1
|
Prades-Sagarra E, Yaromina A, Dubois L. Understanding the impact of radiation-induced lymphopenia: Preclinical and clinical research perspectives. Clin Transl Radiat Oncol 2024; 49:100852. [PMID: 39315059 PMCID: PMC11418132 DOI: 10.1016/j.ctro.2024.100852] [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: 06/10/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy has revolutionized the field of cancer treatment, changing the standard of care to the use of immune checkpoint inhibitors. Radiotherapy can boost anti-tumour immune responses by changing the tumour microenvironment, but it also can cause radiotherapy-induced lymphopenia (RIL), a decrease in circulating lymphocyte counts. RIL has been associated with lower survival in patients undergoing radiotherapy, and new studies have suggested that it can also affect immunotherapy outcome. To study RIL's effects and to explore mitigation treatment strategies, preclinical models closely mimicking the clinical situation are needed. State-of-the-art image-guided small animal irradiators now offer the possibility to target specific organs in small animals to induce RIL, aiding research on its molecular mechanisms and prevention. This review covers the relationship between radiotherapy and RIL, its impact on patient survival, and future directions to generate models to investigate and prevent RIL.
Collapse
Affiliation(s)
- E. Prades-Sagarra
- The M-Lab, Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - A. Yaromina
- The M-Lab, Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - L.J. Dubois
- The M-Lab, Department of Precision Medicine, GROW - Research Institute for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
2
|
Kim J, Choi H, Jeun SS, Ahn S. From lymphopenia to restoration: IL-7 immunotherapy for lymphocyte recovery in glioblastoma. Cancer Lett 2024; 588:216714. [PMID: 38369003 DOI: 10.1016/j.canlet.2024.216714] [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: 12/12/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024]
Abstract
Glioblastoma, the most prevalent malignant primary brain tumor, presents substantial treatment challenges because of its inherent aggressiveness and limited therapeutic options. Lymphopenia, defined as reduced peripheral blood lymphocyte count, commonly occurs as a consequence of the disease and its treatment. Recent studies have associated lymphopenia with a poor prognosis. Factors that contribute to lymphopenia include radiotherapy, chemotherapy, and the tumor itself. Patients who are female, older, using dexamethasone, or receiving higher doses of radiation therapy are particularly vulnerable to this condition. Several preclinical studies have explored the use of interleukin-7, a crucial cytokine for lymphocyte homeostasis, to restore lymphocyte counts and potentially rebuild the immune system to combat glioblastoma cells. With the development of recombinant interleukin-7 for prolonged activity in the body, various clinical trials are underway to explore this treatment in patients with glioblastoma. Our study provides a comprehensive summary of the incidence of lymphopenia, its potential biological background, and the associated clinical risk factors. Furthermore, we reviewed several clinical trials using IL-7 cytokine therapy in glioblastoma patients. We propose IL-7 as a promising immunotherapeutic strategy for glioblastoma treatment. We are optimistic that our study will enhance understanding of the complex interplay between lymphopenia and glioblastoma and will pave the way for the development of more effective treatment modalities.
Collapse
Affiliation(s)
- Joonseok Kim
- College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Haeyoun Choi
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Cajander S, Kox M, Scicluna BP, Weigand MA, Mora RA, Flohé SB, Martin-Loeches I, Lachmann G, Girardis M, Garcia-Salido A, Brunkhorst FM, Bauer M, Torres A, Cossarizza A, Monneret G, Cavaillon JM, Shankar-Hari M, Giamarellos-Bourboulis EJ, Winkler MS, Skirecki T, Osuchowski M, Rubio I, Bermejo-Martin JF, Schefold JC, Venet F. Profiling the dysregulated immune response in sepsis: overcoming challenges to achieve the goal of precision medicine. THE LANCET. RESPIRATORY MEDICINE 2024; 12:305-322. [PMID: 38142698 DOI: 10.1016/s2213-2600(23)00330-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 12/26/2023]
Abstract
Sepsis is characterised by a dysregulated host immune response to infection. Despite recognition of its significance, immune status monitoring is not implemented in clinical practice due in part to the current absence of direct therapeutic implications. Technological advances in immunological profiling could enhance our understanding of immune dysregulation and facilitate integration into clinical practice. In this Review, we provide an overview of the current state of immune profiling in sepsis, including its use, current challenges, and opportunities for progress. We highlight the important role of immunological biomarkers in facilitating predictive enrichment in current and future treatment scenarios. We propose that multiple immune and non-immune-related parameters, including clinical and microbiological data, be integrated into diagnostic and predictive combitypes, with the aid of machine learning and artificial intelligence techniques. These combitypes could form the basis of workable algorithms to guide clinical decisions that make precision medicine in sepsis a reality and improve patient outcomes.
Collapse
Affiliation(s)
- Sara Cajander
- Department of Infectious Diseases, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Matthijs Kox
- Department of Intensive Care Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei hospital, University of Malta, Msida, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Raquel Almansa Mora
- Department of Cell Biology, Genetics, Histology and Pharmacology, University of Valladolid, Valladolid, Spain
| | - Stefanie B Flohé
- Department of Trauma, Hand, and Reconstructive Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ignacio Martin-Loeches
- St James's Hospital, Dublin, Ireland; Hospital Clinic, Institut D'Investigacions Biomediques August Pi i Sunyer, Universidad de Barcelona, Barcelona, Spain
| | - Gunnar Lachmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Anesthesiology and Operative Intensive Care Medicine, Berlin, Germany
| | - Massimo Girardis
- Department of Intensive Care and Anesthesiology, University Hospital of Modena, Modena, Italy
| | - Alberto Garcia-Salido
- Hospital Infantil Universitario Niño Jesús, Pediatric Critical Care Unit, Madrid, Spain
| | - Frank M Brunkhorst
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Antoni Torres
- Pulmonology Department. Hospital Clinic of Barcelona, University of Barcelona, Ciberes, IDIBAPS, ICREA, Barcelona, Spain
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Guillaume Monneret
- Immunology Laboratory, Hôpital E Herriot - Hospices Civils de Lyon, Lyon, France; Université Claude Bernard Lyon-1, Hôpital E Herriot, Lyon, France
| | | | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute of Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | | | - Martin Sebastian Winkler
- Department of Anesthesiology and Intensive Care, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marcin Osuchowski
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Vienna, Austria
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Integrated Research and Treatment Center, Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Jesus F Bermejo-Martin
- Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; School of Medicine, Universidad de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fabienne Venet
- Immunology Laboratory, Hôpital E Herriot - Hospices Civils de Lyon, Lyon, France; Centre International de Recherche en Infectiologie, Inserm U1111, CNRS, UMR5308, Ecole Normale Supeérieure de Lyon, Universiteé Claude Bernard-Lyon 1, Lyon, France.
| |
Collapse
|
4
|
Dinges SS, Amini K, Notarangelo LD, Delmonte OM. Primary and secondary defects of the thymus. Immunol Rev 2024; 322:178-211. [PMID: 38228406 PMCID: PMC10950553 DOI: 10.1111/imr.13306] [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] [Indexed: 01/18/2024]
Abstract
The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.
Collapse
Affiliation(s)
- Sarah S. Dinges
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kayla Amini
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Park JH, Lee SW, Choi D, Lee C, Sung YC. Harnessing the Power of IL-7 to Boost T Cell Immunity in Experimental and Clinical Immunotherapies. Immune Netw 2024; 24:e9. [PMID: 38455462 PMCID: PMC10917577 DOI: 10.4110/in.2024.24.e9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
The cytokine IL-7 plays critical and nonredundant roles in T cell immunity so that the abundance and availability of IL-7 act as key regulatory mechanisms in T cell immunity. Importantly, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells that are limited in their numbers. Thus, T cells depend on cell extrinsic IL-7, and the amount of in vivo IL-7 is considered a major factor in maximizing and maintaining the number of T cells in peripheral tissues. Moreover, IL-7 provides metabolic cues and promotes the survival of both naïve and memory T cells. Thus, IL-7 is also essential for the functional fitness of T cells. In this regard, there has been an extensive effort trying to increase the protein abundance of IL-7 in vivo, with the aim to augment T cell immunity and harness T cell functions in anti-tumor responses. Such approaches started under experimental animal models, but they recently culminated into clinical studies, with striking effects in re-establishing T cell immunity in immunocompromised patients, as well as boosting anti-tumor effects. Depending on the design, glycosylation, and the structure of recombinantly engineered IL-7 proteins and their mimetics, recombinant IL-7 molecules have shown dramatic differences in their stability, efficacy, cellular effects, and overall immune functions. The current review is aimed to summarize the past and present efforts in the field that led to clinical trials, and to highlight the therapeutical significance of IL-7 biology as a master regulator of T cell immunity.
Collapse
Affiliation(s)
- Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Donghoon Choi
- Research Institute of NeoImmune Tech., Co, Ltd., Bio Open Innovation Center, Pohang 37666, Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Young Chul Sung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
6
|
Min H, Valente LA, Xu L, O'Neil SM, Begg LR, Kurtzberg J, Filiano AJ. Improving thymus implantation for congenital athymia with interleukin-7. Clin Transl Immunology 2023; 12:e1475. [PMID: 38020730 PMCID: PMC10665642 DOI: 10.1002/cti2.1475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives Thymus implantation is a recently FDA-approved therapy for congenital athymia. Patients receiving thymus implantation develop a functional but incomplete T cell compartment. Our objective was to develop a mouse model to study clinical thymus implantation in congenital athymia and to optimise implantation procedures to maximise T cell education and expansion of naïve T cells. Methods Using Foxn1 nu athymic mice as recipients, we tested MHC-matched and -mismatched donor thymi that were implanted as fresh tissue or cultured to remove donor T cells. We first implanted thymus under the kidney capsule and then optimised intramuscular implantation. Using competitive adoptive transfer assays, we investigated whether the failure of newly developed T cells to expand into a complete T cell compartment was because of intrinsic deficits or whether there were deficits in engaging MHC molecules in the periphery. Finally, we tested whether recombinant IL-7 would promote the expansion of host naïve T cells educated by the implanted thymus. Results We determined that thymus implants in Foxn1 nu athymic mice mimic many aspects of clinical thymus implants in patients with congenital athymia. When we implanted cultured, MHC-mismatched donor thymus into Foxn1 nu athymic mice, mice developed a limited T cell compartment with notably underdeveloped naïve populations and overrepresented memory-like T cells. Newly generated T cells were predominantly educated by MHC molecules expressed by the donor thymus, thus potentially undergoing another round of selection once in the peripheral circulation. Using competitive adoptive transfer assays, we compared expansion rates of T cells educated on donor thymus versus T cells educated during typical thymopoiesis in MHC-matched and -mismatched environments. Once in the circulation, regardless of the MHC haplotypes, T cells educated on a donor thymus underwent abnormal expansion with initially more robust proliferation coupled with greater cell death, resembling IL-7 independent spontaneous expansion. Treating implanted mice with recombinant interleukin (IL-7) promoted homeostatic expansion that improved T cell development, expanded the T cell receptor repertoire, and normalised the naïve T cell compartment. Conclusion We conclude that implanting cultured thymus into the muscle of Foxn1 nu athymic mice is an appropriate system to study thymus implantation for congenital athymia and immunodeficiencies. T cells are educated by the donor thymus, yet naïve T cells have deficits in expansion. IL-7 greatly improves T cell development after thymus implantation and may offer a novel strategy to improve outcomes of clinical thymus implantation.
