1
|
Graham LV, Fisher JG, Doyle ADP, Sale B, Del Rio L, French AJE, Mayor NP, Turner TR, Marsh SGE, Cragg MS, Forconi F, Khakoo SI, Blunt MD. KIR2DS2+ NK cells in cancer patients demonstrate high activation in response to tumour-targeting antibodies. Front Oncol 2024; 14:1404051. [PMID: 39286025 PMCID: PMC11402612 DOI: 10.3389/fonc.2024.1404051] [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: 03/20/2024] [Accepted: 08/13/2024] [Indexed: 09/19/2024] Open
Abstract
Strategies to mobilise natural killer (NK) cells against cancer include tumour-targeting antibodies, NK cell engagers (NKCEs) and the adoptive transfer of ex vivo expanded healthy donor-derived NK cells. Genetic and functional studies have revealed that expression of the activating killer immunoglobulin-like receptor KIR2DS2 is associated with enhanced function in NK cells from healthy donors and improved outcome in several different malignancies. The optimal strategy to leverage KIR2DS2+ NK cells therapeutically is however currently unclear. In this study, we therefore evaluated the response of KIR2DS2-expressing NK cells to activation against cancer with clinically relevant tumour-targeting antibodies and following ex vivo expansion. We identified that KIR2DS2high NK cells from patients with chronic lymphocytic leukaemia and hepatocellular carcinoma had enhanced activation in response to tumour-targeting antibodies compared to KIR2DS2- NK cells. However, the superior function of healthy donor derived KIR2DS2high NK cells was lost following ex vivo expansion which is required for adoptive transfer-based therapeutic strategies. These data provide evidence that targeting KIR2DS2 directly in cancer patients may allow for the utilisation of their enhanced effector function, however such activity may be lost following their ex vivo expansion.
Collapse
Affiliation(s)
- Lara V Graham
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Jack G Fisher
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Amber D P Doyle
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Ben Sale
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Luis Del Rio
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Albert J E French
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
| | - Neema P Mayor
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- Department of Academic Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Thomas R Turner
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- Department of Academic Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London, United Kingdom
- Department of Academic Haematology, University College London (UCL) Cancer Institute, London, United Kingdom
| | - Mark S Cragg
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Francesco Forconi
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
- Haematology Department, Cancer Care Directorate, University Hospital Southampton National Health Service (NHS) Trust, Southampton, United Kingdom
| | - Salim I Khakoo
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| | - Matthew D Blunt
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
2
|
Blunt MD, Fisher H, Schittenhelm RB, Mbiribindi B, Fulton R, Khan S, Espana-Serrano L, Graham LV, Bastidas-Legarda L, Burns D, Khakoo SM, Mansour S, Essex JW, Ayala R, Das J, Purcell AW, Khakoo SI. The nuclear export protein XPO1 provides a peptide ligand for natural killer cells. SCIENCE ADVANCES 2024; 10:eado6566. [PMID: 39178254 PMCID: PMC11343027 DOI: 10.1126/sciadv.ado6566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024]
Abstract
XPO1 (Exportin-1/CRM1) is a nuclear export protein that is frequently overexpressed in cancer and functions as a driver of oncogenesis. Currently small molecules that target XPO1 are being used in the clinic as anticancer agents. We identify XPO1 as a target for natural killer (NK) cells. Using immunopeptidomics, we have identified a peptide derived from XPO1 that can be recognized by the activating NK cell receptor KIR2DS2 in the context of human leukocyte antigen-C. The peptide can be endogenously processed and presented to activate NK cells specifically through this receptor. Although high XPO1 expression in cancer is commonly associated with a poor prognosis, we show that the outcome of specific cancers, such as hepatocellular carcinoma, can be substantially improved if there is concomitant evidence of NK cell infiltration. We thus identify XPO1 as a bona fide tumor antigen recognized by NK cells that offers an opportunity for a personalized approach to NK cell therapy for solid tumors.
