1
|
Naing A, Infante J, Goel S, Burris H, Black C, Marshall S, Achour I, Barbee S, May R, Morehouse C, Pollizzi K, Song X, Steele K, Elgeioushi N, Walcott F, Karakunnel J, LoRusso P, Weise A, Eder J, Curti B, Oberst M. Anti-PD-1 monoclonal antibody MEDI0680 in a phase I study of patients with advanced solid malignancies. J Immunother Cancer 2019; 7:225. [PMID: 31439037 PMCID: PMC6704567 DOI: 10.1186/s40425-019-0665-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/05/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The safety, efficacy, pharmacokinetics, and pharmacodynamics of the anti-programmed cell death-1 antibody MEDI0680 were evaluated in a phase I, multicenter, dose-escalation study in advanced solid malignancies. METHODS MEDI0680 was administered intravenously once every 2 weeks (Q2W) or once every 3 weeks at 0.1, 0.5, 2.5, 10 or 20 mg/kg. Two cohorts received 20 mg/kg once a week for 2 or 4 weeks, then 20 mg/kg Q2W. All were treated for 12 months or until progression. The primary endpoint was safety. Secondary endpoints were efficacy and pharmacokinetics. Exploratory endpoints included pharmacodynamics. RESULTS Fifty-eight patients were treated. Median age was 62.5 years and 81% were male. Most had kidney cancer (n = 36) or melanoma (n = 9). There were no dose-limiting toxicities. Treatment-related adverse events occurred in 83% and were grade ≥ 3 in 21%. Objective clinical responses occurred in 8/58 patients (14%): 5 with kidney cancer, including 1 with a complete response, and 3 with melanoma. The relationship between dose and serum levels was predictable and linear, with apparent receptor saturation at 10 mg/kg Q2W and all 20 mg/kg cohorts. CONCLUSIONS MEDI0680 induced peripheral T-cell proliferation and increased plasma IFNγ and associated chemokines regardless of clinical response. CD8+ T-cell tumor infiltration and tumoral gene expression of IFNG, CD8A, CXCL9, and granzyme K (GZMK) were also increased following MEDI0680 administration. TRIAL REGISTRATION NCT02013804 ; date of registration December 12, 2013.
Collapse
Affiliation(s)
- Aung Naing
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Jeffrey Infante
- Drug Development Unit, Sarah Cannon Research Institute, Tennessee Oncology, Nashville, TN, USA.,Present Address: Department of Oncology, Janssen, Raritan, NJ, USA
| | - Sanjay Goel
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Howard Burris
- Drug Development Unit, Sarah Cannon Research Institute, Tennessee Oncology, Nashville, TN, USA
| | - Chelsea Black
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA.,Present Address: PRA Health Sciences, Blue Bell, PA, USA
| | - Shannon Marshall
- Department of Research, Amplimmune Inc., Gaithersburg, MD, USA.,Present Address: Early Oncology Clinical, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ikbel Achour
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Susannah Barbee
- Department of Research, Amplimmune Inc., Gaithersburg, MD, USA.,Present Address: Department of Immuno-Oncology Research, FivePrime Therapeutics, Inc., South San Francisco, CA, USA
| | - Rena May
- Department of Research, Amplimmune Inc., Gaithersburg, MD, USA.,Present Address: Late-stage Development, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Chris Morehouse
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kristen Pollizzi
- Discovery Sciences, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Xuyang Song
- Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Keith Steele
- Translational Medicine, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Farzana Walcott
- Early Oncology Clinical, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Joyson Karakunnel
- Early Oncology Clinical, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA.,Present Address: Department of Clinical Development, Arcus Biosciences, Hayward, CA, USA
| | - Patricia LoRusso
- Department of Hematology & Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA.,Present Address: Medical Oncology, Yale Cancer Center, New Haven, CT, USA
| | - Amy Weise
- Department of Hematology & Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Joseph Eder
- Medical Oncology, Yale Cancer Center, New Haven, CT, USA
| | - Brendan Curti
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Michael Oberst
- Discovery Sciences, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
2
|
Li K, Tian H. Development of small-molecule immune checkpoint inhibitors of PD-1/PD-L1 as a new therapeutic strategy for tumour immunotherapy. J Drug Target 2018; 27:244-256. [PMID: 29448849 DOI: 10.1080/1061186x.2018.1440400] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy has been increasingly utilised to treat advanced malignancies. The signalling network of immune checkpoints has attracted considerable attention. Immune checkpoint inhibitors are revolutionising the treatment options and expectations for patients with cancer. The reported clinical success of targeting the T-cell immune checkpoint receptors PD-1/PD-L1 has demonstrated the importance of immune modulation. Indeed, antibodies binding to PD-1 or PD-L1 have shown remarkable efficacy. However, antibody drugs have many disadvantages, such as their production cost, stability, and immunogenicity and, therefore, small-molecule inhibitors of PD-1 and its ligand PD-L1 are being introduced. Small-molecule inhibitors could offer inherent advantages in terms of pharmacokinetics and druggability, thereby providing additional methods for cancer treatment and achieving better therapeutic effects. In this review, we first discuss how PD-1/PD-L1-targeting inhibitors modulate the relationship between immune cells and tumour cells in tumour immunotherapy. Second, we discuss how the immunomodulatory potential of these inhibitors can be exploited via rational combinations with immunotherapy and targeted therapy. Third, this review is the first to summarise the current clinical and preclinical evidence regarding small-molecule inhibitors of the PD-1/PD-L1 immune checkpoint, considering features and responses related to the tumours and to the host immune system.