Collapse
Affiliation(s)
- Hyunjung Min
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Laura A Valente
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PathologyDuke UniversityDurhamNCUSA
| | - Li Xu
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Shane M O'Neil
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Lauren R Begg
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Joanne Kurtzberg
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PediatricsDuke UniversityDurhamNCUSA
| | - Anthony J Filiano
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PathologyDuke UniversityDurhamNCUSA
- Department of NeurosurgeryDuke UniversityDurhamNCUSA
- Department of ImmunologyDuke UniversityDurhamNCUSA
| |
Collapse
|
7
|
Dower WJ, Park AI, Bakker AV, Cwirla SE, Pongtornpipat P, Williams BM, Joshi P, Baxter BA, Needels MC, Barrett RW. A mechanistically novel peptide agonist of the IL-7 receptor that addresses limitations of IL-7 cytokine therapy. PLoS One 2023; 18:e0286834. [PMID: 37874823 PMCID: PMC10597491 DOI: 10.1371/journal.pone.0286834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
Interleukin (IL)-7 is broadly active on T-cell populations, and modified versions have been clinically evaluated for a variety of therapeutic applications, including cancer, lymphopenia, and infectious diseases; and found to be relatively well-tolerated and biologically active. Here we describe novel IL-7R agonists that are unrelated in structure to IL-7, bind to the receptor subunits differently from IL-7, but closely emulate IL-7 biology. The small size, low structural complexity, and the natural amino acid composition of the pharmacologically active peptide MDK1472 allows facile incorporation into protein structures, such as the IgG2-Fc fusion MDK-703. This molecule possesses properties potentially better suited to therapeutic applications than native IL-7 or its derivatives. We compared these compounds with IL-7 for immune cell selectivity, induction of IL-7R signaling, receptor-mediated internalization, proliferation, and generation of immune cell phenotypes in human and non-human primate (NHP) peripheral blood cells in vitro; and found them to be similar in biological activity to IL-7. In cynomolgus macaques, MDK-703 exhibits a circulating half-life of 46 hr and produces sustained T-cell expansion characteristic of IL-7 treatment. In the huCD34+-engrafted NSG mouse model of the human immune system, MDK-703 induces an immune cell profile very similar to that generated by IL-7-derived compounds; including the pronounced expansion of memory T-cells, particularly the population of stem-like memory T-cells (Tscm) which may be important for anti-tumor activities reported with IL-7 treatment. Clinical administration of IL-7 and modified variants has been reported to induce anti-drug antibodies (ADAs), including IL-7 neutralizing antibodies. The novel peptide agonist reported here scores very low in predicted immunogenicity, and because the peptide lacks sequence similarity with IL-7, the problematic immunogenic neutralization of endogenous cytokine should not occur. The properties we report here implicate MDK-703 as a candidate for clinical evaluation in oncology, anti-viral and other infectious disease, vaccine enhancement, and treatment of lymphopenia.
Collapse
Affiliation(s)
- William J. Dower
- Medikine, Inc., Menlo Park, California, United States of America
| | | | - Alice V. Bakker
- Medikine, Inc., Menlo Park, California, United States of America
| | - Steven E. Cwirla
- Medikine, Inc., Menlo Park, California, United States of America
| | | | - Blake M. Williams
- Medikine, Inc., Menlo Park, California, United States of America
- Department of Biomedical Engineering, and Center for Quantitative Bioinformatics and Quantitative Biology, Colleges of Engineering and Medicine, University of Illinois Chicago, IL, United States of America
| | - Prarthana Joshi
- Medikine, Inc., Menlo Park, California, United States of America
| | - Bryan A. Baxter
- Medikine, Inc., Menlo Park, California, United States of America
| | | | | |
Collapse
|
8
|
Leonard WJ, Lin JX. Strategies to therapeutically modulate cytokine action. Nat Rev Drug Discov 2023; 22:827-854. [PMID: 37542128 DOI: 10.1038/s41573-023-00746-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2023] [Indexed: 08/06/2023]
Abstract
Cytokines are secreted or membrane-presented molecules that mediate broad cellular functions, including development, differentiation, growth and survival. Accordingly, the regulation of cytokine activity is extraordinarily important both physiologically and pathologically. Cytokine and/or cytokine receptor engineering is being widely investigated to safely and effectively modulate cytokine activity for therapeutic benefit. IL-2 in particular has been extensively engineered, to create IL-2 variants that differentially exhibit activities on regulatory T cells to potentially treat autoimmune disease versus effector T cells to augment antitumour effects. Additionally, engineering approaches are being applied to many other cytokines such as IL-10, interferons and IL-1 family cytokines, given their immunosuppressive and/or antiviral and anticancer effects. In modulating the actions of cytokines, the strategies used have been broad, including altering affinities of cytokines for their receptors, prolonging cytokine half-lives in vivo and fine-tuning cytokine actions. The field is rapidly expanding, with extensive efforts to create improved therapeutics for a range of diseases.
Collapse
Affiliation(s)
- Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Gusev A. Germline mechanisms of immunotherapy toxicities in the era of genome-wide association studies. Immunol Rev 2023; 318:138-156. [PMID: 37515388 PMCID: PMC11472697 DOI: 10.1111/imr.13253] [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: 03/14/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023]
Abstract
Cancer immunotherapy has revolutionized the treatment of advanced cancers and is quickly becoming an option for early-stage disease. By reactivating the host immune system, immunotherapy harnesses patients' innate defenses to eradicate the tumor. By putatively similar mechanisms, immunotherapy can also substantially increase the risk of toxicities or immune-related adverse events (irAEs). Severe irAEs can lead to hospitalization, treatment discontinuation, lifelong immune complications, or even death. Many irAEs present with similar symptoms to heritable autoimmune diseases, suggesting that germline genetics may contribute to their onset. Recently, genome-wide association studies (GWAS) of irAEs have identified common germline associations and putative mechanisms, lending support to this hypothesis. A wide range of well-established GWAS methods can potentially be harnessed to understand the etiology of irAEs specifically and immunotherapy outcomes broadly. This review summarizes current findings regarding germline effects on immunotherapy outcomes and discusses opportunities and challenges for leveraging germline genetics to understand, predict, and treat irAEs.
Collapse
Affiliation(s)
- Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
- Division of Genetics, Brigham & Women's Hospital, Boston, Massachusetts, USA
- The Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
10
|
Hesari M, Attar Z, Soltani-Shirazi S, Keshavarzian O, Taheri R, Tabrizi R, Fouladseresht H. The Therapeutic Values of IL-7/IL-7R and the Recombinant Derivatives in Glioma: A Narrative Review. J Interferon Cytokine Res 2023; 43:319-334. [PMID: 37566474 DOI: 10.1089/jir.2023.0050] [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: 08/13/2023] Open
Abstract
Interleukin-7 (IL-7) is essential for maintaining the immune system's defense functions by regulating the development and homeostasis of lymphocytes. Findings have shown the high efficacy of IL-7/IL-7 receptor (IL-7R)-based immunotherapy on various malignancies, with confirmation in both animal models and humans. In recent years, the progression-free survival and overall survival of patients suffering from gliomas significantly increased by introducing C7R-expressing chimeric antigen receptor (CAR)-T cells and long-acting IL-7 agonists such as NT-I7 (rhIL-7-hyFc, Efineptakin alfa). However, the effect of IL-7-based immunotherapies on the resistance of tumor cells to chemotherapy (when used simultaneously with chemotherapy agents) is still ambiguous and requires further studies. This article first reviews the pathophysiological roles of IL-7/IL-7R in tumors, focusing on gliomas. Subsequently, it discusses the therapeutic values of IL-7/IL-7R and the recombinant derivatives in gliomas.
Collapse
Affiliation(s)
| | - Zeinab Attar
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Department of Pharmacology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shakiba Soltani-Shirazi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Reza Taheri
- Department of Neurosurgery, Fasa University of Medical Sciences, Fasa, Iran
| | - Reza Tabrizi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
11
|
Fu Y, Tang R, Zhao X. Engineering cytokines for cancer immunotherapy: a systematic review. Front Immunol 2023; 14:1218082. [PMID: 37483629 PMCID: PMC10357296 DOI: 10.3389/fimmu.2023.1218082] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Cytokines are pivotal mediators of cell communication in the tumor microenvironment. Multiple cytokines are involved in the host antitumor response, but the production and function of these cytokines are usually dysregulated during malignant tumor progression. Considering their clinical potential and the early successful use of cytokines in cancer immunotherapy, such as interferon alpha-2b (IFNα-2b; IntronA®) and IL-2 (Proleukin®), cytokine-based therapeutics have been extensively evaluated in many follow-up clinical trials. Following these initial breakthroughs, however, clinical translation of these natural messenger molecules has been greatly limited owing to their high-degree pleiotropic features and complex biological properties in many cell types. These characteristics, coupled with poor pharmacokinetics (a short half-life), have hampered the delivery of cytokines via systemic administration, particularly because of severe dose-limiting toxicities. New engineering approaches have been developed to widen the therapeutic window, prolong pharmacokinetic effects, enhance tumor targeting and reduce adverse effects, thereby improving therapeutic efficacy. In this review, we focus on the recent progress and competitive landscape in cytokine engineering strategies and preclinical/clinical therapeutics for cancer. In addition, aiming to promote engineered cytokine-based cancer immunotherapy, we present a profound discussion about the feasibility of recently developed methods in clinical medicine translation.
Collapse
Affiliation(s)
- Yong Fu
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
- Jiangsu Simcere Pharmaceutical Co, Ltd., Nanjing, China
| | - Renhong Tang
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
- Simcere Zaiming Pharmaceutical Co, Ltd., Nanjing, China
| | - Xiaofeng Zhao
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing, China
- Jiangsu Simcere Pharmaceutical Co, Ltd., Nanjing, China
| |
Collapse
|
12
|
Pandit H, Valentin A, Angel M, Deleage C, Bergamaschi C, Bear J, Sowder R, Felber BK, Pavlakis GN. Step-dose IL-7 treatment promotes systemic expansion of T cells and alters immune cell landscape in blood and lymph nodes. iScience 2023; 26:105929. [PMID: 36685042 PMCID: PMC9852696 DOI: 10.1016/j.isci.2023.105929] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/06/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
We employed a dose-escalation regimen in rhesus macaques to deliver glycosylated IL-7, a cytokine critical for development and maintenance of T lymphocytes. IL-7 increased proliferation and survival of T cells and triggered several chemokines and cytokines. Induction of CXCL13 in lymph nodes (LNs) led to a remarkable increase of B cells in the LNs, proliferation of germinal center follicular T helper cells and elevated IL-21 levels suggesting an increase in follicle activity. Transcriptomics analysis showed induction of IRF-7 and Flt3L, which was linked to increased frequency of circulating plasmacytoid dendritic cells (pDCs) on IL-7 treatment. These pDCs expressed higher levels of CCR7, homed to LNs, and were associated with upregulation of type-1 interferon gene signature and increased production of IFN-α2a on TLR stimulation. Superior effects and dose-sparing advantage was observed by the step-dose regimen. Thus, IL-7 treatment leads to systemic effects involving both lymphoid and myeloid compartments.