Collapse
Affiliation(s)
- Matthew D. Blunt
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hayden Fisher
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Ralf B. Schittenhelm
- Monash Proteomics & Metabolomics Platform, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Berenice Mbiribindi
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Rebecca Fulton
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sajida Khan
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Laura Espana-Serrano
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Lara V. Graham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Leidy Bastidas-Legarda
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
- Monash Proteomics & Metabolomics Platform, Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, and The Department of Pediatrics, Pelotonia Institute for Immuno-Oncology, Ohio State University, Columbus, OH, USA
| | - Daniel Burns
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sophie M.S. Khakoo
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Salah Mansour
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jonathan W. Essex
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Rochelle Ayala
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute, and The Department of Pediatrics, Pelotonia Institute for Immuno-Oncology, Ohio State University, Columbus, OH, USA
| | - Anthony W. Purcell
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Salim I. Khakoo
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
3
|
Mohammad Taheri M, Javan F, Poudineh M, Athari SS. Beyond CAR-T: The rise of CAR-NK cell therapy in asthma immunotherapy. J Transl Med 2024; 22:736. [PMID: 39103889 PMCID: PMC11302387 DOI: 10.1186/s12967-024-05534-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Asthma poses a major public health burden. While existing asthma drugs manage symptoms for many, some patients remain resistant. The lack of a cure, especially for severe asthma, compels exploration of novel therapies. Cancer immunotherapy successes with CAR-T cells suggest its potential for asthma treatment. Researchers are exploring various approaches for allergic diseases including membrane-bound IgE, IL-5, PD-L2, and CTLA-4 for asthma, and Dectin-1 for fungal asthma. NK cells offer several advantages over T cells for CAR-based immunotherapy. They offer key benefits: (1) HLA compatibility, meaning they can be used in a wider range of patients without the need for matching tissue types. (2) Minimal side effects (CRS and GVHD) due to their limited persistence and cytokine profile. (3) Scalability for "off-the-shelf" production from various sources. Several strategies have been introduced that highlight the superiority and challenges of CAR-NK cell therapy for asthma treatment including IL-10, IFN-γ, ADCC, perforin-granzyme, FASL, KIR, NCRs (NKP46), DAP, DNAM-1, TGF-β, TNF-α, CCL, NKG2A, TF, and EGFR. Furthermore, we advocate for incorporating AI for CAR design optimization and CRISPR-Cas9 gene editing technology for precise gene manipulation to generate highly effective CAR constructs. This review will delve into the evolution and production of CAR designs, explore pre-clinical and clinical studies of CAR-based therapies in asthma, analyze strategies to optimize CAR-NK cell function, conduct a comparative analysis of CAR-T and CAR-NK cell therapy with their respective challenges, and finally present established novel CAR designs with promising potential for asthma treatment.
Collapse
Affiliation(s)
| | - Fatemeh Javan
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Seyed Shamseddin Athari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Immunology, Zanjan School of Medicine, Zanjan University of Medical Sciences, 12th Street, Shahrake Karmandan, Zanjan, 45139-561111, Iran.
