graduate School of Medicine, Hokkaido University, Sapporo 068-8638, Japan
| | - Yudai Tabuchi
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
| | - Riku Katsuki
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
| | - Masumi Taki
- Department of Engineering Science, Graduate School of Informatics and Engineering, University of Electro-Communications (UEC), 1-5-1 Chofugaoka, Chofu 182-8585, Japan
- Institute for Advanced Science, UEC, Chofu 182-8585, Japan
| |
Collapse
|
13
|
García Melián MF, Moreno M, Cerecetto H, Calzada V. Aptamer-Based Immunotheranostic Strategies. Cancer Biother Radiopharm 2023; 38:246-255. [PMID: 36603108 DOI: 10.1089/cbr.2022.0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The escape from immune surveillance is a hallmark of cancer progression. The classic immune checkpoint molecules PD-1, PD-L1, CTLA-4, LAG-3, TIM-3 novel ones are part of a sophisticated system of up- and downmodulation of the immune system, which is unregulated in cancer. In recent years, there have been remarkable advances in the development of targeting strategies, focused principally on immunotherapies aiming at blocking those molecules involved in the evasion of the immune system. However, there are still challenges to predicting their efficacy due to the wide heterogeneity of clinical responses. Thus, there is a need to develop new strategies, and theranostics has much to contribute in this field. Besides that, aptamers have emerged as promising molecules with the potential to generate a huge impact in the immunotheranostic field. They are single-stranded oligonucleotides with a unique self-folding tridimensional structure, with high affinity and specificity for the target. In particular, their small size and physicochemical characteristics make them a versatile tool for designing theranostic strategies. Here, we review the progress in theranostic strategies based on aptamers against immune checkpoints, and highlight the potential of those approaches.
Collapse
Affiliation(s)
- María Fernanda García Melián
- Área de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María Moreno
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Hugo Cerecetto
- Área de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Victoria Calzada
- Área de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
14
|
Immune Checkpoint Inhibitors for Vaccine Improvements: Current Status and New Approaches. Pharmaceutics 2022; 14:pharmaceutics14081721. [PMID: 36015348 PMCID: PMC9415890 DOI: 10.3390/pharmaceutics14081721] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/08/2022] [Accepted: 08/12/2022] [Indexed: 12/11/2022] Open
Abstract
In recent years, the use of immune checkpoint inhibitors (ICIs) in combination with approved or experimental vaccines has proven to be a promising approach to improve vaccine immunogenicity and efficacy. This strategy seeks to overcome the immunosuppressive mechanisms associated with the vaccine response, thereby achieving increased immunogenicity and efficacy. Most of the information on the use of ICIs combined with vaccines derives from studies on certain anti-tumor vaccines combined with monoclonal antibodies (mAbs) against either cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), or programmed death-ligand 1 (PD-L1). However, over the past few years, emerging strategies to use new-generation ICIs as molecular adjuvants are paving the way for future advances in vaccine research. Here, we review the current state and future directions of the use of ICIs in experimental and clinical settings, including mAbs and alternative new approaches using antisense oligonucleotides (ASOs), small non-coding RNAs, aptamers, peptides, and other small molecules for improving vaccine efficacy. The scope of this review mainly includes the use of ICIs in therapeutic antitumor vaccines, although recent research on anti-infective vaccines will also be addressed.
Collapse
|
15
|
Gao F, Yin J, Chen Y, Guo C, Hu H, Su J. Recent advances in aptamer-based targeted drug delivery systems for cancer therapy. Front Bioeng Biotechnol 2022; 10:972933. [PMID: 36051580 PMCID: PMC9424825 DOI: 10.3389/fbioe.2022.972933] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
The past decade has become an important strategy in precision medicine for the targeted therapy of many diseases, expecially various types of cancer. As a promising targeted element, nucleic acid aptamers are single-stranded functional oligonucleotides which have specific abilities to bind with various target molecules ranging from small molecules to entire organisms. They are often named ‘chemical antibody’ and have aroused extensive interest in diverse clinical studies on account of their advantages, such as considerable biostability, versatile chemical modification, low immunogenicity and quick tissue penetration. Thus, aptamer-embedded drug delivery systems offer an unprecedented opportunity in bioanalysis and biomedicine. In this short review, we endeavor to discuss the recent advances in aptamer-based targeted drug delivery platforms for cancer therapy. Some perspectives on the advantages, challenges and opportunities are also presented.
Collapse
Affiliation(s)
- Fei Gao
- Institude of Translation Medicine, Shanghai University, Shanghai, China
| | - Jianhui Yin
- Institude of Translation Medicine, Shanghai University, Shanghai, China
| | - Yan Chen
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, Beijing, China
| | - Changyong Guo
- Institude of Translation Medicine, Shanghai University, Shanghai, China
| | - Honggang Hu
- Institude of Translation Medicine, Shanghai University, Shanghai, China
- *Correspondence: Jiacan Su, ; Honggang Hu,
| | - Jiacan Su
- Institude of Translation Medicine, Shanghai University, Shanghai, China
- *Correspondence: Jiacan Su, ; Honggang Hu,
| |
Collapse
|
16
|
Gao YF, Lu YY, Fan XZ, Wang YH, Tian JH, Saed YA, Li RS, Zhou XS. Blockage of TIM-3 relieves lupus nephritis by expanding Treg cells and promoting their suppressive capacity in MRL/lpr mice. Int Immunopharmacol 2022; 110:108971. [PMID: 35777268 DOI: 10.1016/j.intimp.2022.108971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022]
Abstract
T Cell Immunoglobulin and Mucin Containing Protein-3 (TIM-3) is an important immune checkpoint protein that is expressed in Tregs and affects their function. However, the expression and role of TIM-3 in modulating regulatory T cells (Tregs) in lupus nephritis (LN) are still unknown. In this study, we found that the percentage of TIM-3+ cells among spleen lymphocytes, CD4+ T cells and Tregs was higher in MRL/lpr mice than in MpJ mice. TIM-3high CD4+ T cells and TIM-3high Tregs were mainly responsible for the increase. The percentage of Tregs in TIM-3high CD4+ T cells was lower than that in TIM-3low CD4+ T cells, and the expression of CTLA-4 and IL-10 was lower in TIM-3high Tregs than in the TIM-3low Tregs in MRL/lpr mice. Blockade of TIM-3 in vivo significantly increased the Treg population and the expression of CTLA-4 and IL-10 in Tregs, thus relieving the LN symptoms and pathology in MRL/lpr mice. Additionally, bioinformatics analysis indicated that TIM-3 regulates Treg cells in LN mainly through cytokine-cytokine receptor interactions, the PI3K-Akt signaling pathway, the T cell receptor signaling pathway, Th17 cell differentiation and the FoxO signaling pathway. Together, our study has demonstrated that TIM-3 regulates Tregs in LN and that overexpression of TIM-3 in CD4+ T cells and Tregs leads to Treg quantity and quality deficiency in MRL/lpr mice. Blockade of TIM-3 protects against LN by expanding Tregs and enhancing their suppressive capacity. Finally, TIM-3 might be a potential therapeutic target for the treatment of LN.
