1
|
Li L, Pu H, Zhang X, Guo X, Li G, Zhang M. Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol 2025; 209:104683. [PMID: 40024354 DOI: 10.1016/j.critrevonc.2025.104683] [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: 06/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lung cancer is one of the most common malignant tumors, of which non-small cell lung cancer (NSCLC) accounts for about 85 %. Although immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 inhibitors, have significantly improved the prognosis of patients with NSCLC. There are still many patients do not benefit from ICIs. Primary resistance remains a major challenge in advanced NSCLC. The cancer-immunity cycle describes the process from antigen release to T cell recognition and killing of the tumor, which provides a framework for understanding anti-tumor immunity. The classical cycle consists of seven steps, and alterations at each stage can result in resistance. This review examines the current status of PD-1/PD-L1 blockade in the treatment of advanced NSCLC and explores potential mechanisms of resistance. We summarize the latest clinical trials of PD-1/PD-L1 inhibitors combined with other therapies and explore potential targets for overcoming primary resistance to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaoxin Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaotian Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Guangrui Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
2
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [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: 12/10/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
3
|
Johnson M, Hong D, Braña I, Schöffski P, Galvao V, Rangwala F, Ma B, Hernandez R, Kamat A, Kato K, Schreiber TH, Pandite L, Siu LL. First-in-human, phase 1 dose escalation study of SL-279252, a hexameric PD1-Fc-OX40L fusion protein, in patients with advanced solid tumors and lymphoma. Invest New Drugs 2025; 43:284-292. [PMID: 40042547 DOI: 10.1007/s10637-025-01518-7] [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: 01/09/2025] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
SL-279252 is a bifunctional hexameric fusion protein adjoining the extracellular domains of PD-1 and OX40L via an inert IgG4 derived Fc domain. A Phase 1 dose escalation study was conducted in patients (pts) with advanced solid tumors or lymphomas. SL-279252 was administered intravenously across 12 dose levels (range: 0.0001-24 mg/kg). Objectives included evaluation of safety, dose-limiting toxicity (DLT), recommended phase 2 dose, pharmacokinetic and pharmacodynamic (PD) parameters, and anti-tumor activity. Forty-nine pts (48 with solid tumor and 1 with lymphoma) were enrolled (median age 64 years; 53% male; median [range] of 3 [0-5] prior systemic therapies; 61% had been previously treated with PD-1/L1 inhibitors). Most common treatment-related adverse events (AEs) were infusion-related reaction (16%), maculopapular rash (10%), fatigue (6%), and neutropenia (6%). Treatment-related Grade (G) 3 AE was neutropenia (4%). There were no G4 or G5 AEs or DLTs. SL-279252 Cmax and area under the curve (AUC) increased proportionally with dose. T½ was ~ 20 h. Baseline anti-drug antibodies (ADA) were observed in 11/42 pts who had received a PD-1 inhibitor within 250 days. 7/31 pts had a persistent SL-279252 induced ADA response. PD effects consistent with OX40 engagement included dose dependent egress of CD4 + OX40 + cells and increases in Ki67 + CD4 and CD8 central and effector memory cells in the blood. Best response by iRECIST [1] in 46 response evaluable subjects was 1 iPR and 15 iSD. SL-279252 was well tolerated. PD effects consistent with OX40 activation were observed, however, efficacy was limited which may have been due to the disease characteristics, prior treatment with PD-1/L1 inhibitors, neutralization of the PD-1 domain of SL-279252 by circulating PD-1 inhibitors, limited SL-279252 penetration into tumors or other variables. Trial register number NCT03894618. Trial registration date 28-March-2019.
Collapse
Affiliation(s)
| | - David Hong
- MD Anderson Cancer Center, Houston, TX, USA
| | - Irene Braña
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Patrick Schöffski
- University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | | | | | - Bo Ma
- Shattuck Labs, Durham, NC, 27709, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Shree T, Czerwinski D, Haebe S, Sathe A, Grimes S, Martin B, Ozawa M, Hoppe R, Ji H, Levy R. A Phase I Clinical Trial Adding OX40 Agonism to In Situ Therapeutic Cancer Vaccination in Patients with Low-Grade B-cell Lymphoma Highlights Challenges in Translation from Mouse to Human Studies. Clin Cancer Res 2025; 31:868-880. [PMID: 39745391 PMCID: PMC11922159 DOI: 10.1158/1078-0432.ccr-24-2770] [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: 08/23/2024] [Revised: 10/25/2024] [Accepted: 12/30/2024] [Indexed: 03/04/2025]
Abstract
PURPOSE Activating T-cell costimulatory receptors is a promising approach for cancer immunotherapy. In preclinical work, adding an OX40 agonist to in situ vaccination with SD101, a TLR9 agonist, was curative in a mouse model of lymphoma. We sought to test this combination in a phase I clinical trial for patients with low-grade B-cell lymphoma. PATIENTS AND METHODS We treated 14 patients with low-dose radiation, intratumoral SD101, and intratumoral and intravenous BMS986178, an agonistic anti-OX40 antibody. The primary outcome was safety. Secondary outcomes included overall response rate and progression-free survival. RESULTS Adverse events were consistent with prior experience with low-dose radiation and SD101. No synergistic or dose-limiting toxicities were observed. One patient had a partial response, and nine patients had stable disease, a result inferior to our experience with TLR9 agonism and low-dose radiation alone. Flow cytometry and single-cell RNA sequencing of serial tumor biopsies revealed that T and NK cells were activated after treatment. However, high baseline OX40 expression in T follicular helper and T regulatory type 1 cells, as well as high posttreatment soluble OX40, shed from these T cells upon activation, associated with progression-free survival of less than 6 months. CONCLUSIONS Clinical results of T-cell costimulatory receptor agonism have now repeatedly been inferior to the motivating preclinical results. Our study highlights potential barriers to clinical translation, particularly differences in preclinical and clinical reagents and the complex biology of these coreceptors in heterogeneous T cell subpopulations, some of which may antagonize immunotherapy.
Collapse
Affiliation(s)
- Tanaya Shree
- Division of Oncology, Department of Medicine, Stanford University School of Medicine; Stanford, California, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health and Sciences University; Portland, Oregon, USA
| | - Debra Czerwinski
- Division of Oncology, Department of Medicine, Stanford University School of Medicine; Stanford, California, USA
| | - Sarah Haebe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine; Stanford, California, USA
- Medical Department III, LMU University Hospital, LMU Munich; Munich, Germany
| | - Anuja Sathe
- Division of Oncology, Department of Medicine, Stanford University School of Medicine; Stanford, California, USA
| | - Sue Grimes
- Stanford Genome Technology Center, Stanford University; Stanford, California, USA
| | - Brock Martin
- Department of Pathology, Stanford University School of Medicine; Stanford, California, USA
| | - Michael Ozawa
- Department of Pathology, Stanford University School of Medicine; Stanford, California, USA
| | - Richard Hoppe
- Department of Radiation Oncology, Stanford University; Stanford, California, USA
| | - Hanlee Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine; Stanford, California, USA
- Stanford Genome Technology Center, Stanford University; Stanford, California, USA
| | - Ronald Levy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine; Stanford, California, USA
| |
Collapse
|
5
|
Jeong M, Yoon J, Kim K, Wang J, Koo Y, Sailor MJ, Joo J, Park JH. Programmable Porous Silicon Microparticles for Temporally Staged Drug Delivery in Combination Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7395-7405. [PMID: 39847761 DOI: 10.1021/acsami.4c19425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Combination therapies using checkpoint inhibitors with immunostimulatory agonists have attracted great attention due to their synergistic therapeutic effects for cancer treatment. However, such combination immunotherapies require specific timing of doses to show sufficient antitumor efficacy. Sequential treatment usually requires multiple administrations of the individual drugs at specific time points, thus increasing the complexity of the drug regimen and compromising patient compliance. Here, we introduce an injectable porous silicon microparticle (pSiMP) for combination cancer immunotherapy where its multilayered nanopore structure was electrochemically programmed to achieve release of three distinct immunomodulatory drugs in the right sequence at the desired time. We find the optimal sequential treatment timeline of stimulator of interferon genes (STING) agonist, anti-OX40 antibody (aOX40), and anti-PD-1 antibody (aPD-1) for immunosuppressive tumors. We show that a single intratumoral injection of a cocktail of release-programmed pSiMPs coloaded with each antibody and a STING agonist significantly suppresses the tumor growth compared to conventional treatment involving sequential bolus injections, or an injection of pSiMPs configured to release all drugs at the same time, with no delay. With the timely release of immunomodulatory drugs, the programmable pSiMPs offer an effective treatment strategy for combination immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jinmyoung Joo
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| | | |
Collapse
|
6
|
Zhang J, Zhou L, Sun X, Lin Y, Yuan J, Yang C, Liao C. SHR-1806, a robust OX40 agonist to promote T cell-mediated antitumor immunity. Cancer Biol Ther 2024; 25:2426305. [PMID: 39543823 PMCID: PMC11572088 DOI: 10.1080/15384047.2024.2426305] [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: 06/28/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Anti-CTLA-4 and anti-PD-1/PD-L1 antibodies have significantly revolutionized cancer immunotherapy. However, the persistent challenge of low patient response rates necessitates novel approaches to overcome immune tolerance. Targeting immunostimulatory signaling may have a better chance of success for its ability to enhance effector T cell (Teff) function and expansion for antitumor immunity. Among various immunostimulatory pathways, the evidence underscores the potential of activating OX40-OX40L signaling to enhance CD8+ T cell generation and maintenance while suppressing regulatory T cells (Tregs) within the tumor microenvironment (TME). In this study, we introduce a potent agonistic anti-OX40 antibody, SHR-1806, designed to target OX40 receptors on activated T cells and amplify antitumor immune responses. SHR-1806 demonstrates a high affinity and specificity for human OX40 protein, eliciting FcγR-mediated agonistic effects, T cell activation, antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) activities in vitro. In human OX40 knock-in mice bearing MC38 tumor, SHR-1806 shows a trend toward a higher potency than the reference anti-OX40 antibody produced in-house, GPX4, an analog of pogalizumab, the most advanced drug candidate developed by Roche. Furthermore, SHR-1806 displays promising anti-tumor activity alone or in combination with toll-like receptor 7 (TLR7) agonist or PD-L1 inhibitor in mouse models. Evaluation of SHR-1806 in rhesus monkeys indicates a favorable safety profile and typical pharmacokinetic characteristics. Thus, SHR-1806 emerges as a robust OX40 agonist with promising therapeutic potential.
Collapse
Affiliation(s)
- Jun Zhang
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| | - Lei Zhou
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| | - Xing Sun
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| | - Yuan Lin
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| | - Jimin Yuan
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| | - Changyong Yang
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| | - Cheng Liao
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Shanghai Shengdi Pharmaceutical Co., Ltd., Shanghai, China
- Innovative Drug R&D, Pre-clinical Development and Translational Medicine, Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, Jiangsu, China
| |
Collapse
|
7
|
Wei J, Li W, Zhang P, Guo F, Liu M. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer 2024; 23:279. [PMID: 39725966 DOI: 10.1186/s12943-024-02179-5] [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: 09/05/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed the treatment landscape for various malignancies, achieving notable clinical outcomes across a wide range of indications. Despite these advances, resistance to immune checkpoint blockade (ICB) remains a critical clinical challenge, characterized by variable response rates and non-durable benefits. However, growing research into the complex intrinsic and extrinsic characteristics of tumors has advanced our understanding of the mechanisms behind ICI resistance, potentially improving treatment outcomes. Additionally, robust predictive biomarkers are crucial for optimizing patient selection and maximizing the efficacy of ICBs. Recent studies have emphasized that multiple rational combination strategies can overcome immune checkpoint resistance and enhance susceptibility to ICIs. These findings not only deepen our understanding of tumor biology but also reveal the unique mechanisms of action of sensitizing agents, extending clinical benefits in cancer immunotherapy. In this review, we will explore the underlying biology of ICIs, discuss the significance of the tumor immune microenvironment (TIME) and clinical predictive biomarkers, analyze the current mechanisms of resistance, and outline alternative combination strategies to enhance the effectiveness of ICIs, including personalized strategies for sensitizing tumors to ICIs.
Collapse
Grants
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Wenke Li
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Pengfei Zhang
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Ming Liu
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
8
|
Croft M, Salek-Ardakani S, Ware CF. Targeting the TNF and TNFR superfamilies in autoimmune disease and cancer. Nat Rev Drug Discov 2024; 23:939-961. [PMID: 39448880 DOI: 10.1038/s41573-024-01053-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/26/2024]
Abstract
The first anti-tumour necrosis factor (TNF) monoclonal antibody, infliximab (Remicade), celebrated its 25th anniversary of FDA approval in 2023. Inhibitors of TNF have since proved clinically efficacious at reducing inflammation associated with several autoimmune diseases, including rheumatoid arthritis, psoriasis and Crohn's disease. The success of TNF inhibitors raised unrealistic expectations for targeting other members of the TNF superfamily (TNFSF) of ligands and their receptors, with difficulties in part related to their more limited, variable expression and potential redundancy. However, there has been a resurgence of interest and investment, with many of these cytokines or their cognate receptors now under clinical investigation as targets for modulation of autoimmune and inflammatory diseases, as well as cancer. This Review assesses TNFSF-targeted biologics currently in clinical development for immune system-related diseases, highlighting ongoing challenges and future directions.