Collapse
Affiliation(s)
- Hrishikesh Pandit
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Matthew Angel
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Center for Cancer Research Collaborative Bioinformatics Resource, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Claire Deleage
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Raymond Sowder
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| |
Collapse
|
13
|
Peng J, Madduri S, Clontz AD, Stewart DA. Clinical trial-identified inflammatory biomarkers in breast and pancreatic cancers. Front Endocrinol (Lausanne) 2023; 14:1106520. [PMID: 37181043 PMCID: PMC10173309 DOI: 10.3389/fendo.2023.1106520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Breast cancer and pancreatic cancer are two common cancer types characterized by high prevalence and high mortality rates, respectively. However, breast cancer has been more well-studied than pancreatic cancer. This narrative review curated inflammation-associated biomarkers from clinical studies that were systematically selected for both breast and pancreatic cancers and discusses some of the common and unique elements between the two endocrine-regulated malignant diseases. Finding common ground between the two cancer types and specifically analyzing breast cancer study results, we hoped to explore potential feasible methods and biomarkers that may be useful also in diagnosing and treating pancreatic cancer. A PubMed MEDLINE search was used to identify articles that were published between 2015-2022 of different kinds of clinical trials that measured immune-modulatory biomarkers and biomarker changes of inflammation defined in diagnosis and treatment of breast cancer and pancreatic cancer patients. A total of 105 papers (pancreatic cancer 23, breast cancer 82) were input into Covidence for the title and abstract screening. The final number of articles included in this review was 73 (pancreatic cancer 19, breast cancer 54). The results showed some of the frequently cited inflammatory biomarkers for breast and pancreatic cancers included IL-6, IL-8, CCL2, CD8+ T cells and VEGF. Regarding unique markers, CA15-3 and TNF-alpha were two of several breast cancer-specific, and CA19 and IL-18 were pancreatic cancer-specific. Moreover, we discussed leptin and MMPs as emerging biomarker targets with potential use for managing pancreatic cancer based on breast cancer studies in the future, based on inflammatory mechanisms. Overall, the similarity in how both types of cancers respond to or result in further disruptive inflammatory signaling, and that point to a list of markers that have been shown useful in diagnosis and/or treatment method response or efficacy in managing breast cancer could potentially provide insights into developing the same or more useful diagnostic and treatment measurement inflammatory biomarkers for pancreatic cancer. More research is needed to investigate the relationship and associated inflammatory markers between the similar immune-associated biological mechanisms that contribute to breast and pancreatic cancer etiology, drive disease progression or that impact treatment response and reflect survival outcomes.
Collapse
Affiliation(s)
- Jing Peng
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Supradeep Madduri
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Angela D. Clontz
- Department of Nutrition, Meredith College, Raleigh, NC, United States
| | - Delisha A. Stewart
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- *Correspondence: Delisha A. Stewart,
| |
Collapse
|
14
|
Hino C, Xu Y, Xiao J, Baylink DJ, Reeves ME, Cao H. The potential role of the thymus in immunotherapies for acute myeloid leukemia. Front Immunol 2023; 14:1102517. [PMID: 36814919 PMCID: PMC9940763 DOI: 10.3389/fimmu.2023.1102517] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
Understanding the factors which shape T-lymphocyte immunity is critical for the development and application of future immunotherapeutic strategies in treating hematological malignancies. The thymus, a specialized central lymphoid organ, plays important roles in generating a diverse T lymphocyte repertoire during the infantile and juvenile stages of humans. However, age-associated thymic involution and diseases or treatment associated injury result in a decline in its continuous role in the maintenance of T cell-mediated anti-tumor/virus immunity. Acute myeloid leukemia (AML) is an aggressive hematologic malignancy that mainly affects older adults, and the disease's progression is known to consist of an impaired immune surveillance including a reduction in naïve T cell output, a restriction in T cell receptor repertoire, and an increase in frequencies of regulatory T cells. As one of the most successful immunotherapies thus far developed for malignancy, T-cell-based adoptive cell therapies could be essential for the development of a durable effective treatment to eliminate residue leukemic cells (blasts) and prevent AML relapse. Thus, a detailed cellular and molecular landscape of how the adult thymus functions within the context of the AML microenvironment will provide new insights into both the immune-related pathogenesis and the regeneration of a functional immune system against leukemia in AML patients. Herein, we review the available evidence supporting the potential correlation between thymic dysfunction and T-lymphocyte impairment with the ontogeny of AML (II-VI). We then discuss how the thymus could impact current and future therapeutic approaches in AML (VII). Finally, we review various strategies to rejuvenate thymic function to improve the precision and efficacy of cancer immunotherapy (VIII).
Collapse
Affiliation(s)
- Christopher Hino
- Department of Internal Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yi Xu
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Jeffrey Xiao
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - David J Baylink
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Mark E Reeves
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| | - Huynh Cao
- Division of Hematology and Oncology, Department of Medicine, Loma Linda University, Loma Linda, CA, United States.,Loma Linda University Cancer Center, Loma Linda, CA, United States
| |
Collapse
|
15
|
IL-7: Comprehensive review. Cytokine 2022; 160:156049. [DOI: 10.1016/j.cyto.2022.156049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023]
|
16
|
Coupet CA, Dubois C, Evlachev A, Kehrer N, Baldazza M, Hofman S, Vierboom M, Martin P, Inchauspe G. Intravenous injection of a novel viral immunotherapy encoding human interleukin-7 in nonhuman primates is safe and increases absolute lymphocyte count. Hum Vaccin Immunother 2022; 18:2133914. [PMID: 36315906 PMCID: PMC9746448 DOI: 10.1080/21645515.2022.2133914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Persistence of an immunosuppression, affecting both the innate and adaptive arms of the immune system, plays a role in sepsis patients' morbidity and late mortality pointing to the need for broad and effective immune interventions. MVA-hIL-7-Fc is a non-replicative recombinant Modified Vaccinia virus Ankara encoding the human interleukin-7 fused to human IgG2 Fc fragment. We have shown in murine sepsis models the capacity of this new virotherapy to stimulate both arms of the immune system and increase survival. Herein, an exploratory study in nonhuman primates was performed following a single intravenous injection of the MVA-hIL-7-Fc used at the clinical dose to assess its safety and biological activities. Four cynomolgus macaques were followed for 3 weeks post-injection (p.i), without observed acute adverse reactions. Circulating hIL-7-Fc was detected during the first 3-5 days p.i with a detection peaking at 12 h p.i. IL-7 receptor engagement and downstream signal transduction were detected in T cells demonstrating functionality of the expressed IL-7. Expansion of blood lymphocytes, mainly CD4 and CD8 naïve and central memory T cells, was observed on day 7 p.i. together with a transient increase of Ki67 expression on T lymphocytes. In addition, we observed an increase in circulating B and NK cells as well as monocytes were albeit with different kinetics and levels. This study indicates that a vectorized IL-7-Fc, injected by intravenous route at a relevant clinical dose in a large animal model, is active without adverse reactions supporting the clinical development of this novel virotherapy for treatment of sepsis patients.
Collapse
Affiliation(s)
| | | | | | - Nadine Kehrer
- Infectious Diseases Department, Transgene SA, Lyon, France
| | - Marie Baldazza
- Infectious Diseases Department, Transgene SA, Lyon, France
| | - Sam Hofman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Michel Vierboom
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Perrine Martin
- Infectious Diseases Department, Transgene SA, Lyon, France
| | - Geneviève Inchauspe
- Infectious Diseases Department, Transgene SA, Lyon, France,CONTACT Geneviève Inchauspe Infectious Diseases department, Transgene SA, 317 Avenue Jean Jaures, Lyon69007, France
| |
Collapse
|
17
|
A single administration of hIL-7-hyFc induces long-lasting T-cell expansion with maintained effector functions. Blood Adv 2022; 6:6093-6107. [PMID: 36206199 PMCID: PMC9772483 DOI: 10.1182/bloodadvances.2021006591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 07/29/2022] [Accepted: 09/01/2022] [Indexed: 12/15/2022] Open
Abstract
Interleukin-7 (IL-7) is an essential cytokine for T-cell homeostatic proliferation and maintenance. Clinical studies have shown the potential benefits of IL-7 therapy in various diseases associated with lymphopenia. However, the kinetics of the T-cell response to a single administration of IL-7 in humans have not been fully elucidated. Here, we investigated the effects of Fc-fused long-acting recombinant human IL-7 (hIL-7-hyFc, efineptakin alfa) on lymphocytes in healthy adults after a single subcutaneous or intramuscular administration. Administration of hIL-7-hyFc increased the CD8+ and CD4+ T-cell numbers up to 2.5-fold, with corresponding upregulation of Ki-67 and Bcl-2 expression, peaking at day 3 or 7. Regulatory T cells (Tregs) did not expand. Among CD8+ and CD4+ T cells, all T-cell subsets (TN, TEM, TCM, TEMRA, and TSCM) increased for 56 days. The T-cell receptor repertoire diversity of naive CD8+ and CD4+ T cells was increased by hIL-7-hyFc, whereas the memory T-cell subsets did not differ between day 56 and day 0. Transcriptomic analysis revealed that hIL-7-hyFc induced robust T-cell expansion without changes in gene expression profiles associated with T-cell functions or genes related to T-cell exhaustion, senescence, and anergy. The effector functions of antigen-specific CD8+ T cells were preserved after hIL-7-hyFc administration. Our results suggest that hIL-7-hyFc administration induced a sustained increase in the numbers of CD8+ and CD4+ T cells, but not Tregs, without qualitative changes. These results support the potential of hIL-7-hyFc as a treatment for patients with compromised T-cell immunity or as a vaccine adjuvant.
Collapse
|
18
|
Crausaz M, Monneret G, Conti F, Lukaszewicz AC, Marchand JB, Martin P, Inchauspé G, Venet F. A novel virotherapy encoding human interleukin-7 improves ex vivo T lymphocyte functions in immunosuppressed patients with septic shock and critically ill COVID-19. Front Immunol 2022; 13:939899. [PMID: 36045686 PMCID: PMC9422896 DOI: 10.3389/fimmu.2022.939899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/19/2022] [Indexed: 12/21/2022] Open
Abstract
A majority of patients with sepsis surviving the first days in intensive care units (ICU) enter a state of immunosuppression contributing to their worsening. A novel virotherapy based on the non-propagative Modified Virus Ankara (MVA) expressing the human interleukin-7 (hIL-7) cytokine fused to an Fc fragment, MVA-hIL-7-Fc, was developed and shown to enhance innate and adaptive immunity and confer survival advantages in murine sepsis models. Here, we assessed the capacity of hIL-7-Fc produced by the MVA-hIL-7-Fc to improve ex vivo T lymphocyte functions from ICU patients with sepsis. Primary hepatocytes were transduced with the MVA-hIL-7-Fc or an empty MVA, and cell supernatants containing the secreted hIL-7-Fc were harvested for in vitro and ex vivo studies. Whole blood from ICU patients [septic shock = 15, coronavirus disease 2019 (COVID-19) = 30] and healthy donors (n = 36) was collected. STAT5 phosphorylation, cytokine production, and cell proliferation were assessed upon T cell receptor (TCR) stimulation in presence of MVA-hIL-7-Fc-infected cell supernatants. Cells infected by MVA-hIL-7-Fc produced a dimeric, glycosylated, and biologically active hIL-7-Fc. Cell supernatants containing the expressed hIL-7-Fc triggered the IL-7 pathway in T lymphocytes as evidenced by the increased STAT5 phosphorylation in CD3+ cells from patients and healthy donors. The secreted hIL-7-Fc improved Interferon-γ (IFN-γ) and/or Tumor necrosis factor-α (TNF-α) productions and CD4+ and CD8+ T lymphocyte proliferation after TCR stimulation in patients with bacterial and viral sepsis. This study demonstrates the capacity of the novel MVA-hIL-7-Fc-based virotherapy to restore ex vivo T cells immune functions in ICU patients with sepsis and COVID-19, further supporting its clinical development.
