1
|
Shi Q, Luo Y, Xiang Q, Kang X, Feng Z. CD28 Superfamily Costimulatory Molecules in Chronic Pain: Focus on Immunomodulation. Mol Neurobiol 2025; 62:7915-7926. [PMID: 39956885 DOI: 10.1007/s12035-025-04746-3] [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: 09/13/2023] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Chronic pain has substantial effects on patients' quality of life and psychological well-being. It does not respond satisfactorily to available medicinal therapeutics because its mechanism remains unclear. Recent studies have shown a strong relationship between chronic pain and immunomodulation. As important members of the immune response, CD28 superfamily costimulatory molecules were demonstrated to have an analgesic effect on chronic pain. Based on research on the role of these molecules in chronic pain, new and highly effective analgesic medicines are anticipated that could be used in combination with some previous analgesic medicines to reduce substance abuse and side effects. This review of the literature will examine the pain-regulating mechanisms of CD28 superfamily costimulatory molecules, focusing on immunomodulation. In addition, this review will discuss the potential and difficulties of developing novel analgesic medicines targeting CD28 superfamily costimulatory molecules.
Collapse
Affiliation(s)
- Qinglu Shi
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Yujia Luo
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
| | - Qiaomin Xiang
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China
- Department of Anesthesiology, Ninghai First Hospital, Ningbo, Zhejiang, China
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Zhiying Feng
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
2
|
Lovewell RR, Langermann S, Flies DB. Immune inhibitory receptor agonist therapeutics. Front Immunol 2025; 16:1566869. [PMID: 40207220 PMCID: PMC11979287 DOI: 10.3389/fimmu.2025.1566869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
The immune system maintains the health of an organism through complex sensing and communication mechanisms. Receptors on the surface of immune cells respond to stimuli resulting in activity described at its most basic as inhibitory or stimulatory. Significant progress in therapeutic intervention has occurred by modulating these pathways, yet much remains to be accomplished. Therapeutics that antagonize, or block, immune inhibitory receptor (IIR) pathways, such as checkpoint inhibitors in cancer are a key example. Antagonism of immune stimulatory receptors (ISRs) for dysregulated inflammation and autoimmunity have received significant attention. An alternative strategy is to agonize, or induce signaling, in immune pathways to treat disease. Agonism of ISRs has been employed with some success in disease settings, but agonist therapeutics of IIRs have great, untapped potential. This review discusses and highlights recent advances in pre-clinical and clinical therapeutics designed to agonize IIR pathways to treat diseases. In addition, an understanding of IIR agonists based on activity at a cellular level as either agonist suppression of stimulatory cells (SuSt), or a new concept, agonist suppression of suppressive cells (SuSu) is proposed.
Collapse
|
3
|
Jafri Z, Zhang J, O'Meara CH, Joshua AM, Parish CR, Khachigian LM. Interplay between CD28 and PD-1 in T cell immunotherapy. Vascul Pharmacol 2025; 158:107461. [PMID: 39734005 DOI: 10.1016/j.vph.2024.107461] [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: 11/26/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Immune checkpoint therapy targeting the PD-1/PD-L1 axis has revolutionised the treatment of solid tumors. However, T cell exhaustion underpins resistance to current anti-PD-1 therapies, resulting in lower response rates in cancer patients. CD28 is a T cell costimulatory receptor that can influence the PD-1 signalling pathway (and vice versa). CD28 signalling has the potential to counter T cell exhaustion by serving as a potential complementary response to traditional anti-PD-1 therapies. Here we discuss the interplay between PD-1 and CD28 in T cell immunotherapy and additionally how CD28 transcriptionally modulates T cell exhaustion. We also consider clinical attempts at targeting CD28; the challenges faced by past attempts and recent promising developments.
Collapse
Affiliation(s)
- Zuhayr Jafri
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jingwen Zhang
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Connor H O'Meara
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; Division of Head & Neck Oncology and Microvascular Reconstruction, Department of Otolaryngology, Head & Neck Surgery, University of Virginia Health Services, Charlottesville, VA 22903, USA; Department of Otolaryngology, Head & Neck Surgery, Australian National University, Acton, ACT 0200, Australia
| | - Anthony M Joshua
- Kinghorn Cancer Centre, St Vincents Hospital, Sydney and Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Christopher R Parish
- Cancer and Vascular Biology Group, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
4
|
Klein C, Brinkmann U, Reichert JM, Kontermann RE. The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov 2024; 23:301-319. [PMID: 38448606 DOI: 10.1038/s41573-024-00896-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
Bispecific antibodies (bsAbs) enable novel mechanisms of action and/or therapeutic applications that cannot be achieved using conventional IgG-based antibodies. Consequently, development of these molecules has garnered substantial interest in the past decade and, as of the end of 2023, 14 bsAbs have been approved: 11 for the treatment of cancer and 3 for non-oncology indications. bsAbs are available in different formats, address different targets and mediate anticancer function via different molecular mechanisms. Here, we provide an overview of recent developments in the field of bsAbs for cancer therapy. We focus on bsAbs that are approved or in clinical development, including bsAb-mediated dual modulators of signalling pathways, tumour-targeted receptor agonists, bsAb-drug conjugates, bispecific T cell, natural killer cell and innate immune cell engagers, and bispecific checkpoint inhibitors and co-stimulators. Finally, we provide an outlook into next-generation bsAbs in earlier stages of development, including trispecifics, bsAb prodrugs, bsAbs that induce degradation of tumour targets and bsAbs acting as cytokine mimetics.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland.
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
5
|
Wang B, Liu Y, Yuan R, Dou X, Qian N, Pan X, Xu G, Xu Q, Dong B, Yang C, Li H, Wang J, Bai G, Liu L, Gao X. XFab-α4-1BB/CD40L fusion protein activates dendritic cells, improves expansion of antigen-specific T cells, and exhibits antitumour efficacy in multiple solid tumour models. Cancer Immunol Immunother 2023; 72:4015-4030. [PMID: 37863852 PMCID: PMC10991239 DOI: 10.1007/s00262-023-03535-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/21/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Additional immunotherapies are still warranted for non-responders to checkpoint inhibitors with refractory or relapsing cancers, especially for patients with "cold" tumours lacking significant immune infiltration at treatment onset. We developed XFab-α4-1BB/CD40L, a bispecific antibody targeting 4-1BB and CD40 for dendritic cell activation and priming of tumour-reactive T cells to inhibit tumours. METHODS XFab-α4-1BB/CD40L was developed by engineering an anti-4-1BB Fab arm into a CD40L trimer based on XFab® platform. Characterisation of the bispecific antibody was performed by cell-based reporter assays, maturation of dendritic cell assays, and mixed lymphocyte reactions. The abilities of antigen-specific T-cell expansion and antitumour efficacy were assessed in syngeneic mouse tumour models. Toxicological and pharmacodynamic profiles were investigated in non-human primates. RESULTS XFab-α4-1BB/CD40L demonstrated independent CD40 agonistic activity and conditional 4-1BB activity mediated by CD40 crosslinking, leading to dendritic cell maturation and T-cell proliferation in vitro. We confirmed the expansion of antigen-specific T cells in the vaccination model and potent tumour regression induced by the bispecific antibody alone or in combination with gemcitabine in vivo, concomitant with improved tumour-reactive T-cell infiltration. XFab-α4-1BB/CD40L showed no signs of liver toxicity at doses up to 51 mg/kg in a repeated-dose regimen in non-human primates. CONCLUSIONS XFab-α4-1BB/CD40L is capable of enhancing antitumour immunity by modulating dendritic cell and T-cell functions via targeting 4-1BB agonism to areas of CD40 expression. The focused, potent, and safe immune response induced by the bispecific antibody supports further clinical investigations for the treatment of solid tumours.