Collapse
Affiliation(s)
- Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | | | - Carl F Ware
- Laboratory of Molecular Immunology, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
9
|
Serrano A, Zalba S, Lasarte JJ, Troconiz IF, Riva N, Garrido MJ. Quantitative Approach to Explore Regulatory T Cell Activity in Immuno-Oncology. Pharmaceutics 2024; 16:1461. [PMID: 39598584 PMCID: PMC11597491 DOI: 10.3390/pharmaceutics16111461] [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: 10/01/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
The failure of immunotherapies in cancer patients is being widely studied due to the complexities present in the tumor microenvironment (TME), where regulatory T cells (Treg) appear to actively participate in providing an immune escape mechanism for tumors. Therefore, therapies to specifically inhibit tumor-infiltrating Treg represent a challenge, because Treg are distributed throughout the body and provide physiological immune homeostasis to prevent autoimmune diseases. Characterization of immunological and functional profiles could help to identify the mechanisms that need to be inhibited or activated to ensure Treg modulation in the tumor. To address this, quantitative in silico approaches based on mechanistic mathematical models integrating multi-scale information from immune and tumor cells and the effect of different therapies have allowed the building of computational frameworks to simulate different hypotheses, some of which have subsequently been experimentally validated. Therefore, this review presents a list of diverse computational mathematical models that examine the role of Treg as a crucial immune resistance mechanism contributing to the failure of immunotherapy. In addition, this review highlights the relevance of certain molecules expressed in Treg that are associated with the TME immunosuppression, which could be incorporated into the mathematical model for a better understanding of the contribution of Treg modulation. Finally, different preclinical and clinical combinations of molecules are also included to show the trend of new therapies targeting Treg.
Collapse
Affiliation(s)
- Alejandro Serrano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.); (S.Z.); (I.F.T.)
- Navarra Institute for Health Research (IdisNA), 31008 Pamplona, Spain
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.); (S.Z.); (I.F.T.)
- Navarra Institute for Health Research (IdisNA), 31008 Pamplona, Spain
| | - Juan Jose Lasarte
- Navarra Institute for Health Research (IdisNA), 31008 Pamplona, Spain
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain;
| | - Iñaki F. Troconiz
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.); (S.Z.); (I.F.T.)
- Navarra Institute for Health Research (IdisNA), 31008 Pamplona, Spain
- Institute of Data Sciences and Artificial Intelligence (DATAI), University of Navarra, 31008 Pamplona, Spain
| | - Natalia Riva
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.); (S.Z.); (I.F.T.)
- Navarra Institute for Health Research (IdisNA), 31008 Pamplona, Spain
| | - Maria J. Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.); (S.Z.); (I.F.T.)
- Navarra Institute for Health Research (IdisNA), 31008 Pamplona, Spain
| |
Collapse
|
10
|
Thapa B, Kato S, Nishizaki D, Miyashita H, Lee S, Nesline MK, Previs RA, Conroy JM, DePietro P, Pabla S, Kurzrock R. OX40/OX40 ligand and its role in precision immune oncology. Cancer Metastasis Rev 2024; 43:1001-1013. [PMID: 38526805 PMCID: PMC11300540 DOI: 10.1007/s10555-024-10184-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Immune checkpoint inhibitors have changed the treatment landscape for various malignancies; however, their benefit is limited to a subset of patients. The immune machinery includes both mediators of suppression/immune evasion, such as PD-1, PD-L1, CTLA-4, and LAG-3, all of which can be inhibited by specific antibodies, and immune-stimulatory molecules, such as T-cell co-stimulatory receptors that belong to the tumor necrosis factor receptor superfamily (TNFRSF), including OX40 receptor (CD134; TNFRSF4), 4-1BB (CD137; TNFRSF9), and glucocorticoid-induced TNFR-related (GITR) protein (CD357; TNFRSF18). In particular, OX40 and its binding ligand OX40L (CD134L; TNFSF4; CD252) are critical for immunoregulation. When OX40 on activated T cells binds OX40L on antigen-presenting cells, T-cell activation and immune stimulation are initiated via enhanced T-cell survival, proliferation and cytotoxicity, memory T-cell formation, and abrogation of regulatory T cell (Treg) immunosuppressive functions. OX40 agonists are in clinical trials both as monotherapy and in combination with other immunotherapy agents, in particular specific checkpoint inhibitors, for cancer treatment. To date, however, only a minority of patients respond. Transcriptomic profiling reveals that OX40 and OX40L expression vary between and within tumor types, and that only ~ 17% of cancer patients have high OX40 and low OX40L, one of the expression patterns that might be theoretically amenable to OX40 agonist enhancement. Taken together, the data suggest that the OX40/OX40L machinery is a critical part of the immune stimulatory system and that understanding endogenous expression patterns of these molecules and co-existing checkpoints merits further investigation in the context of a precision immunotherapy strategy for cancer therapy.
Collapse
Affiliation(s)
- Bicky Thapa
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Shumei Kato
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | | | - Suzanna Lee
- Center for Personalized Cancer Therapy, University of California San Diego, Moores Cancer Center, La Jolla, CA, USA
| | | | | | | | | | | | - Razelle Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
11
|
Zhao J, Li L, Feng X, Yin H, Fan X, Gao C, Zhao M, Lu Q. Blockade of OX40/OX40L signaling using anti-OX40L alleviates murine lupus nephritis. Eur J Immunol 2024; 54:e2350915. [PMID: 38798163 DOI: 10.1002/eji.202350915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024]
Abstract
Genetic variants of the OX40 ligand (OX40L) locus are associated with the risk of systemic lupus erythematosus (SLE), it is unclear how the OX40L blockade delays the lupus phenotype. Therefore, we examined the effects of an anti-OX40L antibody in MRL/Lpr mice. Next, we investigated the effect of anti-OX40L on immunosuppression in keyhole limpet hemocyanin-immunized C57BL/6J mice. In vitro treatment of anti-OX40L in CD4+ T and B220+ B cells was used to explore the role of OX40L in the pathogenesis of SLE. Anti-OX40L alleviated murine lupus nephritis, accompanied by decreased production of anti-dsDNA and proteinuria, as well as lower frequencies of splenic T helper (Th) 1 and T-follicular helper cells (Tfh). In keyhole limpet hemocyanin-immunized mice, decreased levels of immunoglobulins and plasmablasts were observed in the anti-OX40L group. Anti-OX40L reduced the number and area of germinal centers. Compared with the control IgG group, anti-OX40L downregulated CD4+ T-cell differentiation into Th1 and Tfh cells and upregulated CD4+ T-cell differentiation into regulatory T cells in vitro. Furthermore, anti-OX40L inhibited toll-like receptor 7-mediated differentiation of antibody-secreting cells and antibody production through the regulation of the SPIB-BLIMP1-XBP1 axis in B cells. These results suggest that OX40L is a promising therapeutic target for SLE.
Collapse
Affiliation(s)
- Junpeng Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Liming Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiwei Feng
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Huiqi Yin
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xinyu Fan
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Changxing Gao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
- Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Dai J, Zhu J, Zhu L, Wang X, Bao J, Chen X, Zhou Y, Min L, Qi H, Liu Q, Shen J, Tian M, Shao J, Li R, Liu B. An investigator-initiated clinical study in patients with refractory or recurrent solid tumors: 'R-ISV-FOLactis' trial. Future Oncol 2024; 20:1393-1400. [PMID: 39034683 PMCID: PMC11376417 DOI: 10.1080/14796694.2024.2357063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/15/2024] [Indexed: 07/23/2024] Open
Abstract
Aim: In situ vaccination, a kind of therapeutic cancer vaccine, can be realized by radiotherapy and intratumoral immune injection. This study combines intratumoral injection, radiotherapy and PD-1 blockade for synergistic antitumor effect.Materials & methods: Patients with advanced solid tumors who are unresponsive or intolerant to standard treatment will be treated with hypofractionated radiotherapy, intratumoral injection of FOLactis, PD-1 blockade. The primary end point is to observe the efficacy and safety, with the secondary end point to evaluate abscopal effects and the correlation between the immunological rationale and efficacy.Discussion: The combined regimen will be utilized to trigger antitumor immunity and is expected to be feasible and effective and provide a novel option for the comprehensive treatment of cancer.Clinical Trial Registration: ChiCTR2200060660 (ChiCTR.gov.cn).
Collapse
Affiliation(s)
- Juanjuan Dai
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junmeng Zhu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lijing Zhu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaolu Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jinfeng Bao
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinjie Chen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yingling Zhou
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Limei Min
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haoyue Qi
- Department of Oncology, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qin Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Shen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Manman Tian
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Shao
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rutian Li
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
13
|
Qahwaji R, Ashankyty I, Sannan NS, Hazzazi MS, Basabrain AA, Mobashir M. Pharmacogenomics: A Genetic Approach to Drug Development and Therapy. Pharmaceuticals (Basel) 2024; 17:940. [PMID: 39065790 PMCID: PMC11279827 DOI: 10.3390/ph17070940] [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: 05/06/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The majority of the well-known pharmacogenomics research used in the medical sciences contributes to our understanding of medication interactions. It has a significant impact on treatment and drug development. The broad use of pharmacogenomics is required for the progress of therapy. The main focus is on how genes and an intricate gene system affect the body's reaction to medications. Novel biomarkers that help identify a patient group that is more or less likely to respond to a certain medication have been discovered as a result of recent developments in the field of clinical therapeutics. It aims to improve customized therapy by giving the appropriate drug at the right dose at the right time and making sure that the right prescriptions are issued. A combination of genetic, environmental, and patient variables that impact the pharmacokinetics and/or pharmacodynamics of medications results in interindividual variance in drug response. Drug development, illness susceptibility, and treatment efficacy are all impacted by pharmacogenomics. The purpose of this work is to give a review that might serve as a foundation for the creation of new pharmacogenomics applications, techniques, or strategies.
Collapse
Affiliation(s)
- Rowaid Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ibraheem Ashankyty
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
| | - Naif S. Sannan
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Ar Rimayah, Riyadh 14611, Saudi Arabia;
- King Abdullah International Medical Research Center, Jeddah 22384, Saudi Arabia
| | - Mohannad S. Hazzazi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ammar A. Basabrain
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Mobashir
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| |
Collapse
|
14
|
Willoughby JE, Dou L, Bhattacharya S, Jackson H, Seestaller-Wehr L, Kilian D, Bover L, Voo KS, Cox KL, Murray T, John M, Shi H, Bojczuk P, Jing J, Niederer H, Shepherd AJ, Hook L, Hopley S, Inzhelevskaya T, Penfold CA, Mockridge CI, English V, Brett SJ, Srinivasan R, Hopson C, Smothers J, Hoos A, Paul E, Martin SL, Morley PJ, Yanamandra N, Cragg MS. Impact of isotype on the mechanism of action of agonist anti-OX40 antibodies in cancer: implications for therapeutic combinations. J Immunother Cancer 2024; 12:e008677. [PMID: 38964788 PMCID: PMC11227834 DOI: 10.1136/jitc-2023-008677] [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] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND OX40 has been widely studied as a target for immunotherapy with agonist antibodies taken forward into clinical trials for cancer where they are yet to show substantial efficacy. Here, we investigated potential mechanisms of action of anti-mouse (m) OX40 and anti-human (h) OX40 antibodies, including a clinically relevant monoclonal antibody (mAb) (GSK3174998) and evaluated how isotype can alter those mechanisms with the aim to develop improved antibodies for use in rational combination treatments for cancer. METHODS Anti-mOX40 and anti-hOX40 mAbs were evaluated in a number of in vivo models, including an OT-I adoptive transfer immunization model in hOX40 knock-in (KI) mice and syngeneic tumor models. The impact of FcγR engagement was evaluated in hOX40 KI mice deficient for Fc gamma receptors (FcγR). Additionally, combination studies using anti-mouse programmed cell death protein-1 (mPD-1) were assessed. In vitro experiments using peripheral blood mononuclear cells (PBMCs) examining possible anti-hOX40 mAb mechanisms of action were also performed. RESULTS Isotype variants of the clinically relevant mAb GSK3174998 showed immunomodulatory effects that differed in mechanism; mIgG1 mediated direct T-cell agonism while mIgG2a acted indirectly, likely through depletion of regulatory T cells (Tregs) via activating FcγRs. In both the OT-I and EG.7-OVA models, hIgG1 was the most effective human isotype, capable of acting both directly and through Treg depletion. The anti-hOX40 hIgG1 synergized with anti-mPD-1 to improve therapeutic outcomes in the EG.7-OVA model. Finally, in vitro assays with human peripheral blood mononuclear cells (hPBMCs), anti-hOX40 hIgG1 also showed the potential for T-cell stimulation and Treg depletion. CONCLUSIONS These findings underline the importance of understanding the role of isotype in the mechanism of action of therapeutic mAbs. As an hIgG1, the anti-hOX40 mAb can elicit multiple mechanisms of action that could aid or hinder therapeutic outcomes, dependent on the microenvironment. This should be considered when designing potential combinatorial partners and their FcγR requirements to achieve maximal benefit and improvement of patient outcomes.