Collapse
Affiliation(s)
- Morgane Crausaz
- Department of Infectious Diseases, Transgene SA, Lyon, France
- EA 7426 Pathophysiology of injury-induced immunosuppression (PI3), Lyon 1 University/Hospices Civils de Lyon/bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Guillaume Monneret
- EA 7426 Pathophysiology of injury-induced immunosuppression (PI3), Lyon 1 University/Hospices Civils de Lyon/bioMérieux, Hôpital Edouard Herriot, Lyon, France
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, Lyon, France
| | - Filippo Conti
- EA 7426 Pathophysiology of injury-induced immunosuppression (PI3), Lyon 1 University/Hospices Civils de Lyon/bioMérieux, Hôpital Edouard Herriot, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 Pathophysiology of injury-induced immunosuppression (PI3), Lyon 1 University/Hospices Civils de Lyon/bioMérieux, Hôpital Edouard Herriot, Lyon, France
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service d'anesthésie-réanimation, Lyon, France
| | | | - Perrine Martin
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | | | - Fabienne Venet
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Laboratoire d'Immunologie, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France
| |
Collapse
|
19
|
Lélu K, Dubois C, Evlachev A, Crausaz M, Baldazza M, Kehrer N, Brandely R, Schlesinger Y, Silvestre N, Marchand JB, Bastien B, Leung-Theung-Long S, Unsinger J, Martin P, Inchauspé G. Viral Delivery of IL-7 Is a Potent Immunotherapy Stimulating Innate and Adaptive Immunity and Confers Survival in Sepsis Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:99-117. [PMID: 35667841 DOI: 10.4049/jimmunol.2101145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/25/2022] [Indexed: 12/29/2022]
Abstract
Persistence of an immunosuppressive state plays a role in septic patient morbidity and late mortality. Both innate and adaptive pathways are impaired, pointing toward the need for immune interventions targeting both arms of the immune system. We developed a virotherapy using the nonpropagative modified vaccinia virus Ankara (MVA), which harbors the intrinsic capacity to stimulate innate immunity, to deliver IL-7, a potent activator of adaptive immunity. The rMVA-human IL-7 (hIL-7)-Fc encoding the hIL-7 fused to the human IgG2-Fc was engineered and shown to express a dimeric, glycosylated, and biologically active cytokine. Following a single i.v. injection in naive mice, the MVA-hIL-7-Fc increased the number of total and activated B, T, and NK cells but also myeloid subpopulations (Ly6Chigh, Ly6Cint, and Ly6Cneg cells) in both lung and spleen. It triggered differentiation of T cells in central memory, effector memory, and acute effector phenotypes and enhanced polyfunctionality of T cells, notably the number of IFN-γ-producing cells. The MVA vector contributed significantly to immune cell activation, particularly of NK cells. The MVA-hIL-7-Fc conferred a significant survival advantage in the cecal ligation and puncture (CLP) and Candida albicans sepsis models. It significantly increased cell numbers and activation in both spleen and lung of CLP mice. Comparatively, in naive and CLP mice, the rhIL-7-Fc soluble counterpart overall induced less vigorous, shorter lasting, and narrower immune activities than did the MVA-hIL-7-Fc and favored TNF-α-producing cells. The MVA-hIL-7-Fc represents a novel class of immunotherapeutic with clinical potential for treatment of septic patients.
Collapse
Affiliation(s)
- Karine Lélu
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Clarisse Dubois
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Alexei Evlachev
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Morgane Crausaz
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Marie Baldazza
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Nadine Kehrer
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | - Renée Brandely
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | - Yasmin Schlesinger
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | - Nathalie Silvestre
- Department of Vectorology, Transgene SA, Illkirch-Graffenstraden, France
| | | | - Bérangère Bastien
- Department of Medical Affairs, Transgene SA, Illkirch-Graffenstraden, France
| | | | - Jacqueline Unsinger
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO.,Department of Medicine, Washington University School of Medicine, St. Louis, MO; and.,Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Perrine Martin
- Department of Infectious Diseases, Transgene SA, Lyon, France
| | | |
Collapse
|
20
|
Kim MY, Jayasinghe R, Devenport JM, Ritchey JK, Rettig MP, O'Neal J, Staser KW, Kennerly KM, Carter AJ, Gao F, Lee BH, Cooper ML, DiPersio JF. A long-acting interleukin-7, rhIL-7-hyFc, enhances CAR T cell expansion, persistence, and anti-tumor activity. Nat Commun 2022; 13:3296. [PMID: 35697686 PMCID: PMC9192727 DOI: 10.1038/s41467-022-30860-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/23/2022] [Indexed: 12/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is routinely used to treat patients with refractory hematologic malignancies. However, a significant proportion of patients experience suboptimal CAR T cell cytotoxicity and persistence that can permit tumor cell escape and disease relapse. Here we show that a prototype pro-lymphoid growth factor is able to enhance CAR T cell efficacy. We demonstrate that a long-acting form of recombinant human interleukin-7 (IL-7) fused with hybrid Fc (rhIL-7-hyFc) promotes proliferation, persistence and cytotoxicity of human CAR T cells in xenogeneic mouse models, and murine CAR T cells in syngeneic mouse models, resulting in long-term tumor-free survival. Thus, rhIL-7-hyFc represents a tunable clinic-ready adjuvant for improving suboptimal CAR T cell activity.
Collapse
Affiliation(s)
- Miriam Y Kim
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Reyka Jayasinghe
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica M Devenport
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie K Ritchey
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie O'Neal
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Karl W Staser
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Division of Dermatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Krista M Kennerly
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Alun J Carter
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Matthew L Cooper
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
21
|
Leilei Z, Kewen Z, Biao H, Fang H, Yigang W. The Role of Chemokine IL-7 in Tumor and Its Potential Antitumor Immunity. J Interferon Cytokine Res 2022; 42:243-250. [PMID: 35613386 DOI: 10.1089/jir.2021.0236] [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: 11/12/2022] Open
Abstract
Interleukin-7 (IL-7) is a cytokine belonging to the chemokine family. It plays a key role in the differentiation, development, and maturation of T lymphocytes and B lymphocytes, which is pivotal to adaptive immunity. In addition to its role in lymphocyte development, recent studies have indicated the antitumor functions of IL-7 in the tumor microenvironment. In this review, we discuss the role of IL-7 in tumors and summarize its antitumor potential and clinical application in lymphoma, leukemia, breast cancer, colon cancer, and so on. Furthermore, the combinational strategies of IL-7 and other antitumor drugs have been also discussed.
Collapse
Affiliation(s)
- Zhang Leilei
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
- Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China
| | - Zhou Kewen
- Department of Immunology, University of Toronto, Bachelor of Science, Toronto, Canada
| | - Huang Biao
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Huang Fang
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Wang Yigang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China
| |
Collapse
|
22
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
23
|
Campian JL, Ghosh S, Kapoor V, Yan R, Thotala S, Jash A, Hu T, Mahadevan A, Rifai K, Page L, Lee BH, Ferrando-Martinez S, Wolfarth AA, Yang SH, Hallahan D, Chheda MG, Thotala D. Long-acting recombinant human interleukin-7, NT-I7, increases cytotoxic CD8 + T cells and enhances survival in mouse glioma models. Clin Cancer Res 2022; 28:1229-1239. [PMID: 35031547 DOI: 10.1158/1078-0432.ccr-21-0947] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/27/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with glioblastoma (GBM) are treated with radiation therapy (RT) and temozolomide (TMZ). These treatments may cause prolonged systemic lymphopenia, which itself is associated with poor outcomes. NT-I7 is a long-acting IL-7 that expands CD4 and CD8 T cell numbers in humans and mice. We tested whether NT-I7 prevents systemic lymphopenia and improves survival in mouse models of GBM. EXPERIMENTAL DESIGN C57BL/6 mice bearing intracranial tumors (GL261 or CT2A) were treated with RT (1.8 Gy/day x 5 days), TMZ (33 mg/kg/day x 5 days), and/or NT-I7 (10 mg/kg on the final day of RT). We followed the mice for survival while serially analyzing levels of circulating T lymphocytes. We assessed regulatory T cells (Treg) and cytotoxic T lymphocytes in the tumor microenvironment, cervical lymph nodes, spleen, and thymus; and hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. RESULTS GBM tumor-bearing mice treated with RT+NT-I7 increased T lymphocytes in the lymph nodes, thymus, and spleen, enhanced IFNγ production, and decreased Treg cells in the tumor which was associated with a significant increase in survival. NT-I7 also enhanced central memory and effector memory CD8 T cells in lymphoid organs and tumor. Depleting CD8 T cells abrogated the effects of NT-I7. Furthermore, NT-I7 treatment decreased progenitor cells in the bone marrow. CONCLUSION In orthotopic glioma-bearing mice, NT-I7 mitigates radiation-related lymphopenia, increases cytotoxic CD8 T lymphocytes systemically and in the tumor, and improves survival. A phase I/II trial to evaluate NT-I7 in patients with high-grade gliomas is ongoing (NCT03687957).
Collapse
Affiliation(s)
| | - Subhajit Ghosh
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Vaishali Kapoor
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Ran Yan
- Washington University in St. Louis
| | | | | | - Tong Hu
- Medicine, Washington University in St. Louis
| | - Anita Mahadevan
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Kasem Rifai
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Logan Page
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Byung Ha Lee
- Translational Research Division, NeoImmuneTech, Inc
| | | | | | | | - Dennis Hallahan
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | | | - Dinesh Thotala
- Radiation Oncology, Washington University in St. Louis School of Medicine
| |
Collapse
|
24
|
Wolfarth AA, Dhar S, Goon JB, Ezeanya UI, Ferrando-Martínez S, Lee BH. Advancements of Common Gamma-Chain Family Cytokines in Cancer Immunotherapy. Immune Netw 2022; 22:e5. [PMID: 35291658 PMCID: PMC8901704 DOI: 10.4110/in.2022.22.e5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 12/01/2022] Open
Affiliation(s)
| | - Swati Dhar
- NeoImmuneTech, Inc., Rockville, MD 20850, USA
| | | | | | | | | |
Collapse
|
25
|
Chen D, Tang TX, Deng H, Yang XP, Tang ZH. Interleukin-7 Biology and Its Effects on Immune Cells: Mediator of Generation, Differentiation, Survival, and Homeostasis. Front Immunol 2021; 12:747324. [PMID: 34925323 PMCID: PMC8674869 DOI: 10.3389/fimmu.2021.747324] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Interleukin-7 (IL-7), a molecule known for its growth-promoting effects on progenitors of B cells, remains one of the most extensively studied cytokines. It plays a vital role in health maintenance and disease prevention, and the congenital deficiency of IL-7 signaling leads to profound immunodeficiency. IL-7 contributes to host defense by regulating the development and homeostasis of immune cells, including T lymphocytes, B lymphocytes, and natural killer (NK) cells. Clinical trials of recombinant IL-7 have demonstrated safety and potent immune reconstitution effects. In this article, we discuss IL-7 and its functions in immune cell development, drawing on a substantial body of knowledge regarding the biology of IL-7. We aim to answer some remaining questions about IL-7, providing insights essential for designing new strategies of immune intervention.