Collapse
Affiliation(s)
- Bochun Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Yujie Liu
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Ruofei Yuan
- Capital Medical University, Beijing, 100069, People's Republic of China
| | - Xiaoqian Dou
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Niliang Qian
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Xiujie Pan
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Guili Xu
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Qinzhi Xu
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Bo Dong
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Cuima Yang
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Hongjie Li
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Jingjing Wang
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Guijun Bai
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China
| | - Liangfa Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, People's Republic of China.
| | - Xin Gao
- Beijing Immunoah Pharma Tech Co., Ltd., Beijing, 100141, People's Republic of China.
| |
Collapse
|
6
|
Li Q, He Y, Pan J. CrossFuse-XGBoost: accurate prediction of the maximum recommended daily dose through multi-feature fusion, cross-validation screening and extreme gradient boosting. Brief Bioinform 2023; 25:bbad511. [PMID: 38216539 PMCID: PMC10786712 DOI: 10.1093/bib/bbad511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024] Open
Abstract
In the drug development process, approximately 30% of failures are attributed to drug safety issues. In particular, the first-in-human (FIH) trial of a new drug represents one of the highest safety risks, and initial dose selection is crucial for ensuring safety in clinical trials. With traditional dose estimation methods, which extrapolate data from animals to humans, catastrophic events have occurred during Phase I clinical trials due to interspecies differences in compound sensitivity and unknown molecular mechanisms. To address this issue, this study proposes a CrossFuse-extreme gradient boosting (XGBoost) method that can directly predict the maximum recommended daily dose of a compound based on existing human research data, providing a reference for FIH dose selection. This method not only integrates multiple features, including molecular representations, physicochemical properties and compound-protein interactions, but also improves feature selection based on cross-validation. The results demonstrate that the CrossFuse-XGBoost method not only improves prediction accuracy compared to that of existing local weighted methods [k-nearest neighbor (k-NN) and variable k-NN (v-NN)] but also solves the low prediction coverage issue of v-NN, achieving full coverage of the external validation set and enabling more reliable predictions. Furthermore, this study offers a high level of interpretability by identifying the importance of different features in model construction. The 241 features with the most significant impact on the maximum recommended daily dose were selected, providing references for optimizing the structure of new compounds and guiding experimental research. The datasets and source code are freely available at https://github.com/cqmu-lq/CrossFuse-XGBoost.
Collapse
Affiliation(s)
- Qiang Li
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yu He
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
7
|
Pereira PDC, Diniz DG, da Costa ER, Magalhães NGDM, da Silva ADJF, Leite JGS, Almeida NIP, Cunha KDN, de Melo MAD, Vasconcelos PFDC, Diniz JAP, Brites D, Anthony DC, Diniz CWP, Guerreiro-Diniz C. Genes, inflammatory response, tolerance, and resistance to virus infections in migratory birds, bats, and rodents. Front Immunol 2023; 14:1239572. [PMID: 37711609 PMCID: PMC10497949 DOI: 10.3389/fimmu.2023.1239572] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Normally, the host immunological response to viral infection is coordinated to restore homeostasis and protect the individual from possible tissue damage. The two major approaches are adopted by the host to deal with the pathogen: resistance or tolerance. The nature of the responses often differs between species and between individuals of the same species. Resistance includes innate and adaptive immune responses to control virus replication. Disease tolerance relies on the immune response allowing the coexistence of infections in the host with minimal or no clinical signs, while maintaining sufficient viral replication for transmission. Here, we compared the virome of bats, rodents and migratory birds and the molecular mechanisms underlying symptomatic and asymptomatic disease progression. We also explore the influence of the host physiology and environmental influences on RNA virus expression and how it impacts on the whole brain transcriptome of seemingly healthy semipalmated sandpiper (Calidris pusilla) and spotted sandpiper (Actitis macularius). Three time points throughout the year were selected to understand the importance of longitudinal surveys in the characterization of the virome. We finally revisited evidence that upstream and downstream regulation of the inflammatory response is, respectively, associated with resistance and tolerance to viral infections.
Collapse
Affiliation(s)
- Patrick Douglas Corrêa Pereira
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Seção de Hepatologia, Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Pará, Brazil
- Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Emanuel Ramos da Costa
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
- Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Nara Gyzely de Morais Magalhães
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Anderson de Jesus Falcão da Silva
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Jéssica Gizele Sousa Leite
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Natan Ibraim Pires Almeida
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Kelle de Nazaré Cunha
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Mauro André Damasceno de Melo
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Centro de Ciências Biológicas e da Saúde, Universidade do Estado do Pará, Belém, Pará, Brazil
- Seção de Arbovirologia e Febres Hemorrágicas, Instituto Evandro Chagas, Ananindeua, Pará, Brazil
| | - José Antonio Picanço Diniz
- Seção de Hepatologia, Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém, Pará, Brazil
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Daniel Clive Anthony
- Department of Pharmacology, Laboratory of Experimental Neuropathology, University of Oxford, Oxford, United Kingdom
| | - Cristovam Wanderley Picanço Diniz
- Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Cristovam Guerreiro-Diniz
- Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal de Educação, Bragança, Pará, Brazil
| |
Collapse
|
8
|
Tapia-Galisteo A, Álvarez-Vallina L, Sanz L. Bi- and trispecific immune cell engagers for immunotherapy of hematological malignancies. J Hematol Oncol 2023; 16:83. [PMID: 37501154 PMCID: PMC10373336 DOI: 10.1186/s13045-023-01482-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Immune cell engagers are engineered antibodies with at least one arm binding a tumor-associated antigen and at least another one directed against an activating receptor in immune effector cells: CD3 for recruitment of T cells and CD16a for NK cells. The first T cell engager (the anti-CD19 blinatumomab) was approved by the FDA in 2014, but no other one hit the market until 2022. Now the field is gaining momentum, with three approvals in 2022 and 2023 (as of May): the anti-CD20 × anti-CD3 mosunetuzumab and epcoritamab and the anti-B cell maturation antigen (BCMA) × anti-CD3 teclistamab, and another three molecules in regulatory review. T cell engagers will likely revolutionize the treatment of hematological malignancies in the short term, as they are considerably more potent than conventional monoclonal antibodies recognizing the same tumor antigens. The field is thriving, with a plethora of different formats and targets, and around 100 bispecific T cell engagers more are already in clinical trials. Bispecific NK cell engagers are also in early-stage clinical studies and may offer similar efficacy with milder side effects. Trispecific antibodies (engaging either T cell or NK cell receptors) raise the game even further with a third binding moiety, which allows either the targeting of an additional tumor-associated antigen to increase specificity and avoid immune escape or the targeting of additional costimulatory receptors on the immune cell to improve its effector functions. Altogether, these engineered molecules may change the paradigm of treatment for relapsed or refractory hematological malignancies.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Luis Álvarez-Vallina
- Immuno-Oncology and Immunotherapy Group, Biomedical Research Institute Hospital Universitario, 12 de Octubre, Madrid, Spain.