Collapse
Affiliation(s)
- Jane E Willoughby
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Lang Dou
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Heather Jackson
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Laura Seestaller-Wehr
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - David Kilian
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Laura Bover
- Immunology Department/ Genomics Medicine Department, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Kui S Voo
- ORBIT, Institute of Applied Cancer Science, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Kerry L Cox
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Tom Murray
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mel John
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hong Shi
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Paul Bojczuk
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Junping Jing
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Heather Niederer
- Biopharm Discovery, GlaxoSmithKline Research & Development Limited, Stevenage, UK
| | - Andrew J Shepherd
- Protein, Cellular and Structural Sciences, GlaxoSmithKline Research & Development Limited, Gunnels Wood Road, Stevenage, UK
| | - Laura Hook
- Biopharm Discovery, GlaxoSmithKline Research & Development Limited, Stevenage, UK
| | - Stephanie Hopley
- Biopharm Discovery, GlaxoSmithKline Research & Development Limited, Stevenage, UK
| | - Tatyana Inzhelevskaya
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Chris A Penfold
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - C Ian Mockridge
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Vikki English
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Sara J Brett
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Roopa Srinivasan
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Christopher Hopson
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - James Smothers
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Axel Hoos
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Elaine Paul
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
- GlaxoSmithKline, Durham, North Carolina, USA
| | - Stephen L Martin
- Biopharm Discovery, GlaxoSmithKline Research & Development Limited, Stevenage, UK
| | - Peter J Morley
- Immunology Research Unit, GlaxoSmithKline Research & Development Limited, Gunnels Wood Road, Stevenage, UK
| | - Niranjan Yanamandra
- Immuno-Oncology and Combinations RU, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
15
|
Butterfield LH, Najjar YG. Immunotherapy combination approaches: mechanisms, biomarkers and clinical observations. Nat Rev Immunol 2024; 24:399-416. [PMID: 38057451 PMCID: PMC11460566 DOI: 10.1038/s41577-023-00973-8] [Citation(s) in RCA: 76] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 12/08/2023]
Abstract
The approval of the first immune checkpoint inhibitors provided a paradigm shift for the treatment of malignancies across a broad range of indications. Whereas initially, single-agent immune checkpoint inhibition was used, increasing numbers of patients are now treated with combination immune checkpoint blockade, where non-redundant mechanisms of action of the individual agents generally lead to higher response rates. Furthermore, immune checkpoint therapy has been combined with various other therapeutic modalities, including chemotherapy, radiotherapy and other immunotherapeutics such as vaccines, adoptive cellular therapies, cytokines and others, in an effort to maximize clinical efficacy. Currently, a large number of clinical trials test combination therapies with an immune checkpoint inhibitor as a backbone. However, proceeding without inclusion of broad, if initially exploratory, biomarker investigations may ultimately slow progress, as so far, few combinations have yielded clinical successes based on clinical data alone. Here, we present the rationale for combination therapies and discuss clinical data from clinical trials across the immuno-oncology spectrum. Moreover, we discuss the evolution of biomarker approaches and highlight the potential new directions that comprehensive biomarker studies can yield.
Collapse
Affiliation(s)
- Lisa H Butterfield
- University of California San Francisco, Microbiology and Immunology, San Francisco, CA, USA.
| | | |
Collapse
|
16
|
Khan B, Qahwaji RM, Alfaifi MS, Mobashir M. Nivolumab and Ipilimumab Acting as Tormentors of Advanced Tumors by Unleashing Immune Cells and Associated Collateral Damage. Pharmaceutics 2024; 16:732. [PMID: 38931856 PMCID: PMC11207028 DOI: 10.3390/pharmaceutics16060732] [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: 03/18/2024] [Revised: 04/22/2024] [Accepted: 05/01/2024] [Indexed: 06/28/2024] Open
Abstract
Combining immune checkpoint inhibitors, specifically nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4), holds substantial promise in revolutionizing cancer treatment. This review explores the transformative impact of these combinations, emphasizing their potential for enhancing therapeutic outcomes across various cancers. Immune checkpoint proteins, such as PD1 and CTLA4, play a pivotal role in modulating immune responses. Blocking these checkpoints unleashes anticancer activity, and the synergy observed when combining multiple checkpoint inhibitors underscores their potential for enhanced efficacy. Nivolumab and ipilimumab harness the host's immune system to target cancer cells, presenting a powerful approach to prevent tumor development. Despite their efficacy, immune checkpoint inhibitors are accompanied by a distinct set of adverse effects, particularly immune-related adverse effects affecting various organs. Understanding these challenges is crucial for optimizing treatment strategies and ensuring patient well-being. Ongoing clinical trials are actively exploring the combination of checkpoint inhibitory therapies, aiming to decipher their synergistic effects and efficacy against diverse cancer types. This review discusses the mechanisms, adverse effects, and various clinical trials involving nivolumab and ipilimumab across different cancers, emphasizing their transformative impact on cancer treatment.
Collapse
Affiliation(s)
- Bushra Khan
- Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Rowaid M. Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22233, Saudi Arabia;
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mashael S. Alfaifi
- Department of Epidemiology, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Mohammad Mobashir
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institute, Solnavägen 9, 171 65 Solna, Sweden
| |
Collapse
|
17
|
Lim SH, Beers SA, Al-Shamkhani A, Cragg MS. Agonist Antibodies for Cancer Immunotherapy: History, Hopes, and Challenges. Clin Cancer Res 2024; 30:1712-1723. [PMID: 38153346 PMCID: PMC7615925 DOI: 10.1158/1078-0432.ccr-23-1014] [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: 08/08/2023] [Revised: 10/31/2023] [Accepted: 12/11/2023] [Indexed: 12/29/2023]
Abstract
Immunotherapy is among the most promising new treatment modalities to arise over the last two decades; antibody drugs are delivering immunotherapy to millions of patients with many different types of cancer. Initial success with antibody therapeutics came in the form of direct targeting or cytotoxic antibodies, such as rituximab and trastuzumab, which bind directly to tumor cells to elicit their destruction. These were followed by immunomodulatory antibodies that elicit antitumor responses by either stimulating immune cells or relieving tumor-mediated suppression. By far the most successful approach in the clinic to date has been relieving immune suppression, with immune checkpoint blockade now a standard approach in the treatment of many cancer types. Despite equivalent and sometimes even more impressive effects in preclinical models, agonist antibodies designed to stimulate the immune system have lagged behind in their clinical translation. In this review, we document the main receptors that have been targeted by agonist antibodies, consider the various approaches that have been evaluated to date, detail what we have learned, and consider how their anticancer potential can be unlocked.
Collapse
Affiliation(s)
- Sean H. Lim
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
| | - Stephen A. Beers
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
| | - Aymen Al-Shamkhani
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
| | - Mark S. Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, University of Southampton Faculty of Medicine, Southampton, SO16 6YD, UK
- Institute for Life Sciences, University of Southampton, Southampton, SO17 1BJ, UK
| |
Collapse
|
18
|
Min L, Wang X, Chen A, Zhou Y, Ge Y, Dai J, Chang X, Sun W, Liu Q, Zhou X, Tian M, Kong W, Zhu J, Shen J, Liu B, Li R. Design of a single-center, phase II trial to explore the efficacy and safety of 'R-ISV-RO' treatment in advanced tumors. Future Oncol 2024; 20:1139-1149. [PMID: 38445361 PMCID: PMC11318728 DOI: 10.2217/fon-2023-0962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
Background: The authors' preclinical study has confirmed that RO adjuvant (composed of TLR 7 agonists [imiquimod/R837] and OX40 agonists) injected into local lesions induces the regression of both primary tumor and distant metastasis. The authors propose to realize local control and exert abscopal effect through an 'R-ISV-RO' in situ strategy plus anti-PD-1 monoclonal antibody in advanced tumors. Methods: This study is a single-center, exploratory, phase II trial to evaluate the efficacy and safety of R-ISV-RO plus anti-PD-1 monoclonal antibody in advanced tumors. 30 patients with one or more measurable extracerebral lesions that are accessible for radiation or injection will be enrolled. The primary endpoint is the objective response rate of target lesions. Discussion/Conclusion: The efficacy and safety of the novel strategy will be further validated through this clinical trial.Clinical trial registration: ChiCTR2100053870 (www.chictr.org.cn/).
Collapse
Affiliation(s)
- Limei Min
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Xiaolu Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Anni Chen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese & Western Medicine, Nanjing University of Chinese Medicine, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Yingling Zhou
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Traditional Chinese & Western Medicine, Nanjing University of Chinese Medicine, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Yuchen Ge
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Juanjuan Dai
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Xiaofeng Chang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Wu Sun
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Qin Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Xia Zhou
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Manman Tian
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Wentao Kong
- Department of Ultrasound Diagnosis, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Junmeng Zhu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Jie Shen
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| | - Rutian Li
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, No. 321 Zhongshan Road, Gulou District, Nanjing City, Jiangsu Province, 210008, China
| |
Collapse
|
19
|
Rakké YS, Buschow SI, IJzermans JNM, Sprengers D. Engaging stimulatory immune checkpoint interactions in the tumour immune microenvironment of primary liver cancers - how to push the gas after having released the brake. Front Immunol 2024; 15:1357333. [PMID: 38440738 PMCID: PMC10910082 DOI: 10.3389/fimmu.2024.1357333] [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: 12/17/2023] [Accepted: 01/31/2024] [Indexed: 03/06/2024] Open
Abstract
Hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) are the first and second most common primary liver cancer (PLC). For decades, systemic therapies consisting of tyrosine kinase inhibitors (TKIs) or chemotherapy have formed the cornerstone of treating advanced-stage HCC and CCA, respectively. More recently, immunotherapy using immune checkpoint inhibition (ICI) has shown anti-tumour reactivity in some patients. The combination regimen of anti-PD-L1 and anti-VEGF antibodies has been approved as new first-line treatment of advanced-stage HCC. Furthermore, gemcibatine plus cisplatin (GEMCIS) with an anti-PD-L1 antibody is awaiting global approval for the treatment of advanced-stage CCA. As effective anti-tumour reactivity using ICI is achieved in a minor subset of both HCC and CCA patients only, alternative immune strategies to sensitise the tumour microenvironment of PLC are waited for. Here we discuss immune checkpoint stimulation (ICS) as additional tool to enhance anti-tumour reactivity. Up-to-date information on the clinical application of ICS in onco-immunology is provided. This review provides a rationale of the application of next-generation ICS either alone or in combination regimen to potentially enhance anti-tumour reactivity in PLC patients.
Collapse
Affiliation(s)
- Yannick S. Rakké
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Sonja I. Buschow
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| | - Jan N. M. IJzermans
- Department of Surgery, Erasmus MC-Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus MC-Cancer Institute-University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
20
|
Galle P, Finn RS, Mitchell CR, Ndirangu K, Ramji Z, Redhead GS, Pinato DJ. Treatment-emergent antidrug antibodies related to PD-1, PD-L1, or CTLA-4 inhibitors across tumor types: a systematic review. J Immunother Cancer 2024; 12:e008266. [PMID: 38238030 PMCID: PMC10806538 DOI: 10.1136/jitc-2023-008266] [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] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Increased understanding of how the immune system regulates tumor growth has innovated the use of immunotherapeutics to treat various cancers. The impact of such therapies, including programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, on the production of antidrug antibodies (ADAs) and their impact on outcomes, is poorly understood. This study aims to evaluate the clinical trial evidence on ADA incidence associated with PD-1, PD-L1, and CTLA-4 inhibitors in the treatment of cancer and to assess associations between treatment administered, ADA incidence, and treatment outcomes. METHODS Embase®, Medline®, and EBM Reviews were searched via the OVID® platform on February 15, 2022. Conference proceedings, clinical trial registries, and global regulatory and reimbursement body websites were also searched. Eligible publications included clinical trials enrolling patients receiving cancer treatment with either PD-1, PD-L1, or CTLA-4 reporting outcomes including incidence or prevalence of ADAs and the impact of immunogenicity on treatment safety and efficacy. Reference lists of eligible publications were also searched. The review was conducted and reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and evidence quality assessment was conducted using the appropriate Joanna Briggs Institute Critical Appraisal tool. RESULTS After screening 4160 records and reviewing 97 full publications, a total of 34 publications reporting on 68 trials were included. A further 41 relevant clinical trials were identified on ClinicalTrials.gov and a further 32 from searches of packaging inserts. In total, 141 relevant trials covering 15 different checkpoint inhibitors and 16 different tumor types were included. Across the included trials, atezolizumab was associated with the highest incidence of ADAs (29.6% of 639 patients), followed by nivolumab (11.2% of 2,085 patients). Combination checkpoint inhibitor treatment appeared to increase the rate of ADAs versus monotherapy. Only 17 trials reported on the impact of ADAs on treatment outcomes with mixed results for the impact of ADAs on treatment efficacy, safety, and pharmacokinetics. CONCLUSIONS Checkpoint inhibitors for the treatment of cancer are immunogenic, with the incidence of treatment-emergent ADAs varying between individual therapies. It remains unclear what impact ADAs have on treatment outcomes.