Collapse
Affiliation(s)
- Deng Chen
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting-Xuan Tang
- Class 1901, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Hai Deng
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang-Ping Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Division of Trauma and Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Smith PL, Piadel K, Dalgleish AG. Directing T-Cell Immune Responses for Cancer Vaccination and Immunotherapy. Vaccines (Basel) 2021; 9:1392. [PMID: 34960140 PMCID: PMC8708201 DOI: 10.3390/vaccines9121392] [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: 10/12/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/21/2022] Open
Abstract
Cancer vaccination and immunotherapy revolutionised the treatment of cancer, a result of decades of research into the immune system in health and disease. However, despite recent breakthroughs in treating otherwise terminal cancer, only a minority of patients respond to cancer immunotherapy and some cancers are largely refractive to immunotherapy treatment. This is due to numerous issues intrinsic to the tumour, its microenvironment, or the immune system. CD4+ and CD8+ αβ T-cells emerged as the primary effector cells of the anti-tumour immune response but their function in cancer patients is often compromised. This review details the mechanisms by which T-cell responses are hindered in the setting of cancer and refractive to immunotherapy, and details many of the approaches under investigation to direct T-cell function and improve the efficacy of cancer vaccination and immunotherapy.
Collapse
Affiliation(s)
- Peter Lawrence Smith
- Institute of Infection and Immunity, St. Georges University of London, London SW17 0RE, UK; (K.P.); (A.G.D.)
| | | | | |
Collapse
|
27
|
Borgoni S, Kudryashova KS, Burka K, de Magalhães JP. Targeting immune dysfunction in aging. Ageing Res Rev 2021; 70:101410. [PMID: 34280555 DOI: 10.1016/j.arr.2021.101410] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/28/2021] [Accepted: 07/14/2021] [Indexed: 12/23/2022]
Abstract
Human aging is a multifactorial phenomenon that affects numerous organ systems and cellular processes, with the immune system being one of the most dysregulated. Immunosenescence, the gradual deterioration of the immune system, and inflammaging, a chronic inflammatory state that persists in the elderly, are among the plethora of immune changes that occur during aging. Almost all populations of immune cells change with age in terms of numbers and/or activity. These alterations are in general highly detrimental, resulting in an increased susceptibility to infections, reduced healing abilities, and altered homeostasis that promote the emergence of age-associated diseases such as cancer, diabetes, and other diseases associated with inflammation. Thanks to recent developments, several strategies have been proposed to target central immunological processes or specific immune subpopulations affected by aging. These therapeutic approaches could soon be applied in the clinic to slow down or even reverse specific age-induced immune changes in order to rejuvenate the immune system and prevent or reduce the impact of various diseases. Due to its systemic nature and interconnection with all the other systems in the body, the immune system is an attractive target for aging intervention because relatively targeted modifications to a small set of cells have the potential to improve the health of multiple organ systems. Therefore, anti-aging immune targeting therapies could represent a potent approach for improving healthspan. Here, we review aging changes in the major components of the immune system, we summarize the current immune-targeting therapeutic approaches in the context of aging and discuss the future directions in the field of immune rejuvenation.
Collapse
|
28
|
Nakao S, Arai Y, Tasaki M, Yamashita M, Murakami R, Kawase T, Amino N, Nakatake M, Kurosaki H, Mori M, Takeuchi M, Nakamura T. Intratumoral expression of IL-7 and IL-12 using an oncolytic virus increases systemic sensitivity to immune checkpoint blockade. Sci Transl Med 2021; 12:12/526/eaax7992. [PMID: 31941828 DOI: 10.1126/scitranslmed.aax7992] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/16/2019] [Accepted: 12/16/2019] [Indexed: 12/18/2022]
Abstract
The immune status of the tumor microenvironment is a key indicator in determining the antitumor effectiveness of immunotherapies. Data support the role of activation and expansion of tumor-infiltrating lymphocytes (TILs) in increasing the benefit of immunotherapies in patients with solid tumors. We found that intratumoral injection of a tumor-selective oncolytic vaccinia virus encoding interleukin-7 (IL-7) and IL-12 into tumor-bearing immunocompetent mice activated the inflammatory immune status of previously poorly immunogenic tumors and resulted in complete tumor regression, even in distant tumor deposits. Mice achieving complete tumor regression resisted rechallenge with the same tumor cells, suggesting establishment of long-term tumor-specific immune memory. Combining this virotherapy with anti-programmed cell death-1 (PD-1) or anti-cytotoxic T lymphocyte antigen 4 (CTLA4) antibody further increased the antitumor activity as compared to virotherapy alone, in tumor models unresponsive to either of the checkpoint inhibitor monotherapies. These findings suggest that administration of an oncolytic vaccinia virus carrying genes encoding for IL-7 and IL-12 has antitumor activity in both directly injected and distant noninjected tumors through immune status changes rendering tumors sensitive to immune checkpoint blockade. The benefit of intratumoral IL-7 and IL-12 expression was also observed in humanized mice bearing human cancer cells. These data support further investigation in patients with non-inflamed solid tumors.
Collapse
Affiliation(s)
- Shinsuke Nakao
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan.
| | - Yukinori Arai
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Mamoru Tasaki
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Midori Yamashita
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Ryuji Murakami
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Tatsuya Kawase
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Nobuaki Amino
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Motomu Nakatake
- Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Hajime Kurosaki
- Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| | - Masamichi Mori
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Masahiro Takeuchi
- Drug Discovery Research, Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba 305-8585, Japan
| | - Takafumi Nakamura
- Department of Biomedical Science, Graduate School of Medical Sciences, Tottori University, 86 Nishi-cho, Yonago 683-8503, Japan
| |
Collapse
|
29
|
Abstract
Clinical trials have demonstrated that an increased number of effector cells, especially tumor-specific T cells, is positively linked with patients’ prognosis. Although the discovery of checkpoint inhibitors (CPIs) has led to encouraging progress in cancer immunotherapy, the lack of either T cells or targets for CPIs is a limitation for patients with poor prognosis. Since interleukin (IL)-2 and IL-7 are cytokines that target many aspects of T-cell responses, they have been used to treat cancers. In this review, we focus on the basic biology of how these cytokines regulate T-cell response and on the clinical trials using the cytokines against cancer. Further, we introduce several recent studies that aim to improve cytokines’ biological activities and find the strategy for combination with other therapeutics.
Collapse
Affiliation(s)
- Ji-Hae Kim
- Department of Life Sciences, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| | - Kun-Joo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| |
Collapse
|
30
|
Koukourakis MI, Giatromanolaki A. Lymphopenia and intratumoral lymphocytic balance in the era of cancer immuno-radiotherapy. Crit Rev Oncol Hematol 2021; 159:103226. [PMID: 33482348 DOI: 10.1016/j.critrevonc.2021.103226] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/15/2020] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION The immune response has been recognized as a major tumor-eradication component of radiotherapy. OBJECTIVE This review studies, under a clinical perspective, two contrasting effects of radiotherapy, namely immunosuppression and radiovaccination. MATERIALS AND METHODS We critically reviewed the available clinical and experimental experience on radiotherapy-induced lymphopenia. RESULTS Radiation-induced tumor damage promotes radio-vaccination, enhances cytotoxic immune responses, and potentiates immunotherapy. Nevertheless, radiotherapy induces systemic and intratumoral lymphopenia. The above effects are directly related to radiotherapy fractionation and field size/location, and tumor characteristics. DISCUSSION Hypofractionated stereotactic and accelerated irradiation better promotes radio-vaccination and produces less severe lymphopenia. Adopting cytoprotective policies and combining lympho-stimulatory agents or agents blocking regulatory lymphocyte activity are awaited to unmask the radio-vaccination effect, enhancing the efficacy immuno-radiotherapy. CONCLUSION Radiation-induced lymphopenia and immunosuppression are important issues that should be considered in the design of immuno-radiotherapy clinical trials.
Collapse
Affiliation(s)
- Michael I Koukourakis
- Department of Radiotherapy/Oncology, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece.
| | - Alexandra Giatromanolaki
- Department of Pathology, Medical School, Democritus University of Thrace, Alexandroupolis 68100, Greece
| |
Collapse
|
31
|
Abstract
Following periods of haematopoietic cell stress, such as after chemotherapy, radiotherapy, infection and transplantation, patient outcomes are linked to the degree of immune reconstitution, specifically of T cells. Delayed or defective recovery of the T cell pool has significant clinical consequences, including prolonged immunosuppression, poor vaccine responses and increased risks of infections and malignancies. Thus, strategies that restore thymic function and enhance T cell reconstitution can provide considerable benefit to individuals whose immune system has been decimated in various settings. In this Review, we focus on the causes and consequences of impaired adaptive immunity and discuss therapeutic strategies that can recover immune function, with a particular emphasis on approaches that can promote a diverse repertoire of T cells through de novo T cell formation.
Collapse
|
32
|
Xu H, Lin M, Hu Y, Zhang L, Li Q, Zhu J, Wang S, Xi M. Lymphopenia During Definitive Chemoradiotherapy in Esophageal Squamous Cell Carcinoma: Association with Dosimetric Parameters and Patient Outcomes. Oncologist 2020; 26:e425-e434. [PMID: 32960471 DOI: 10.1002/onco.13533] [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] [Received: 02/05/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The objective of this study was to investigate the relationship between clinical characteristics, as well as dosimetric parameters, and the risk of treatment-related lymphopenia in esophageal squamous cell carcinoma (ESCC) treated with definitive chemoradiotherapy (CRT). MATERIALS AND METHODS Clinical characteristics and dosimetric parameters were collected from 436 patients with ESCC who received definitive CRT from 2010 through 2017. Absolute lymphocyte counts (ALCs) were obtained before, during, and 1 month after CRT. Grade 4 (G4) lymphopenia was defined as ALC <0.2 × 109 /L during CRT. Logistic regression analysis was used to evaluate the effect of each factor on predicting G4 lymphopenia. The relationship between lymphopenia and overall survival (OS) was examined, and a nomogram was developed to predict OS. RESULTS G4 lymphopenia was observed in 103 patients (23.6%) during CRT. Multivariate analysis indicated that planning target volume (PTV), lung V10 , heart V10 , performance status, and pretreatment lymphopenia were significant risk factors for G4 lymphopenia. Patients with G4 lymphopenia had significantly worse survival than those without. Based on multivariate analysis, clinical TNM stage, radiotherapy modality, pretreatment ALC, and G4 lymphopenia were predictive of OS and were incorporated into the nomogram, yielding a concordance index of 0.71. CONCLUSIONS G4 lymphopenia during definitive CRT was associated with larger PTVs, higher lung V10 and heart V10 , and worse survival. IMPLICATIONS FOR PRACTICE The purpose of this study was to investigate the relationship between clinical characteristics, as well as dosimetric parameters, and the risk of treatment-related lymphopenia in 436 patients with esophageal squamous cell carcinoma who received definitive chemoradiotherapy. Grade 4 (G4) lymphopenia was observed in 23.6% of patients during radiotherapy. G4 lymphopenia was associated with larger planning target volumes, higher lung V10 and heart V10 , and worse survival. Then, a nomogram was built based on multivariate analysis, yielding excellent performance to predict overall survival. Prospective studies are needed to investigate potential approaches for mitigating severe lymphopenia, which may ultimately convert into survival benefits.