Collapse
Affiliation(s)
- Yan-Fang Gao
- Department of Nephrology, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuan-Yue Lu
- Department of Nephrology, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiu-Zhao Fan
- Department of Nephrology, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China; Department of Nephrology, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China
| | - Yan-Hong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ji-Hua Tian
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yasin-Abdi Saed
- Department of Nephrology, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rong-Shan Li
- Department of Nephrology, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China; Department of Nephrology, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China.
| | - Xiao-Shuang Zhou
- Department of Nephrology, The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China; Department of Nephrology, Shanxi Provincial People's Hospital, Shanxi Kidney Disease Institute, Taiyuan, Shanxi, China.
| |
Collapse
|
17
|
Liu H, Huang Y, Huang M, Huang Z, Wang Q, Qing L, Li L, Xu S, Jia B. Current Status, Opportunities, and Challenges of Exosomes in Oral Cancer Diagnosis and Treatment. Int J Nanomedicine 2022; 17:2679-2705. [PMID: 35733418 PMCID: PMC9208818 DOI: 10.2147/ijn.s365594] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/01/2022] [Indexed: 12/13/2022] Open
Abstract
Oral cancer is one of the most common cancers in the world, with more than 300,000 cases diagnosed each year, of which oral squamous cell carcinoma accounts for more than 90%, with a 5-year survival rate of only 40–60%, and poor prognosis. Exploring new strategies for the early diagnosis and treatment of oral cancer is key to improving the survival rate. Exosomes are nanoscale lipid bilayer membrane vesicles that are secreted by almost all cell types. During the development of oral cancer, exosomes can transport their contents (DNA, RNA, proteins, etc) to target cells and promote or inhibit the proliferation, invasion, and metastasis of oral cancer cells by influencing the host immune response, drug-resistant metastasis, and tumour angiogenesis. Therefore, exosomes have great potential and advantages as biomarkers for oral cancer diagnosis, and as drug delivery vehicles or targets for oral cancer therapy. In this review, we first describe the biogenesis, biological functions, and isolation methods of exosomes, followed by their relationship with oral cancer. Here, we focused on the potential of exosomes as oral cancer biomarkers, drug carriers, and therapeutic targets. Finally, we provide an insightful discussion of the opportunities and challenges of exosome application in oral cancer diagnosis and treatment, intending to offer new ideas for the clinical management of oral cancer.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yisheng Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Mingshu Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhijie Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qin Wang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ling Qing
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Li Li
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Bo Jia
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
18
|
Thomas BJ, Porciani D, Burke DH. Cancer immunomodulation using bispecific aptamers. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:894-915. [PMID: 35141049 PMCID: PMC8803965 DOI: 10.1016/j.omtn.2022.01.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Evasion of immune destruction is a major hallmark of cancer. Recent US Food and Drug Administration (FDA) approvals of various immunomodulating therapies underline the important role that reprogramming the immune system can play in combating this disease. However, a wide range of side effects still limit the therapeutic potential of immunomodulators, suggesting a need for more precise reagents with negligible off-target and on-target/off-tumor effects. Aptamers are single-chained oligonucleotides that bind their targets with high specificity and affinity owing to their three-dimensional (3D) structures, and they are one potential way to address this need. In particular, bispecific aptamers (bsApts) have been shown to induce artificial immune synapses that promote T cell activation and subsequent tumor cell lysis in various in vitro and in vivo pre-clinical models. We discuss these advances here, along with gaps in bsApt biology at both the cellular and resident tissue levels that should be addressed to accelerate their translation into the clinic. The broad application, minimal production cost, and relative lack of immunogenicity of bsApts give them some ideal qualities for manipulating the immune system. Building upon lessons from other novel therapies, bsApts could soon provide clinicians with an immunomodulating toolbox that is not only potent and efficacious but exercises a wide therapeutic index.
Collapse
Affiliation(s)
- Brian J. Thomas
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65201, USA
| | - David Porciani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65201, USA
| | - Donald H. Burke
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212, USA
- Bond Life Sciences Center, University of Missouri, Columbia, MO 65201, USA
| |
Collapse
|
19
|
Chen BJ, Zhao JW, Zhang DH, Zheng AH, Wu GQ. Immunotherapy of Cancer by Targeting Regulatory T cells. Int Immunopharmacol 2022; 104:108469. [PMID: 35008005 DOI: 10.1016/j.intimp.2021.108469] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 01/23/2023]
Abstract
Regulatory T (Treg) cells maintain immune homeostasis by inhibiting abnormal/overactive immune responses to both autogenic and nonautogenic antigens. Treg cells play an important role in immune tolerance, autoimmune diseases, infectious diseases, organ transplantation, and tumor diseases. Treg cells have two functional characteristics: T cell anergy and immunosuppression. Treg cells remain immune unresponsive to high concentrations of interleukin-2 and anti-CD3 monoclonal antibodies. In addition, the activation of Treg cells after TCR-mediated signal stimulation inhibits the activation and proliferation of effector T cells. In the process of tumor development, Treg cells accumulate locally in the tumor and lead to tumor escape by inducing anergy and immunosuppression. It is believed that targeted elimination of Treg cells can activate tumor-specific effector T cells and improve the efficiency of cancer immunotherapy. Therefore, inhibition/clearance of Treg cells is a promising strategy for enhancing antitumor immunity. Here, we review studies of cancer immunotherapies targeting Treg cells.