| |
Collapse
|
4
|
Marques-Mejias A, Bartha I, Ciaccio CE, Chinthrajah RS, Chan S, Hershey GKK, Hui-Beckman JW, Kost L, Lack G, Layhadi JA, Leung DYM, Marshall HF, Nadeau KC, Radulovic S, Rajcoomar R, Shamji MH, Sindher S, Brough HA. Skin as the target for allergy prevention and treatment. Ann Allergy Asthma Immunol 2024; 133:133-143. [PMID: 38253125 DOI: 10.1016/j.anai.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/27/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024]
Abstract
The fact that genetic and environmental factors could trigger disruption of the epithelial barrier and subsequently initiate a TH2 inflammatory cascade conversely proposes that protecting the same barrier and promoting adequate interactions with other organs, such as the gut, may be crucial for lowering the risk and preventing atopic diseases, particularly, food allergies. In this review, we provide an overview of structural characteristics that support the epithelial barrier hypothesis in patients with atopic dermatitis, including the most relevant filaggrin gene mutations, the recent discovery of the role of the transient receptor potential vanilloid 1, and the role involvement of the microbiome in healthy and damaged skin. We present experimental and human studies that support the mechanisms of allergen penetration, particularly the dual allergen exposure and the outside-in, inside-out, and outside-inside-outside hypotheses. We discuss classic skin-targeted therapies for food allergy prevention, including moisturizers, steroids, and topical calcineurin inhibitors, along with pioneering trials proposed to change their current use (Prevention of Allergy via Cutaneous Intervention and Stopping Eczema and ALlergy). We provide an overview of the novel therapies that enhance the skin barrier, such as probiotics and prebiotics topical application, read-through drugs, direct and indirect FLG replacement, and interleukin and janus kinases inhibitors. Last, we discuss the newer strategies for preventing and treating food allergies in the form of epicutaneous immunotherapy and the experimental use of single-dose of adeno-associated virus vector gene immunotherapy.
Collapse
Affiliation(s)
- Andreina Marques-Mejias
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom
| | - Irene Bartha
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom
| | - Christina E Ciaccio
- Department of Pediatrics, The University of Chicago, Chicago, Illinois; Department of Medicine, The University of Chicago, Chicago, Illinois
| | - R Sharon Chinthrajah
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Susan Chan
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Laurie Kost
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Gideon Lack
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Janice A Layhadi
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Hannah F Marshall
- Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom
| | - Kari C Nadeau
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Suzana Radulovic
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Reena Rajcoomar
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sayantani Sindher
- Department of Medicine, and Sean N Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, California
| | - Helen A Brough
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London, Guy's and St Thomas', NHS Foundation Trust, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
5
|
Song Y, Lei L, Cai X, Wei H, Yu CY. Immunomodulatory Peptides for Tumor Treatment. Adv Healthc Mater 2024:e2400512. [PMID: 38657003 DOI: 10.1002/adhm.202400512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/07/2024] [Indexed: 04/26/2024]
Abstract
Peptides exhibit various biological activities, including biorecognition, cell targeting, and tumor penetration, and can stimulate immune cells to elicit immune responses for tumor immunotherapy. Peptide self-assemblies and peptide-functionalized nanocarriers can reduce the effect of various biological barriers and the degradation by peptidases, enhancing the efficiency of peptide delivery and improving antitumor immune responses. To date, the design and development of peptides with various functionalities have been extensively reviewed for enhanced chemotherapy; however, peptide-mediated tumor immunotherapy using peptides acting on different immune cells, to the knowledge, has not yet been summarized. Thus, this work provides a review of this emerging subject of research, focusing on immunomodulatory anticancer peptides. This review introduces the role of peptides in the immunomodulation of innate and adaptive immune cells, followed by a link between peptides in the innate and adaptive immune systems. The peptides are discussed in detail, following a classification according to their effects on different innate and adaptive immune cells, as well as immune checkpoints. Subsequently, two delivery strategies for peptides as drugs are presented: peptide self-assemblies and peptide-functionalized nanocarriers. The concluding remarks regarding the challenges and potential solutions of peptides for tumor immunotherapy are presented.