Collapse
Affiliation(s)
- Peter Galle
- University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Richard S Finn
- University of California Los Angeles, Los Angeles, California, USA
| | | | | | | | | | - David J Pinato
- Surgery and Cancer, Imperial College London, London, UK
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
21
|
Jang A, Lichterman JN, Zhong JY, Shoag JE, Garcia JA, Zhang T, Barata PC. Immune approaches beyond traditional immune checkpoint inhibitors for advanced renal cell carcinoma. Hum Vaccin Immunother 2023; 19:2276629. [PMID: 37947202 PMCID: PMC10653627 DOI: 10.1080/21645515.2023.2276629] [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: 08/05/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Renal cell carcinoma (RCC), especially clear cell RCC, is generally considered an immunotherapy-responsive cancer. Recently, the prognosis for patients with locally advanced and metastatic RCC has significantly improved with the regulatory approvals of anti-PD-1/PD-L1/CTLA-4 immune checkpoint inhibitor (ICI)-based regimens. Yet in most cases, RCC will remain initially unresponsive to treatment or will develop resistance over time. Hence, there remains an unmet need to understand what leads to ICI resistance and to develop novel immune and nonimmune treatments to enhance the response to ICIs. In this review, we highlight recently published studies and the latest clinical studies investigating the next generation of immune approaches to locally advanced and metastatic RCC beyond traditional ICIs. These trials include cytokines, gut microbiota-based therapies, novel immune checkpoint agents, vaccines, and chimeric antigen receptor T cells. These agents are being evaluated as monotherapy or in combination with traditional ICIs and will hopefully provide improved outcomes to patients with RCC soon.
Collapse
Affiliation(s)
- Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jake N. Lichterman
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey Y. Zhong
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Jonathan E. Shoag
- Department of Urology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jorge A. Garcia
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Tian Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pedro C. Barata
- Division of Solid Tumor Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
22
|
Bredel D, Tihic E, Mouraud S, Danlos FX, Susini S, Aglave M, Alfaro A, Mohamed-Djalim C, Rouanne M, Halse H, Bigorgne A, Tselikas L, Dalle S, Hartl DM, Baudin E, Guettier C, Vibert E, Rosmorduc O, Robert C, Ferlicot S, Parier B, Albiges L, de Montpreville VT, Besse B, Mercier O, Even C, Breuskin I, Classe M, Radulescu C, Lebret T, Pautier P, Gouy S, Scoazec JY, Zitvogel L, Marabelle A, Bonvalet M. Immune checkpoints are predominantly co-expressed by clonally expanded CD4 +FoxP3 + intratumoral T-cells in primary human cancers. J Exp Clin Cancer Res 2023; 42:333. [PMID: 38057799 DOI: 10.1186/s13046-023-02897-6] [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: 07/14/2023] [Accepted: 11/11/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND In addition to anti-PD(L)1, anti-CTLA-4 and anti-LAG-3, novel immune checkpoint proteins (ICP)-targeted antibodies have recently failed to demonstrate significant efficacy in clinical trials. In these trials, patients were enrolled without screening for drug target expression. Although these novel ICP-targeted antibodies were expected to stimulate anti-tumor CD8 + T-cells, the rationale for their target expression in human tumors relied on pre-clinical IHC stainings and transcriptomic data, which are poorly sensitive and specific techniques for assessing membrane protein expression on immune cell subsets. Our aim was to describe ICP expression on intratumoral T-cells from primary solid tumors to better design upcoming neoadjuvant cancer immunotherapy trials. METHODS We prospectively performed multiparameter flow cytometry and single-cell RNA sequencing (scRNA-Seq) paired with TCR sequencing on freshly resected human primary tumors of various histological types to precisely determine ICP expression levels within T-cell subsets. RESULTS Within a given tumor type, we found high inter-individual variability for tumor infiltrating CD45 + cells and for T-cells subsets. The proportions of CD8+ T-cells (~ 40%), CD4+ FoxP3- T-cells (~ 40%) and CD4+ FoxP3+ T-cells (~ 10%) were consistent across patients and indications. Intriguingly, both stimulatory (CD25, CD28, 4-1BB, ICOS, OX40) and inhibitory (PD-1, CTLA-4, PD-L1, CD39 and TIGIT) checkpoint proteins were predominantly co-expressed by intratumoral CD4+FoxP3+ T-cells. ScRNA-Seq paired with TCR sequencing revealed that T-cells with high clonality and high ICP expressions comprised over 80% of FoxP3+ cells among CD4+ T-cells. Unsupervised clustering of flow cytometry and scRNAseq data identified subsets of CD8+ T-cells and of CD4+ FoxP3- T-cells expressing certain checkpoints, though these expressions were generally lower than in CD4+ FoxP3+ T-cell subsets, both in terms of proportions among total T-cells and ICP expression levels. CONCLUSIONS Tumor histology alone does not reveal the complete picture of the tumor immune contexture. In clinical trials, assumptions regarding target expression should rely on more sensitive and specific techniques than conventional IHC or transcriptomics. Flow cytometry and scRNAseq accurately characterize ICP expression within immune cell subsets. Much like in hematology, flow cytometry can better describe the immune contexture of solid tumors, offering the opportunity to guide patient treatment according to drug target expression rather than tumor histological type.
Collapse
Affiliation(s)
- Delphine Bredel
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
| | - Edi Tihic
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Séverine Mouraud
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - François-Xavier Danlos
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Innovation Thérapeutique Et d'Essais Précoces (DITEP), 94805, Villejuif, France
| | - Sandrine Susini
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Marine Aglave
- Gustave Roussy, Plateforme de bioinformatique, F-94805, Villejuif, France
| | - Alexia Alfaro
- Gustave Roussy, Université Paris-Saclay, UMS 23/3655, Plateforme Imagerie Et Cytométrie, Villejuif, France
| | - Chifaou Mohamed-Djalim
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| | - Mathieu Rouanne
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, USA
| | - Héloise Halse
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1163, Institut Imagine, Université Paris Descartes, 75015, Paris, France
| | - Amélie Bigorgne
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1163, Institut Imagine, Université Paris Descartes, 75015, Paris, France
| | - Lambros Tselikas
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Stéphane Dalle
- Department of Dermatology, HCL Cancer Institute, Lyon Cancer Research Center, 69495, Lyon, France
| | - Dana M Hartl
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Eric Baudin
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Catherine Guettier
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service d'Anatomie Pathologique, Hôpital Bicêtre, AP-HP, 94270, Le Kremlin-Bicêtre, France
- UMR-S 1193, Hôpital Paul Brousse Université Paris Saclay, 94800, Villejuif, France
| | - Eric Vibert
- UMR-S 1193, Hôpital Paul Brousse Université Paris Saclay, 94800, Villejuif, France
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, 94800, Villejuif, France
| | - Olivier Rosmorduc
- Centre Hépato-Biliaire, Hôpital Paul Brousse, AP-HP, 94800, Villejuif, France
| | - Caroline Robert
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U981, Gustave Roussy, 94805, Villejuif, France
| | - Sophie Ferlicot
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service d'Anatomie Pathologique, Hôpital Bicêtre, AP-HP, 94270, Le Kremlin-Bicêtre, France
- Centre National de Recherche Scientifique (CNRS), Gustave Roussy, Université Paris-Saclay, UMR 9019, 94805, Villejuif, France
| | - Bastien Parier
- Service de Chirurgie Urologique, Hôpital Bicêtre, AP-HP, Le Kremlin-Bicêtre, France
| | - Laurence Albiges
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | | | - Benjamin Besse
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Olaf Mercier
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
- Service de Chirurgie Thoracique Et Transplantation Cardio-Pulmonaire, Hôpital Marie-Lannelongue, UMR_S 999 INSERM, Université Paris-Saclay, GHPSJ, 92350, Le Plessis-Robinson, France
| | - Caroline Even
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Ingrid Breuskin
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Marion Classe
- Gustave Roussy, Département de Biopathologie, F-94805, Villejuif, France
| | - Camélia Radulescu
- Département de Pathologie, Hôpital Foch, UVSQ, Université Paris-Saclay, 92150, Suresnes, France
| | - Thierry Lebret
- Département d'Urologie, Hôpital Foch, UVSQ-Université Paris-Saclay, 92150, Suresnes, France
| | - Patricia Pautier
- Gustave Roussy, Département d'Oncologie Médicale, F-94805, Villejuif, France
| | - Sébastien Gouy
- Gustave Roussy, Université Paris Saclay, Département d'Anesthésie, Chirurgie et Imagerie Interventionnelle, F-94805, Villejuif, France
| | - Jean-Yves Scoazec
- Gustave Roussy, Département de Biopathologie, F-94805, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Aurélien Marabelle
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France.
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France.
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France.
- Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France.
- Gustave Roussy, Département d'Innovation Thérapeutique Et d'Essais Précoces (DITEP), 94805, Villejuif, France.
| | - Mélodie Bonvalet
- Gustave Roussy, 114 Rue Édouard Vaillant, 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) U1015, Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), 94805, Villejuif, France
- Institut National de La Santé Et de La Recherche Médicale (INSERM) CIC1428, Centre d'Investigation Clinique BIOTHERIS, 94805, Villejuif, France
| |
Collapse
|
23
|
Fujikawa K, Saito T, Kurose K, Kojima T, Funakoshi T, Sato E, Kakimi K, Iida S, Doki Y, Oka M, Ueda R, Wada H. Integrated analysis of phase 1a and 1b randomized controlled trials; Treg-targeted cancer immunotherapy with the humanized anti-CCR4 antibody, KW-0761, for advanced solid tumors. PLoS One 2023; 18:e0291772. [PMID: 37729184 PMCID: PMC10511099 DOI: 10.1371/journal.pone.0291772] [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: 03/29/2023] [Accepted: 08/03/2023] [Indexed: 09/22/2023] Open
Abstract
INTRODUCTION Regulatory T cells (Tregs) have attracted attention as a novel therapeutic target to augment the clinical efficacy of immunotherapy. We conducted phase Ia and Ib trials to examine the safety and efficacy of the anti-CCR4 antibody, KW-0761 (mogamulizumab), which may eliminate effector Tregs (eTregs). We herein overviewed the results of these trials, presented cases with a durable clinical response, and investigated factors associated with the clinical effects of KW-0761. METHODS Forty-nine patients with CCR4-negative solid cancers were enrolled in the phase Ia and Ib trials on KW-0761. An integral analysis of safety, clinical responses, prognosis, blood laboratory data, and cancer testis antigen-specific immune responses was performed. RESULTS Grade 3-4 treatment-related adverse events were reported in 21 (42.9%) out of 49 patients, all of which were manageable. A partial response and stable disease were observed in 1 and 9 patients, respectively. A durable clinical response was noted in 2 esophageal and 2 lung cancer patients. eTreg depletion in peripheral blood was confirmed in most patients, and eTreg depletion was sustained during the KW-0761 treatment. High lymphocyte levels at baseline and 2 weeks after the initiation of KW-0761 were associated with a favorable clinical outcome. CONCLUSIONS A durable clinical response was noted in some patients, and high lymphocyte levels before treatment initiation may be a biomarker for the efficacy of KW-0761. The synergistic effect of KW-0761 for depleting Tregs and other immunotherapies is expected in the future.
Collapse
Affiliation(s)
- Kaoru Fujikawa
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takuro Saito
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Koji Kurose
- Department of Respiratory Medicine, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Takashi Kojima
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Eiichi Sato
- Department of Pathology, Institute of Medical Science (Medical Research Center), Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-Ku, Tokyo, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Mikio Oka
- Department of Immuno-Oncology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Ryuzo Ueda
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
24
|
Cirella A, Bolaños E, Luri-Rey C, Di Trani CA, Olivera I, Gomis G, Glez-Vaz J, Pinci B, Garasa S, Sánchez-Gregorio S, Azpilikueta A, Eguren-Santamaria I, Valencia K, Palencia B, Alvarez M, Ochoa MC, Teijeira Á, Berraondo P, Melero I. Intratumoral immunotherapy with mRNAs encoding chimeric protein constructs encompassing IL-12, CD137 agonists, and TGF-β antagonists. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:668-682. [PMID: 37650116 PMCID: PMC10462790 DOI: 10.1016/j.omtn.2023.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/25/2023] [Indexed: 09/01/2023]
Abstract
Intratumoral immunotherapy strategies for cancer based on interleukin-12 (IL-12)-encoding cDNA and mRNA are under clinical development in combination with anti-PD-(L)1 monoclonal antibodies. To make the most of these approaches, we have constructed chimeric mRNAs encoding single-chain IL-12 fused to single-chain fragment variable (scFv) antibodies that bind to transforming growth factor β (TGF-β) and CD137 (4-1BB). Several neutralizing TGF-β agents and CD137 agonists are also undergoing early-phase clinical trials. To attain TGF-β and CD137 binding by the constructions, we used bispecific tandem scFv antibodies (taFvs) derived from the specific 1D11 and 1D8 monoclonal antibodies (mAbs), respectively. Transfection of mRNAs encoding the chimeric constructs achieved functional expression of the proteins able to act on their targets. Upon mRNA intratumoral injections in the transplantable mouse cancer models CT26, MC38, and B16OVA, potent therapeutic effects were observed following repeated injections into the tumors. Efficacy was dependent on the number of CD8+ T cells able to recognize tumor antigens that infiltrated the malignant tissue. Although the abscopal effects on concomitant uninjected lesions were modest, such distant effects on untreated lesions were markedly increased when combined with systemic PD-1 blockade.