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario, 12 de Octubre, Madrid, Spain.
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
| |
Collapse
|
9
|
Melo V, Nelemans LC, Vlaming M, Lourens HJ, Wiersma VR, Bilemjian V, Huls G, de Bruyn M, Bremer E. EGFR-selective activation of CD27 co-stimulatory signaling by a bispecific antibody enhances anti-tumor activity of T cells. Front Immunol 2023; 14:1191866. [PMID: 37545491 PMCID: PMC10399592 DOI: 10.3389/fimmu.2023.1191866] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023] Open
Abstract
A higher density of tumor infiltrating lymphocytes (TILs) in the tumor microenvironment, particularly cytotoxic CD8+ T cells, is associated with improved clinical outcome in various cancers. However, local inhibitory factors can suppress T cell activity and hinder anti-tumor immunity. Notably, TILs from various cancer types express the co-stimulatory Tumor Necrosis Factor receptor CD27, making it a potential target for co-stimulation and re-activation of tumor-infiltrated and tumor-reactive T cells. Anti-cancer therapeutics based on exploiting CD27-mediated T cell co-stimulation have proven safe, but clinical responses remain limited. This is likely because current monoclonal antibodies fail to effectively activate CD27 signaling, as this receptor requires higher-order receptor cross-linking. Here, we report on a bispecific antibody, CD27xEGFR, that targets both CD27 and the tumor antigen, epidermal growth factor receptor (EGFR). By targeting EGFR, which is commonly expressed on carcinomas, CD27xEGFR induced cancer cell-localized crosslinking and activation of CD27. The design of CD27xEGFR includes an Fc-silent domain, which is designed to minimize potential toxicity by reducing Fc gamma receptor-mediated binding and activation of immune cells. CD27xEGFR bound to both of its targets simultaneously and triggered EGFR-restricted co-stimulation of T cells as measured by T cell proliferation, T cell activation markers, cytotoxicity and IFN-γ release. Further, CD27xEGFR augmented T cell cytotoxicity in a panel of artificial antigen-presenting carcinoma cell line models, leading to Effector-to-Target ratio-dependent elimination of cancer cells. Taken together, we present the in vitro characterization of a novel bispecific antibody that re-activates T cell immunity in EGFR-expressing cancers through targeted co-stimulation of CD27.
Collapse
Affiliation(s)
- Vinicio Melo
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Levi Collin Nelemans
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martijn Vlaming
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Harm Jan Lourens
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Valerie R. Wiersma
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Vrouyr Bilemjian
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gerwin Huls
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marco de Bruyn
- Department of Obstetrics & Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Edwin Bremer
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
10
|
Promsote W, Xu L, Hataye J, Fabozzi G, March K, Almasri CG, DeMouth ME, Lovelace SE, Talana CA, Doria-Rose NA, McKee K, Hait SH, Casazza JP, Ambrozak D, Beninga J, Rao E, Furtmann N, Birkenfeld J, McCarthy E, Todd JP, Petrovas C, Connors M, Hebert AT, Beck J, Shen J, Zhang B, Levit M, Wei RR, Yang ZY, Pegu A, Mascola JR, Nabel GJ, Koup RA. Trispecific antibody targeting HIV-1 and T cells activates and eliminates latently-infected cells in HIV/SHIV infections. Nat Commun 2023; 14:3719. [PMID: 37349337 PMCID: PMC10287722 DOI: 10.1038/s41467-023-39265-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 05/30/2023] [Indexed: 06/24/2023] Open
Abstract
Agents that can simultaneously activate latent HIV, increase immune activation and enhance the killing of latently-infected cells represent promising approaches for HIV cure. Here, we develop and evaluate a trispecific antibody (Ab), N6/αCD3-αCD28, that targets three independent proteins: (1) the HIV envelope via the broadly reactive CD4-binding site Ab, N6; (2) the T cell antigen CD3; and (3) the co-stimulatory molecule CD28. We find that the trispecific significantly increases antigen-specific T-cell activation and cytokine release in both CD4+ and CD8+ T cells. Co-culturing CD4+ with autologous CD8+ T cells from ART-suppressed HIV+ donors with N6/αCD3-αCD28, results in activation of latently-infected cells and their elimination by activated CD8+ T cells. This trispecific antibody mediates CD4+ and CD8+ T-cell activation in non-human primates and is well tolerated in vivo. This HIV-directed antibody therefore merits further development as a potential intervention for the eradication of latent HIV infection.
Collapse
Affiliation(s)
- Wanwisa Promsote
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ling Xu
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
- ModeX Therapeutics Inc., 22 Strathmore Road, Natick, MA, 01760, USA
| | - Jason Hataye
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Giulia Fabozzi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kylie March
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cassandra G Almasri
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan E DeMouth
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sarah E Lovelace
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Chloe Adrienna Talana
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicole A Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Krisha McKee
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sabrina Helmold Hait
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joseph P Casazza
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Ambrozak
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Ercole Rao
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
| | | | - Joerg Birkenfeld
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
- Perspix Biotech GmbH, FiZ Frankfurt Innovation Center Biotechnology, Altenhoeferallee 3, 60438, Frankfurt, Germany
| | - Elizabeth McCarthy
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John-Paul Todd
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Constantinos Petrovas
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital (chuv) and University of Lausanne, Lausanne, Switzerland
| | | | | | - Jeremy Beck
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
| | - Junqing Shen
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
| | - Bailin Zhang
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
| | | | - Ronnie R Wei
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
- ModeX Therapeutics Inc., 22 Strathmore Road, Natick, MA, 01760, USA
| | - Zhi-Yong Yang
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA
- ModeX Therapeutics Inc., 22 Strathmore Road, Natick, MA, 01760, USA
| | - Amarendra Pegu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- ModeX Therapeutics Inc., 22 Strathmore Road, Natick, MA, 01760, USA
| | - Gary J Nabel
- Sanofi, 640 Memorial Dr., Cambridge, MA, 02139, USA.