Collapse
Affiliation(s)
- Yang Song
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Longtianyang Lei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xingyu Cai
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, 410013, China
| |
Collapse
|
6
|
Foti Randazzese S, Panasiti I, Caminiti L, Catamerò F, Landi M, De Filippo M, Votto M, Olcese R, Favuzza F, Giovannini M, Barberi S. Current state and advances in desensitization for peanut allergy in pediatric age. Pediatr Allergy Immunol 2024; 35:e14127. [PMID: 38646959 DOI: 10.1111/pai.14127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/15/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
Peanut allergy affects about 1%-3% of the pediatric population in the world, with an important increase in the last decades. Nowadays, international guidelines recommend the early introduction of peanuts in the infant diet, with poor information about the quantity and the frequency of the intake. Allergen immunotherapy may represent the only therapeutic strategy able to modify the natural history of peanut allergy. In particular, oral immunotherapy showed the most promising results in terms of efficacy, but with significant rates of adverse reactions, mostly gastrointestinal. In 2020, the Food and Drug Administration and the European Medicines Agency approved Palforzia®, an oral drug for patients aged 4-17 years. Several studies are ongoing to improve the tolerability of oral immunotherapy and standardize the desensitization protocols. Sublingual immunotherapy permits to offer much lower doses than oral immunotherapy, but fewer adverse events are shown. Subcutaneous immunotherapy is associated with the greatest systemic adverse effects. Epicutaneous immunotherapy, for which Viaskin® patch was approved, has the highest safety profile. Innovative studies are evaluating the use of biological drugs, such as omalizumab or dupilumab, and probiotics, such as Lactobacillus rhamnosus, in monotherapy or associated with oral immunotherapy. Therapy for peanut allergy is constantly evolving, and new perspectives are ongoing to develop.
Collapse
Affiliation(s)
- Simone Foti Randazzese
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Ilenia Panasiti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Lucia Caminiti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Francesco Catamerò
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Massimo Landi
- Institute of Biomedicine and Molecular Immunology, Pediatric National Healthcare System, Turin, Italy
| | - Maria De Filippo
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Martina Votto
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Filippo Favuzza
- Pediatric Unit, Hospital Holy Family Fatebenefratelli Company, Erba, Italy
| | - Mattia Giovannini
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | | |
Collapse
|
7
|
Graham LV, Fisher JG, Khakoo SI, Blunt MD. Targeting KIR as a novel approach to improve CAR-NK cell function. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2023; 7:230-235. [PMID: 38229912 PMCID: PMC7615527 DOI: 10.20517/jtgg.2023.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Chimeric antigen receptor (CAR) NK cells are demonstrating promising activity in clinical trials and possess a favorable safety profile compared to CAR-T cells. The Killer cell Immunoglobulin-like Receptors (KIR) have a critical role in the control of NK cell function, and recently, this family of activating and inhibitory receptors have been targeted to improve CAR-NK function. These strategies include the utilisation of inhibitory KIR to reduce trogocytosis-associated NK cell fratricide, the downregulation of inhibitory KIR on CAR-NK cells to alleviate HLA mediated suppression, the selection of CAR-NK cell donors enriched for activating KIR, and the use of activating KIR intracellular domains within novel CAR constructs. These pre-clinical studies demonstrate the potential utility of targeting the KIR to improve CAR-NK cell efficacy and patient outcomes.
Collapse
Affiliation(s)
- Lara V Graham
- School of Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Jack G Fisher
- School of Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Salim I Khakoo
- School of Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Matthew D Blunt
- School of Clinical and Experimental Sciences, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
8
|
Zahedipour F, Jamialahmadi K, Zamani P, Reza Jaafari M. Improving the efficacy of peptide vaccines in cancer immunotherapy. Int Immunopharmacol 2023; 123:110721. [PMID: 37543011 DOI: 10.1016/j.intimp.2023.110721] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Peptide vaccines have shown great potential in cancer immunotherapy by targeting tumor antigens and activating the patient's immune system to mount a specific response against cancer cells. However, the efficacy of peptide vaccines in inducing a sustained immune response and achieving clinical benefit remains a major challenge. In this review, we discuss the current status of peptide vaccines in cancer immunotherapy and strategies to improve their efficacy. We summarize the recent advancements in the development of peptide vaccines in pre-clinical and clinical settings, including the use of novel adjuvants, neoantigens, nano-delivery systems, and combination therapies. We also highlight the importance of