Collapse
Affiliation(s)
- Bo-Jin Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing-Wen Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Da-Hong Zhang
- Department of Urology Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ai-Hong Zheng
- Department of Oncology Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Guo-Qing Wu
- Department of Oncology Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Bailly C, Thuru X, Quesnel B. Modulation of the Gal-9/TIM-3 Immune Checkpoint with α-Lactose. Does Anomery of Lactose Matter? Cancers (Basel) 2021; 13:cancers13246365. [PMID: 34944985 PMCID: PMC8699133 DOI: 10.3390/cancers13246365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The disaccharide lactose is a common excipient in pharmaceutical products. In addition, the two anomers α- and β-lactose can exert immuno-modulatory effects. α-Lactose functions as a major regulator of the T-cell immunoglobulin mucin-3 (Tim-3)/Galectin-9 (Gal-9) immune checkpoint, through direct binding to the β-galactoside-binding lectin galectin-9. The blockade of TIM-3 with monoclonal antibodies or small molecules represents a promising approach to combat onco-hematological diseases, in particular myelodysplastic syndromes, and acute myeloid leukemia. Alternatively, the activity of the checkpoint can be modulated via targeting of Gal-9 with both α- and β-lactose. In fact, lactose is a quasi-pan-galectin ligand, capable of modulating the functions of most of the 16 galectin molecules. This review discusses the capacity of lactose and Gal-9 to modulate the TIM-3/Gal-9 and PD-1/PD-L1 immune checkpoints in oncology. The immuno-regulatory roles of lactose and Gal-9 are highlighted. Abstract The disaccharide lactose is an excipient commonly used in pharmaceutical products. The two anomers, α- and β-lactose (α-L/β-L), differ by the orientation of the C-1 hydroxyl group on the glucose unit. In aqueous solution, a mutarotation process leads to an equilibrium of about 40% α-L and 60% β-L at room temperature. Beyond a pharmaceutical excipient in solid products, α-L has immuno-modulatory effects and functions as a major regulator of TIM-3/Gal-9 immune checkpoint, through direct binding to the β-galactoside-binding lectin galectin-9. The blockade of the co-inhibitory checkpoint TIM-3 expressed on T cells with anti-TIM-3 antibodies represents a promising approach to combat different onco-hematological diseases, in particular myelodysplastic syndromes and acute myeloid leukemia. In parallel, the discovery and development of anti-TIM-3 small molecule ligands is emerging, including peptides, RNA aptamers and a few specifically designed heterocyclic molecules. An alternative option consists of targeting the different ligands of TIM-3, notably Gal-9 recognized by α-lactose. Modulation of the TIM-3/Gal-9 checkpoint can be achieved with both α- and β-lactose. Moreover, lactose is a quasi-pan-galectin ligand, capable of modulating the functions of most of the 16 galectin molecules. The present review provides a complete analysis of the pharmaceutical and galectin-related biological functions of (α/β)-lactose. A focus is made on the capacity of lactose and Gal-9 to modulate both the TIM-3/Gal-9 and PD-1/PD-L1 immune checkpoints in oncology. Modulation of the TIM-3/Gal-9 checkpoint is a promising approach for the treatment of cancers and the role of lactose in this context is discussed. The review highlights the immuno-regulatory functions of lactose, and the benefit of the molecule well beyond its use as a pharmaceutical excipient.
Collapse
Affiliation(s)
- Christian Bailly
- OncoWitan, Scientific Consulting Office, 59290 Lille, France
- Correspondence:
| | - Xavier Thuru
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020—UMR1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France; (X.T.); (B.Q.)
| | - Bruno Quesnel
- University of Lille, CNRS, Inserm, CHU Lille, UMR9020—UMR1277—Canther—Cancer Heterogeneity, Plasticity and Resistance to Therapies, 59000 Lille, France; (X.T.); (B.Q.)
| |
Collapse
|
21
|
Wan Q, Liu X, Zu Y. Oligonucleotide aptamers for pathogen detection and infectious disease control. Theranostics 2021; 11:9133-9161. [PMID: 34522231 PMCID: PMC8419047 DOI: 10.7150/thno.61804] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/04/2021] [Indexed: 12/21/2022] Open
Abstract
During an epidemic or pandemic, the primary task is to rapidly develop precise diagnostic approaches and effective therapeutics. Oligonucleotide aptamer-based pathogen detection assays and control therapeutics are promising, as aptamers that specifically recognize and block pathogens can be quickly developed and produced through simple chemical synthesis. This work reviews common aptamer-based diagnostic techniques for communicable diseases and summarizes currently available aptamers that target various pathogens, including the SARS-CoV-2 virus. Moreover, this review discusses how oligonucleotide aptamers might be leveraged to control pathogen propagation and improve host immune system responses. This review offers a comprehensive data source to the further develop aptamer-based diagnostics and therapeutics specific for infectious diseases.
Collapse
Affiliation(s)
| | | | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
22
|
Ștefan G, Hosu O, De Wael K, Lobo-Castañón MJ, Cristea C. Aptamers in biomedicine: Selection strategies and recent advances. Electrochim Acta 2021. [DOI: 10.1016/j.electacta.2021.137994] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Emrani S, Lamar M, Price CC, Baliga S, Wasserman V, Matusz E, Swenson R, Baliga G, Libon DJ. Assessing the capacity for mental manipulation in patients with statically-determined mild cognitive impairment using digital technology. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aims: Prior research employing a standard backward digit span test has been successful in operationally defining neurocognitive constructs associated with the Fuster’s model of executive attention. The current research sought to test if similar behavior could be obtained using a cross-modal mental manipulation test.