- ModeX Therapeutics Inc., 22 Strathmore Road, Natick, MA, 01760, USA.
| | - Richard A Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Shah D, Soper B, Shopland L. Cytokine release syndrome and cancer immunotherapies - historical challenges and promising futures. Front Immunol 2023; 14:1190379. [PMID: 37304291 PMCID: PMC10248525 DOI: 10.3389/fimmu.2023.1190379] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Cancer is the leading cause of death worldwide. Cancer immunotherapy involves reinvigorating the patient's own immune system to fight against cancer. While novel approaches like Chimeric Antigen Receptor (CAR) T cells, bispecific T cell engagers, and immune checkpoint inhibitors have shown promising efficacy, Cytokine Release Syndrome (CRS) is a serious adverse effect and remains a major concern. CRS is a phenomenon of immune hyperactivation that results in excessive cytokine secretion, and if left unchecked, it may lead to multi-organ failure and death. Here we review the pathophysiology of CRS, its occurrence and management in the context of cancer immunotherapy, and the screening approaches that can be used to assess CRS and de-risk drug discovery earlier in the clinical setting with more predictive pre-clinical data. Furthermore, the review also sheds light on the potential immunotherapeutic approaches that can be used to overcome CRS associated with T cell activation.
Collapse
Affiliation(s)
- Deep Shah
- In vivo Services, The Jackson Laboratory, Sacramento, CA, United States
| | - Brian Soper
- Technical Information Services, The Jackson Laboratory, Bar Harbor, ME, United States
| | - Lindsay Shopland
- In vivo Services, The Jackson Laboratory, Sacramento, CA, United States
| |
Collapse
|
12
|
Yao Y, Hu Y, Wang F. Trispecific antibodies for cancer immunotherapy. Immunology 2023. [PMID: 36855956 DOI: 10.1111/imm.13636] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Despite the clinical success of monoclonal and bispecific antibodies, there are still limitations in the therapeutic effect of malignant tumours, such as low response rate, treatment resistance, and so on, inspiring the exploration of trispecific antibodies (TsAbs). TsAbs further improve the safety and efficacy and has great clinical potential through three targets combination and formats optimization. This article reviews the development history and the target combination features of TsAbs. Although there are still great challenges in the clinical application of TsAbs, it is undeniable that TsAbs may be a breakthrough in the development of antibody drugs.
Collapse
Affiliation(s)
- Yin Yao
- Department of Pharmacy, Fourth People's Hospital of Gui Yang, Guiyang, China
| | - Yiyin Hu
- Department of Nursing, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Fei Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Ma K, Que W, Hu X, Guo WZ, Zhong L, Ueda D, Gu EL, Li XK. Combinations of anti-GITR antibody and CD28 superagonist ameliorated dextran sodium sulfate-induced mouse colitis. Clin Exp Immunol 2022; 208:340-350. [PMID: 35511600 PMCID: PMC9226153 DOI: 10.1093/cei/uxac039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 11/12/2022] Open
Abstract
Ulcerative colitis (UC) is one of the two main forms of inflammatory bowel disease (IBD) and is an idiopathic, chronic inflammatory disease of the colonic mucosa with an unclear etiology. Interleukin (IL)-10 has been reported to play a crucial role in the maintenance of immune homeostasis in the intestinal environment. Type 1 regulatory T (Tr1) cells are a subset of CD4+Foxp3- T cells able to secrete high amounts of IL-10 with potent immunosuppressive properties. In this study, we found that the combination of anti-GITR antibody (G3c) and CD28 superagonist (D665) treatment stimulated the generation of a large amount of Tr1 cells. Furthermore, G3c/D665 treatment not only significantly relieved severe mucosal damage but also reduced the incidence of colonic shortening, weight loss, and hematochezia. Dextran sodium sulfate (DSS) upregulated the mRNA levels of IL-6, IL-1β, IL-17, IL-12, tumor necrosis factor-alpha, C-C chemokine receptor type 5, and Bax in splenic lymphocytes (SPLs) and colon tissues, while G3c/D665 treatment conversely inhibited the increase in mRNA levels of these genes. In addition, G3c/D665 treatment altered the proportion of CD4+ and CD8+ T cells and increased CD4+CD25+Foxp3+ regulatory T cells in SPLs, mesenteric lymph nodes (MLNs), and lamina propria lymphocytes (LPLs). Thus, the combination of G3c and D665 treatment showed efficacy against DSS-induced UC in mice by inducing a large amount of Tr1 cell generation via the musculoaponeurotic fibrosarcoma pathways in vivo and relieving inflammatory responses both systematically and locally.
Collapse
Affiliation(s)
- Kuai Ma
- Department of Gastroenterology and Hepatology, Jing'an District Central Hospital, Jing'an Branch of Huashan Hospital, Fudan University, Shanghai, China.,Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Weitao Que
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Xin Hu
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Wen-Zhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liang Zhong
- Department of Gastroenterology, Huashan Hospital, Fudan University, Shanghai, China
| | - Daisuke Ueda
- Division of Hepato-Pancreato-Biliary Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Er-Li Gu
- Department of Gastroenterology and Hepatology, Jing'an District Central Hospital, Jing'an Branch of Huashan Hospital, Fudan University, Shanghai, China
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Fukaya-Shiba A, Otsuka K, Sasaki H, Shikano M, Wakao R. Identification of Novel Modalities Through Bibliometric Analysis for Timely Development of Regulatory Guidance: A Case Study of T Cell Immunity. Front Med (Lausanne) 2021; 8:756870. [PMID: 34708061 PMCID: PMC8544749 DOI: 10.3389/fmed.2021.756870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Background: The mission of medicines regulatory agencies is to ensure the timely access of innovative products for patients to improve public health. Thus, regulators should foresee evolving technologies and build expertise prior to reviewing innovative products. Novel modalities and new classes of therapeutics in biological or cell-based products represent a regulatory challenge because of knowledge gaps, as exemplified by the unexpected cytokine release syndrome in the first-in-human clinical trial of the CD28 super-agonist. Meanwhile, recent treatments harnessing T cell co-signaling pathways provide an opportunity for investigation. Therefore, this study aimed to systematically identify and evaluate novel modalities for T cell immunity to assess the need for regulatory guidance. Methods: A PubMed search was carried out using the query, "immun* AND t lymph*" to select publications. Subsequently, a citation network was created, followed by clustering and text mining to identify the modalities and classes of therapeutics under development. Results and Discussion: Analysis of the top 20 clusters revealed research domains characterized by keywords such as immune checkpoint antibody, chimeric antigen receptor (CAR)-T cells, microbiota, exosome, regulatory T cells, unconventional T cells, and vaccines. After reviewing the pharmacological concepts, clinical trial information, and available guidance, we presented a perspective on the future development of guidance for these domains. Conclusion: Bibliometric analyses identified a set of innovative modalities targeted for drug development with which regulatory guidance is going to catch up. This strategy could help in the successful development of upcoming modalities to ensure readiness for clinical application as part of horizon scanning.