Collapse
Affiliation(s)
- Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Claudia Augusta Di Trani
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Gabriel Gomis
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Javier Glez-Vaz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Beatrice Pinci
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Saray Garasa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Sandra Sánchez-Gregorio
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Arantza Azpilikueta
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Iñaki Eguren-Santamaria
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
| | - Karmele Valencia
- Program of Solid Tumors, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Belén Palencia
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
| | - Maite Alvarez
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Maria C. Ochoa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Álvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, 31008 Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- Department of Oncology, Clínica Universidad de Navarra, 28027 Madrid, Spain
- Centro Del Cancer de La Universidad de Navarra (CCUN), 31008 Pamplona, Spain
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
25
|
Cortellino S, Quagliariello V, Delfanti G, Blaževitš O, Chiodoni C, Maurea N, Di Mauro A, Tatangelo F, Pisati F, Shmahala A, Lazzeri S, Spagnolo V, Visco E, Tripodo C, Casorati G, Dellabona P, Longo VD. Fasting mimicking diet in mice delays cancer growth and reduces immunotherapy-associated cardiovascular and systemic side effects. Nat Commun 2023; 14:5529. [PMID: 37684243 PMCID: PMC10491752 DOI: 10.1038/s41467-023-41066-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Immune checkpoint inhibitors cause side effects ranging from autoimmune endocrine disorders to severe cardiotoxicity. Periodic Fasting mimicking diet (FMD) cycles are emerging as promising enhancers of a wide range of cancer therapies including immunotherapy. Here, either FMD cycles alone or in combination with anti-OX40/anti-PD-L1 are much more effective than immune checkpoint inhibitors alone in delaying melanoma growth in mice. FMD cycles in combination with anti-OX40/anti-PD-L1 also show a trend for increased effects against a lung cancer model. As importantly, the cardiac fibrosis, necrosis and hypertrophy caused by immune checkpoint inhibitors are prevented/reversed by FMD treatment in both cancer models whereas immune infiltration of CD3+ and CD8+ cells in myocardial tissues and systemic and myocardial markers of oxidative stress and inflammation are reduced. These results indicate that FMD cycles in combination with immunotherapy can delay cancer growth while reducing side effects including cardiotoxicity.
Collapse
Affiliation(s)
- S Cortellino
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero in Vulture, Italy
| | - V Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - G Delfanti
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - O Blaževitš
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - C Chiodoni
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - N Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - A Di Mauro
- Pathology and Cytopathology Unit, Department of Support to Cancer Pathways Diagnostics Area, Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale", 80131, Naples, Italy
| | - F Tatangelo
- Pathology and Cytopathology Unit, Department of Support to Cancer Pathways Diagnostics Area, Istituto Nazionale Tumori-IRCCS "Fondazione G. Pascale", 80131, Naples, Italy
| | - F Pisati
- Histopathology Unit, Cogentech Società Benefit srl, 20139, Milan, Italy
| | - A Shmahala
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - S Lazzeri
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - V Spagnolo
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - E Visco
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - C Tripodo
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- University of Palermo School of Medicine, Palermo, Italy
| | - G Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - P Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - V D Longo
- IFOM, The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
26
|
Sun Q, Hong Z, Zhang C, Wang L, Han Z, Ma D. Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends. Signal Transduct Target Ther 2023; 8:320. [PMID: 37635168 PMCID: PMC10460796 DOI: 10.1038/s41392-023-01522-4] [Citation(s) in RCA: 190] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/11/2023] [Accepted: 05/28/2023] [Indexed: 08/29/2023] Open
Abstract
Immune-checkpoint inhibitors (ICBs), in addition to targeting CTLA-4, PD-1, and PD-L1, novel targeting LAG-3 drugs have also been approved in clinical application. With the widespread use of the drug, we must deeply analyze the dilemma of the agents and seek a breakthrough in the treatment prospect. Over the past decades, these agents have demonstrated dramatic efficacy, especially in patients with melanoma and non-small cell lung cancer (NSCLC). Nonetheless, in the field of a broad concept of solid tumours, non-specific indications, inseparable immune response and side effects, unconfirmed progressive disease, and complex regulatory networks of immune resistance are four barriers that limit its widespread application. Fortunately, the successful clinical trials of novel ICB agents and combination therapies, the advent of the era of oncolytic virus gene editing, and the breakthrough of the technical barriers of mRNA vaccines and nano-delivery systems have made remarkable breakthroughs currently. In this review, we enumerate the mechanisms of each immune checkpoint targets, associations between ICB with tumour mutation burden, key immune regulatory or resistance signalling pathways, the specific clinical evidence of the efficacy of classical targets and new targets among different tumour types and put forward dialectical thoughts on drug safety. Finally, we discuss the importance of accurate triage of ICB based on recent advances in predictive biomarkers and diagnostic testing techniques.
Collapse
Affiliation(s)
- Qian Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhenya Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Cong Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liangliang Wang
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhiqiang Han
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Ding Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
27
|
Redmond WL. Challenges and opportunities in the development of combination immunotherapy with OX40 agonists. Expert Opin Biol Ther 2023; 23:901-912. [PMID: 37587644 PMCID: PMC10530613 DOI: 10.1080/14712598.2023.2249396] [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: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Costimulatory members of the tumor necrosis factor receptor family, such as OX40 (CD134), provide essential survival and differentiation signals that enhance T cell function. Specifically, OX40 (CD134) agonists stimulate potent anti-tumor immunity in a variety of preclinical models but their therapeutic impact in patients with advanced malignancies has been limited thus far. AREAS COVERED In this review, we discuss the current state of combination immunotherapy with OX40 agonists including preclinical studies and recent clinical trials. We also discuss the strengths and limitations of these approaches and provide insight into alternatives that may help enhance the efficacy of combination OX40 agonist immunotherapy. EXPERT OPINION OX40 agonist immunotherapy has not yet demonstrated significant clinical activity as a monotherapy or in combination with immune checkpoint blockade (ICB), likely due to several factors including the timing of administration, drug potency, and selection of agents for combination therapy clinical trials. We believe that careful consideration of the biological mechanisms regulating OX40 expression and function may help inform new approaches, particularly in combination with novel agents, capable of increasing the therapeutic efficacy of this approach.
Collapse
Affiliation(s)
- William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213
| |
Collapse
|
28
|
Postel-Vinay S, Lam VK, Ros W, Bauer TM, Hansen AR, Cho DC, Stephen Hodi F, Schellens JHM, Litton JK, Aspeslagh S, Autio KA, Opdam FL, McKean M, Somaiah N, Champiat S, Altan M, Spreafico A, Rahma O, Paul EM, Ahlers CM, Zhou H, Struemper H, Gorman SA, Watmuff M, Yablonski KM, Yanamandra N, Chisamore MJ, Schmidt EV, Hoos A, Marabelle A, Weber JS, Heymach JV. First-in-human phase I study of the OX40 agonist GSK3174998 with or without pembrolizumab in patients with selected advanced solid tumors (ENGAGE-1). J Immunother Cancer 2023; 11:jitc-2022-005301. [PMID: 36927527 PMCID: PMC10030671 DOI: 10.1136/jitc-2022-005301] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND The phase I first-in-human study ENGAGE-1 evaluated the humanized IgG1 OX40 agonistic monoclonal antibody GSK3174998 alone (Part 1 (P1)) or in combination with pembrolizumab (Part 2 (P2)) in patients with advanced solid tumors. METHODS GSK3174998 (0.003-10 mg/kg) ± pembrolizumab (200 mg) was administered intravenously every 3 weeks using a continuous reassessment method for dose escalation. Primary objectives were safety and tolerability; secondary objectives included pharmacokinetics, immunogenicity, pharmacodynamics, and clinical activity. RESULTS 138 patients were enrolled (45 (P1) and 96 (P2, including 3 crossovers)). Treatment-related adverse events occurred in 51% (P1) and 64% (P2) of patients, fatigue being the most common (11% and 24%, respectively). No dose-toxicity relationship was observed, and maximum-tolerated dose was not reached. Dose-limiting toxicities (P2) included Grade 3 (G3) pleural effusion and G1 myocarditis with G3 increased troponin. GSK3174998 ≥0.3 mg/kg demonstrated pharmacokinetic linearity and >80% receptor occupancy on circulating T cells; 0.3 mg/kg was selected for further evaluation. Limited clinical activity was observed for GSK3174998 (P1: disease control rate (DCR) ≥24 weeks 9%) and was not greater than that expected for pembrolizumab alone (P2: overall response rate 8%, DCR ≥24 weeks 28%). Multiplexed immunofluorescence data from paired biopsies suggested that increased infiltration of natural killer (NK)/natural killer T (NKT) cells and decreased regulatory T cells (Tregs) in the tumor microenvironment may contribute to clinical responses: CD16+CD56-CD134+ NK /NKT cells and CD3+CD4+FOXP3+CD134+ Tregs exhibited the largest magnitude of change on treatment, whereas CD3+CD8+granzyme B+PD-1+CD134+ cytotoxic T cells were the least variable. Tumor gene expression profiling revealed an upregulation of inflammatory responses, T-cell proliferation, and NK cell function on treatment with some inflammatory cytokines upregulated in peripheral blood. However, target engagement, evidenced by pharmacologic activity in peripheral blood and tumor tissue, did not correlate with clinical efficacy. The low number of responses precluded identifying a robust biomarker signature predictive of response. CONCLUSIONS GSK3174998±pembrolizumab was well tolerated over the dose range tested and demonstrated target engagement. Limited clinical activity does not support further development of GSK3174998±pembrolizumab in advanced cancers. TRIAL REGISTRATION NUMBER NCT02528357.
Collapse
Affiliation(s)
- Sophie Postel-Vinay
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Vincent K Lam
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Willeke Ros
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Todd M Bauer
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Aaron R Hansen
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Daniel C Cho
- New York Medical College, Valhalla, New York, USA
| | - F Stephen Hodi
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jan H M Schellens
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Jennifer K Litton
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sandrine Aspeslagh
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Karen A Autio
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Frans L Opdam
- Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | - Neeta Somaiah
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Mehmet Altan
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anna Spreafico
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Osama Rahma
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Elaine M Paul
- GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | | | - Helen Zhou
- GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | | | | | | | | | | | | | - Axel Hoos
- GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Aurelien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Jeffrey S Weber
- Perlmutter Cancer Center, NYU Langone Health, New York, New York, USA
| | - John V Heymach
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
29
|
Abstract
Cancer immunotherapy with immune-checkpoint blockade has improved the outcomes of patients with various malignancies, yet a majority do not benefit or develop resistance. To address this unmet need, efforts across the field are targeting additional coinhibitory receptors, costimulatory proteins, and intracellular mediators that could prevent or bypass anti-PD1 resistance mechanisms. The CD28 costimulatory pathway is necessary for antigen-specific T cell activation, though prior CD28 agonists did not translate successfully to clinic due to toxicity. Casitas B lymphoma-b (Cbl-b) is a downstream, master regulator of both CD28 and CTLA-4 signaling. This E3 ubiquitin ligase regulates both innate and adaptive immune cells, ultimately promoting an immunosuppressive tumor microenvironment (TME) in the absence of CD28 costimulation. Recent advances in pharmaceutical screening and computational biology have enabled the development of novel platforms to target this once 'undruggable' protein. These platforms include DNA encoded library screening, allosteric drug targeting, small-interfering RNA inhibition, CRISPR genome editing, and adoptive cell therapy. Both genetic knock-out models and Cbl-b inhibitors have been shown to reverse immunosuppression in the TME, stimulate cytotoxic T cell activity, and promote tumor regression, findings augmented with PD1 blockade in experimental models. In translating Cbl-b inhibitors to clinic, we propose specific gene expression profiles that may identify patient populations most likely to benefit. Overall, novel Cbl-b inhibitors provide antigen-specific immune stimulation and are a promising therapeutic tool in the field of immuno-oncology.