personalized cancer vaccines, which consider the unique genetic and immunological profiles of individual patients. We also discuss the strategies to enhance the immunogenicity of peptide vaccines such as multivalent peptides, conjugated peptides, fusion proteins, and self-assembled peptides. Although, peptide vaccines alone are weak immunogens, combining peptide vaccines with other immunotherapeutic approaches and developing novel approaches such as personalized vaccines can be promising methods to significantly enhance their efficacy and improve the clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Fatemeh Zahedipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Falcone N, Ermis M, Tamay DG, Mecwan M, Monirizad M, Mathes TG, Jucaud V, Choroomi A, de Barros NR, Zhu Y, Vrana NE, Kraatz HB, Kim HJ, Khademhosseini A. Peptide Hydrogels as Immunomaterials and Their Use in Cancer Immunotherapy Delivery. Adv Healthc Mater 2023; 12:e2301096. [PMID: 37256647 PMCID: PMC10615713 DOI: 10.1002/adhm.202301096] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/15/2023] [Indexed: 06/01/2023]
Abstract
Peptide-based hydrogel biomaterials have emerged as an excellent strategy for immune system modulation. Peptide-based hydrogels are supramolecular materials that self-assemble into various nanostructures through various interactive forces (i.e., hydrogen bonding and hydrophobic interactions) and respond to microenvironmental stimuli (i.e., pH, temperature). While they have been reported in numerous biomedical applications, they have recently been deemed promising candidates to improve the efficacy of cancer immunotherapies and treatments. Immunotherapies seek to harness the body's immune system to preemptively protect against and treat various diseases, such as cancer. However, their low efficacy rates result in limited patient responses to treatment. Here, the immunomaterial's potential to improve these efficacy rates by either functioning as immune stimulators through direct immune system interactions and/or delivering a range of immune agents is highlighted. The chemical and physical properties of these peptide-based materials that lead to immuno modulation and how one may design a system to achieve desired immune responses in a controllable manner are discussed. Works in the literature that reports peptide hydrogels as adjuvant systems and for the delivery of immunotherapies are highlighted. Finally, the future trends and possible developments based on peptide hydrogels for cancer immunotherapy applications are discussed.
Collapse
Affiliation(s)
- Natashya Falcone
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
| | - Dilara Goksu Tamay
- BIOMATEN, Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara, 06800, Turkey
- Department of Biotechnology, Middle East Technical University, Ankara, 06800, Turkey
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Mahsa Monirizad
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Tess Grett Mathes
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| | - Nihal Engin Vrana
- SPARTHA Medical, CRBS 1 Rue Eugene Boeckel, Strasbourg, 67000, France
| | - Heinz-Bernhard Kraatz
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, M5S 3E5, Canada
- Department of Physical and Environmental Sciences, University of Toronto Scarborough, Toronto, ON, M1C 1A4, Canada
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, 1018 Westwood Blvd, Los Angeles, CA, 90034, USA
| |
Collapse
|
10
|
Blunt MD, Vallejo Pulido A, Fisher JG, Graham LV, Doyle ADP, Fulton R, Carter MJ, Polak M, Johnson PWM, Cragg MS, Forconi F, Khakoo SI. KIR2DS2 Expression Identifies NK Cells With Enhanced Anticancer Activity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:379-390. [PMID: 35768150 PMCID: PMC7613074 DOI: 10.4049/jimmunol.2101139] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 05/08/2022] [Indexed: 12/13/2022]
Abstract
NK cells are promising cellular therapeutics against hematological and solid malignancies. Immunogenetic studies have identified that various activating killer cell Ig-like receptors (KIRs) are associated with cancer outcomes. Specifically, KIR2DS2 has been associated with reduced incidence of relapse following transplant in hematological malignancies and improved outcomes in solid tumors, but the mechanism remains obscure. Therefore, we investigated how KIR2DS2 expression impacts NK cell function. Using a novel flow cytometry panel, we show that human NK cells with high KIR2DS2 expression have enhanced spontaneous activation against malignant B cell lines, liver cancer cell lines, and primary chronic lymphocytic leukemia cells. Surface expression of CD16 was increased on KIR2DS2high NK cells, and, accordingly, KIR2DS2high NK cells had increased activation against lymphoma cells coated with the clinically relevant anti-CD20 Abs rituximab and obinutuzumab. Bulk RNA sequencing revealed that KIR2DS2high NK cells have upregulation of NK-mediated cytotoxicity, translation, and FCGR gene pathways. We developed a novel single-cell RNA-sequencing technique to identify KIR2DS2+ NK cells, and this confirmed that KIR2DS2 is associated with enhanced NK cell-mediated cytotoxicity. This study provides evidence that KIR2DS2 marks a population of NK cells primed for anticancer activity and indicates that KIR2DS2 is an attractive target for NK-based therapeutic strategies.