Collapse
Affiliation(s)
- Ai Fukaya-Shiba
- Center for Regulatory Science, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Kouhei Otsuka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Hajime Sasaki
- Institute for Future Initiatives, The University of Tokyo, Tokyo, Japan
| | - Mayumi Shikano
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Rika Wakao
- Center for Regulatory Science, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| |
Collapse
|
15
|
Orrù V, Steri M, Cucca F, Fiorillo E. Application of Genetic Studies to Flow Cytometry Data and Its Impact on Therapeutic Intervention for Autoimmune Disease. Front Immunol 2021; 12:714461. [PMID: 34531863 PMCID: PMC8438121 DOI: 10.3389/fimmu.2021.714461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/13/2021] [Indexed: 12/03/2022] Open
Abstract
In recent years, systematic genome-wide association studies of quantitative immune cell traits, represented by circulating levels of cell subtypes established by flow cytometry, have revealed numerous association signals, a large fraction of which overlap perfectly with genetic signals associated with autoimmune diseases. By identifying further overlaps with association signals influencing gene expression and cell surface protein levels, it has also been possible, in several cases, to identify causal genes and infer candidate proteins affecting immune cell traits linked to autoimmune disease risk. Overall, these results provide a more detailed picture of how genetic variation affects the human immune system and autoimmune disease risk. They also highlight druggable proteins in the pathogenesis of autoimmune diseases; predict the efficacy and side effects of existing therapies; provide new indications for use for some of them; and optimize the research and development of new, more effective and safer treatments for autoimmune diseases. Here we review the genetic-driven approach that couples systematic multi-parametric flow cytometry with high-resolution genetics and transcriptomics to identify endophenotypes of autoimmune diseases for the development of new therapies.
Collapse
Affiliation(s)
- Valeria Orrù
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Sardinia, Italy
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Sardinia, Italy
| | - Francesco Cucca
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Sardinia, Italy.,Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Sardinia, Italy
| |
Collapse
|
16
|
Chi LH, Burrows AD, Anderson RL. Can preclinical drug development help to predict adverse events in clinical trials? Drug Discov Today 2021; 27:257-268. [PMID: 34469805 DOI: 10.1016/j.drudis.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/03/2021] [Accepted: 08/24/2021] [Indexed: 12/23/2022]
Abstract
The development of novel therapeutics is associated with high rates of attrition, with unexpected adverse events being a major cause of failure. Serious adverse events have led to organ failure, cancer development and deaths that were not expected outcomes in clinical trials. These life-threatening events were not identified during therapeutic development due to the lack of preclinical safety tests that faithfully represented human physiology. We highlight the successful application of several novel technologies, including high-throughput screening, organs-on-chips, microbiome-containing drug-testing platforms and humanised mouse models, for mechanistic studies and prediction of toxicity. We propose the incorporation of similar preclinical tests into future drug development to reduce the likelihood of hazardous therapeutics entering later-stage clinical trials.
Collapse
Affiliation(s)
- Lap Hing Chi
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Allan D Burrows
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Robin L Anderson
- Translational Breast Cancer Program, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia; School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
17
|
Salvador R, Zhang A, Horai R, Caspi RR. Microbiota as Drivers and as Therapeutic Targets in Ocular and Tissue Specific Autoimmunity. Front Cell Dev Biol 2021; 8:606751. [PMID: 33614621 PMCID: PMC7893107 DOI: 10.3389/fcell.2020.606751] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022] Open
Abstract
Autoimmune uveitis is a major cause of blindness in humans. Activation of retina-specific autoreactive T cells by commensal microbiota has been shown to trigger uveitis in mice. Although a culprit microbe and/or its immunogenic antigen remains to be identified, studies from inducible and spontaneous mouse models suggest the potential of microbiota-modulating therapies for treating ocular autoimmune disease. In this review, we summarize recent findings on the contribution of microbiota to T cell-driven, tissue-specific autoimmunity, with an emphasis on autoimmune uveitis, and analyze microbiota-altering interventions, including antibiotics, probiotics, and microbiota-derived metabolites (e.g., short-chain fatty acids), which have been shown to be effective in other autoimmune diseases. We also discuss the need to explore more translational animal models as well as to integrate various datasets (microbiomic, transcriptomic, proteomic, metabolomic, and other cellular measurements) to gain a better understanding of how microbiota can directly or indirectly modulate the immune system and contribute to the onset of disease. It is hoped that deeper understanding of these interactions may lead to more effective treatment interventions.
Collapse
Affiliation(s)
- Ryan Salvador
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Amy Zhang
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Reiko Horai
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
18
|
Yong KSM, Her Z, Tan SY, Tan WWS, Liu M, Lai F, Heng SM, Fan Y, Chang KTE, Wang CI, Chan JKY, Chen J, Chen Q. Humanized Mouse as a Tool to Predict Immunotoxicity of Human Biologics. Front Immunol 2020; 11:553362. [PMID: 33193321 PMCID: PMC7604536 DOI: 10.3389/fimmu.2020.553362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 09/28/2020] [Indexed: 11/24/2022] Open
Abstract
Advancements in science enable researchers to constantly innovate and create novel biologics. However, the use of non-human animal models during the development of biologics impedes identification of precise in vivo interactions between the human immune system and treatments. Due to lack of this understanding, adverse effects are frequently observed in healthy volunteers and patients exposed to potential biologics during clinical trials. In this study, we evaluated and compared the effects of known immunotoxic biologics, Proleukin®/IL-2 and OKT3 in humanized mice (reconstituted with human fetal cells) to published clinical outcomes. We demonstrated that humanized mice were able to recapitulate in vivo pathological changes and human-specific immune responses, such as elevated cytokine levels and modulated lymphocytes and myeloid subsets. Given the high similarities of immunological side effects observed between humanized mice and clinical studies, this model could be used to assess immunotoxicity of biologics at a pre-clinical stage, without placing research participants and/or patients at risk.