Methods: Memory clinic patients were studied. Using Jak-Bondi criteria, 24 patients were classified with mild cognitive impairment (MCI), and 33 memory clinic patients did not meet criteria for MCI (i.e. non-MCI). All patients were assessed with the digital version of the WRAML-2 Symbolic Working Memory Test-Part 1, a cross-modal mental manipulation task where patients hear digits, but respond by touching digits from lowest to highest on an answer key. Only 4 and 5-span trials were analyzed. Using an iPad, all test stimuli were played; and, all responses were obtained with a touch key. Only correct trials were analyzed. Average time to complete trials and latency for each digit was recorded.
Results: Groups did not differ when average time to complete 4-span trials was calculated. MCI patients displayed slower latency, or required more time to re-order the 1st and 3rd digits. Regression analyses, primarily involving initial and latter response latencies, were associated with better, but different underlying neuropsychological abilities. Almost no 5-span analyses were significant.
Conclusions: This cross-modal test paradigm found no difference for total average time. MCI patients generated slower 1st and 3rd response latency, suggesting differences in time allocation to achieve correct serial order recall. Moreover, different neuropsychological abilities were associated with different time-based test components. These data extend prior findings using a standard backward digit span test. Differences in time epochs are consistent with constructs underlying the model of executive attention and help explain mental manipulation deficits in MCI. These latency measures could constitute neurocognitive biomarkers that track emergent disease.
Collapse
Affiliation(s)
- Sheina Emrani
- Department of Psychology, Rowan University, Glassboro, NJ 08028, USA
| | - Melissa Lamar
- Department of Behavioral Sciences and the Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Catherine C. Price
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL 32610, USA
| | - Satya Baliga
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Victor Wasserman
- Department of Psychology, Rowan University, Glassboro, NJ 08028, USA
| | - Emily Matusz
- 5New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Rod Swenson
- Department Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, Grand Fork, Fargo, ND 58103, USA
| | - Ganesh Baliga
- Department of Computer Science, Rowan University, Glassboro, NJ 08028, USA
| | - David J. Libon
- Department of Psychology, Rowan University, Glassboro, NJ 08028, USA 5New Jersey Institute for Successful Aging, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| |
Collapse
|
24
|
Agnello L, Camorani S, Fedele M, Cerchia L. Aptamers and antibodies: rivals or allies in cancer targeted therapy? EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:107-121. [PMID: 36046085 PMCID: PMC9400792 DOI: 10.37349/etat.2021.00035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/01/2021] [Indexed: 12/29/2022] Open
Abstract
The goal of an efficacious cancer therapy is to specifically target diseased cells at high accuracy while sparing normal, healthy cells. Over the past three decades, immunotherapy, based on the use of monoclonal antibodies (mAbs) directed against tumor-associated antigens, to inhibit their oncogenic function, or against immune checkpoints, to modulate specific T cell responses against cancer, has proven to be an important strategy for cancer therapy. Nevertheless, the number of mAbs approved for clinical use is still limited because of significant drawbacks to their applicability. Oligonucleotide aptamers, similarly to antibodies, form high-affinity bonds with their specific protein targets, thus representing an effective tool for active cancer targeting. Compared to antibodies, aptamers’ use as therapeutic agents benefits from their low size, low/no immunogenicity, simple synthesis and design flexibility for improving efficacy and stability. This review intends to highlight recently emerged applications of aptamers as recognition elements, from biomarker discovery to targeted drug delivery and targeted treatment, showing aptamers’ potential to work in conjunction with antibodies for attacking cancer from multiple flanks.
Collapse
Affiliation(s)
- Lisa Agnello
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| | - Simona Camorani
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| | - Laura Cerchia
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore”, National Research Council (CNR), Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
25
|
Zhou J, Wang W, Li Q. Potential therapeutic targets in the tumor microenvironment of hepatocellular carcinoma: reversing the protumor effect of tumor-associated macrophages. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:73. [PMID: 33596985 PMCID: PMC7890827 DOI: 10.1186/s13046-021-01873-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
In hepatocellular carcinoma patients, due to the microenvironmental specificity of liver, the tumor microenvironment exhibits high immunosuppression and drug resistance, resulting in excessive or insufficient responses to immunotherapy. The dynamic interactions between tumor cells and immune modulators in the TME significantly impact the occurrence and development of tumors, efficacy, and drug resistance, which can create a much more positive response to immunotherapy. Moreover, with the wide application of single-cell sequencing technology in the TME, increasing evidence shows an interaction network among cells. Sequencing results suggest that specific tumor-associated macrophages are a hub node, connecting different cell populations in the cell interaction network, and can could regulate tumor generation and antitumor immunity. This review focused on therapeutic targets that could be targeted to remodel the tumor microenvironment and reprogram the tumor-associated macrophage phenotype in hepatocellular carcinoma patients, thereby improving immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Jingyi Zhou
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, People's Republic of China
| | - Weiyu Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, People's Republic of China
| | - Qi Li
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
26
|
Saleh R, Toor SM, Elkord E. Targeting TIM-3 in solid tumors: innovations in the preclinical and translational realm and therapeutic potential. Expert Opin Ther Targets 2020; 24:1251-1262. [PMID: 33103506 DOI: 10.1080/14728222.2020.1841750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have shown a great therapeutic efficacy in cancer patients. However, a significant proportion of cancer patients remain unresponsive or show limited response. T cell immunoglobulin and mucin-domain containing protein-3 (TIM-3) is a co-inhibitory receptor expressed on various cell types and is involved in the attenuation of immune responses. TIM-3 and its ligands are highly expressed in various solid malignancies and some studies have reported its association with worse disease outcomes. Thus, targeting TIM-3 could be a promising therapeutic approach to treat cancer patients. AREAS COVERED This review describes the role of TIM-3 and its ligands in regulating anti-tumor immunity and their contribution to cancer progression. Moreover, this review focuses on the preclinical models and translational data from important studies published in PubMed till October 2020, which demonstrate the therapeutic benefits of targeting TIM-3 signaling. EXPERT OPINION Despite the promising data obtained from targeting TIM-3 in preclinical models, precise mechanisms underlying the anti-tumor effects of TIM-3 inhibition are not fully elucidated. Therefore, mechanistic studies are required to provide better insights into the anti-tumor effects of targeting TIM-3, and clinical data are necessary to determine the safety profiles and therapeutic efficacy of TIM-3 inhibition in cancer patients.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Eyad Elkord