Collapse
Affiliation(s)
- Ryan C Augustin
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Riyue Bao
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason J Luke
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
30
|
Zhang Y, Yao Q, Pan Y, Fang X, Xu H, Zhao T, Zhu G, Jiang T, Li S, Cao H. Efficacy and Safety of PD-1/PD-L1 Checkpoint Inhibitors versus Anti-PD-1/PD-L1 Combined with Other Therapies for Tumors: A Systematic Review. Cancers (Basel) 2023; 15:682. [PMID: 36765640 PMCID: PMC9913120 DOI: 10.3390/cancers15030682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/29/2022] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE In recent years, the anti-programmed cell death protein-1 and its ligand (PD-1/PD-L1) or combination therapies have been recommended as an alternative emerging choice of treatment for oncology patients. However, the efficacy and adverse events of different combination strategies for the treatment of tumors remain controversial. METHODS PubMed, Embase, Cochrane Library, the American Society of Clinical Oncology (ASCO), and the European Society of Medicine Oncology (ESMO) were searched from database inception until 16 February 2022. The endpoints of objective response rate (ORR), disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and adverse events (AEs) were analyzed from different treatment schemes and tumor types. The protocol was registered in PROSPERO (CRD42022328927). RESULTS This meta-analysis included forty-eight eligible studies. Combination therapy has improved ORR (RR = 1.40, p < 0.001), DCR (RR = 1.22, p < 0.001), and PFS (the median survival ratio (MSR) was estimated to be 1.475 p < 0.001) compared to anti-PD-1/PD-L1 but had no significant benefit on OS (MSR was estimated to be 1.086 p = 0.117). Besides, combination treatment strategies are more toxic in any grade AEs (RR = 1.13, p < 0.001) and grade 3-5 AEs (RR = 1.81, p < 0.001). CONCLUSIONS Treatment with PD-1/PD-L1 inhibitors in combination with other antitumor therapies improve patients' ORR, DCR, and PFS compared to anti-PD-1/PD-L1. However, it is regrettable that there is no benefit to OS and an increased risk of AEs in combinatorial therapies.
Collapse
Affiliation(s)
- Yiru Zhang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, 739 Dingshen Rd., Zhoushan 316021, China
| | - Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Yong Pan
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, 739 Dingshen Rd., Zhoushan 316021, China
| | - Xinru Fang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Haoying Xu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Tingxiao Zhao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, 739 Dingshen Rd., Zhoushan 316021, China
| | - Guangqi Zhu
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, 739 Dingshen Rd., Zhoushan 316021, China
| | - Tianan Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, 79 Qingchun Rd., Hangzhou 310003, China
| | - Shibo Li
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, 739 Dingshen Rd., Zhoushan 316021, China
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-Chemical Injury Diseases of Zhejiang Province, 79 Qingchun Rd., Hangzhou 310003, China
| |
Collapse
|
31
|
Li Q, Lu J, Li J, Zhang B, Wu Y, Ying T. Antibody-based cancer immunotherapy by targeting regulatory T cells. Front Oncol 2023; 13:1157345. [PMID: 37182149 PMCID: PMC10174253 DOI: 10.3389/fonc.2023.1157345] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023] Open
Abstract
Regulatory T cells (Tregs) are among the most abundant suppressive cells, which infiltrate and accumulate in the tumor microenvironment, leading to tumor escape by inducing anergy and immunosuppression. Their presence has been correlated with tumor progression, invasiveness and metastasis. Targeting tumor-associated Tregs is an effective addition to current immunotherapy approaches, but it may also trigger autoimmune diseases. The major limitation of current therapies targeting Tregs in the tumor microenvironment is the lack of selective targets. Tumor-infiltrating Tregs express high levels of cell surface molecules associated with T-cell activation, such as CTLA4, PD-1, LAG3, TIGIT, ICOS, and TNF receptor superfamily members including 4-1BB, OX40, and GITR. Targeting these molecules often attribute to concurrent depletion of antitumor effector T-cell populations. Therefore, novel approaches need to improve the specificity of targeting Tregs in the tumor microenvironment without affecting peripheral Tregs and effector T cells. In this review, we discuss the immunosuppressive mechanisms of tumor-infiltrating Tregs and the status of antibody-based immunotherapies targeting Tregs.
Collapse
Affiliation(s)
- Quanxiao Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Baohong Zhang
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yanling Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Tianlei Ying, ; Yanling Wu,
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai Engineering Research Center for Synthetic Immunology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Tianlei Ying, ; Yanling Wu,
| |
Collapse
|
32
|
Jhajj HS, Lwo TS, Yao EL, Tessier PM. Unlocking the potential of agonist antibodies for treating cancer using antibody engineering. Trends Mol Med 2023; 29:48-60. [PMID: 36344331 PMCID: PMC9742327 DOI: 10.1016/j.molmed.2022.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/22/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022]
Abstract
Agonist antibodies that target immune checkpoints, such as those in the tumor necrosis factor receptor (TNFR) superfamily, are an important class of emerging therapeutics due to their ability to regulate immune cell activity, especially for treating cancer. Despite their potential, to date, they have shown limited clinical utility and further antibody optimization is urgently needed to improve their therapeutic potential. Here, we discuss key antibody engineering approaches for improving the activity of antibody agonists by optimizing their valency, specificity for different receptors (e.g., bispecific antibodies) and epitopes (e.g., biepitopic or biparatopic antibodies), and Fc affinity for Fcγ receptors (FcγRs). These powerful approaches are being used to develop the next generation of cancer immunotherapeutics with improved efficacy and safety.
Collapse
Affiliation(s)
- Harkamal S Jhajj
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Timon S Lwo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily L Yao
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter M Tessier
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
33
|
Martin C, Enrico D. Current and novel therapeutic strategies for optimizing immunotherapy outcomes in advanced non-small cell lung cancer. Front Oncol 2022; 12:962947. [PMID: 36568253 PMCID: PMC9772042 DOI: 10.3389/fonc.2022.962947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
During the past decade, immunotherapy has dramatically improved the outcomes of patients with non-small cell lung cancer (NSCLC). The development of specific antibodies against the programmed death (PD1) receptor and its ligand PD-L1 (programmed death ligand-1) has demonstrated substantial efficacy in advanced NSCLC either in the first or in the second line. However, the success of immune checkpoint inhibitors (ICIs) as monotherapy did not reach all patients and long-term responders still represent a small subset of cases. Under these circumstances, different strategies have been and are being tested to optimize clinical outcomes. Here, we reviewed the current evidence and the more promising perspectives of ICI combination approaches, such as the addition of chemotherapy, antiangiogenic agents, other co-inhibitory or co-stimulatory checkpoints, and targeted therapies.
Collapse
Affiliation(s)
- Claudio Martin
- Department of Medical Oncology, Thoracic Oncology Section, Alexander Fleming Cancer Institute, Buenos Aires, Argentina
- Department of Clinical Research, Alexander Fleming Cancer Institute, Buenos Aires, Argentina
| | - Diego Enrico
- Department of Medical Oncology, Thoracic Oncology Section, Alexander Fleming Cancer Institute, Buenos Aires, Argentina
- Department of Clinical Research, Alexander Fleming Cancer Institute, Buenos Aires, Argentina
| |
Collapse
|
34
|
Davis EJ, Martin-Liberal J, Kristeleit R, Cho DC, Blagden SP, Berthold D, Cardin DB, Vieito M, Miller RE, Hari Dass P, Orcurto A, Spencer K, Janik JE, Clark J, Condamine T, Pulini J, Chen X, Mehnert JM. First-in-human phase I/II, open-label study of the anti-OX40 agonist INCAGN01949 in patients with advanced solid tumors. J Immunother Cancer 2022; 10:jitc-2021-004235. [PMID: 36316061 PMCID: PMC9628691 DOI: 10.1136/jitc-2021-004235] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND OX40 is a costimulatory receptor upregulated on antigen-activated T cells and constitutively expressed on regulatory T cells (Tregs). INCAGN01949, a fully human immunoglobulin G1κ anti-OX40 agonist monoclonal antibody, was designed to promote tumor-specific immunity by effector T-cell activation and Fcγ receptor-mediated Treg depletion. This first-in-human study was conducted to determine the safety, tolerability, and preliminary efficacy of INCAGN01949. METHODS Phase I/II, open-label, non-randomized, dose-escalation and dose-expansion study conducted in patients with advanced or metastatic solid tumors. Patients received INCAGN01949 monotherapy (7-1400 mg) in 14-day cycles while deriving benefit. Safety measures, clinical activity, pharmacokinetics, and pharmacodynamic effects were assessed and summarized with descriptive statistics. RESULTS Eighty-seven patients were enrolled; most common tumor types were colorectal (17.2%), ovarian (8.0%), and non-small cell lung (6.9%) cancers. Patients received a median three (range 1-9) prior therapies, including immunotherapy in 24 patients (27.6%). Maximum tolerated dose was not reached; one patient (1.1%) receiving 350 mg dose reported dose-limiting toxicity of grade 3 colitis. Treatment-related adverse events were reported in 45 patients (51.7%), with fatigue (16 (18.4%)), rash (6 (6.9%)), and diarrhea (6 (6.9%)) being most frequent. One patient (1.1%) with metastatic gallbladder cancer achieved a partial response (duration of 6.3 months), and 23 patients (26.4%) achieved stable disease (lasting >6 months in one patient). OX40 receptor occupancy was maintained over 90% among all patients receiving doses of ≥200 mg, while no treatment-emergent antidrug antibodies were detected across all dose levels. Pharmacodynamic results demonstrated that treatment with INCAGN01949 did not enhance proliferation or activation of T cells in peripheral blood or reduce circulating Tregs, and analyses of tumor biopsies did not demonstrate any consistent increase in effector T-cell infiltration or function, or decrease in infiltrating Tregs. CONCLUSION No safety concerns were observed with INCAGN01949 monotherapy in patients with metastatic or advanced solid tumors. However, tumor responses and pharmacodynamic effects on T cells in peripheral blood and post-therapy tumor biopsies were limited. Studies evaluating INCAGN01949 in combination with other therapies are needed to further evaluate the potential of OX40 agonism as a therapeutic approach in patients with advanced solid tumors. TRIAL REGISTRATION NUMBER NCT02923349.
Collapse
Affiliation(s)
| | | | | | - Daniel C Cho
- Perlmutter Cancer Center, NYU Langone Health, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Dominik Berthold
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Dana B Cardin
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maria Vieito
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Prashanth Hari Dass
- Early Phase Clinical Trials Unit, Churchill Hospital, University of Oxford, Oxford, UK
| | - Angela Orcurto
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | | | | | - Jason Clark
- Incyte Corporation, Wilmington, Delaware, USA
| | | | | | - Xuejun Chen
- Incyte Corporation, Wilmington, Delaware, USA
| | | |
Collapse
|
35
|
Chen YW, Rini BI, Beckermann KE. Emerging Targets in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2022; 14:4843. [PMID: 36230766 PMCID: PMC9561986 DOI: 10.3390/cancers14194843] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
The dual immune checkpoint blockade targeting CTLA-4 and PD-1 (ipilimumab/nivolumab) or the IO combinations targeting PD-1 and anti-VEGF TKIs (pembrolizumab/axitinib, nivolumab/cabozantinib, pembrolizumab/lenvatinib) have demonstrated an overall survival benefit in advanced clear cell renal cell carcinoma (ccRCC). Despite this significant improvement in clinical outcomes in the frontline setting from IO/IO or the IO/TKI combinations, there is a subset of patients of advanced ccRCC that do not respond to such combinations or will lose the initial efficacy and have disease progression. Therefore, a remarkable unmet need exists to develop new therapeutics to improve outcomes. With an enhanced understanding of ccRCC biology and its interaction with the tumor microenvironment, several new therapies are under development targeting ccRCC metabolism, cytokine-signaling, alternative immune checkpoint proteins, and novel biological pathways. In addition, microbiome products enhancing IO response, antibody-drug conjugates, and targeted radionuclides are also being investigated. This review summarizes selected emerging agents that are under development in ccRCC.
Collapse
Affiliation(s)
- Yu-Wei Chen
- Division of Hematology Oncology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Ave, 777 Preston Research Building, Nashville, TN 37232, USA
| | - Brian I. Rini
- Division of Hematology Oncology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Ave, 777 Preston Research Building, Nashville, TN 37232, USA
| | - Kathryn E. Beckermann
- Division of Hematology Oncology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, USA
- Vanderbilt-Ingram Cancer Center, 2220 Pierce Ave, 777 Preston Research Building, Nashville, TN 37232, USA
| |
Collapse
|
36
|
Zhang Q, Rui W, Jiang Y, Yuan F, Chen Y, Guo X, Zhou Y, Wu Z, Wang C, Ding X. Tumor-infiltrating OX40 + lymphocytes is an independent positive prognostic factor for patients with pancreatic ductal adenocarcinoma. Clin Transl Oncol 2022; 24:2029-2038. [PMID: 35731350 DOI: 10.1007/s12094-022-02864-1] [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: 12/02/2021] [Accepted: 05/19/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OX40 signaling pathway occupies a vital place in anti-tumor immunity; however, the role of tumor-infiltrating OX40+ lymphocytes in pancreatic ductal adenocarcinoma (PDAC) remains to be identified. METHODS A total of 325 sequential PDAC patients who received curative tumor resection between January 2014 and December 2016 were enrolled. Tissues of these patients were immunohistochemically assessed for tumor infiltration of CD4+ T cells, CD8+ cytotoxic T cells (CTLs), and OX40+ lymphocytes. The frequency of OX40+ tumor-infiltrating lymphocytes (TILs) was then analyzed to various clinicopathological features, densities of tumor infiltration of CD4+ T cells and CTLs, and survival analysis was conducted using Kaplan-Meier (KM) curves. The risk scores of associated markers were calculated by the Cox proportional-hazards model. RESULTS Our results showed that higher OX40+ lymphocytes infiltration was significantly correlated with superior median overall survival (OS) (25.8 vs 13.4 months, P < 0.001). Additionally, using univariate and multivariate Cox proportional hazards analyses, this study revealed that together with tumor differentiation, tumor size, serum CA199 levels, serum CA125 levels, and the infiltration of intratumoral CD8+ T cells. The abundance of OX40+ lymphocytes within the tumor was continued to be an independent predictor for OS (P = 0.023, HR = 0.713, 95% CI: 0.532-0.954). CONCLUSIONS This study demonstrated that intratumoral infiltration by a high number of OX40+ lymphocytes is a novel biomarker for favorable prognosis in resected PDAC patients, which implies that OX40-agonist-based immunotherapy might be a potential target in PDAC patients.