Collapse
Affiliation(s)
- Kylie Su Mei Yong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Zhisheng Her
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Wilson Wei Sheng Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Min Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Fritz Lai
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Shi Min Heng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Pathology, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianzhu Chen
- Interdisciplinary Research Group in Infectious Diseases, Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore.,The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Di J, Xie F, Xu Y. When liposomes met antibodies: Drug delivery and beyond. Adv Drug Deliv Rev 2020; 154-155:151-162. [PMID: 32926944 DOI: 10.1016/j.addr.2020.09.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Drug encapsulated liposomes and monoclonal antibodies (Mabs) are two distinctively different classes of therapeutics, but both aim to become the ultimate "magic bullet". While PEGylated liposomes rely on the enhanced permeability and retention (EPR) effect for accumulation in solid tumor tissues, Mabs are designed to bind tightly to specific surface antigens on target cells to exert effector functions. Immunoliposome (IL) refers to the structural combination of liposomes and antibodies, whereas the antibodies are usually decorated on the liposome surface. ILs can therefore take advantage of interactions between antibodies and cancer cells for more efficient endocytosis and intracellular drug delivery. The antibody structure, affinity, density, as well as the liposome surface properties and drug to lipid ratios all contribute to the IL pharmacokinetic(PK) and pharmacodynamic(PD) behaviors. The optimal formulation parameters may vary for different target cells and tissues. Furthermore, besides the delivery of cytotoxic drugs to cancer cells, new ILs are being developed to interact with multiple target receptors, multiple target cells and trigger multiple therapeutic effects. We envision that the IL format can be a great platform for the molecular engineering of multi-valent, multi-specific interactions to achieve complex biological functions for therapeutic benefits, especially in the area of cancer immunotherapy.
Collapse
Affiliation(s)
- Jiaxing Di
- School of Pharmacy, Shanghai Jiao Tong University, China
| | - Fang Xie
- Department of Biomedical Engineering, Johns Hopkins University, United States of America
| | - Yuhong Xu
- College of Pharmacy and Chemistry, Dali University, China.
| |
Collapse
|
20
|
Luckey MA, Kim TH, Prakhar P, Keller HR, Crossman A, Choi S, Love PE, Walsh STR, Park JH. SOCS3 is a suppressor of γc cytokine signaling and constrains generation of murine Foxp3 + regulatory T cells. Eur J Immunol 2020; 50:986-999. [PMID: 32144749 DOI: 10.1002/eji.201948307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 01/16/2020] [Accepted: 03/05/2020] [Indexed: 12/18/2022]
Abstract
SOCS3 is a cytosolic inhibitor of cytokine signaling that suppresses the activation of cytokine receptor-associated JAK kinases. Mechanistically, SOCS3 is recruited to a site in the cytokine receptors known as the SOCS3-interaction motif, and then binds JAK molecules to inhibit their kinase activity. The SOCS3-interaction motif is found in receptors of the gp130 cytokine family but mostly absent from other cytokine receptors, including γc. Thus, SOCS3 has been considered a selective suppressor of gp130 family cytokines, but not γc cytokines. Considering that γc signaling induces SOCS3 expression in T cells, here we revisited the role of SOCS3 on γc signaling. Using SOCS3 transgenic mice, we found that increased abundance of SOCS3 not only suppressed signaling of the gp130 family cytokine IL-6, but also signaling of the γc family cytokine IL-7. Consequently, SOCS3 transgenic mice were impaired in IL-7-dependent T cell development in the thymus and the homeostasis of mature T cells in peripheral tissues. Moreover, enforced SOCS3 expression interfered with the generation of Foxp3+ regulatory T cells that requires signaling by the γc family cytokine IL-2. Collectively, we report an underappreciated role for SOCS3 in suppressing γc cytokine signaling, effectively expanding its scope of target cytokines in T cell immunity.
Collapse
Affiliation(s)
- Megan A Luckey
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| | - Tae-Hyoun Kim
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| | - Praveen Prakhar
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| | - Hilary R Keller
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD.,Department of Surgery, Guthrie Robert Packer Hospital, Sayre, PA
| | - Assiatu Crossman
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| | - Seeyoung Choi
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - Paul E Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | - Scott T R Walsh
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD
| |
Collapse
|
21
|
Wu L, Seung E, Xu L, Rao E, Lord DM, Wei RR, Cortez-Retamozo V, Ospina B, Posternak V, Ulinski G, Piepenhagen P, Francesconi E, El-Murr N, Beil C, Kirby P, Li A, Fretland J, Vicente R, Deng G, Dabdoubi T, Cameron B, Bertrand T, Ferrari P, Pouzieux S, Lemoine C, Prades C, Park A, Qiu H, Song Z, Zhang B, Sun F, Chiron M, Rao S, Radošević K, Yang ZY, Nabel GJ. Trispecific antibodies enhance the therapeutic efficacy of tumor-directed T cells through T cell receptor co-stimulation. ACTA ACUST UNITED AC 2019; 1:86-98. [DOI: 10.1038/s43018-019-0004-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
|
22
|
Cavero I, Holzgrefe HH. 18th world congress of basic and clinical pharmacology: thought-provoking lectures on drug safety issues. Expert Opin Drug Saf 2019; 18:1145-1148. [PMID: 31566014 DOI: 10.1080/14740338.2019.1673726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Introduction: Pharmacology of the Future for Science, Drug Development and Therapeutics was the leitmotif which guided the presentations at the 18th World Congress of Basic and Clinical Pharmacology held in Kyoto in July 2018 (WPC2018).Areas covered: Of the 380 invited lectures, this report reviews the opening presentation on immune checkpoint inhibitors and three talks dealing with drug safety issues (irreproducibility of nonclinical data, clinical Phase 1 catastrophes by TGN1214 and BIA-102,474-101 in healthy volunteers, and Phase I sentinel dosing to reduce risk to clinical study participants).Expert opinion: The nonclinical safety assessment of drug candidates preceding clinical investigations requires the adoption of more human predictable biological assays and a careful and critical analysis of all available knowledge on a candidate to ensure the safety of clinical trial participants.
Collapse
|
23
|
Sleep Matters: CD4 + T Cell Memory Formation and the Central Nervous System. Trends Immunol 2019; 40:674-686. [PMID: 31262652 DOI: 10.1016/j.it.2019.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 11/23/2022]
Abstract
The mechanisms of CD4+ T-cell memory formation in the immune system are debated. With the well-established concept of memory formation in the central nervous system (CNS), we propose that formation of CD4+ T-cell memory depends on the interaction of two different cell systems handling two types of stored information. First, information about antigen (event) and challenge (context) is taken up by antigen-presenting cells, as initial storage. Second, event and context information is transferred to CD4+ T cells. During activation, two categories of CD4+ T cell develop: effector CD4+ T cells, carrying event and context information, enabling them to efficiently focus their response to tissues under attack; and persisting CD4+ T cells, providing context-independent antigen-specific memories and long-term storage. This novel hypothesis is supported by the observation that mammalian sleep can improve both CNS and CD4+ T-cell memory.