Collapse
Affiliation(s)
- Matthew D Blunt
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Andres Vallejo Pulido
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Jack G Fisher
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Lara V Graham
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Amber D P Doyle
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Rebecca Fulton
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Matthew J Carter
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Marta Polak
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| | - Peter W M Johnson
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Mark S Cragg
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Francesco Forconi
- School of Cancer Sciences, University of Southampton, Southampton, United Kingdom
| | - Salim I Khakoo
- School of Clinical and Experimental Sciences, University of Southampton, Southampton, United Kingdom; and
| |
Collapse
|
11
|
Reinwald S, Rolland JM, O’Hehir RE, van Zelm MC. Peanut oral immunotherapy – current trends in clinical trials. IMMUNOTHERAPY ADVANCES 2022; 2:ltac004. [PMID: 35919493 PMCID: PMC9327116 DOI: 10.1093/immadv/ltac004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/27/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
Immunotherapy for allergy has been practised for over 100 years. Low-dose repeated exposure to specific allergen extracts over several months to years can successfully induce clinical tolerance in patients with allergy to insect venoms, pollen, house dust mite and domestic animals. Different regimens and routes for immunotherapy include subcutaneous, sublingual, oral and intralymphatic. Food allergies have been difficult to treat in this way due to high anaphylactic potential and only recently the first immunotherapy for peanut allergy has received regulatory approval. Several clinical trials have indicated high efficacy in desensitisation of peanut-allergic individuals using oral immunotherapy, which allows for safer administration of relatively high allergen concentrations. Still, the risk of adverse events including serious allergic reactions and high anxiety levels for patients remains, demonstrating the need for further optimisation of treatment protocols. Here we discuss the design and outcomes of recent clinical trials with traditional oral immunotherapy, and consider alternative protocols and formulations for safer and more effective oral treatment strategies for peanut allergy.
Collapse
Affiliation(s)
- Simone Reinwald
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Respiratory Medicine, Central Clinical School, Monash University, and Alfred Hospital, Melbourne, VIC, Australia
| | - Jennifer M Rolland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Respiratory Medicine, Central Clinical School, Monash University, and Alfred Hospital, Melbourne, VIC, Australia
| | - Robyn E O’Hehir
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Respiratory Medicine, Central Clinical School, Monash University, and Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Respiratory Medicine, Central Clinical School, Monash University, and Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
12
|
Wang K, Wang C, Jiang H, Zhang Y, Lin W, Mo J, Jin C. Combination of Ablation and Immunotherapy for Hepatocellular Carcinoma: Where We Are and Where to Go. Front Immunol 2022; 12:792781. [PMID: 34975896 PMCID: PMC8714655 DOI: 10.3389/fimmu.2021.792781] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide and is increasing in incidence. Local ablative therapy plays a leading role in HCC treatment. Radiofrequency (RFA) is one of the first-line therapies for early local ablation. Other local ablation techniques (e.g., microwave ablation, cryoablation, irreversible electroporation, phototherapy.) have been extensively explored in clinical trials or cell/animal studies but have not yet been established as a standard treatment or applied clinically. On the one hand, single treatment may not meet the needs. On the other hand, ablative therapy can stimulate local and systemic immune effects. The combination strategy of immunotherapy and ablation is reasonable. In this review, we briefly summarized the current status and progress of ablation and immunotherapy for HCC. The immune effects of local ablation and the strategies of combination therapy, especially synergistic strategies based on biomedical materials, were discussed. This review is hoped to provide references for future researches on ablative immunotherapy to arrive to a promising new era of HCC treatment.