Collapse
Affiliation(s)
- Kui Li
- a Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Science & Peking Union Medical College , Tianjin , China
| | - Hongqi Tian
- a Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine , Chinese Academy of Medical Science & Peking Union Medical College , Tianjin , China
| |
Collapse
|
3
|
Tackling Cancer Resistance by Immunotherapy: Updated Clinical Impact and Safety of PD-1/PD-L1 Inhibitors. Cancers (Basel) 2018; 10:cancers10020032. [PMID: 29370105 PMCID: PMC5836064 DOI: 10.3390/cancers10020032] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/21/2018] [Accepted: 01/23/2018] [Indexed: 02/07/2023] Open
Abstract
Cancer therapy has been constantly evolving with the hope of finding the most effective agents with the least toxic effects to eradicate tumors. Cancer immunotherapy is currently among the most promising options, fulfilling this hope in a wide range of tumors. Immunotherapy aims to activate immunity to fight cancer in a very specific and targeted manner; however, some abnormal immune reactions known as immune-related adverse events (IRAEs) might occur. Therefore, many researchers are aiming to define the most proper protocols for managing these complications without interfering with the anticancer effect. One of these targeted approaches is the inhibition of the interaction between the checkpoint protein, programmed death-receptor 1 (PD-1), and its ligand, programmed death-ligand 1 (PD-L1), via a class of antibodies known as PD-1/PD-L1 inhibitors. These antibodies achieved prodigious success in a wide range of malignancies, including those where optimal treatment is not yet fully identified. In this review, we have critically explored and discussed the outcome of the latest PD-1 and PD-L1 inhibitor studies in different malignancies compared to standard chemotherapeutic alternatives with a special focus on the clinical efficacy and safety. The approval of the clinical applications of nivolumab, pembrolizumab, atezolizumab, avelumab, and durvalumab in the last few years clearly highlights the hopeful future of PD-1/PD-L1 inhibitors for cancer patients. These promising results of PD-1/PD-L1 inhibitors have encouraged many ongoing preclinical and clinical trials to explore the extent of antitumor activity, clinical efficacy and safety as well as to extend their applications.
Collapse
|
4
|
Salama AKS, Moschos SJ. Next steps in immuno-oncology: enhancing antitumor effects through appropriate patient selection and rationally designed combination strategies. Ann Oncol 2017; 28:57-74. [PMID: 28177433 PMCID: PMC6887913 DOI: 10.1093/annonc/mdw534] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background Cancers escape immune surveillance via distinct mechanisms that involve central (negative selection within the thymus) or peripheral (lack of costimulation, receipt of death/anergic signals by tumor, immunoregulatory cell populations) immune tolerance. During the 1990s, moderate clinical benefit was seen using several cytokine therapies for a limited number of cancers. Over the past 20 years, extensive research has been performed to understand the role of various components of peripheral immune tolerance, with the co-inhibitory immune checkpoint molecules cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed death 1 (PD-1), and its ligand (PD-L1) being the most well-characterized at preclinical and clinical levels. Patients and methods We used PubMed and Google Scholar searches to identify key articles published reporting preclinical and clinical studies investigating CTLA-4 and PD-1/PD-L1, frequently cited review articles, and clinical studies of CTLA-4 and PD-1/PD-L1 pathway inhibitors, including combination therapy strategies. We also searched recent oncology congress presentations and clinicaltrials.gov to cover the most up-to-date clinical trial data and ongoing clinical trials of immune checkpoint inhibitor (ICI) combinations. Results Inhibiting CTLA-4 and PD-1 using monoclonal antibody therapies administered as single agents has been associated with clinical benefit in distinct patient subgroups across several malignancies. Concurrent blockade of CTLA-4 and components of the PD-1/PD-L1 system using various schedules has shown synergy and even higher incidence of durable antitumor responses at the expense of increased rates of immune-mediated adverse events, which can be life-threatening, but are rarely fatal and are reversible in most cases using established treatment guidelines. Conclusions Dual immune checkpoint blockade has demonstrated promising clinical benefit in numerous solid tumor types. This example of concurrent modulation of multiple components of the immune system is currently being investigated in other cancers using various immunomodulatory strategies.
Collapse
Affiliation(s)
- A. K. S. Salama
- Division of Medical Oncology, Duke University Medical Center, Durham
| | - S. J. Moschos
- Division of Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, USA
| |
Collapse
|