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford , Manchester, United Kingdom
| |
Collapse
|
27
|
Role of extracellular vesicles in tumour microenvironment. Cell Commun Signal 2020; 18:163. [PMID: 33081785 PMCID: PMC7574205 DOI: 10.1186/s12964-020-00643-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/10/2020] [Indexed: 12/16/2022] Open
Abstract
In recent years, it has been demonstrated that extracellular vesicles (EVs) can be released by almost all cell types, and detected in most body fluids. In the tumour microenvironment (TME), EVs serve as a transport medium for lipids, proteins, and nucleic acids. EVs participate in various steps involved in the development and progression of malignant tumours by initiating or suppressing various signalling pathways in recipient cells. Although tumour-derived EVs (T-EVs) are known for orchestrating tumour progression via systemic pathways, EVs from non-malignant cells (nmEVs) also contribute substantially to malignant tumour development. Tumour cells and non-malignant cells typically communicate with each other, both determining the progress of the disease. In this review, we summarise the features of both T-EVs and nmEVs, tumour progression, metastasis, and EV-mediated chemoresistance in the TME. The physiological and pathological effects involved include but are not limited to angiogenesis, epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) remodelling, and immune escape. We discuss potential future directions of the clinical application of EVs, including diagnosis (as non-invasive biomarkers via liquid biopsy) and therapeutic treatment. This may include disrupting EV biogenesis and function, thus utilising the features of EVs to repurpose them as a therapeutic tool in immunotherapy and drug delivery systems. We also discuss the overall findings of current studies, identify some outstanding issues requiring resolution, and propose some potential directions for future research. Video abstract.
Collapse
|
28
|
Tran PHL, Xiang D, Tran TTD, Yin W, Zhang Y, Kong L, Chen K, Sun M, Li Y, Hou Y, Zhu Y, Duan W. Exosomes and Nanoengineering: A Match Made for Precision Therapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904040. [PMID: 31531916 DOI: 10.1002/adma.201904040] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/02/2019] [Indexed: 05/28/2023]
Abstract
Targeted exosomal delivery systems for precision nanomedicine attract wide interest across areas of molecular cell biology, pharmaceutical sciences, and nanoengineering. Exosomes are naturally derived 50-150 nm nanovesicles that play important roles in cell-to-cell and/or cell-to-tissue communications and cross-species communication. Exosomes are also a promising class of novel drug delivery vehicles owing to their ability to shield their payload from chemical and enzymatic degradations as well as to evade recognition by and subsequent removal by the immune system. Combined with a new class of affinity ligands known as aptamers or chemical antibodies, molecularly targeted exosomes are poised to become the next generation of smartly engineered nanovesicles for precision medicine. Here, recent advances in targeted exosomal delivery systems engineered by aptamer for future strategies to promote human health using this class of human-derived nanovesicles are summarized.
Collapse
Affiliation(s)
- Phuong H L Tran
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
| | - Dongxi Xiang
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, 77 Avenue Louise Pasteur, Boston, MA, 02115, USA
| | - Thao T D Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Wang Yin
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
| | - Yumei Zhang
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
| | - Lingxue Kong
- Institute for Frontier Materials, Deakin University, Geelong, Victoria, 3216, Australia
| | - Kuisheng Chen
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, He'nan Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Miaomiao Sun
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, He'nan Key Laboratory of Tumor Pathology, Zhengzhou, 450052, China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, and St George and Sutherland Clinical School, University of New South Wales, Kensington, NSW, 2217, Australia
| | - Yingchun Hou
- Laboratory of Tumor Molecular and Cellular Biology, College of Life Sciences, Shaanxi Normal University, 620 West Chang'an Avenue, Xi'an, Shaanxi, 710119, China
| | - Yimin Zhu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Geelong, Victoria, 3216, Australia
- GenePharma-Deakin Joint Laboratory of Aptamer Medicine, Suzhou, 215123, China
- GenePharma-Deakin Joint Laboratory of Aptamer Medicine, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
29
|
Cui X, Song M, Liu Y, Yuan Y, Huang Q, Cao Y, Lu F. Identifying conformational changes of aptamer binding to theophylline: A combined biolayer interferometry, surface-enhanced Raman spectroscopy, and molecular dynamics study. Talanta 2020; 217:121073. [PMID: 32498900 DOI: 10.1016/j.talanta.2020.121073] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 11/20/2022]
Abstract
Theophylline is a potent bronchodilator for the treatment of asthma, bronchitis, and emphysema. Its narrow therapeutic window (20-100 μM) demands that the blood concentration of theophylline be monitored carefully, which can be achieved by aptamer capture. Thus, an understanding of what occurs when aptamers bind to theophylline is critical for identifying a high-affinity and high-specificity aptamer, which improve the sensitivity and selectivity of theophylline detection. Consequently, there is an urgent need to develop a simple, convenient, and nondestructive method to monitor conformational changes during the binding process. Here, we report the determination of the affinity of a selected aptamer and theophylline via biolayer interferometry (BLI) experiments. Additionally, using surface-enhanced Raman spectroscopy (SERS), the conformational changes on theophylline-aptamer binding were identified from differences in the SER spectra. Finally, molecular dynamics (MD) simulations were used to identify the specific conformational changes of the aptamer during the binding process. Such a combined BLI-SERS-MD method provides an in-depth understanding of the theophylline-aptamer binding processes and a comprehensive explanation for conformational changes, which helps to select, design, and modify an aptamer with high affinity and specificity. It can also be used as a scheme for the study of other aptamer-ligand interactions, which can be applied to the detection, sensing, clinical diagnosis, and treatment of diseases.