Collapse
Affiliation(s)
- Qiwei Zhang
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Weiwei Rui
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Yongsheng Jiang
- Department of General Surgery, Pancreatic Disease Center, Research Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoxia Guo
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Yu Zhou
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China
| | - Zhiyuan Wu
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China.
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China.
| | - Xiaoyi Ding
- Department of Interventional Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No. 197, Ruijin Er Road, Shanghai, 200025, China.
| |
Collapse
|
37
|
Ruiz D, Haynes C, Marable J, Pundkar C, Nance RL, Bedi D, Agarwal P, Suryawanshi AS, Mishra A, Smith BF, Sandey M. Development of OX40 agonists for canine cancer immunotherapy. iScience 2022; 25:105158. [PMID: 36217551 PMCID: PMC9547195 DOI: 10.1016/j.isci.2022.105158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/19/2022] [Accepted: 09/15/2022] [Indexed: 01/13/2023] Open
Abstract
Recent breakthroughs in cancer immunotherapy have provided unprecedented clinical benefits to human cancer patients. Cancer is also one of the most common causes of death in pet dogs. Thus, canine-specific immune therapies targeting similar signaling pathways can provide better treatment options for canine cancer patients. Here, we describe the development and characterization of two canine-specific anti-OX40 agonists to activate OX40 signaling. We show that canine OX40, like human OX40, is not expressed on resting T cells, and its expression is markedly increased on canine CD4 T cells and Tregs after stimulation with concanavalin A (Con-A). cOX40 is also expressed on tumor-infiltrating lymphocytes (TILs) in canine osteosarcoma patients. The canine-specific OX40 agonists strongly activates cPBMCs by increasing IFN-γ expression and do not require Fc receptor-mediated cross-linking for OX40 agonism. Together, these results suggest that cFcOX40L proteins are potent OX40 agonists and have the potential to enhance antitumor immunity in canine cancer patients.
Collapse
Affiliation(s)
- Damien Ruiz
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Chloe Haynes
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Jonathan Marable
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Chetan Pundkar
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Rebecca L. Nance
- Scott Ritchy Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Deepa Bedi
- Biomedical Sciences, Tuskegee University, Tuskegee, AL, USA
| | - Payal Agarwal
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
- Scott Ritchy Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Amol S. Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Amarjit Mishra
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Bruce F. Smith
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
- Scott Ritchy Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Maninder Sandey
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
- Corresponding author
| |
Collapse
|
38
|
Mussafi O, Mei J, Mao W, Wan Y. Immune checkpoint inhibitors for PD-1/PD-L1 axis in combination with other immunotherapies and targeted therapies for non-small cell lung cancer. Front Oncol 2022; 12:948405. [PMID: 36059606 PMCID: PMC9430651 DOI: 10.3389/fonc.2022.948405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/22/2022] [Indexed: 12/13/2022] Open
Abstract
It has been widely acknowledged that the use of immune checkpoint inhibitors (ICI) is an effective therapeutic treatment in many late-stage cancers. However, not all patients could benefit from ICI therapy. Several biomarkers, such as high expression of PD-L1, high mutational burden, and higher number of tumor infiltration lymphocytes have shown to predict clinical benefit from immune checkpoint therapies. One approach using ICI in combination with other immunotherapies and targeted therapies is now being investigated to enhance the efficacy of ICI alone. In this review, we summarized the use of other promising immunotherapies and targeted therapies in combination with ICI in treatment of lung cancers. The results from multiple animals and clinical trials were reviewed. We also briefly discussed the possible outlooks for future treatment.
Collapse
Affiliation(s)
- Ofek Mussafi
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY, United States
| | - Jie Mei
- Department of Oncology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Wenjun Mao
- Department of Cardiothoracic Surgery, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
39
|
Kim TW, Burris HA, de Miguel Luken MJ, Pishvaian MJ, Bang YJ, Gordon M, Awada A, Camidge DR, Hodi FS, McArthur GA, Miller WH, Cervantes A, Chow LQ, Lesokhin AM, Rutten A, Sznol M, Rishipathak D, Chen SC, Stefanich E, Pourmohamad T, Anderson M, Kim J, Huseni M, Rhee I, Siu LL. First-In-Human Phase I Study of the OX40 Agonist MOXR0916 in Patients with Advanced Solid Tumors. Clin Cancer Res 2022; 28:3452-3463. [PMID: 35699599 PMCID: PMC9662912 DOI: 10.1158/1078-0432.ccr-21-4020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/27/2022] [Accepted: 06/10/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE OX40, a receptor transiently expressed by T cells upon antigen recognition, is associated with costimulation of effector T cells and impairment of regulatory T-cell function. This first-in-human study evaluated MOXR0916, a humanized effector-competent agonist IgG1 monoclonal anti-OX40 antibody. PATIENTS AND METHODS Eligible patients with locally advanced or metastatic refractory solid tumors were treated with MOXR0916 intravenously once every 3 weeks (Q3W). A 3+3 dose-escalation stage (0.2-1,200 mg; n = 34) was followed by expansion cohorts at 300 mg (n = 138) for patients with melanoma, renal cell carcinoma, non-small cell lung carcinoma, urothelial carcinoma, and triple-negative breast cancer. RESULTS MOXR0916 was well tolerated with no dose-limiting toxicities observed. An MTD was not reached. Most patients (95%) experienced at least one adverse event (AE); 56% of AEs, mostly grade 1-2, were related to MOXR0916. Most common treatment-related AEs included fatigue (17%), diarrhea (8%), myalgia (7%), nausea (6%), decreased appetite (6%), and infusion-related reaction (5%). Pharmacokinetic (PK) parameters were dose proportional between 80 and 1,200 mg and supported Q3W administration. The recommended expansion dose based on PK and OX40 receptor saturation was 300 mg Q3W. Immune activation and upregulation of PD-L1 was observed in a subset of paired tumor biopsies. One renal cell carcinoma patient experienced a confirmed partial response. Overall, 33% of patients achieved stable disease. CONCLUSIONS Although objective responses were rarely observed with MOXR0916 monotherapy, the favorable safety profile and evidence of tumor immune activation in a subset of patients support further investigation in combination with complementary agents such as PD-1/PD-L1 antagonists.
Collapse
Affiliation(s)
- Tae Won Kim
- Asan Medical Center, University of Ulsan, Seoul, Korea
| | | | | | | | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, Korea
| | | | | | - D. Ross Camidge
- University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Grant A. McArthur
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Wilson H. Miller
- Jewish General Hospital and Segal Cancer Centre, McGill University, Montréal, Canada
| | - Andres Cervantes
- Biomedical Research Institute INCLIVA, University of Valencia, Valencia Spain
| | | | - Alexander M. Lesokhin
- Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Medical College, New York, New York
| | - Annemie Rutten
- GasthuisZusters Antwerpen Sint-Augustinus, Antwerp, Belgium
| | - Mario Sznol
- Yale School of Medicine, New Haven, Connecticut
| | | | | | | | | | | | - Jeong Kim
- Genentech, Inc., South San Francisco, California
| | | | - Ina Rhee
- Genentech, Inc., South San Francisco, California
| | - Lillian L. Siu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Chocarro L, Bocanegra A, Blanco E, Fernández-Rubio L, Arasanz H, Echaide M, Garnica M, Ramos P, Piñeiro-Hermida S, Vera R, Escors D, Kochan G. Cutting-Edge: Preclinical and Clinical Development of the First Approved Lag-3 Inhibitor. Cells 2022; 11:2351. [PMID: 35954196 PMCID: PMC9367598 DOI: 10.3390/cells11152351] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized medical practice in oncology since the FDA approval of the first ICI 11 years ago. In light of this, Lymphocyte-Activation Gene 3 (LAG-3) is one of the most important next-generation immune checkpoint molecules, playing a similar role as Programmed cell Death protein 1 (PD-1) and Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4). 19 LAG-3 targeting molecules are being evaluated at 108 clinical trials which are demonstrating positive results, including promising bispecific molecules targeting LAG-3 simultaneously with other ICIs. Recently, a new dual anti-PD-1 (Nivolumab) and anti-LAG-3 (Relatimab) treatment developed by Bristol Myers Squibb (Opdualag), was approved by the Food and Drug Administration (FDA) as the first LAG-3 blocking antibody combination for unresectable or metastatic melanoma. This novel immunotherapy combination more than doubled median progression-free survival (PFS) when compared to nivolumab monotherapy (10.1 months versus 4.6 months). Here, we analyze the large clinical trial responsible for this historical approval (RELATIVITY-047), and discuss the preclinical and clinical developments that led to its jump into clinical practice. We will also summarize results achieved by other LAG-3 targeting molecules with promising anti-tumor activities currently under clinical development in phases I, I/II, II, and III. Opdualag will boost the entry of more LAG-3 targeting molecules into clinical practice, supporting the accumulating evidence highlighting the pivotal role of LAG-3 in cancer.
Collapse
Affiliation(s)
- Luisa Chocarro
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Ana Bocanegra
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Ester Blanco
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), 31001 Pamplona, Spain
| | - Leticia Fernández-Rubio
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Hugo Arasanz
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
- Medical Oncology Unit, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain;
| | - Miriam Echaide
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Maider Garnica
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Pablo Ramos
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Sergio Piñeiro-Hermida
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Ruth Vera
- Medical Oncology Unit, Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain;
| | - David Escors
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| | - Grazyna Kochan
- Oncoimmunology Research Unit, Navarrabiomed-Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), Hospital Universitario de Navarra (HUN), Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31001 Pamplona, Spain; (E.B.); (L.F.-R.); (H.A.); (M.E.); (M.G.); (P.R.); (S.P.-H.); (D.E.); (G.K.)
| |
Collapse
|
41
|
Rittig SM, Lutz MS, Clar KL, Zhou Y, Kropp KN, Koch A, Hartkopf AD, Hinterleitner M, Zender L, Salih HR, Maurer S, Hinterleitner C. Controversial Role of the Immune Checkpoint OX40L Expression on Platelets in Breast Cancer Progression. Front Oncol 2022; 12:917834. [PMID: 35875148 PMCID: PMC9304936 DOI: 10.3389/fonc.2022.917834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/08/2022] [Indexed: 11/22/2022] Open
Abstract
In conventional T cells, OX40 has been identified as a major costimulating receptor augmenting survival and clonal expansion of effector and memory T cell populations. In regulatory T cells, (Treg) OX40 signaling suppresses cellular activity and differentiation. However, clinical trials investigating OX40 agonists to enhance anti-tumor immunity, showed only limited success so far. Here we show that platelets from breast cancer patients express relevant levels of OX40L and platelet OX40L (pOX40L) inversely correlates with platelet-expressed immune checkpoint molecules GITRL (pGITRL) and TACI (pTACI). While high expression of pOX40L correlates with T and NK cell activation, elevated pOX40L levels identify patients with higher tumor grades, the occurrence of metastases, and shorter recurrence-free survival (RFS). Of note, OX40 mRNA levels in breast cancer correlate with enhanced expression of anti-apoptotic, immune-suppressive, and tumor-promoting mRNA gene signatures. Our data suggest that OX40L on platelets might play counteracting roles in cancer and anti-tumor immunity. Since pOX40L reflects disease relapse better than the routinely used predictive markers CA15-3, CEA, and LDH, it could serve as a novel biomarker for refractory disease in breast cancer.