Collapse
Affiliation(s)
- Hui Xu
- Department of Head and Neck Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, People's Republic of China
| | - Maosheng Lin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, People's Republic of China.,Departments of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yingying Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, People's Republic of China.,Department of Nuclear Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, People's Republic of China.,Departments of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Qiaoqiao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, People's Republic of China.,Departments of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jinhan Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, People's Republic of China.,Departments of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Shi Wang
- Department of Engineering Physics, Tsinghua University, Beijing, People's Republic of China
| | - Mian Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, People's Republic of China.,Departments of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| |
Collapse
|
33
|
Kim JH, Kim YM, Choi D, Jo SB, Park HW, Hong SW, Park S, Kim S, Moon S, You G, Kang YW, Park Y, Lee BH, Lee SW. Hybrid Fc-fused interleukin-7 induces an inflamed tumor microenvironment and improves the efficacy of cancer immunotherapy. Clin Transl Immunology 2020; 9:e1168. [PMID: 32994996 PMCID: PMC7507498 DOI: 10.1002/cti2.1168] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/07/2020] [Accepted: 08/02/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Emerging oncotherapeutic strategies require the induction of an immunostimulatory tumor microenvironment (TME) containing numerous tumor‐reactive CD8+ T cells. Interleukin‐7 (IL‐7), a T‐cell homeostatic cytokine, induces an antitumor response; however, the detailed mechanisms underlying the contributions of the IL‐7 to TME remain unclear. Here, we aimed to investigate the mechanism underlying the induction of antitumor response by hybrid Fc‐fused long‐acting recombinant human IL‐7 (rhIL‐7‐hyFc) through regulation of both adaptive and innate immune cells in the TME. Methods We evaluated rhIL‐7‐hyFc‐mediated antitumor responses in murine syngeneic tumor models. We analysed the cellular and molecular features of tumor‐infiltrating lymphocytes (TILs) and changes in the TME after rhIL‐7‐hyFc treatment. Furthermore, we evaluated the antitumor efficacy of rhIL‐7‐hyFc combined with chemotherapy and checkpoint inhibitors (CPIs). Results Systemic delivery of rhIL‐7‐hyFc induced significant therapeutic benefits by expanding CD8+ T cells with enhanced tumor tropism. In tumors, rhIL‐7‐hyFc increased both tumor‐reactive and bystander CD8+ TILs, all of which displayed enhanced effector functions but less exhausted phenotypes. Moreover, rhIL‐7‐hyFc suppressed the generation of immunosuppressive myeloid cells in the bone marrow of tumor‐bearing mice, resulting in the immunostimulatory TME. Combination therapy with chemotherapy and CPIs, rhIL‐7‐hyFc elicited a strong antitumor response and even under a T lymphopenic condition by restoring CD8+ T cells. When combined with chemotherapy and CPIs, rhIL‐7‐hyFc administration enhanced antitumor response under intact andlymphopenic conditions by restoring CD8+ T cells. Conclusion Taken together, these data demonstrate that rhIL‐7‐hyFc induces antitumor responses by generating T‐cell‐inflamed TME and provide a preclinical proof of concept of immunotherapy with rhIL‐7‐hyFc to enhance therapeutic responses in the clinic.
Collapse
Affiliation(s)
- Ji-Hae Kim
- Laboratory of Cellular Immunology Department of Life Sciences Pohang University of Science and Technology Pohang Korea
| | - Young-Min Kim
- Laboratory of Cellular Immunology Department of Life Sciences Pohang University of Science and Technology Pohang Korea
| | - Donghoon Choi
- Research Institute of NeoImmuneTech, Inc. Rockville MD USA
| | - Saet-Byeol Jo
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Han Wook Park
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Sung-Wook Hong
- Laboratory of T Cell Biology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea.,Department of Microbiology and Immunology Center for Immunology University of Minnesota Medical School Minneapolis MN USA
| | - Sujeong Park
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Sora Kim
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Sookjin Moon
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Gihoon You
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Yeon-Woo Kang
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Yunji Park
- Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| | - Byung Ha Lee
- Research Institute of NeoImmuneTech, Inc. Rockville MD USA
| | - Seung-Woo Lee
- Laboratory of Cellular Immunology Department of Life Sciences Pohang University of Science and Technology Pohang Korea.,Laboratory of Cellular Immunology Division of Integrative Biosciences and Biotechnology Pohang University of Science and Technology Pohang Korea
| |
Collapse
|
34
|
Courivaud C, Bamoulid J, Crepin T, Gaiffe E, Laheurte C, Saas P, Ducloux D. Pre-transplant Thymic Function Predicts Is Associated With Patient Death After Kidney Transplantation. Front Immunol 2020; 11:1653. [PMID: 32903778 PMCID: PMC7438875 DOI: 10.3389/fimmu.2020.01653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Accelerated thymic involution is a main feature of end-stage renal disease (ESRD)-associated immune senescence. Recent evidences suggest that ESRD-associated immune senescence is associated with adverse outcomes in dialysis patients. However, no study focused on the association between pre-transplant thymic function and patient survival after transplantation. We conducted a prospective, multicenter study to assess whether pre-transplant thymic function measured by recent thymic emigrants (RTE) may predict death after first kidney transplantation. Results were tested in a validation cohort. Nine hundred and sixty-seven incident kidney transplant recipients were included in the prospective study. Mean follow up was 5.1 + 2.9 years. Eighty two patients (8.5%) died during follow up. Lower RTE levels were associated with a higher risk of death (2.53; 95%CI, 1.54–4.39 for each decrease of 1 log in RTE; p < 0.001). Cancer-related death was particularly increased in patients with low RTE levels (4.23; 95%CI, 1.43–12.13; p = 0.007). One hundred and thirty-six patients having received a first kidney transplantation were included in the validation cohort. Lower TREC levels were associated with higher risk of death (1.90; 95%CI, 1.11–3.51 for each decrease of 1 log in RTE; p = 0.025). RTE were not associated with death-censored graft loss. Pre-transplant thymic function is strongly associated with death after transplantation. Attempt to reverse ESRD-related thymic loss may prevent premature death.
Collapse
Affiliation(s)
- Cécile Courivaud
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Jamal Bamoulid
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Thomas Crepin
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France
| | - Emilie Gaiffe
- CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France.,CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France
| | - Caroline Laheurte
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Philippe Saas
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France.,EFS Bourgogne Franche-Comté, Plateforme de Biomonitoring, CIC 1431/UMR1098, Besançon, France
| | - Didier Ducloux
- Inserm, UMR1098, Federation Hospitalo-Universitaire INCREASE, Besançon, France.,Univ. Bourgogne Franche-Comté, Faculté de Médecine et de Pharmacie, LabEx LipSTIC, Besançon, France.,Structure Fédérative de Recherche, SFR FED4234, Besançon, France.,CHU Besançon, Department of Nephrology, Dialysis, and Renal Transplantation, Besançon, France.,CHU Besançon, CIC Biothérapie, INSERM CIC1431, Besançon, France
| |
Collapse
|
35
|
Marković I, Savvides SN. Modulation of Signaling Mediated by TSLP and IL-7 in Inflammation, Autoimmune Diseases, and Cancer. Front Immunol 2020; 11:1557. [PMID: 32849527 PMCID: PMC7396566 DOI: 10.3389/fimmu.2020.01557] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/12/2020] [Indexed: 12/30/2022] Open
Abstract
Thymic Stromal Lymphopoietin (TSLP) and Interleukin-7 (IL-7) are widely studied cytokines within distinct branches of immunology. On one hand, TSLP is crucially important for mediating type 2 immunity at barrier surfaces and has been linked to widespread allergic and inflammatory diseases of the airways, skin, and gut. On the other hand, IL-7 operates at the foundations of T-cell and innate lymphoid cell (ILC) development and homeostasis and has been associated with cancer. Yet, TSLP and IL-7 are united by key commonalities in their structure and the structural basis of the receptor assemblies they mediate to initiate cellular signaling, in particular their cross-utilization of IL-7Rα. As therapeutic targeting of TSLP and IL-7 via diverse approaches is reaching advanced stages and in light of the plethora of mechanistic and structural data on receptor signaling mediated by the two cytokines, the time is ripe to provide integrated views of such knowledge. Here, we first discuss the major pathophysiological roles of TSLP and IL-7 in autoimmune diseases, inflammation and cancer. Subsequently, we curate structural and mechanistic knowledge about receptor assemblies mediated by the two cytokines. Finally, we review therapeutic avenues targeting TSLP and IL-7 signaling. We envision that such integrated view of the mechanism, structure, and modulation of signaling assemblies mediated by TSLP and IL-7 will enhance and fine-tune the development of more effective and selective approaches to further interrogate the role of TSLP and IL-7 in physiology and disease.
Collapse
Affiliation(s)
- Iva Marković
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Savvas N Savvides
- VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
36
|
Delrieu L, Anota A, Trédan O, Freyssenet D, Maire A, Canada B, Fournier B, Febvey-Combes O, Pilleul F, Bouhamama A, Caux C, Joly F, Fervers B, Pialoux V, Pérol D, Pérol O. Design and methods of a national, multicenter, randomized and controlled trial to assess the efficacy of a physical activity program to improve health-related quality of life and reduce fatigue in women with metastatic breast cancer: ABLE02 trial. BMC Cancer 2020; 20:622. [PMID: 32620149 PMCID: PMC7333295 DOI: 10.1186/s12885-020-07093-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Patients with a metastatic breast cancer suffer from a deteriorated health-related quality of life and numerous symptoms such as pain, severe fatigue and a decrease of their physical fitness. As the feasibility of a physical activity program has been demonstrated in this population, ABLE02 aims to assess the efficacy of a 6 month-physical activity program using connected devices to improve health-related quality of life and to reduce fatigue in women with metastatic breast cancer. METHODS ABLE02 is a prospective, national, multicenter, randomized, controlled and open-label study. A total of 244 patients with a metastatic breast cancer, with at least one positive hormone receptor and a first-line chemotherapy planned, will be randomly assigned (1:1 ratio) to: (i) the intervention arm to receive physical activity recommendations, an activity tracker to wear 24 h a day during the whole intervention (6 months) with at least three weekly walking sessions and quizzes each week on physical activity and nutrition (ii) the control arm to receive physical activity recommendations only. Health-related quality of life will be assessed every 6 weeks and main assessments will be conducted at baseline, M3, M6, M12 and M18 to evaluate the clinical, physical, biological and psychological parameters and survival of participants. All questionnaires will be completed on a dedicated application. DISCUSSION An activity program based on a smartphone application linked to an activity tracker may help to improve quality of life and reduce fatigue of patients with a metastatic breast cancer. The growth of e-health offers the opportunity to get real-time data as well as improving patient empowerment in order to change long-term behaviors. TRIAL REGISTRATION NCT number: NCT04354233 .
Collapse
Affiliation(s)
- Lidia Delrieu
- Department of Cancer and Environment, Léon Bérard Cancer Center, 28 rue Laennec, 69008, Lyon, France
- Inter-University Laboratory of Human Movement Biology EA7424, University Claude Bernard Lyon 1, University of Lyon, Villeurbanne, France
| | - Amélie Anota
- Methodology and Quality of Life in Oncology unit (INSERM UMR 1098), University Hospital of Besançon, Besançon, France
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, F-25000, Besançon, France
- French National Platform Quality of Life and Cancer, Besançon, France
| | - Olivier Trédan
- Department of Medical Oncology, Léon Bérard Cancer Center, Lyon, France
| | - Damien Freyssenet
- Inter-University Laboratory of Human Movement Biology EA7424, Univ Lyon, University Jean Monnet Saint-Etienne, Saint-Etienne, France
| | - Aurélia Maire
- Department of Cancer and Environment, Léon Bérard Cancer Center, 28 rue Laennec, 69008, Lyon, France
| | - Brice Canada
- Laboratory on Vulnerabilities and Innovations in Sport, University Claude Bernard Lyon 1, University of Lyon, Villeurbanne, France
| | - Baptiste Fournier
- Department of Cancer and Environment, Léon Bérard Cancer Center, 28 rue Laennec, 69008, Lyon, France
| | - Olivia Febvey-Combes
- Department of Clinical Research and Innovation, Léon Bérard Cancer Center, Lyon, France
| | - Frank Pilleul
- Department of Interventional Radiology, Léon Bérard Cancer Center, Lyon, France
| | - Amine Bouhamama
- Department of Interventional Radiology, Léon Bérard Cancer Center, Lyon, France
| | - Christophe Caux
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon (CRCL), Léon Bérard Cancer Center, Lyon, France
| | - Florence Joly
- Medical Oncology Department, Centre François Baclesse, Caen, France
- INSERM, U1086, ANTICIPE, Caen, France
- Cancer & Cognition, Platform, Ligue Contre le Cancer, CHU de Caen, Caen, France
| | - Béatrice Fervers
- Department of Cancer and Environment, Léon Bérard Cancer Center, 28 rue Laennec, 69008, Lyon, France
- INSERM UA8, Léon Bérard Cancer Center, Lyon, France
| | - Vincent Pialoux
- Inter-University Laboratory of Human Movement Biology EA7424, University Claude Bernard Lyon 1, University of Lyon, Villeurbanne, France
| | - David Pérol
- Department of Clinical Research and Innovation, Léon Bérard Cancer Center, Lyon, France
| | - Olivia Pérol
- Department of Cancer and Environment, Léon Bérard Cancer Center, 28 rue Laennec, 69008, Lyon, France.