Collapse
Affiliation(s)
- Xiaolin Cui
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Menghua Song
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yan Liu
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Yifan Yuan
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China
| | - Qiang Huang
- State Key Laboratory of Genetic Engineering, MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Yongbing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200082, China
| | - Feng Lu
- School of Pharmacy, Second Military Medical University, Shanghai, 200433, China; Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Shanghai, 200433, China.
| |
Collapse
|
30
|
Fu Z, Xiang J. Aptamers, the Nucleic Acid Antibodies, in Cancer Therapy. Int J Mol Sci 2020; 21:ijms21082793. [PMID: 32316469 PMCID: PMC7215806 DOI: 10.3390/ijms21082793] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/09/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
The arrival of the monoclonal antibody (mAb) technology in the 1970s brought with it the hope of conquering cancers to the medical community. However, mAbs, on the whole, did not achieve the expected wonder in cancer therapy although they do have demonstrated successfulness in the treatment of a few types of cancers. In 1990, another technology of making biomolecules capable of specific binding appeared. This technique, systematic evolution of ligands by exponential enrichment (SELEX), can make aptamers, single-stranded DNAs or RNAs that bind targets with high specificity and affinity. Aptamers have some advantages over mAbs in therapeutic uses particularly because they have little or no immunogenicity, which means the feasibility of repeated use and fewer side effects. In this review, the general properties of the aptamer, the advantages and limitations of aptamers, the principle and procedure of aptamer production with SELEX, particularly the undergoing studies in aptamers for cancer therapy, and selected anticancer aptamers that have entered clinical trials or are under active investigations are summarized.
Collapse
Affiliation(s)
- Zhaoying Fu
- Department of Biochemistry and Molecular Biology, College of Medicine, Yanan University, Yanan 716000, China
- Correspondence: (Z.F.); (J.X.)
| | - Jim Xiang
- Division of Oncology, University of Saskatchewan, Saskatoon, SA S7N 4H4, Canada
- Correspondence: (Z.F.); (J.X.)
| |
Collapse
|
31
|
Abstract
Cancer immunotherapy has shown great potential as witnessed by an increasing number of immuno-oncology drug approvals in the past few years. Meanwhile, the field of nucleic acid therapeutics has made significant advancement. Nucleic acid therapeutics, such as plasmids, antisense oligonucleotides (ASO), small interfering RNA (siRNA) and microRNA, messenger RNA (mRNA), immunomodulatory DNA/RNA, and gene-editing guide RNA (gRNA) are attractive due to their versatile abilities to alter the expression of target endogenous genes or even synthetic genes, and modulate the immune responses. These abilities can play vital roles in the development of novel immunotherapy strategies. However, limited by the intrinsic physicochemical properties such as negative charges, hydrophilicity, as well as susceptibility to enzymatic degradation, the delivery of nucleic acid therapeutics faces multiple challenges. It is therefore pivotal to develop drug delivery systems that can carry, protect, and specifically deliver and release nucleic acid therapeutics to target tissues and cells. In this review, we attempted to summarize recent advances in nucleic acid therapeutics and the delivery systems for these therapeutics in cancer immunotherapy.
Collapse
Affiliation(s)
- Shurong Zhou
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Wenjie Chen
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Janet Cole
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| | - Guizhi Zhu
- Department of Pharmaceutics, Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology, Drug Discovery and Development (ISB3D), School of Pharmacy, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23219, USA
| |
Collapse
|
32
|
Park M, Kang KW. Phosphatidylserine receptor-targeting therapies for the treatment of cancer. Arch Pharm Res 2019; 42:617-628. [PMID: 31172440 DOI: 10.1007/s12272-019-01167-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/01/2019] [Indexed: 02/07/2023]
Abstract
Asymmetric distribution of phospholipids across the plasma membrane is a unique characteristic of eukaryotic cells. Phosphatidylcholine and sphingomyelin are exposed in the outer leaflet, and phosphatidylserine (PS) is predominantly located in the inner leaflet. Redistribution of PS to the cell surface can be observed in several physiological conditions, such as apoptosis and platelet activation, or in pathological conditions, such as the release of microvesicles/exosomes from tumor tissues. PS binding to the phosphatidylserine receptor (PSR) on immune cells initiates immunosuppressive pathways that can lead to immune evasion by cancer cells. Conversely, PSR activation of cancer cells plays an important role in their survival, proliferation and metastasis. Herein, we briefly summarize both recent advances in our understanding of the pathological roles of PS and its receptor in cancer biology, as well as relevant pharmacological approaches.
Collapse
Affiliation(s)
- Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
33
|
Yang S, Wen J, Li H, Xu L, Liu Y, Zhao N, Zeng Z, Qi J, Jiang W, Han W, Zu Y. Aptamer-Engineered Natural Killer Cells for Cell-Specific Adaptive Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1900903. [PMID: 31026116 PMCID: PMC6541510 DOI: 10.1002/smll.201900903] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/04/2019] [Indexed: 05/28/2023]
Abstract
Natural killer (NK) cells are a key component of the innate immune system as they can attack cancer cells without prior sensitization. However, due to lack of cell-specific receptors, NK cells are not innately able to perform targeted cancer immunotherapy. Aptamers are short single-stranded oligonucleotides that specifically recognize their targets with high affinity in a similar manner to antibodies. To render NK cells with target-specificity, synthetic CD30-specific aptamers are anchored on cell surfaces to produce aptamer-engineered NK cells (ApEn-NK) without genetic alteration or cell damage. Under surface-anchored aptamer guidance, ApEn-NK specifically bind to CD30-expressing lymphoma cells but do not react to off-target cells. The resulting specific cell binding of ApEn-NK triggers higher apoptosis/death rates of lymphoma cells compared to parental NK cells. Additionally, experiments with primary human NK cells demonstrate the potential of ApEn-NK to specifically target and kill lymphoma cells, thus presenting a potential new approach for targeted immunotherapy by NK cells.