Collapse
Affiliation(s)
- Kunpeng Wang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Cong Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Hao Jiang
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Yaqiong Zhang
- Department of Clinical Laboratory, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Weidong Lin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Jinggang Mo
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Chong Jin
- Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
13
|
Natural Killer Cells and Type 1 Innate Lymphoid Cells in Hepatocellular Carcinoma: Current Knowledge and Future Perspectives. Int J Mol Sci 2021; 22:ijms22169044. [PMID: 34445750 PMCID: PMC8396475 DOI: 10.3390/ijms22169044] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023] Open
Abstract
Natural killer (NK) cells and type 1 innate lymphoid cells (ILC1) are specific innate lymphoid cell subsets that are key for the detection and elimination of pathogens and cancer cells. In liver, while they share a number of characteristics, they differ in many features. These include their developmental pathways, tissue distribution, phenotype and functions. NK cells and ILC1 contribute to organ homeostasis through the production of key cytokines and chemokines and the elimination of potential harmful bacteria and viruses. In addition, they are equipped with a wide range of receptors, allowing them to detect “stressed cells’ such as cancer cells. Our understanding of the role of innate lymphoid cells in hepatocellular carcinoma (HCC) is growing owing to the development of mouse models, the progress in immunotherapeutic treatment and the recent use of scRNA sequencing analyses. In this review, we summarize the current understanding of NK cells and ILC1 in hepatocellular carcinoma and discuss future strategies to take advantage of these innate immune cells in anti-tumor immunity. Immunotherapies hold great promise in HCC, and a better understanding of the role and function of NK cells and ILC1 in liver cancer could pave the way for new NK cell and/or ILC1-targeted treatment.
Collapse
|
14
|
Relevance of Polymorphic KIR and HLA Class I Genes in NK-Cell-Based Immunotherapies for Adult Leukemic Patients. Cancers (Basel) 2021; 13:cancers13153767. [PMID: 34359667 PMCID: PMC8345033 DOI: 10.3390/cancers13153767] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Immunotherapies are promising approaches to curing different acute leukemias. Natural killer (NK) cells are lymphocytes that are efficient in the elimination of leukemic cells. NK-cell-based immunotherapies are particularly attractive, but the landscape of the heterogeneity of NK cells must be deciphered. This review provides an overview of the polymorphic KIR and HLA class I genes that modulate the NK cell repertoire and how these markers can improve the outcomes of patients with acute leukemia. A better knowledge of these genetic markers that are linked to NK cell subsets that are efficient against hematological diseases will optimize hematopoietic stem-cell donor selection and NK immunotherapy design. Abstract Since the mid-1990s, the biology and functions of natural killer (NK) cells have been deeply investigated in healthy individuals and in people with diseases. These effector cells play a particularly crucial role after allogeneic hematopoietic stem-cell transplantation (HSCT) through their graft-versus-leukemia (GvL) effect, which is mainly mediated through polymorphic killer-cell immunoglobulin-like receptors (KIRs) and their cognates, HLA class I ligands. In this review, we present how KIRs and HLA class I ligands modulate the structural formation and the functional education of NK cells. In particular, we decipher the current knowledge about the extent of KIR and HLA class I gene polymorphisms, as well as their expression, interaction, and functional impact on the KIR+ NK cell repertoire in a physiological context and in a leukemic context. In addition, we present the impact of NK cell alloreactivity on the outcomes of HSCT in adult patients with acute leukemia, as well as a description of genetic models of KIRs and NK cell reconstitution, with a focus on emergent T-cell-repleted haplo-identical HSCT using cyclosphosphamide post-grafting (haplo-PTCy). Then, we document how the immunogenetics of KIR/HLA and the immunobiology of NK cells could improve the relapse incidence after haplo-PTCy. Ultimately, we review the emerging NK-cell-based immunotherapies for leukemic patients in addition to HSCT.
Collapse
|