- INSERM UA8, Léon Bérard Cancer Center, Lyon, France.
| |
Collapse
|
37
|
Lambin P, Lieverse RIY, Eckert F, Marcus D, Oberije C, van der Wiel AMA, Guha C, Dubois LJ, Deasy JO. Lymphocyte-Sparing Radiotherapy: The Rationale for Protecting Lymphocyte-rich Organs When Combining Radiotherapy With Immunotherapy. Semin Radiat Oncol 2020; 30:187-193. [PMID: 32381298 PMCID: PMC8412054 DOI: 10.1016/j.semradonc.2019.12.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is now strong clinical and preclinical evidence that lymphocytes, for example, CD8+ T cells, are key effectors of immunotherapy and that irradiation of large blood vessels, the heart, and lymphoid organs (including nodes, spleen, bones containing bone marrow, and thymus in children) causes transient or persistent lymphopenia. Furthermore, there is extensive clinical evidence, across multiple cancer sites and treatment modalities, that lymphopenia correlates strongly with decreased overall survival. At the moment, we lack quantitative evidence to establish the relationship between dose-volume and dose-rate to critical normal structures and lymphopenia. Therefore, we propose that data should be systematically recorded to characterise a possible quantitative relationship. This might enable us to improve the efficacy of radiotherapy and develop strategies to predict and prevent treatment-related lymphopenia. In anticipation of more quantitative data, we recommend the application of the principle of As Low As Reasonably Achievable to lymphocyte-rich regions for radiotherapy treatment planning to reduce the radiation doses to these structures, thus moving toward "Lymphocyte-Sparing Radiotherapy."
Collapse
Affiliation(s)
- Philippe Lambin
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands.
| | - Relinde I Y Lieverse
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Franziska Eckert
- Department of Radiation Oncology, University Hospital and Medical Faculty Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Damiënne Marcus
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Cary Oberije
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Alexander M A van der Wiel
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center, Bronx, NY
| | - Ludwig J Dubois
- The D-Lab and The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
38
|
Boyer-Suavet S, Cremoni M, Dupeyrat T, Zorzi K, Brglez V, Benzaken S, Esnault V, Seitz-Polski B. Functional immune assay using interferon-gamma could predict infectious events in end-stage kidney disease. Clin Chim Acta 2020; 502:287-292. [DOI: 10.1016/j.cca.2019.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 12/19/2022]
|
39
|
Flip the coin: IL-7 and IL-7R in health and disease. Nat Immunol 2019; 20:1584-1593. [PMID: 31745336 DOI: 10.1038/s41590-019-0479-x] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
The cytokine IL-7 and its receptor, IL-7R, are critical for T cell and, in the mouse, B cell development, as well as differentiation and survival of naive T cells, and generation and maintenance of memory T cells. They are also required for innate lymphoid cell (ILC) development and maintenance, and consequently for generation of lymphoid structures and barrier defense. Here we discuss the central role of IL-7 and IL-7R in the lymphoid system and highlight the impact of their deregulation, placing a particular emphasis on their 'dark side' as promoters of cancer development. We also explore therapeutic implications and opportunities associated with either positive or negative modulation of the IL-7-IL-7R signaling axis.
Collapse
|
40
|
Ménétrier-Caux C, Ray-Coquard I, Blay JY, Caux C. Lymphopenia in Cancer Patients and its Effects on Response to Immunotherapy: an opportunity for combination with Cytokines? J Immunother Cancer 2019; 7:85. [PMID: 30922400 PMCID: PMC6437964 DOI: 10.1186/s40425-019-0549-5] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/27/2019] [Indexed: 12/12/2022] Open
Abstract
Quantitative lymphocyte alterations are frequent in patients with cancer, and strongly impact prognosis and survival. The development of cancers in immunosuppressed patients has demonstrated the contribution of different T cell populations, including CD4+ cells, in the control of cancer occurrence.Whereas absolute numbers of neutrophils, platelets and red blood cells are routinely monitored in clinic following treatments, because of possible short-term complications, absolute lymphocyte counts (ALC), their subpopulations or diversity (phenotype, TCR) are rarely analyzed and never used to choose therapy or as prognostic criteria. The recent identification of immune checkpoint inhibitors (ICPi) as powerful therapeutic agents has revitalized immunotherapy of cancer in a broader group of diseases than anticipated. The status of the immune system is now recognized as an important biomarker for response to these novel treatments. Blood ALC values, along with tumor infiltration by CD8+T cells, and ICPi and ICPi-ligand expression, are likely to be a potential marker of sensitivity to anti-ICPi therapy.In this article, we review the current knowledge on the incidence and significance of lymphopenia in cancer patients, and discuss therapeutic strategies to restore lymphocyte numbers.
Collapse
Affiliation(s)
- Christine Ménétrier-Caux
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon (CRCL), Centre Léon Bérard, F-69008, Lyon, France. .,Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Centre Léon Bérard, F-69008, Lyon, France.
| | | | - Jean-Yves Blay
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon (CRCL), Centre Léon Bérard, F-69008, Lyon, France.,Medical Oncology department, Centre Léon Bérard, F-69008, Lyon, France
| | - Christophe Caux
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Cancer Research Center of Lyon (CRCL), Centre Léon Bérard, F-69008, Lyon, France.,Innovation in Immuno-monitoring and Immunotherapy Platform (PI3), Centre Léon Bérard, F-69008, Lyon, France
| |
Collapse
|
41
|
Therapeutic Vaccines for Genitourinary Malignancies. Vaccines (Basel) 2018; 6:vaccines6030055. [PMID: 30103542 PMCID: PMC6161030 DOI: 10.3390/vaccines6030055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022] Open
Abstract
The field of genitourinary malignancies has been a showcase for therapeutic cancer vaccine success since the application of intravesicular Bacillus Calmette-Guerin (BCG) for bladder cancer in the 1970s and enjoyed a renaissance in 2010 with the US Food and Drug Administration (FDA) approval of sipuleucel-T for prostate cancer. Several vaccine strategies have emerged, such as autologous or allogeneic whole-tumor vaccines, DNA vaccines, use of viral vectors, and peptides as immunostimulatory adjuvants. Despite impressive early trials, vaccine monotherapy has achieved limited success in the clinical world; however, combinations of vaccine and immune checkpoint inhibition or vaccine and cytokine stimulation are expected to move the field forward. This article reviews pivotal trials of cancer vaccines in prostate, renal, and bladder cancer and ongoing trials combining vaccines with other immune therapy agents.
Collapse
|
42
|
Van Acker HH, Campillo-Davo D, Roex G, Versteven M, Smits EL, Van Tendeloo VF. The role of the common gamma-chain family cytokines in γδ T cell-based anti-cancer immunotherapy. Cytokine Growth Factor Rev 2018; 41:54-64. [PMID: 29773448 DOI: 10.1016/j.cytogfr.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 12/28/2022]
Abstract
Cytokines of the common gamma-chain receptor family, comprising interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15 and IL-21, are vital with respect to organizing and sustaining healthy immune cell functions. Supporting the anti-cancer immune response, these cytokines inspire great interest for their use as vaccine adjuvants and cancer immunotherapies. It is against this background that gamma delta (γδ) T cells, as special-force soldiers and natural contributors of the tumor immunosurveillance, also received a lot of attention the last decade. As γδ T cell-based cancer trials are coming of age, this present review focusses on the effects of the different cytokines of the common gamma-chain receptor family on γδ T cells with respect to boosting γδ T cells as a therapeutic target in cancer immunotherapy. This review also gathers data that IL-15 in particular exhibits key features for augmenting the anti-tumor activity of effector killer γδ T cells whilst overcoming the myriad of immune escape mechanisms used by cancer cells.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Gils Roex
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Maarten Versteven
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium; Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| |
Collapse
|
43
|
Wrangle JM, Patterson A, Johnson CB, Neitzke DJ, Mehrotra S, Denlinger CE, Paulos CM, Li Z, Cole DJ, Rubinstein MP. IL-2 and Beyond in Cancer Immunotherapy. J Interferon Cytokine Res 2018; 38:45-68. [PMID: 29443657 PMCID: PMC5815463 DOI: 10.1089/jir.2017.0101] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/26/2017] [Indexed: 12/11/2022] Open
Abstract
The development of the T- and natural killer (NK) cell growth factor IL-2 has been a sentinel force ushering in the era of immunotherapy in cancer. With the advent of clinical grade recombinant IL-2 in the mid-1980s, oncologists could for the first time directly manipulate lymphocyte populations with systemic therapy. By itself, recombinant IL-2 can induce clinical responses in up to 15% of patients with metastatic cancer or renal cell carcinoma. When administered with adoptively transferred tumor-reactive lymphocytes, IL-2 promotes T cell engraftment and response rates of up to 50% in metastatic melanoma patients. Importantly, these IL-2-driven responses can yield complete and durable responses in a subset of patients. However, the use of IL-2 is limited by toxicity and concern of the expansion of T regulatory cells. To overcome these limitations and improve response rates, other T cell growth factors, including IL-15 and modified forms of IL-2, are in clinical development. Administering T cell growth factors in combination with other agents, such as immune checkpoint pathway inhibitors, may also improve efficacy. In this study, we review the development of T- and NK cell growth factors and highlight current combinatorial approaches based on these reagents.
Collapse
Affiliation(s)
- John M. Wrangle
- Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Alicia Patterson
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - C. Bryce Johnson
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Daniel J. Neitzke
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Chadrick E. Denlinger
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Chrystal M. Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| | - David J. Cole
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Mark P. Rubinstein
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
44
|
Aspinall R, Lang PO. Interventions to restore appropriate immune function in the elderly. IMMUNITY & AGEING 2018; 15:5. [PMID: 29416551 PMCID: PMC5785902 DOI: 10.1186/s12979-017-0111-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/28/2017] [Indexed: 01/03/2023]
Abstract
Advanced age is one indicator of likely immune dysfunction. As worldwide, the global population contains progressively more and more older individuals there is likelihood of an increased prevalence and incidence of infectious diseases due to common and emergent pathogens. The resultant increase in mortality and morbidity would be matched by the risk of functional decline and disability. Maintaining immune function at a plateau throughout life may therefore be associated with considerable cost savings. The aim of improving immune function in older individuals may be achieved through considering a therapeutic approach to rejuvenate, stimulate or support the indigenous immune system to perform in a more optimal manner. In terms of cost effectiveness a therapeutic approach may prove difficult because of issues associated with; identifying those who would benefit the most from this treatment, identifying the type of treatment which would suit them and identifying whether the treatment was successful. The alternative of supporting or providing a stronger stimulus through vaccination, whilst more cost effective, may be a more valuable option in the short term. Both approaches will be addressed in this review.