Collapse
Affiliation(s)
- Shuanghui Yang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jianguo Wen
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Huan Li
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ling Xu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yanting Liu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Nianxi Zhao
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Zihua Zeng
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Jianjun Qi
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Wenqi Jiang
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Wei Han
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Hematology, First Hospital of Jilin University, Changchun 130021, Jilin, China
| | - Youli Zu
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
34
|
Soldevilla MM, Meraviglia-Crivelli de Caso D, Menon AP, Pastor F. Aptamer-iRNAs as Therapeutics for Cancer Treatment. Pharmaceuticals (Basel) 2018; 11:E108. [PMID: 30340426 PMCID: PMC6315413 DOI: 10.3390/ph11040108] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Aptamers are single-stranded oligonucleotides (ssDNA or ssRNA) that bind and recognize their targets with high affinity and specificity due to their complex tertiary structure. Aptamers are selected by a method called SELEX (Systematic Evolution of Ligands by EXponential enrichment). This method has allowed the selection of aptamers to different types of molecules. Since then, many aptamers have been described for the potential treatment of several diseases including cancer. It has been described over the last few years that aptamers represent a very useful tool as therapeutics, especially for cancer therapy. Aptamers, thanks to their intrinsic oligonucleotide nature, present inherent advantages over other molecules, such as cell-based products. Owing to their higher tissue penetrability, safer profile, and targeting capacity, aptamers are likely to become a novel platform for the delivery of many different types of therapeutic cargos. Here we focus the review on interfering RNAs (iRNAs) as aptamer-based targeting delivered agents. We have gathered the most reliable information on aptamers as targeting and carrier agents for the specific delivery of siRNAs, shRNA, microRNAs, and antisense oligonucleotides (ASOs) published in the last few years in the context of cancer therapy.
Collapse
Affiliation(s)
- Mario M Soldevilla
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| | - Daniel Meraviglia-Crivelli de Caso
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| | - Ashwathi P Menon
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| | - Fernando Pastor
- Molecular Therapy Program, Aptamer Core, Center for the Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain.
- Navarre Health Research Institute (IdiSNA), 31008 Pamplona, Spain.
| |
Collapse
|
35
|
Liu F, Liu Y, Chen Z. Tim-3 expression and its role in hepatocellular carcinoma. J Hematol Oncol 2018; 11:126. [PMID: 30309387 PMCID: PMC6182863 DOI: 10.1186/s13045-018-0667-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/19/2018] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common tumors in the world, and its mortality is still on the rise. Limited treatments and low chemotherapy sensitivity of HCC make new therapeutic strategies urgently needed. With the rise of immune checkpoint blockade, anti-CTLA-4 antibodies and anti-PD-1 antibodies have shown therapeutic effects in various tumors. T cell immunoglobulin mucin-3 (Tim-3), a newly discovered immune checkpoint molecule, plays a major role in the development of HCC. Tim-3 can be used to evaluate the prognosis and therapeutic effects in HCC, and Tim-3 intervention has shown anti-tumor effects in preclinical experiments. This review summarizes findings regarding Tim-3 and HCC in recent years and discusses the rationale of Tim-3 as a therapeutic target for HCC.
Collapse
Affiliation(s)
- Feifei Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Yanning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China
| | - Zhi Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79# Qingchun Road, 6A-17, Hangzhou, 310003, China.
| |
Collapse
|
36
|
Pastor F, Berraondo P, Etxeberria I, Frederick J, Sahin U, Gilboa E, Melero I. An RNA toolbox for cancer immunotherapy. Nat Rev Drug Discov 2018; 17:751-767. [DOI: 10.1038/nrd.2018.132] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
37
|
Zhu G, Chen X. Aptamer-based targeted therapy. Adv Drug Deliv Rev 2018; 134:65-78. [PMID: 30125604 PMCID: PMC6239901 DOI: 10.1016/j.addr.2018.08.005] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 07/12/2018] [Accepted: 08/16/2018] [Indexed: 12/13/2022]
Abstract
Precision medicine holds great promise to harness genetic and epigenetic cues for targeted treatment of a variety of diseases, ranging from many types of cancers, neurodegenerative diseases, to cardiovascular diseases. The proteomic profiles resulting from the unique genetic and epigenetic signatures represent a class of relatively well accessible molecular targets for both interrogation (e.g., diagnosis, prognosis) and intervention (e.g., targeted therapy) of these diseases. Aptamers are promising for such applications by specific binding with cognate disease biomarkers. Nucleic acid aptamers are a class of DNA or RNA with unique three-dimensional conformations that allow them to specifically bind with target molecules. Aptamers can be relatively easily screened, reproducibly manufactured, programmably designed, and chemically modified for various biomedical applications, including targeted therapy. Aptamers can be chemically modified to resist enzymatic degradation or optimize their pharmacological behaviors, which ensured their chemical integrity and bioavailability under physiological conditions. In this review, we will focus on recent progress and discuss the challenges and opportunities in the research areas of aptamer-based targeted therapy in the forms of aptamer therapeutics and aptamer-drug conjugates (ApDCs).