Collapse
|
24
|
Medler J, Wajant H. Tumor necrosis factor receptor-2 (TNFR2): an overview of an emerging drug target. Expert Opin Ther Targets 2019; 23:295-307. [PMID: 30856027 DOI: 10.1080/14728222.2019.1586886] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Tumor necrosis factor (TNF) receptor 2 (TNFR2) is one of two receptors of the cytokines, TNF and lymphotoxin-α. TNFR1 is a strong inducer of proinflammatory activities. TNFR2 has proinflammatory effects too, but it also elicits strong anti-inflammatory activities and has protective effects on oligodendrocytes, cardiomyocytes, and keratinocytes. The protective and anti-inflammatory effects of TNFR2 may explain why TNF inhibitors failed to be effective in diseases such as heart failure or multiple sclerosis, where TNF has been strongly implicated as a driving force. Stimulatory and inhibitory TNFR2 targeting hence attracts considerable interest for the treatment of autoimmune diseases and cancer. Areas covered: Based on a brief description of the pathophysiological importance of the TNF-TNFR1/2 system, we discuss the potential applications of TNFR2 targeting therapies. We also debate TNFR2 activation as a way forward in the search for TNFR2-specific agents. Expert opinion: The use of TNFR2 to target regulatory T-cells is attractive, but this approach is just one amongst many suitable targets. With respect to its preference for Treg stimulation and protection of non-immune cells, TNFR2 is more unique and thus offers opportunities for translational success.
Collapse
Affiliation(s)
- Juliane Medler
- a Division for Molecular Internal Medicine, Department of Internal Medicine II , University Hospital Würzburg , Würzburg , Germany
| | - Harald Wajant
- a Division for Molecular Internal Medicine, Department of Internal Medicine II , University Hospital Würzburg , Würzburg , Germany
| |
Collapse
|
25
|
Talotta R, Rucci F, Canti G, Scaglione F. Pros and cons of the immunogenicity of monoclonal antibodies in cancer treatment: a lesson from autoimmune diseases. Immunotherapy 2019; 11:241-254. [DOI: 10.2217/imt-2018-0081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The aim of this review is to report the current evidence on immunogenicity of monoclonal antibodies (moAbs) used in cancer compared with autoimmune diseases, focusing on local microenvironment. English abstracts were identified in Medline and www.clinicaltrials.gov . A total of 82 papers were selected. The percentage of immunogenicity of moAbs used for cancer therapy, evaluated as the serum concentration of antidrug antibodies, is significantly lower than that of moAbs used for the treatment of autoimmune diseases. This condition may rely on a different immunologic background characterized by a hyperactivation of immune cells in autoimmune diseases. The formation of complexes between antidrug antibodies and non-neutralizing moAbs bound to neoplastic antigens may allow more efficient elimination of cancer cells, but additional studies are needed.
Collapse
Affiliation(s)
- Rossella Talotta
- Postgraduate School of Clinical Pharmacology & Toxicology, University of Milan, 20162, Milan, Italy
- Laboratory of Genetics, ASST ‘Grande Ospedale Metropolitano Niguarda’, 20162, Milan, Italy
| | - Francesco Rucci
- Postgraduate School of Clinical Pharmacology & Toxicology, University of Milan, 20162, Milan, Italy
- Laboratory of Genetics, ASST ‘Grande Ospedale Metropolitano Niguarda’, 20162, Milan, Italy
| | - Gianfranco Canti
- Department of Medical Biotechnology & Traslational Medicine, University of Milan, 20129, Milan, Italy
| | - Francesco Scaglione
- Department of Oncology & Onco-Hematology, University of Milan, 20162, Milan, Italy
- Clinical Pharmacology Unit, ASST ‘Grande Ospedale Metropolitano Niguarda’, 20162, Milan, Italy
| |
Collapse
|
26
|
Han X, Vesely MD. Stimulating T Cells Against Cancer With Agonist Immunostimulatory Monoclonal Antibodies. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 342:1-25. [PMID: 30635089 DOI: 10.1016/bs.ircmb.2018.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Elimination of cancer cells through antitumor immunity has been a long-sought after goal since Sir F. Macfarlane Burnet postulated the theory of immune surveillance against tumors in the 1950s. Finally, the use of immunotherapeutics against established cancer is becoming a reality in the past 5years. Most notable are the monoclonal antibodies (mAbs) directed against inhibitory T-cell receptors cytotoxic T lymphocyte antigen-4 and programmed death-1. The next generation of mAbs targeting T cells is designed to stimulate costimulatory receptors on T cells. Here we review the recent progress on these immunostimulatory agonist antibodies against the costimulatory receptors CD137, GITR, OX40, and CD27.
Collapse
Affiliation(s)
- Xue Han
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Matthew D Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
27
|
Abstract
Regulation of immune responses is critical for ensuring pathogen clearance and for preventing reaction against self-antigens. Failure or breakdown of immunological tolerance results in autoimmunity. CD28 is an important co-stimulatory receptor expressed on T cells that, upon specific ligand binding, delivers signals essential for full T-cell activation and for the development and homeostasis of suppressive regulatory T cells. Many
in vivo mouse models have been used for understanding the role of CD28 in the maintenance of immune homeostasis, thus leading to the development of CD28 signaling modulators that have been approved for the treatment of some autoimmune diseases. Despite all of this progress, a deeper understanding of the differences between the mouse and human receptor is required to allow a safe translation of pre-clinical studies in efficient therapies. In this review, we discuss the role of CD28 in tolerance and autoimmunity and the clinical efficacy of drugs that block or enhance CD28 signaling, by highlighting the success and failure of pre-clinical studies, when translated to humans.
Collapse
Affiliation(s)
- Nicla Porciello
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| |
Collapse
|
28
|
Jameson SC, Masopust D. What Is the Predictive Value of Animal Models for Vaccine Efficacy in Humans? Reevaluating the Potential of Mouse Models for the Human Immune System. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029132. [PMID: 28348039 DOI: 10.1101/cshperspect.a029132] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Much of what we understand about immunology, including the response to vaccines, come from studies in mice because they provide many practical advantages compared with research in higher mammals and humans. Nevertheless, modalities for preventing or treating disease do not always translate from mouse to humans, which has led to increasing scrutiny of the continued merits of mouse research. Here, we summarize the pros and cons of current laboratory mouse models for immunology research and discuss whether overreliance on nonphysiological, ultra-hygienic animal husbandry approaches has limited the ultimate translation potential of mouse-derived data to humans. Alternative approaches are discussed that may extend the use of the mouse model for preclinical studies.