Collapse
Affiliation(s)
- Susanne M. Rittig
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitaetsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin, Germany
| | - Martina S. Lutz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Kim L. Clar
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Yanjun Zhou
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Korbinian N. Kropp
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of Mainz, Mainz, Germany
| | - André Koch
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, Tuebingen, Germany
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Gynecology and Obstetrics, University Hospital of Ulm, Ulm, Germany
| | - Martina Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Stefanie Maurer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Precision Immunology Institute, Department of Oncological Sciences, and The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Stefanie Maurer,
| | - Clemens Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
42
|
Lao J, Cao C, Niu X, Deng S, Ming S, Liang S, Shang Y, Yuan Y, Shi X, Liang Z, Wu M, Wu Y. OX40 enhances T cell immune response to PD-1 blockade therapy in non-small cell lung cancer. Int Immunopharmacol 2022; 108:108813. [DOI: 10.1016/j.intimp.2022.108813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/14/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022]
|
43
|
Goldman JW, Piha-Paul SA, Curti B, Pedersen KS, Bauer TM, Groenland SL, Carvajal RD, Chhaya V, Kirby G, McGlinchey K, Hammond SA, Streicher KL, Townsley D, Chae YK, Voortman J, Marabelle A, Powderly J. Safety and tolerability of MEDI0562, an OX40 agonist monoclonal antibody, in combination with durvalumab or tremelimumab in adult patients with advanced solid tumors. Clin Cancer Res 2022; 28:3709-3719. [PMID: 35699623 DOI: 10.1158/1078-0432.ccr-21-3016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/03/2021] [Accepted: 06/10/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Combination therapies targeting immunological checkpoints have shown promise in treating multiple tumor types. We report safety and tolerability of MEDI0562, a humanized IgG1K OX40 monoclonal antibody, in combination with durvalumab (anti-PD-L1), or tremelimumab (anti-CTLA-4), in adult patients with previously treated advanced solid tumors. EXPERIMENTAL DESIGN In this phase 1, multicenter, open-label study, patients received escalating doses of MEDI0562 (2.25, 7.5, or 22.5 mg) every two weeks (Q2W) in combination with durvalumab (1500 mg) or tremelimumab (75 or 225 mg) Q4W, intravenously, until unacceptable toxicity or progressive disease. Tumor assessments were performed Q8W. The primary objective was to evaluate safety and tolerability. RESULTS Among the 27 and 31 patients who received MEDI0562 + durvalumab or MEDI0562 + tremelimumab, 74.1% and 67.7% reported a treatment-related adverse event (AE), and 22.2% and 19.4% experienced a treatment‑emergent AE that led to discontinuation, respectively. The maximum tolerated dose of MEDI0562 + durvalumab was 7.5 mg MEDI0562 + 1500 mg durvalumab; the maximum administered dose of MEDI0562 + tremelimumab was 22.5 mg MEDI0562 + 225 mg tremelimumab. Three patients in the MEDI0562 + durvalumab arm had a partial response. The mean percentage of Ki67+CD4+ and Ki67+CD8+ memory T cells increased by >100% following the first dose of MEDI0562 + durvalumab or tremelimumab in all dose cohorts. A decrease in OX40+FOXP3 T regulatory cells was observed in a subset of patients with available paired biopsies. CONCLUSIONS Following dose escalation, moderate toxicity was observed in both treatment arms, with no clear efficacy signals demonstrated.
Collapse
Affiliation(s)
| | - Sarina A Piha-Paul
- The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brendan Curti
- Providence Cancer Center and Earle A. Chiles Research Institute, Portland, OR, United States
| | | | - Todd M Bauer
- Sarah Cannon Research Institute / Tennessee Oncology, PLLC., Nashville, TN, United States
| | | | | | - Vaishali Chhaya
- AstraZeneca (United States), Gaithersburg, MD, United States
| | - Gray Kirby
- AstraZeneca (United States), Gaithersburg, MD, United States
| | - Kelly McGlinchey
- AstraZeneca (United Kingdom), Gaithersburg, Maryland, United States
| | | | | | | | - Young Kwang Chae
- Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jens Voortman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - John Powderly
- Carolina BioOncology Institute, Huntersville, NC, United States
| |
Collapse
|
44
|
Sanmamed MF, Berraondo P, Rodriguez-Ruiz ME, Melero I. Charting roadmaps towards novel and safe synergistic immunotherapy combinations. NATURE CANCER 2022; 3:665-680. [PMID: 35764745 DOI: 10.1038/s43018-022-00401-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
Checkpoint inhibitor-based cancer immunotherapy is often combined in the clinic with other immunotherapy strategies, targeted therapies, chemotherapy or standard-of-care treatments to achieve superior therapeutic efficacy. The large number of immunotherapy combinations that are currently undergoing clinical testing necessitate the establishment of faithful criteria to prioritize optimal combinations with evidence of synergy, to determine their safety and optimal sequence of administration and to identify biomarkers of therapy resistance and response. In this review, we focus on recent developments in immunotherapy combinations and reflect on how combinations should be optimized to maximize the impact of immunotherapy in clinical oncology.
Collapse
Affiliation(s)
- Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Maria E Rodriguez-Ruiz
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Departments of Oncology and Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
| |
Collapse
|
45
|
Yadav R, Redmond WL. Current Clinical Trial Landscape of OX40 Agonists. Curr Oncol Rep 2022; 24:951-960. [PMID: 35352295 DOI: 10.1007/s11912-022-01265-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Despite the efficacy of immune checkpoint blockade (ICB) immunotherapy, most cancer patients still develop progressive disease necessitating additional treatment options. One approach is ligation of the OX40 (CD134) costimulatory receptor which promotes T cell activation, effector function, and the generation of long-lived memory cells. RECENT FINDINGS Numerous preclinical studies have demonstrated that OX40 agonists alone or in combination with ICB (e.g., anti-PD-1, anti-PD-L1, and anti-CTLA-4) augment anti-tumor immunity. In this review, we discuss the impact of OX40 agonists on T cell function and the therapeutic potential of OX40 agonists alone or in conjunction with ICB for patients with advanced malignancies.
Collapse
Affiliation(s)
- Rashi Yadav
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, 4805 NE Glisan St., 2N35, Portland, OR, 97213, USA.
| |
Collapse
|
46
|
Lee J, Kim D, Min B. Tissue Resident Foxp3+ Regulatory T Cells: Sentinels and Saboteurs in Health and Disease. Front Immunol 2022; 13:865593. [PMID: 35359918 PMCID: PMC8963273 DOI: 10.3389/fimmu.2022.865593] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/22/2022] [Indexed: 01/04/2023] Open
Abstract
Foxp3+ regulatory T (Treg) cells are a CD4 T cell subset with unique immune regulatory function that are indispensable in immunity and tolerance. Their indisputable importance has been investigated in numerous disease settings and experimental models. Despite the extensive efforts in determining the cellular and molecular mechanisms operating their functions, our understanding their biology especially in vivo remains limited. There is emerging evidence that Treg cells resident in the non-lymphoid tissues play a central role in regulating tissue homeostasis, inflammation, and repair. Furthermore, tissue-specific properties of those Treg cells that allow them to express tissue specific functions have been explored. In this review, we will discuss the potential mechanisms and key cellular/molecular factors responsible for the homeostasis and functions of tissue resident Treg cells under steady-state and inflammatory conditions.
Collapse
Affiliation(s)
- Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Dongkyun Kim
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Booki Min
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- *Correspondence: Booki Min,
| |
Collapse
|
47
|
Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat Rev Clin Oncol 2022; 19:37-50. [PMID: 34580473 DOI: 10.1038/s41571-021-00552-7] [Citation(s) in RCA: 458] [Impact Index Per Article: 152.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 02/08/2023]
Abstract
The discovery and clinical implementation of immune-checkpoint inhibitors (ICIs) targeting CTLA4, PD-1 and PD-L1 has revolutionized the treatment of cancer, as recognized by the 2018 Nobel Prize for Medicine and Physiology. This groundbreaking new approach has improved the outcomes of patients with various forms of advanced-stage cancer; however, the majority of patients receiving these therapies, even in combination, do not derive clinical benefit. Further development of agents targeting additional immune checkpoints, co-stimulatory receptors and/or co-inhibitory receptors that control T cell function is therefore critical. In this Review, we discuss the translational potential and clinical development of agents targeting both co-stimulatory and co-inhibitory T cell receptors. Specifically, we describe their mechanisms of action, and provide an overview of ongoing clinical trials involving novel ICIs including those targeting LAG3, TIM3, TIGIT and BTLA as well as agonists of the co-stimulatory receptors GITR, OX40, 41BB and ICOS. We also discuss several additional approaches, such as harnessing T cell metabolism, in particular via adenosine signalling, inhibition of IDO1, and targeting changes in glucose and fatty acid metabolism. We conclude that further efforts are needed to optimize the timing of combination ICI approaches and, most importantly, to individualize immunotherapy based on both patient-specific and tumour-specific characteristics.
Collapse
|
48
|
Corke L, Sacher A. New Strategies and Combinations to Improve Outcomes in Immunotherapy in Metastatic Non-Small-Cell Lung Cancer. Curr Oncol 2021; 29:38-55. [PMID: 35049678 PMCID: PMC8774728 DOI: 10.3390/curroncol29010004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors have transformed the treatment of metastatic non-small-cell lung cancer, yielding marked improvements in survival and the potential for durable clinical responses. Primary and acquired resistance to current immune checkpoint inhibitors constitute a key challenge despite the remarkable responses observed in a subset of patients. Multiple novel combination immunotherapy and adoptive cell therapy strategies are presently being developed to address treatment resistance. The success of these strategies hinges upon rational clinical trial design as well as careful consideration of the immunologic mechanisms within the variable tumor immune microenvironment (TIME) which underpin resistance to immunotherapy. Further research is needed to facilitate a deeper understanding of these complex mechanisms within the TIME, which may ultimately provide the key to restoring and enhancing an effective anti-tumor immune response. This review aims to provide an introduction to some of the recent and notable combination immunotherapy and cell therapy strategies used in advanced non-small-cell lung cancer (NSCLC), and the rationale for their use based on current understanding of the anti-tumor immune response and mechanisms of resistance within the TIME.
Collapse
Affiliation(s)
- Lucy Corke
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada;
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
49
|
Abstract
Melanoma is the most lethal skin cancer that originates from the malignant transformation of melanocytes. Although melanoma has long been regarded as a cancerous malignancy with few therapeutic options, increased biological understanding and unprecedented innovations in therapies targeting mutated driver genes and immune checkpoints have substantially improved the prognosis of patients. However, the low response rate and inevitable occurrence of resistance to currently available targeted therapies have posed the obstacle in the path of melanoma management to obtain further amelioration. Therefore, it is necessary to understand the mechanisms underlying melanoma pathogenesis more comprehensively, which might lead to more substantial progress in therapeutic approaches and expand clinical options for melanoma therapy. In this review, we firstly make a brief introduction to melanoma epidemiology, clinical subtypes, risk factors, and current therapies. Then, the signal pathways orchestrating melanoma pathogenesis, including genetic mutations, key transcriptional regulators, epigenetic dysregulations, metabolic reprogramming, crucial metastasis-related signals, tumor-promoting inflammatory pathways, and pro-angiogenic factors, have been systemically reviewed and discussed. Subsequently, we outline current progresses in therapies targeting mutated driver genes and immune checkpoints, as well as the mechanisms underlying the treatment resistance. Finally, the prospects and challenges in the development of melanoma therapy, especially immunotherapy and related ongoing clinical trials, are summarized and discussed.
Collapse
Affiliation(s)
- Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Huina Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, No. 127 of West Changle Road, 710032, Xi'an, Shaanxi, China.
| |
Collapse
|
50
|
Porciuncula A, Morgado M, Gupta R, Syrigos K, Meehan R, Zacharek SJ, Frederick JP, Schalper KA. Spatial Mapping and Immunomodulatory Role of the OX40/OX40L Pathway in Human Non-Small Cell Lung Cancer. Clin Cancer Res 2021; 27:6174-6183. [PMID: 34518312 DOI: 10.1158/1078-0432.ccr-21-0987] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/12/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE To evaluate the tissue distribution and clinical significance of OX40 and OX40L in human non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN Using multiplexed quantitative immunofluorescence, we conducted simultaneous and localized measurements of OX40 and OX40L proteins, major T-cell subsets, and conventional type 1 dendritic cells (cDC1) in 614 primary NSCLCs from three independent cohorts represented in tissue microarrays. We also measured OX40L protein in samples from a phase I clinical trial of intratumor administration of a lipid nanoparticle encapsulated mRNA encoding OX40L (mRNA-2416) in human solid tumors. Finally, we studied the OX40 pathway in 212 uterine/ovarian serous carcinomas. RESULTS OX40 protein was expressed in approximately 90% of NSCLCs, and OX40L was detected in approximately 10% of cases. Increased expression of OX40 was associated with higher CD4+ and CD8+ T lymphocytes, as well as cDC1s. Elevated expression of OX40L was consistently associated with increased CD4+ tumor-infiltrating lymphocytes and longer overall survival. No association was found between OX40 or OX40L levels and oncogenic driver mutations in EGFR and KRAS in lung adenocarcinomas. Delivering OX40L mRNA using intratumor mRNA-2416 injection mediated increased local OX40L protein levels that was most prominent in a patient with ovarian serous carcinoma. Detectable OX40L protein levels were observed in 15% of primary uterine/ovarian serous malignancies and associated with longer survival. CONCLUSIONS The OX40 pathway is expressed in a fraction of NSCLCs and is associated with a favorable immune contexture. Although OX40L is uncommonly expressed in NSCLC and serous malignancies, it is associated with better prognosis and can be introduced using exogenous mRNA.
Collapse
Affiliation(s)
- Angelo Porciuncula
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Micaela Morgado
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Richa Gupta
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Kostas Syrigos
- Oncology Unit, Department of Medicine, Athens University, Athens, Greece
| | | | | | | | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|