Collapse
Affiliation(s)
- Guizhi Zhu
- Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Granier C, Gey A, Dariane C, Mejean A, Timsit MO, Blanc C, Verkarre V, Radulescu C, Fabre E, Vano Y, Oudard S, Badoual C, Tartour É. [Tim-3: a novel biomarker and therapeutic target in oncology]. Med Sci (Paris) 2018; 34:231-237. [PMID: 29547109 DOI: 10.1051/medsci/20183403011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
T cells harboring multiple co-inhibitory molecules lose their anti-tumoral functionality. PD-1 is a clinically approved target in cancer therapy, but its expression alone does not mean dysfunctionality. The expression of Tim-3 on numerous cell types (T cell, Treg, dendritic cell, myeloid cells) favors tumor escape to immune cells. Within many tumors, PD-1/Tim-3 coexpressing CD8-T cells lose their ability to secrete cytokines (IFNγ, IL-2, TNFα) and their intratumoral infiltration correlates with a bad prognosis. Tim-3 recently appeared as a potential biomarker of anti-PD-1 resistance. Combined blockade of PD-1 and Tim-3 axis demonstrated potent clinical efficacy in preclinical models and reinforced the rationale of using an anti-Tim-3 to override tumor resistance.
Collapse
Affiliation(s)
- Clémence Granier
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France
| | - Alain Gey
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Hôpital européen Georges Pompidou, service d'immunologie biologique, 20, rue Leblanc, 75015 Paris, France
| | - Charles Dariane
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France - Hôpital européen Georges Pompidou, Service de chirurgie urologique, APHP, Paris, France
| | - Arnaud Mejean
- Hôpital européen Georges Pompidou, Service de chirurgie urologique, APHP, Paris, France
| | - Marc-Olivier Timsit
- Hôpital européen Georges Pompidou, Service de chirurgie urologique, APHP, Paris, France
| | - Charlotte Blanc
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France
| | - Virginie Verkarre
- Hôpital européen Georges Pompidou, Service d'anatomie pathologique, APHP, Paris, France
| | | | - Elisabeth Fabre
- Hôpital européen Georges Pompidou, Service d'oncologie médicale, APHP Paris, France
| | - Yann Vano
- Hôpital européen Georges Pompidou, Service d'oncologie médicale, APHP Paris, France - Inserm U1138, université Paris Descartes Sorbonne Paris-Cité, Paris, France
| | - Stéphane Oudard
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Hôpital européen Georges Pompidou, Service d'oncologie médicale, APHP Paris, France
| | - Cécile Badoual
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France - Hôpital européen Georges Pompidou, Service d'anatomie pathologique, APHP, Paris, France
| | - Éric Tartour
- Inserm U970, université Paris Descartes Sorbonne Paris-Cité, Paris, France - Équipe labellisée Ligue contre le cancer, Paris, France - Hôpital européen Georges Pompidou, service d'immunologie biologique, 20, rue Leblanc, 75015 Paris, France
| |
Collapse
|
39
|
Soldevilla MM, Hervas S, Villanueva H, Lozano T, Rabal O, Oyarzabal J, Lasarte JJ, Bendandi M, Inoges S, López-Díaz de Cerio A, Pastor F. Identification of LAG3 high affinity aptamers by HT-SELEX and Conserved Motif Accumulation (CMA). PLoS One 2017; 12:e0185169. [PMID: 28934318 PMCID: PMC5608357 DOI: 10.1371/journal.pone.0185169] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/07/2017] [Indexed: 12/11/2022] Open
Abstract
LAG3 receptor belongs to a family of immune-checkpoints expressed in T lymphocytes and other cells of the immune system. It plays an important role as a rheostat of the immune response. Focus on this receptor as a potential therapeutic target in cancer immunotherapy has been underscored after the success of other immune-checkpoint blockade strategies in clinical trials. LAG3 showcases the interest in the field of autoimmunity as several studies show that LAG3-targeting antibodies can also be used for the treatment of autoimmune diseases. In this work we describe the identification of a high-affinity LAG3 aptamer by High Throughput Sequencing SELEX in combination with a study of potential conserved binding modes according to sequence conservation by using 2D-structure prediction and 3D-RNA modeling using Rosetta. The aptamer with the highest accumulation of these conserved sequence motifs displays the highest affinity to LAG3 recombinant soluble proteins and binds to LAG3-expressing lymphocytes. The aptamer described herein has the potential to be used as a therapeutic agent, as it enhances the threshold of T-cell activation. Nonetheless, in future applications, it could also be engineered for treatment of autoimmune diseases by target depletion of LAG3-effector T lymphocytes.
Collapse
Affiliation(s)
- Mario Martínez Soldevilla
- Aptamer Platform, Molecular Therapeutics Program, Center for Applied Medical Research, (CIMA), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Sandra Hervas
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
- Program Immunology and Immunotherapy, Center for Applied Medical Research, (CIMA), University of Navarra, Pamplona, Spain
| | - Helena Villanueva
- Aptamer Platform, Molecular Therapeutics Program, Center for Applied Medical Research, (CIMA), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Teresa Lozano
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
- Program Immunology and Immunotherapy, Center for Applied Medical Research, (CIMA), University of Navarra, Pamplona, Spain
| | - Obdulia Rabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Julen Oyarzabal
- Small Molecule Discovery Platform, Molecular Therapeutics Program, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Juan José Lasarte
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
- Program Immunology and Immunotherapy, Center for Applied Medical Research, (CIMA), University of Navarra, Pamplona, Spain
| | - Maurizio Bendandi
- Section on Hematology/Oncology, Department of Internal Medicine, Comprehensive Cancer Center, Wake Forest University Baptist Healthcare Center, Winston-Salem, NC, United States of America
- Section of Hematology/Oncology, Department of Internal Medicine, W.G Hefner VA Medical Center, Salisbury/Charlotte, NC, United States of America
| | - Susana Inoges
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
- Department of Immunology and Immunotherapy, University Clinic of Navarra, Pamplona, Spain
| | - Ascensión López-Díaz de Cerio
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
- Department of Immunology and Immunotherapy, University Clinic of Navarra, Pamplona, Spain
| | - Fernando Pastor
- Aptamer Platform, Molecular Therapeutics Program, Center for Applied Medical Research, (CIMA), Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra (IDISNA), Recinto de Complejo Hospitalario de Navarra, Pamplona, Spain
| |
Collapse
|