Collapse
Affiliation(s)
- Stephen C Jameson
- University of Minnesota, Center for Immunology, Minneapolis, Minnesota 55414
| | - David Masopust
- University of Minnesota, Center for Immunology, Minneapolis, Minnesota 55414
| |
Collapse
|
29
|
Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018; 131:58-67. [PMID: 29118008 PMCID: PMC6317697 DOI: 10.1182/blood-2017-06-741033] [Citation(s) in RCA: 816] [Impact Index Per Article: 116.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/31/2017] [Indexed: 02/08/2023] Open
Abstract
CD28 and CTLA-4 are members of a family of immunoglobulin-related receptors that are responsible for various aspects of T-cell immune regulation. The family includes CD28, CTLA-4, and ICOS as well as other proteins, including PD-1, BTLA, and TIGIT. These receptors have both stimulatory (CD28, ICOS) and inhibitory roles (CTLA-4, PD-1, BTLA, and TIGIT) in T-cell function. Increasingly, these pathways are targeted as part of immune modulatory strategies to treat cancers, referred to generically as immune checkpoint blockade, and conversely to treat autoimmunity and CTLA-4 deficiency. Here, we focus on the biology of the CD28/CTLA-4 pathway as a framework for understanding the impacts of therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Behzad Rowshanravan
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Neil Halliday
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
30
|
Martini E, Stirparo GG, Kallikourdis M. Immunotherapy for cardiovascular disease. J Leukoc Biol 2017; 103:493-500. [PMID: 29345361 DOI: 10.1002/jlb.5mr0717-306r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/16/2017] [Accepted: 11/24/2017] [Indexed: 12/11/2022] Open
Abstract
Heart failure (HF), the final stage of pathological cardiac hypertrophy, is a major cause of hospitalization and mortality. The role of inflammation in the pathogenesis of HF has been extensively studied, with great emphasis on proinflammatory cytokines. Yet, clinical trials targeting these cytokines failed to become a credible therapeutic strategy for HF. More recent studies are increasingly highlighting an active role for T cells in the progression of HF pathology. As a result, a number of novel immunotherapy strategies are emerging for the treatment of HF and other cardiovascular diseases, via the targeting of adaptive immunity. Here we provide an overview of the background, details, and expected outcomes of these attempts.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan, Italy
| | - Giuliano Giuseppe Stirparo
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan, Italy
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory, Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano, Milan, Italy.,Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, Italy
| |
Collapse
|
31
|
Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 2017; 6:e1371896. [PMID: 29209572 PMCID: PMC5706611 DOI: 10.1080/2162402x.2017.1371896] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The goal of cancer immunotherapy is to establish new or boost pre-existing anticancer immune responses that eradicate malignant cells while generating immunological memory to prevent disease relapse. Over the past few years, immunomodulatory monoclonal antibodies (mAbs) that block co-inhibitory receptors on immune effectors cells - such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) - or their ligands - such as CD274 (best known as PD-L1) - have proven very successful in this sense. As a consequence, many of such immune checkpoint blockers (ICBs) have already entered the clinical practice for various oncological indications. Considerable attention is currently being attracted by a second group of immunomodulatory mAbs, which are conceived to activate co-stimulatory receptors on immune effector cells. Here, we discuss the mechanisms of action of these immunostimulatory mAbs and summarize recent progress in their preclinical and clinical development.
Collapse
Affiliation(s)
- Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- DKFZ-Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
32
|
Ehret T, Torelli F, Klotz C, Pedersen AB, Seeber F. Translational Rodent Models for Research on Parasitic Protozoa-A Review of Confounders and Possibilities. Front Cell Infect Microbiol 2017. [PMID: 28638807 PMCID: PMC5461347 DOI: 10.3389/fcimb.2017.00238] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Rodents, in particular Mus musculus, have a long and invaluable history as models for human diseases in biomedical research, although their translational value has been challenged in a number of cases. We provide some examples in which rodents have been suboptimal as models for human biology and discuss confounders which influence experiments and may explain some of the misleading results. Infections of rodents with protozoan parasites are no exception in requiring close consideration upon model choice. We focus on the significant differences between inbred, outbred and wild animals, and the importance of factors such as microbiota, which are gaining attention as crucial variables in infection experiments. Frequently, mouse or rat models are chosen for convenience, e.g., availability in the institution rather than on an unbiased evaluation of whether they provide the answer to a given question. Apart from a general discussion on translational success or failure, we provide examples where infections with single-celled parasites in a chosen lab rodent gave contradictory or misleading results, and when possible discuss the reason for this. We present emerging alternatives to traditional rodent models, such as humanized mice and organoid primary cell cultures. So-called recombinant inbred strains such as the Collaborative Cross collection are also a potential solution for certain challenges. In addition, we emphasize the advantages of using wild rodents for certain immunological, ecological, and/or behavioral questions. The experimental challenges (e.g., availability of species-specific reagents) that come with the use of such non-model systems are also discussed. Our intention is to foster critical judgment of both traditional and newly available translational rodent models for research on parasitic protozoa that can complement the existing mouse and rat models.
Collapse
Affiliation(s)
- Totta Ehret
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany.,Department of Molecular Parasitology, Humboldt-Universität zu BerlinBerlin, Germany
| | - Francesca Torelli
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Christian Klotz
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| | - Amy B Pedersen
- School of Biological Sciences, University of EdinburghEdinburgh, United Kingdom
| | - Frank Seeber
- FG16 - Mycotic and Parasitic Agents and Mycobacteria, Robert Koch InstituteBerlin, Germany
| |
Collapse
|
33
|
O'Donnell JS, Smyth MJ, Teng MWL. PD1 functions by inhibiting CD28-mediated co-stimulation. Clin Transl Immunology 2017; 6:e138. [PMID: 28690844 PMCID: PMC5493584 DOI: 10.1038/cti.2017.15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Jake S O'Donnell
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
34
|
Aravalli RN, Steer CJ. Immune-Mediated Therapies for Liver Cancer. Genes (Basel) 2017; 8:E76. [PMID: 28218682 PMCID: PMC5333065 DOI: 10.3390/genes8020076] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023] Open
Abstract
In recent years, immunotherapy has gained renewed interest as an alternative therapeutic approach for solid tumors. Its premise is based on harnessing the power of the host immune system to destroy tumor cells. Development of immune-mediated therapies, such as vaccines, adoptive transfer of autologous immune cells, and stimulation of host immunity by targeting tumor-evasive mechanisms have advanced cancer immunotherapy. In addition, studies on innate immunity and mechanisms of immune evasion have enhanced our understanding on the immunology of liver cancer. Preclinical and clinical studies with immune-mediated therapies have shown potential benefits in patients with liver cancer. In this review, we summarize current knowledge and recent developments in tumor immunology by focusing on two main primary liver cancers: hepatocellular carcinoma and cholangiocarcinoma.
Collapse
Affiliation(s)
- Rajagopal N Aravalli
- Department of Electrical and Computer Engineering, University of Minnesota, 200 Union Street S.E., Minneapolis, MN 55455, USA.
| | - Clifford J Steer
- Departments of Medicine and Genetics, Cell Biology and Development, University of Minnesota, 420 Delaware Street S.E., Minneapolis, MN 55455, USA.
| |
Collapse
|
35
|
Lymphocytes at the Heart of Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1003:225-250. [DOI: 10.1007/978-3-319-57613-8_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
36
|
Chenette EJ. The FEBS Journalpast … and present. FEBS J 2016; 283:4408-4411. [DOI: 10.1111/febs.13974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|