Collapse
Affiliation(s)
- Richard Aspinall
- Rivock Ltd, Bury St Edmunds, UK.,3Anglia Ruskin University, Cambridge, UK
| | - Pierre Olivier Lang
- 2Geriatric and Geriatric Rehabilitation Division, Department of Medicine, University Hospital of Lausanne, Lausanne, Switzerland.,3Anglia Ruskin University, Cambridge, UK
| |
Collapse
|
45
|
Interleukin-7 in the transition of bone marrow progenitors to the thymus. Immunol Cell Biol 2017; 95:916-924. [PMID: 28811625 DOI: 10.1038/icb.2017.68] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/03/2023]
Abstract
Interleukin-7 (IL-7) is essential for the development of T cells in humans and mice where deficiencies in IL-7 signaling result in severe immunodeficiency. T cells require IL-7 at multiple points during development; however, it is unclear when IL-7 is first necessary. We observed that mice with impaired IL-7 signaling had a large reduction in the number of early thymic progenitors (ETPs) while mice that overexpress IL-7 had greatly increased numbers of ETPs. These results indicated that the development of ETPs is sensitive to IL-7. Bone marrow progenitors of ETP are present in normal numbers in mice with impaired IL-7 signaling (IL-7Rα449F) and were efficiently recruited to the thymus. Furthermore, ETPs and their progenitors from IL-7Rα449F mice did not undergo increased apoptosis and proliferate normally compared to WT cells. Mixed bone marrow chimeras demonstrated that IL-7 signaling has a cell-intrinsic role in ETP development but was not required for development of bone marrow progenitors. We have shown a novel role for IL-7 signaling in the development of ETPs that is distinct from classic mechanisms of IL-7 regulating survival and proliferation.
Collapse
|
46
|
Hendriks D, Choi G, de Bruyn M, Wiersma VR, Bremer E. Antibody-Based Cancer Therapy: Successful Agents and Novel Approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:289-383. [PMID: 28325214 DOI: 10.1016/bs.ircmb.2016.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their discovery, antibodies have been viewed as ideal candidates or "magic bullets" for use in targeted therapy in the fields of cancer, autoimmunity, and chronic inflammatory disorders. A wave of antibody-dedicated research followed, which resulted in the clinical approval of a first generation of monoclonal antibodies for cancer therapy such as rituximab (1997) and cetuximab (2004), and infliximab (2002) for the treatment of autoimmune diseases. More recently, the development of antibodies that prevent checkpoint-mediated inhibition of T cell responses invigorated the field of cancer immunotherapy. Such antibodies induced unprecedented long-term remissions in patients with advanced stage malignancies, most notably melanoma and lung cancer, that do not respond to conventional therapies. In this review, we will recapitulate the development of antibody-based therapy, and detail recent advances and new functions, particularly in the field of cancer immunotherapy. With the advent of recombinant DNA engineering, a number of rationally designed molecular formats of antibodies and antibody-derived agents have become available, and we will discuss various molecular formats including antibodies with improved effector functions, bispecific antibodies, antibody-drug conjugates, antibody-cytokine fusion proteins, and T cells genetically modified with chimeric antigen receptors. With these exciting advances, new antibody-based treatment options will likely enter clinical practice and pave the way toward more successful control of malignant diseases.
Collapse
Affiliation(s)
- D Hendriks
- Department of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - G Choi
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - M de Bruyn
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - V R Wiersma
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - E Bremer
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands; University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
47
|
d'Engremont C, Vernerey D, Pointet AL, Simone G, Fein F, Heyd B, Koch S, Vuitton L, Kim S, Jary M, Lamfichek N, Turco C, Lakkis Z, Berger A, Bonnetain F, Taieb J, Bachellier P, Borg C. Additive value of pre-operative and one-month post-operative lymphocyte count for death-risk stratification in patients with resectable pancreatic cancer: a multicentric study. BMC Cancer 2016; 16:823. [PMID: 27782813 PMCID: PMC5080693 DOI: 10.1186/s12885-016-2860-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 10/20/2016] [Indexed: 12/18/2022] Open
Abstract
Background Pancreatic adenocarcinoma (PDAC) incidence is increasing worldwide. Several studies have shown that lymphopenia was correlated with a poor prognosis but the potential interest to measure lymphopenia in the pre and post-operative setting as well as its added value among conventional prognostic factors was never investigated. Methods Data from two independent cohorts in whom patients underwent resection for pancreatic carcinoma were retrospectively recorded. We examined the association between perioperative findings, pre and post-operative lymphocyte counts and overall survival (OS) in univariate and multivariate analyses. Performance assessment and internal validation of the final model were evaluated with Harrell’s C-index, calibration plot and bootstrap sample procedures. Results Three hundred ninety patients were included in the analysis between 2000 and 2011. Pre and post-operative lymphocyte counts were independent prognostic factors associated with OS in multivariate analysis (p = 0.0128 and p = 0.0764, respectively). The addition of lymphocyte count variable to the conventional parameters identified in multivariate analysis (metastatic lymph node ratio, veinous emboli and adjuvant chemotherapy) significantly improved the model discrimination capacity (bootstrap mean difference = 0.04; 95 % CI, 0.01–0.06). The use of a threshold and combining the categorical (≥1000; <1000) information in pre and post lymphocyte counts permitted the identification of 4 subgroups of patients with different prognosis (p < 0.0001). Finally, the description of patients in long-term remission showed that only 3 of 65 (4.6 %) patients with post-operative lymphocyte count under 1000/mm3 were alive 4 years after surgery contrary to 54 of 236 (22.8 %) patients with a post-operative lymphocyte count above 1000/mm3. Conclusion Pre and post-operative lymphopenia are independent prognostic factors for OS and they have an additive value regarding conventional prognostic factors for death-risk stratification and to predict long-term survival. Lymphopenia should be included as stratification factors in future clinical trial assessing overall survival in pancreatic cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2860-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Dewi Vernerey
- Methodology and Quality of Life in Oncology Unit, University Hospital of Besançon, Besançon, France
| | - Anne-Laure Pointet
- Department of Gastroenterology and GI oncology, Paris Descartes University, Georges Pompidou European Hospital, Paris, France
| | - Gaël Simone
- Department of Digestive Surgery and Liver Transplantation, University hospital of Strasbourg, Strasbourg, France
| | - Francine Fein
- Department of Gastroenterology, University Hospital of Besançon, Besançon, France
| | - Bruno Heyd
- Department of Digestive Surgery and Liver Transplantation, University Hospital of Besançon, Besançon, France
| | - Stéphane Koch
- Department of Gastroenterology, University Hospital of Besançon, Besançon, France
| | - Lucine Vuitton
- Department of Gastroenterology, University Hospital of Besançon, Besançon, France
| | - Stefano Kim
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Marine Jary
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Najib Lamfichek
- Department of Digestive Surgery, Hospital of Belfort-Montbeliard, Montbeliard, France
| | - Celia Turco
- Department of Digestive Surgery and Liver Transplantation, University Hospital of Besançon, Besançon, France
| | - Zaher Lakkis
- Department of Digestive Surgery and Liver Transplantation, University Hospital of Besançon, Besançon, France
| | - Anne Berger
- Department of GI Surgery, Paris Descartes University, Georges Pompidou European Hospital, Paris, France
| | - Franck Bonnetain
- Methodology and Quality of Life in Oncology Unit, University Hospital of Besançon, Besançon, France
| | - Julien Taieb
- Department of Gastroenterology and GI oncology, Paris Descartes University, Georges Pompidou European Hospital, Paris, France
| | - Philippe Bachellier
- Department of Digestive Surgery and Liver Transplantation, University hospital of Strasbourg, Strasbourg, France
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besançon, France. .,Centre investigation Clinique en biothérapie, CIC-1431, Besançon, France. .,UMR1098 INSERM/Université de Franche Comté/Etablissement Français du Sang, Besançon, France. .,Department of Oncology, University Hospital of Besançon, 3 Boulevard Alexander Fleming, Besancon, F-25030, France.
| |
Collapse
|
48
|
Merchant MS, Bernstein D, Amoako M, Baird K, Fleisher TA, Morre M, Steinberg SM, Sabatino M, Stroncek DF, Venkatasan AM, Wood BJ, Wright M, Zhang H, Mackall CL. Adjuvant Immunotherapy to Improve Outcome in High-Risk Pediatric Sarcomas. Clin Cancer Res 2016; 22:3182-91. [PMID: 26823601 DOI: 10.1158/1078-0432.ccr-15-2550] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/02/2016] [Indexed: 01/28/2023]
Abstract
PURPOSE Patients with metastatic or relapsed pediatric sarcomas receive cytotoxic regimens that induce high remission rates associated with profound lymphocyte depletion, but ultimately few survive long term. We administered adjuvant immunotherapy to patients with metastatic and recurrent pediatric sarcomas in an effort to improve outcomes. EXPERIMENTAL DESIGN Mononuclear cells were collected via apheresis, and tumor lysate was acquired via percutaneous biopsy at enrollment. Participants received standard antineoplastic therapy, followed by autologous lymphocytes, tumor lysate/keyhole limpet hemocyanin-pulsed dendritic cell vaccinations ± recombinant human IL7. Primary outcomes were toxicity and vaccine responses. Secondary outcomes were immune reconstitution, event-free survival, and overall survival (OS). RESULTS Forty-three patients enrolled and 29 received immunotherapy. The regimen was well tolerated. Intent-to-treat analysis demonstrated 5-year OS of 51% with significant differences based upon histologic group (63% vs. 0% for Ewing/rhabdomyosarcoma vs. other sarcomas) and response to standard therapy (74% no residual disease vs. 0% residual disease). Five-year intent-to-treat OS of patients with newly diagnosed metastatic Ewing/rhabdomyosarcoma was 77%, higher than previously reported in this population and higher than observed in a similar group treated with an earlier adjuvant immunotherapy regimen (25% 5-year OS). T-cell responses to autologous tumor lysate were identified in 62% of immunotherapy recipients, and survival was higher in those patients (73% 5-year OS with vs. 37% without immune response, P = 0.017). Immune reconstitution, measured by CD4 count recovery, was significantly enhanced in subjects treated with recombinant human IL7. CONCLUSIONS Adjuvant immunotherapy may improve survival in patients with metastatic pediatric sarcoma. Clin Cancer Res; 22(13); 3182-91. ©2016 AACR.
Collapse
Affiliation(s)
- Melinda S Merchant
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Donna Bernstein
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Martha Amoako
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Kristin Baird
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Thomas A Fleisher
- Department of Laboratory Medicine, NIH Clinical Center, Bethesda, Maryland
| | | | - Seth M Steinberg
- Department of Biostatistics and Data Management, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Marianna Sabatino
- Department of Transfusion Medicine, NIH Clinical Center, Bethesda, Maryland
| | - Dave F Stroncek
- Department of Transfusion Medicine, NIH Clinical Center, Bethesda, Maryland
| | | | - Bradford J Wood
- Radiology and Imaging Science, NIH Clinical Center, Bethesda, Maryland
| | - Matthew Wright
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Hua Zhang
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland
| | - Crystal L Mackall
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH Clinical Center, Bethesda, Maryland.
| |
Collapse
|